1
|
Luksch H, Schulze F, Geißler-Lösch D, Sprott D, Höfs L, Szegö EM, Tonnus W, Winkler S, Günther C, Linkermann A, Behrendt R, Teichmann LL, Falkenburger BH, Rösen-Wolff A. Tissue inflammation induced by constitutively active STING is mediated by enhanced TNF signaling. eLife 2025; 14:e101350. [PMID: 40111902 PMCID: PMC11996172 DOI: 10.7554/elife.101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 03/17/2025] [Indexed: 03/22/2025] Open
Abstract
Constitutive activation of STING by gain-of-function mutations triggers manifestation of the systemic autoinflammatory disease STING-associated vasculopathy with onset in infancy (SAVI). In order to investigate the role of signaling by tumor necrosis factor (TNF) in SAVI, we used genetic inactivation of TNF receptors 1 and 2 in murine SAVI, which is characterized by T cell lymphopenia, inflammatory lung disease, and neurodegeneration. Genetic inactivation of TNFR1 and TNFR2, however, rescued the loss of thymocytes, reduced interstitial lung disease, and neurodegeneration. Furthermore, genetic inactivation of TNFR1 and TNFR2 blunted transcription of cytokines, chemokines, and adhesions proteins, which result from chronic STING activation in SAVI mice. In addition, increased transendothelial migration of neutrophils was ameliorated. Taken together, our results demonstrate a pivotal role of TNFR signaling in the pathogenesis of SAVI in mice and suggest that available TNFR antagonists could ameliorate SAVI in patients.
Collapse
Affiliation(s)
- Hella Luksch
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Felix Schulze
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - David Geißler-Lösch
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - David Sprott
- Department of Physiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Lennart Höfs
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Eva M Szegö
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, Faculty of Medicine and University Hospital Carl GustavDresdenGermany
| | - Stefan Winkler
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Claudia Günther
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, Faculty of Medicine and University Hospital Carl GustavDresdenGermany
| | - Rayk Behrendt
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital BonnBonnGermany
| | | | - Björn H Falkenburger
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
- Deutsches Zentrum für Neurodegenerative ErkrankungenDresdenGermany
| | - Angela Rösen-Wolff
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| |
Collapse
|
2
|
Johnson EA, Nowar R, Viola KL, Huang W, Zhou S, Bicca MA, Zhu W, Kranz DL, Klein WL, Silverman RB. Inhibition of amyloid beta oligomer accumulation by NU-9: A unifying mechanism for the treatment of neurodegenerative diseases. Proc Natl Acad Sci U S A 2025; 122:e2402117122. [PMID: 40030015 PMCID: PMC11912461 DOI: 10.1073/pnas.2402117122] [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: 01/30/2024] [Accepted: 12/18/2024] [Indexed: 03/19/2025] Open
Abstract
Protein aggregation is a hallmark of neurodegenerative diseases, which connects these neuropathologies by a common phenotype. Various proteins and peptides form aggregates that are poorly degraded, and their ensuing pathological accumulation underlies these neurodegenerative diseases. Similarities may exist in the mechanisms responsible for the buildup of these aggregates. Therefore, therapeutics designed to treat one neurodegenerative disease may be beneficial to others. In ALS models, the compound NU-9 was previously shown to block neurodegeneration produced by aggregation-inducing mutations of SOD-1 and TDP-43 [B. Genç et al., Clin. Transl. Med. 11, e336 (2021)]. Here, we report that NU-9 also prevents the accumulation of amyloid beta oligomers (AβOs), small peptide aggregates that are instigators of Alzheimer's disease neurodegeneration [M. Tolar et al., Int. J. Mol. Sci. 22, 6355 (2021)]. AβO buildup was measured by immunofluorescence imaging of cultured hippocampal neurons exposed to exogenous monomeric Aβ. In this model, AβO buildup occurs via cathepsin L- and dynamin-dependent trafficking. This is prevented by NU-9 through a cellular mechanism that is cathepsin B- and lysosome-dependent, suggesting that NU-9 enhances the ability of endolysosomal trafficking to protect against AβO buildup. This possibility is strongly supported by a quantitative assay for autophagosomes that shows robust stimulation by NU-9. These results contribute additional understanding to the mechanisms of protein aggregation and suggest that multiple neurodegenerative diseases might be treatable by targeting common pathogenic mechanisms responsible for protein aggregation.
Collapse
Affiliation(s)
- Elizabeth A. Johnson
- Department of Chemistry, Northwestern University, Evanston, IL60208
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL60208
| | - Raghad Nowar
- Department of Neurobiology, Northwestern University, Evanston, IL60208
| | - Kirsten L. Viola
- Department of Neurobiology, Northwestern University, Evanston, IL60208
| | - Weijian Huang
- The Master of Biotechnology Program, McCormick School of Engineering, Northwestern University, Evanston, IL60208
| | - Sihang Zhou
- The Master of Biotechnology Program, McCormick School of Engineering, Northwestern University, Evanston, IL60208
| | - Maíra A. Bicca
- Department of Neurobiology, Northwestern University, Evanston, IL60208
| | - Wei Zhu
- Department of Chemistry, Northwestern University, Evanston, IL60208
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL60208
| | - Daniel L. Kranz
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL60208
| | - William L. Klein
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL60208
- Department of Neurobiology, Northwestern University, Evanston, IL60208
- Department of Neurology, Northwestern University, Chicago, IL60611
| | - Richard B. Silverman
- Department of Chemistry, Northwestern University, Evanston, IL60208
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL60208
- Department of Molecular Biosciences, Northwestern University, Evanston, IL60208
- Department of Pharmacology, Northwestern University, Chicago, IL60611
| |
Collapse
|
3
|
Wang X, Wang B, Du X, Liu P, Yang F, Su J, Zhang Y. Associations between neutrophil-lymphocyte ratio and risk of cognitive impairment among Chinese older adults. BMC Geriatr 2025; 25:114. [PMID: 39972309 PMCID: PMC11841232 DOI: 10.1186/s12877-025-05763-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Associations between the neutrophil-lymphocyte ratio (NLR) and cognitive performance in older population are rarely reported. We investigated the associations between NLR and risk of cognitive impairment in Chinese community-dwelling older adults. METHODS Individuals aged ≥ 65 years from the 2011 and 2014 waves of the Chinese Longitudinal Healthy Longevity Survey were enrolled. We used the Chinese version of the Mini-Mental State Examination to evaluate cognitive function, with a score <18 indicating cognitive impairment. NLR was expressed as derived NLR (white blood cell count - lymphocyte count]/lymphocyte count). Logistic regression was used to evaluate the association between NLR levels and risk of cognitive impairment. RESULTS The study enrolled 2375 cognitively healthy participants and 838 with cognitive impairment. Significantly higher NLR values were noted in the latter than in the former group. In the cross-sectional analysis, NLR values in the highest than in the lowest quartile indicated significantly increased risk of cognitive impairment, after controlling for all confounding factors. During follow-up, 134 of the 1173 healthy participants at baseline developed cognitive impairment. NLR values in the highest two quartiles indicated higher risk of cognitive impairment than those in the lowest quartile. When NLR was classified into dichotomous groups, the risk of cognitive impairment was significantly higher in the high-inflammation than in the noninflammatory status group, regardless of the analysis used (cross-sectional or prospective). CONCLUSIONS Elevated NLR status is associated with increased risk of cognitive impairment in Chinese community-dwelling older adults.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurology, Shanxi Bethune Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Binbin Wang
- School of Life Science, Shanxi Normal University, Taiyuan, China
| | - Xueqing Du
- Department of Neurology, Shanxi Bethune Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Peng Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Shanxi Medical University, Shanxi Cardiovascular Hospital (Institute), Taiyuan, China
| | - Fuwen Yang
- Department of Neurology, Shanxi Bethune Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jiao Su
- Department of Neurology, Shanxi Bethune Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yue Zhang
- School of Public Health, Department of Epidemiology, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Ministry Education, Taiyuan, China.
| |
Collapse
|
4
|
Alaei M, Koushki K, Taebi K, Yousefi Taba M, Keshavarz Hedayati S, Keshavarz Shahbaz S. Metal nanoparticles in neuroinflammation: impact on microglial dynamics and CNS function. RSC Adv 2025; 15:5426-5451. [PMID: 39967886 PMCID: PMC11833603 DOI: 10.1039/d4ra07798a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Microglia, the primary immune cells of the central nervous system (CNS), are crucial in maintaining brain homeostasis and responding to pathological changes. While they play protective roles, their activation can lead to neuroinflammation and the progression of neurodegenerative diseases. Metal nanoparticles (NPs), due to their unique ability to cross the blood-brain barrier (BBB), have emerged as promising agents for drug delivery to the CNS. In this way, we aim to review the dual role of metal-containing NPs, gold (AuNPs), silver (AgNPs), iron oxide (IONPs), zinc oxide (ZnONPs), cobalt (CoNPs), titanium dioxide (TiO2NPs), and silica (SiO2NPs) in modulating microglial activity. Some NPs promote anti-inflammatory effects, while others exacerbate neuroinflammation. We examine how these NPs influence microglial activation, focusing on their potential therapeutic benefits and risks. A deeper understanding of NP-microglia interactions is crucial for developing safe and efficient treatments for neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Masood Alaei
- Student Research Committee, Qazvin University of Medical Sciences Qazvin Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| | - Khadijeh Koushki
- Department of Neurosurgery, University of Texas Houston Health Science Center (UTHealth) Houston TX USA
| | - Kimia Taebi
- Student Research Committee, Qazvin University of Medical Sciences Qazvin Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| | - Mahdieh Yousefi Taba
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | | | - Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences Qazvin 34197-59811 Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| |
Collapse
|
5
|
Maggiore A, Latina V, D'Erme M, Amadoro G, Coccurello R. Non-canonical pathways associated to Amyloid beta and tau protein dyshomeostasis in Alzheimer's disease: A narrative review. Ageing Res Rev 2024; 102:102578. [PMID: 39542177 DOI: 10.1016/j.arr.2024.102578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Alzheimer's Disease (AD) is the most common form of dementia among elderly people. This disease imposes a significant burden on the healthcare system, society, and economy due to the increasing global aging population. Current trials with drugs or bioactive compounds aimed at reducing cerebral Amyloid beta (Aβ) plaques and tau protein neurofibrillary tangles, which are the two main hallmarks of this devastating neurodegenerative disease, have not provided significant results in terms of their neuropathological outcomes nor met the expected clinical end-points. Ageing, genetic and environmental risk factors, along with different clinical symptoms suggest that AD is a complex and heterogeneous disorder with multiple interconnected pathological pathways rather than a single disease entity. In the present review, we highlight and discuss various non-canonical, Aβ-independent mechanisms, like gliosis, unhealthy dietary intake, lipid and sugar signaling, and cerebrovascular damage that contribute to the onset and development of AD. We emphasize that challenging the traditional "amyloid cascade hypothesis" may improve our understanding of this age-related complex syndrome and help fight the progressive cognitive decline in AD.
Collapse
Affiliation(s)
- Anna Maggiore
- Department of Biochemical Sciences, Sapienza University, P.le Aldo Moro 5, Rome 00185, Italy; Department of Brain Sciences, Imperial College, London, UK
| | - Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, Rome 00161, Italy; Institute of Translational Pharmacology (IFT) CNR, Via Fosso del Cavaliere 100, Rome 00133, Italy
| | - Maria D'Erme
- Department of Biochemical Sciences, Sapienza University, P.le Aldo Moro 5, Rome 00185, Italy
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, Rome 00161, Italy; Institute of Translational Pharmacology (IFT) CNR, Via Fosso del Cavaliere 100, Rome 00133, Italy.
| | - Roberto Coccurello
- Institute for Complex System (ISC) CNR, Via dei Taurini 19, Rome 00185, Italy; IRCSS Santa Lucia Foundation, European Center for Brain Research, Via Fosso del Fiorano 64-65, Rome 00143, Italy.
| |
Collapse
|
6
|
Ge X, Zhang L, Ye M, Fei A, Wang L, Yuan A. Electroacupuncture improves diabetic cognitive impairment rats by suppressing NLRP3 inflammasome activation via induction of mitophagy. Metab Brain Dis 2024; 40:14. [PMID: 39560708 PMCID: PMC11576777 DOI: 10.1007/s11011-024-01464-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/15/2024] [Indexed: 11/20/2024]
Abstract
Diabetic cognitive impairment pose a significant threat to public health in our aging society. However, the underlying molecular mechanisms have not been fully elucidated, which warrants further investigation. This study aimed to investigate the effects of electroacupuncture on cognitive impairment and its associated mechanisms. The diabetes model was induced via intraperitoneal injection of streptozotocin (STZ) into Sprague-Dawley rats combined with a high-fat and high-sugar diet. The learning and memory abilities of the rats were assessed using behavioral tests. Electron microscopy and hematoxylin-eosin (H&E) staining were used to identify the histological changes of neurons in the CA1 area of the rat hippocampal CA1. An examination of related indicators was performed by Western blotting including NLRP3 inflammasome-associated proteins Caspase-1, IL-18, IL1β, NLRP3, and P62, and mitophagy-related proteins Pink1, LC3, and Parkin. After modeling, rats displayed impaired learning and memory functions. The administration of electroacupuncture treatment alleviated diabetic cognitive impairment, described as shorter escape latency and an increased frequency of platform crossings. The damaged morphological and ultrastructural changes of neurons in rat hippocampal CA1 area can be alleviated through electroacupuncture treatment. Furthermore, in-depth studies suggest that electroacupuncture treatment can suppress NLRP3 inflammasome activation through induction of Pink1, LC3 and Parkin expression. Electroacupuncture treatment can attenuate NLRP3 inflammasome activation by promoting mitophagy, eventually improving cognitive impairment in (STZ)-treated rats with a high-fat die.
Collapse
Affiliation(s)
- Xia Ge
- Department of Endocrinology, The Second Affiliated Hospital of Anhui, University of Chinese Medicine, Hefei, 230001, Anhui, China
| | - Lele Zhang
- Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Min Ye
- Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Aihua Fei
- Department of Endocrinology, The Second Affiliated Hospital of Anhui, University of Chinese Medicine, Hefei, 230001, Anhui, China
| | - Ling Wang
- Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, Anhui, China
| | - Aihong Yuan
- Department of acupuncture and rehabilitation, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China.
| |
Collapse
|
7
|
Jiang X, Wang Y, Lin Z, Li C, Wang Q, Zhang J, Liu X, Li Z, Cui C. Polygonatum sibiricum polysaccharides: A promising strategy in the treatment of neurodegenerative disease. Neurochem Int 2024; 181:105902. [PMID: 39542041 DOI: 10.1016/j.neuint.2024.105902] [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: 02/07/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Neurodegenerative diseases (NDDs), as a neurological disorder characterised by neuronal degeneration and death, are a serious threat to human health and have long attracted attention due to their complex pathogenesis and the ineffectiveness of therapeutic drugs. Existing studies have shown that Polygonatum Sibiricum polysaccharides (PSP) have immunoregulatory, antioxidant, anti-inflammatory and other pharmacological effects, and their neuroprotective effects have been demonstrated in several scientific studies. This paper reviews the main pharmacological effects and mechanisms of PSP in the protection and treatment of NDDs, to provide a reference for the clinical application and basic research of PSP in NDDs.
Collapse
Affiliation(s)
- Xue Jiang
- Shandong Medicine Technician College, 271000, Taian, China; Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, 271000, Taian, China
| | - Yumei Wang
- Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, 271000, Taian, China
| | - Zhaochen Lin
- Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, 271000, Taian, China
| | - Chao Li
- Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, 271000, Taian, China
| | - Qian Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Junyan Zhang
- College of Life Sciences, Northwest A & F University, 710000, Xi'an, China
| | - Xiuhua Liu
- Shandong Taishan Sealwort Biotechnology Limited Liability Company, 271000, Taian, China
| | - Ziye Li
- Xiangya School of Public Health, Central South University, 410078, Changsha, China
| | - Chao Cui
- Qilu Hospital of Shandong University Dezhou Hospital, 253000, Dezhou, China; Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, 271000, Taian, China.
| |
Collapse
|
8
|
Ikezu T, Yang Y, Verderio C, Krämer-Albers EM. Extracellular Vesicle-Mediated Neuron-Glia Communications in the Central Nervous System. J Neurosci 2024; 44:e1170242024. [PMID: 39358029 PMCID: PMC11450539 DOI: 10.1523/jneurosci.1170-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 10/04/2024] Open
Abstract
Communication between neurons and glia significantly influences the development maturation, plasticity, and disease progressions of the nervous system. As a new signaling modality, extracellular vesicles display a diverse role for robust functional regulation of neurons through their protein and nucleic acid cargoes. This review highlights recent breakthroughs in the research of signaling mechanisms between glia and neurons mediated by extracellular vesicles that are important for neural development, axonal maintenance, synaptic functions, and disease progression in the mammalian nervous system. We will discuss the biological roles of extracellular vesicles released from neurons, astroglia, microglia, and oligodendroglia in the nervous system and their implications in neurodegenerative disorders.
Collapse
Affiliation(s)
- Tsuneya Ikezu
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224
| | - Yongjie Yang
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Claudia Verderio
- Department of Biomedical Sciences, CNR Institute of Neuroscience, Università Milano-Bicocca, 20854 Vedano al Lambro (MB), Italy
| | - Eva-Maria Krämer-Albers
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, 55128 Mainz, Rhineland Palatinate, Germany
| |
Collapse
|
9
|
Lin H, Wang Z, Liao Y, Yu Z, Xu H, Qin T, Tang J, Yang X, Chen S, Chen X, Zhang X, Shen Y. Super-resolution ultrasound imaging reveals temporal cerebrovascular changes with disease progression in female 5×FAD mouse model of Alzheimer's disease: correlation with pathological impairments. EBioMedicine 2024; 108:105355. [PMID: 39293213 PMCID: PMC11424966 DOI: 10.1016/j.ebiom.2024.105355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Vascular dysfunction is closely associated with the progression of Alzheimer's disease (AD). A critical research gap exists that no studies have explored the in vivo temporal changes of cerebrovascular alterations with AD progression in mouse models, encompassing both structure and flow dynamics at micron-scale resolution across the early, middle, and late stages of the disease. METHODS In this study, ultrasound localisation microscopy (ULM) was applied to image the cerebrovascular alterations of the transgenic female 5×FAD mouse model across different stages of disease progression: early (4 months), moderate (7 months), and late (12 months). Age-matched non-transgenic (non-Tg) littermates were used as controls. Immunohistology examinations were performed to evaluate the influence of disease progression on the β-amyloid (Aβ) load and microvascular alterations, including morphological changes and the blood-brain barrier (BBB) leakage. FINDINGS Our findings revealed a significant decline in both vascular density and flow velocity in the retrosplenial cortex of 5×FAD mice at an early stage, which subsequently became more pronounced in the visual cortex and hippocampus as the disease progressed. Additionally, we observed a reduction in vascular length preceding diminished flow velocities in cortical penetrating arterioles during the early stages. The quantification of vascular metrics derived from ULM imaging showed significant correlations with those obtained from vascular histological images. Immunofluorescence staining identified early vascular abnormalities in the retrosplenial cortex. As the disease advanced, there was an exacerbation of Aβ accumulation and BBB disruption in a regionally variable manner. The vascular changes observed through ULM imaging exhibited a negative correlation with amyloid load and were associated with the compromise of the BBB integrity. INTERPRETATION Through high-resolution, in vivo imaging of cerebrovasculature, this study reveals significant spatiotemporal dysfunction in cerebrovascular dynamics accompanying disease progression in a mouse model of AD, enhancing our understanding of its pathophysiology. FUNDING This study is supported by grants from National Key Research and Development Program of China (2020YFA0908800), National Natural Science Foundation of China (12074269, 82272014, 82327804, 62071310), Shenzhen Basic Science Research (20220808185138001, JCYJ20220818095612027, JCYJ20210324093006017), STI 2030-Major Projects (2021ZD0200500) and Guangdong Natural Science Foundation (2024A1515012591, 2024A1515011342).
Collapse
Affiliation(s)
- Haoming Lin
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Zidan Wang
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Yingtao Liao
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China; Department of Radiation Oncology, Huizhou Central People's Hospital, Huizhou, 516001, Guangdong, China
| | - Zhifan Yu
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Huiqin Xu
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Ting Qin
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Jianbo Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518071, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Centre for Disease Control and Prevention, Shenzhen, 518055, China
| | - Siping Chen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Xin Chen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Xinyu Zhang
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Yuanyuan Shen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China.
| |
Collapse
|
10
|
Liu C, Zhang W, Zhang H, Zhao C, Du X, Ren J, Qu X. Biomimetic engineering of a neuroinflammation-targeted MOF nanozyme scaffolded with photo-trigger released CO for the treatment of Alzheimer's disease. Chem Sci 2024; 15:13201-13208. [PMID: 39183930 PMCID: PMC11339965 DOI: 10.1039/d4sc02598a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most fatal and irreversible neurodegenerative diseases, which causes a huge emotional and financial burden on families and society. Despite the progress made with recent clinical use of inhibitors of acetylcholinesterase and amyloid-β (Aβ) antibodies, the curative effects of AD treatment remain unsatisfactory, which is probably due to the complexity of pathogenesis and the multiplicity of therapeutic targets. Thus, modulating complex pathological networks could be an alternative approach to treat AD. Here, a neutrophil membrane-coated MOF nanozyme (denoted as Neu-MOF/Fla) is biomimetically engineered to disturb the malignant Aβ deposition-inflammation cycle and ameliorate the pathological network for effective AD treatment. Neu-MOF/Fla could recognize the pathological inflammatory signals of AD, and deliver the photo-triggered anti-inflammatory CO and MOF based hydrolytic nanozymes to the lesion area of the brain in a spontaneous manner. Based on the in vitro and in vivo studies, Neu-MOF/Fla significantly suppresses neuroinflammation, mitigates the Aβ burden, beneficially modulates the pro-inflammatory microglial phenotypes and improves the cognitive defects of AD mice models. Our work presents a good example for developing biomimetic multifunctional nanotherapeutics against AD by means of amelioration of multiple symptoms and improvement of cognitive defects.
Collapse
Affiliation(s)
- Chun Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Wenting Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Haochen Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Chuanqi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Xiubo Du
- College of Life Sciences and Oceanography, Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen University Shenzhen 518060 China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| |
Collapse
|
11
|
Qian K, Yang P, Li Y, Meng R, Cheng Y, Zhou L, Wu J, Xu S, Bao X, Guo Q, Wang P, Xu M, Sheng D, Zhang Q. Rational fusion design inspired by cell-penetrating peptide: SS31/S-14 G Humanin hybrid peptide with amplified multimodal efficacy and bio-permeability for the treatment of Alzheimer's disease. Asian J Pharm Sci 2024; 19:100938. [PMID: 39253611 PMCID: PMC11382307 DOI: 10.1016/j.ajps.2024.100938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/26/2023] [Accepted: 05/16/2024] [Indexed: 09/11/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative disease induced by multiple interconnected mechanisms. Peptide drug candidates with multi-modal efficacy generated from fusion strategy are suitable for addressing multi-facet pathology. However, clinical translation of peptide drugs is greatly hampered by their low permeability into brain. Herein, a hybrid peptide HNSS is generated by merging two therapeutic peptides (SS31 and S-14 G Humanin (HNG)), using a different approach from the classical shuttle-therapeutic peptide conjugate design. HNSS demonstrated increased bio-permeability, with a 2-fold improvement in brain distribution over HNG, thanks to its structure mimicking the design of signal peptide-derived cell-penetrating peptides. HNSS efficiently alleviated mitochondrial dysfunction through the combined effects of mitochondrial targeting, ROS scavenging and p-STAT3 activation. Meanwhile, HNSS with increased Aβ affinity greatly inhibited Aβ oligomerization/fibrillation, and interrupted Aβ interaction with neuron/microglia by reducing neuronal mitochondrial Aβ deposition and promoting microglial phagocytosis of Aβ. In 3× Tg-AD transgenic mice, HNSS treatment efficiently inhibited brain neuron loss and improved the cognitive performance. This work validates the rational fusion design-based strategy for bio-permeability improvement and efficacy amplification, providing a paradigm for developing therapeutic peptide candidates against neurodegenerative disease.
Collapse
Affiliation(s)
- Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuting Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiaoyan Bao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qian Guo
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Pengzhen Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
12
|
Do Carmo S, Kautzmann MAI, Bhattacharjee S, Jun B, Steinberg C, Emmerson JT, Malcolm JC, Bonomo Q, Bazan NG, Cuello AC. Differential effect of an evolving amyloid and tau pathology on brain phospholipids and bioactive lipid mediators in rat models of Alzheimer-like pathology. J Neuroinflammation 2024; 21:185. [PMID: 39080670 PMCID: PMC11290283 DOI: 10.1186/s12974-024-03184-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Brain inflammation contributes significantly to the pathophysiology of Alzheimer's disease, and it is manifested by glial cell activation, increased production of cytokines/chemokines, and a shift in lipid mediators from a pro-homeostatic to a pro-inflammatory profile. However, whether the production of bioactive lipid mediators is affected at earlier stages, prior to the deposition of Aβ plaques and tau hyperphosphorylation, is unknown. The differential contribution of an evolving amyloid and tau pathology on the composition and abundance of membrane phospholipids and bioactive lipid mediators also remains unresolved. METHODS In this study, we examined the cortical levels of DHA- and AA-derived bioactive lipid mediators and of membrane phospholipids by liquid chromatography with tandem mass spectrometry in transgenic rat models of the Alzheimer's-like amyloid and tau pathologies at early and advanced pathological stages. RESULTS Our findings revealed a complex balance between pro-inflammatory and pro-resolving processes in which tau pathology has a more pronounced effect compared to amyloid pathology. At stages preceding tau misfolding and aggregation, there was an increase in pro-resolving lipid mediators (RVD6 and NPD1), DHA-containing phospholipids and IFN-γ levels. However, in advanced tau pathology displaying NFT-like inclusions, neuronal death, glial activation and cognitive deficits, there was an increase in cytokine and PGD2, PGE2, and PGF2α generation accompanied by a drop in IFN-γ levels. This pathology also resulted in a marked increase in AA-containing phospholipids. In comparison, pre-plaque amyloid pathology already presented high levels of cytokines and AA-containing phospholipids together with elevated RVD6 and NPD1 levels. Finally, Aβ plaque deposition was accompanied by a modest increase in prostaglandins, increased AA-containing phospholipids and reduced DHA-containing phospholipids. CONCLUSIONS Our findings suggest a dynamic trajectory of inflammatory and lipid mediators in the evolving amyloid and tau pathologies and support their differing roles on membrane properties and, consequentially, on signal transduction.
Collapse
Affiliation(s)
- Sonia Do Carmo
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada.
| | - Marie-Audrey I Kautzmann
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Carolyn Steinberg
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada
| | - Joshua T Emmerson
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada
| | - Janice C Malcolm
- Department of Cell Anatomy and Cell Biology, McGill University, Montreal, H3A 0C7, Canada
| | - Quentin Bonomo
- Department of Neurology and Neurosurgery, McGill University, Montreal, H3G 1Y6, Canada
| | - Nicolas G Bazan
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada.
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA.
| | - A Claudio Cuello
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada.
- Department of Cell Anatomy and Cell Biology, McGill University, Montreal, H3A 0C7, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, H3G 1Y6, Canada.
- Department of Pharmacology, Oxford University, Oxford, OX1 3QT, UK.
| |
Collapse
|
13
|
Foret MK, Orciani C, Welikovitch LA, Huang C, Cuello AC, Do Carmo S. Early oxidative stress and DNA damage in Aβ-burdened hippocampal neurons in an Alzheimer's-like transgenic rat model. Commun Biol 2024; 7:861. [PMID: 39004677 PMCID: PMC11247100 DOI: 10.1038/s42003-024-06552-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Oxidative stress is a key contributor to AD pathology. However, the earliest role of pre-plaque neuronal oxidative stress, remains elusive. Using laser microdissected hippocampal neurons extracted from McGill-R-Thy1-APP transgenic rats we found that intraneuronal amyloid beta (iAβ)-burdened neurons had increased expression of genes related to oxidative stress and DNA damage responses including Ercc2, Fancc, Sod2, Gsr, and Idh1. DNA damage was further evidenced by increased neuronal levels of XPD (Ercc2) and γH2AX foci, indicative of DNA double stranded breaks (DSBs), and by increased expression of Ercc6, Rad51, and Fen1, and decreased Sirt6 in hippocampal homogenates. We also found increased expression of synaptic plasticity genes (Grin2b (NR2B), CamkIIα, Bdnf, c-fos, and Homer1A) and increased protein levels of TOP2β. Our findings indicate that early accumulation of iAβ, prior to Aβ plaques, is accompanied by incipient oxidative stress and DSBs that may arise directly from oxidative stress or from maladaptive synaptic plasticity.
Collapse
Affiliation(s)
- Morgan K Foret
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Chiara Orciani
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | - Chunwei Huang
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.
- Department of Pharmacology, Oxford University, Oxford, UK.
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
14
|
Gaire BP, Koronyo Y, Fuchs DT, Shi H, Rentsendorj A, Danziger R, Vit JP, Mirzaei N, Doustar J, Sheyn J, Hampel H, Vergallo A, Davis MR, Jallow O, Baldacci F, Verdooner SR, Barron E, Mirzaei M, Gupta VK, Graham SL, Tayebi M, Carare RO, Sadun AA, Miller CA, Dumitrascu OM, Lahiri S, Gao L, Black KL, Koronyo-Hamaoui M. Alzheimer's disease pathophysiology in the Retina. Prog Retin Eye Res 2024; 101:101273. [PMID: 38759947 PMCID: PMC11285518 DOI: 10.1016/j.preteyeres.2024.101273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid β-protein (Aβ) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aβ deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Danziger
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jean-Philippe Vit
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Filippo Baldacci
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | | | - Ernesto Barron
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia; Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Shouri Lahiri
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Gao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Huuha AM, Norevik CS, Coombes JS, Røsbjørgen RN, Miguel-dos-Santos R, Moreira JBN, Kobro-Flatmoen A, Scrimgeour N, Tari AR. Effects of Intravenously Administered Plasma from Exercise-Trained Donors on Microglia and Cytokines in a Transgenic Rat Model of Alzheimer's Disease. Brain Plast 2024; 9:21-41. [PMID: 38993579 PMCID: PMC11234670 DOI: 10.3233/bpl-230154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 07/13/2024] Open
Abstract
Background Microglia and inflammation play a significant role in Alzheimer's disease (AD). Physical exercise and peripheral signals can influence microglial activity in the brain. Modulating the inflammatory response in the brain may provide therapeutic approaches for AD. Objective To assess the effects of intravenously administered blood plasma from exercise-trained donor rats on cognitive function, microglia, and cytokine levels in an AD rat model at two different pathological stages; an early pre-plaque stage and a later stage closer to the emergence of extracellular plaques. Methods Male transgenic McGill-R-Thy1-APP rats aged 2 and 5 months received 14 injections over 6 weeks: 1) plasma from exercise-trained rats (ExPlas), 2) plasma from sedentary rats (SedPlas), or 3) saline. Cognitive function was evaluated in a novel object recognition task. Microglia count and morphology were analyzed in cornu ammonis, dentate gyrus, entorhinal cortex, and subiculum. Amyloid plaque number and size were assessed in the rats with the later treatment start. A multiplex assay was used to measure 23 cytokines in cornu ammonis. Results In rats treated from 2 months of age, ExPlas and SedPlas increased number and length of microglial branches in cornu ammonis and dentate gyrus compared to saline. Only ExPlas-treated rats exhibited similar changes in subiculum, while entorhinal cortex showed no differences across treatments. Microglia count remained unaffected. In rats treated from 5 months of age, there were no significant differences in microglia count or morphology or the number or size of amyloid plaques in any brain region. Compared to both other treatments in early pre-plaque stage rats, SedPlas increased TNF-α levels. ExPlas upregulated GM-CSF, IL-18, and VEGF, while SedPlas increased IL-10 compared to saline. In later-stage rats, ExPlas upregulated IL-17, and SedPlas upregulated TNF-α compared to saline. There were no effects of treatments on recognition memory. Conclusions Intravenous injections of blood plasma from exercise-trained and sedentary donors differentially modulated microglial morphology and cytokine levels in the AD rat model at an early pre-plaque stage of pathology. Exercised plasma may reduce proinflammatory TNF-α signaling and promote microglial responses to early Aβ accumulation but the lack of treatment effects in the later-stage rats emphasizes the potential importance of treatment timing.
Collapse
Affiliation(s)
- Aleksi M. Huuha
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Cecilie Skarstad Norevik
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Jeff S. Coombes
- Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Australia
| | - Ragnhild N. Røsbjørgen
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rodrigo Miguel-dos-Santos
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Physiology, Federal University of Sergipe, Sergipe, Brazil
| | - José Bianco N. Moreira
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Asgeir Kobro-Flatmoen
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, and Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Centre for Alzheimer’s Disease, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nathan Scrimgeour
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Atefe R. Tari
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
16
|
Bocheva G, Bakalov D, Iliev P, Tafradjiiska-Hadjiolova R. The Vital Role of Melatonin and Its Metabolites in the Neuroprotection and Retardation of Brain Aging. Int J Mol Sci 2024; 25:5122. [PMID: 38791160 PMCID: PMC11121732 DOI: 10.3390/ijms25105122] [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: 03/29/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
While primarily produced in the pineal gland, melatonin's influence goes beyond its well-known role in regulating sleep, nighttime metabolism, and circadian rhythms, in the field of chronobiology. A plethora of new data demonstrates melatonin to be a very powerful molecule, being a potent ROS/RNS scavenger with anti-inflammatory, immunoregulatory, and oncostatic properties. Melatonin and its metabolites exert multiple beneficial effects in cutaneous and systemic aging. This review is focused on the neuroprotective role of melatonin during aging. Melatonin has an anti-aging capacity, retarding the rate of healthy brain aging and the development of age-related neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, amyotrophic lateral sclerosis, etc. Melatonin, as well as its metabolites, N1-acetyl-N2-formyl-5-methoxykynuramine (AFMK) and N1-acetyl-5-methoxykynuramine (AMK), can reduce oxidative brain damage by shielding mitochondria from dysfunction during the aging process. Melatonin could also be implicated in the treatment of neurodegenerative conditions, by modifying their characteristic low-grade neuroinflammation. It can either prevent the initiation of inflammatory responses or attenuate the ongoing inflammation. Drawing on the current knowledge, this review discusses the potential benefits of melatonin supplementation in preventing and managing cognitive impairment and neurodegenerative diseases.
Collapse
Affiliation(s)
- Georgeta Bocheva
- Department of Pharmacology and Toxicology, Medical University of Sofia, 1431 Sofia, Bulgaria
| | - Dimitar Bakalov
- Department of Physiology and Pathophysiology, Medical University of Sofia, 1431 Sofia, Bulgaria
| | - Petar Iliev
- Department of Physiology and Pathophysiology, Medical University of Sofia, 1431 Sofia, Bulgaria
| | | |
Collapse
|
17
|
Kaul D, Ehret F, Roohani S, Jendrach M, Buthut M, Acker G, Anwar M, Zips D, Heppner F, Prüss H. Radiation Therapy in Alzheimer's Disease: A Systematic Review. Int J Radiat Oncol Biol Phys 2024; 119:23-41. [PMID: 38042449 DOI: 10.1016/j.ijrobp.2023.11.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/11/2023] [Accepted: 11/19/2023] [Indexed: 12/04/2023]
Abstract
PURPOSE Pathophysiological hallmarks of Alzheimer's disease (AD) include extracellular amyloid plaques and intracellular neurofibrillary tangles. Recent studies also demonstrated a role of neuroinflammation in the progression of the disease. Clinical trials and animal studies using low-dose radiation therapy (LDRT) have shown therapeutic potential for AD. This systematic review summarizes the current evidence on the use of LDRT for the treatment of AD, outlines potential mechanisms of action, and discusses current challenges in the planning of future trials. METHODS AND MATERIALS A systematic review of human and animal studies as well as registered clinical trials describing outcomes for RT in the treatment of AD was conducted. We followed the 2020 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Articles published until July 1, 2023, were included. RESULTS The initial search yielded 993 articles. After the removal of duplicates and ineligible publications, a total of 16 (12 animal, 4 human) studies were included. Various dose regimens were utilized in both animal and human trials. The results revealed that LDRT reduced the number of amyloid plaques and neurofibrillary tangles, and it has a role in the regulation of genes and protein expression involved in the pathological progression of AD. LDRT has demonstrated reduced astro- and microgliosis, anti-inflammatory and neuroprotective effects, and an alleviation of symptoms of cognitive deficits in animal models. Most studies in humans suggested improvements in cognition and behavior. None of the trials or studies described significant (>grade 2) toxicity. CONCLUSIONS Preclinical studies, animal studies, and early clinical trials in humans have shown a promising role for LDRT in the treatment of AD pathologies, although the underlying mechanisms are yet to be fully explored. Phase I/II/III trials are needed to assess the long-term safety, efficacy, and optimal treatment parameters of LDRT in AD treatment.
Collapse
Affiliation(s)
- David Kaul
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Felix Ehret
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Siyer Roohani
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Marina Jendrach
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Maria Buthut
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Güliz Acker
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Muneeba Anwar
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Heppner
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Harald Prüss
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| |
Collapse
|
18
|
Davoody S, Asgari Taei A, Khodabakhsh P, Dargahi L. mTOR signaling and Alzheimer's disease: What we know and where we are? CNS Neurosci Ther 2024; 30:e14463. [PMID: 37721413 PMCID: PMC11017461 DOI: 10.1111/cns.14463] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023] Open
Abstract
Despite the great body of research done on Alzheimer's disease, the underlying mechanisms have not been vividly investigated. To date, the accumulation of amyloid-beta plaques and tau tangles constitutes the hallmark of the disease; however, dysregulation of the mammalian target of rapamycin (mTOR) seems to be significantly involved in the pathogenesis of the disease as well. mTOR, as a serine-threonine protein kinase, was previously known for controlling many cellular functions such as cell size, autophagy, and metabolism. In this regard, mammalian target of rapamycin complex 1 (mTORC1) may leave anti-aging impacts by robustly inhibiting autophagy, a mechanism that inhibits the accumulation of damaged protein aggregate and dysfunctional organelles. Formation and aggregation of neurofibrillary tangles and amyloid-beta plaques seem to be significantly regulated by mTOR signaling. Understanding the underlying mechanisms and connection between mTOR signaling and AD may suggest conducting clinical trials assessing the efficacy of rapamycin, as an mTOR inhibitor drug, in managing AD or may help develop other medications. In this literature review, we aim to elaborate mTOR signaling network mainly in the brain, point to gaps of knowledge, and define how and in which ways mTOR signaling can be connected with AD pathogenesis and symptoms.
Collapse
Affiliation(s)
- Samin Davoody
- Student Research Committee, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Afsaneh Asgari Taei
- Neuroscience Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Pariya Khodabakhsh
- Department of NeurophysiologyInstitute of Physiology, Eberhard Karls University of TübingenTübingenGermany
| | - Leila Dargahi
- Neurobiology Research CenterShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
19
|
De Francesco MA. Herpesviridae, Neurodegenerative Disorders and Autoimmune Diseases: What Is the Relationship between Them? Viruses 2024; 16:133. [PMID: 38257833 PMCID: PMC10818483 DOI: 10.3390/v16010133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/06/2023] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Alzheimer's disease and Parkinson's disease represent the most common forms of cognitive impairment. Multiple sclerosis is a chronic inflammatory disease of the central nervous system responsible for severe disability. An aberrant immune response is the cause of myelin destruction that covers axons in the brain, spinal cord, and optic nerves. Systemic lupus erythematosus is an autoimmune disease characterized by alteration of B cell activation, while Sjögren's syndrome is a heterogeneous autoimmune disease characterized by altered immune responses. The etiology of all these diseases is very complex, including an interrelationship between genetic factors, principally immune associated genes, and environmental factors such as infectious agents. However, neurodegenerative and autoimmune diseases share proinflammatory signatures and a perturbation of adaptive immunity that might be influenced by herpesviruses. Therefore, they might play a critical role in the disease pathogenesis. The aim of this review was to summarize the principal findings that link herpesviruses to both neurodegenerative and autoimmune diseases; moreover, briefly underlining the potential therapeutic approach of virus vaccination and antivirals.
Collapse
Affiliation(s)
- Maria Antonia De Francesco
- Department of Molecular and Translational Medicine, Institute of Microbiology, University of Brescia-ASST Spedali Civili, 25123 Brescia, Italy
| |
Collapse
|
20
|
Anderton E, Chamoli M, Bhaumik D, King CD, Xie X, Foulger A, Andersen JK, Schilling B, Lithgow GJ. Amyloid β accelerates age-related proteome-wide protein insolubility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.13.548937. [PMID: 37503138 PMCID: PMC10369951 DOI: 10.1101/2023.07.13.548937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Loss of proteostasis is a highly conserved feature of aging across model organisms and typically results in the accumulation of insoluble protein aggregates. Protein insolubility is a central feature of major age-related neurodegenerative diseases, including Alzheimer's Disease (AD), where hundreds of insoluble proteins associate with aggregated amyloid beta (Aβ) in senile plaques. Moreover, proteins that become insoluble during aging in model organisms are capable of accelerating Aβ aggregation in vitro. Despite the connection between aging and AD risk, therapeutic approaches to date have overlooked aging-driven protein insolubility as a contributory factor. Here, using an unbiased proteomics approach, we questioned the relationship between Aβ and age-related protein insolubility. We demonstrate that Aβ expression drives proteome-wide protein insolubility in C. elegans and this insoluble proteome closely resembles the insoluble proteome driven by normal aging, suggesting the possibility of a vicious feedforward cycle of aggregation in the context of AD. Importantly, using human genome-wide association studies (GWAS), we show that the CIP is replete with biological processes implicated not only in neurodegenerative diseases but also across a broad array of chronic, age-related diseases (CARDs). This provides suggestive evidence that age-related loss of proteostasis could play a role in general CARD risk. Finally, we show that the CIP is enriched with proteins that modulate the toxic effects of Aβ and that the gut-derived metabolite, Urolithin A, relieves Aβ toxicity, supporting its use in clinical trials for dementia and other age-related diseases.
Collapse
Affiliation(s)
- Edward Anderton
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
- USC Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave., Los Angeles, CA 90191
| | - Manish Chamoli
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | - Dipa Bhaumik
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | - Christina D. King
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | - Xueshu Xie
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | - Anna Foulger
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | - Julie K. Andersen
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | - Birgit Schilling
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | - Gordon J. Lithgow
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| |
Collapse
|
21
|
Yang Y, Seok MJ, Kim YE, Choi Y, Song JJ, Sulistio YA, Kim SH, Chang MY, Oh SJ, Nam MH, Kim YK, Kim TG, Im HI, Koh SH, Lee SH. Adeno-associated virus (AAV) 9-mediated gene delivery of Nurr1 and Foxa2 ameliorates symptoms and pathologies of Alzheimer disease model mice by suppressing neuro-inflammation and glial pathology. Mol Psychiatry 2023; 28:5359-5374. [PMID: 35902630 DOI: 10.1038/s41380-022-01693-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 06/30/2022] [Indexed: 12/16/2022]
Abstract
There is a compelling need to develop disease-modifying therapies for Alzheimer's disease (AD), the most common neuro-degenerative disorder. Together with recent progress in vector development for efficiently targeting the central nervous system, gene therapy has been suggested as a potential therapeutic modality to overcome the limited delivery of conventional types of drugs to and within the damaged brain. In addition, given increasing evidence of the strong link between glia and AD pathophysiology, therapeutic targets have been moving toward those addressing glial cell pathology. Nurr1 and Foxa2 are transcription/epigenetic regulators that have been reported to cooperatively regulate inflammatory and neurotrophic response in glial cells. In this study, we tested the therapeutic potential of Nurr1 and Foxa2 gene delivery to treat AD symptoms and pathologies. A series of functional, histologic, and transcriptome analyses revealed that the combined expression of Nurr1 and Foxa2 substantially ameliorated AD-associated amyloid β and Tau proteinopathy, cell senescence, synaptic loss, and neuro-inflammation in multiple in vitro and in vivo AD models. Intra-cranial delivery of Nurr1 and Foxa2 genes using adeno-associated virus (AAV) serotype 9 improved the memory and cognitive function of AD model mice. The therapeutic benefits of gene delivery were attained mainly by correcting pathologic glial function. These findings collectively indicate that AAV9-mediated Nurr1 and Foxa2 gene transfer could be an effective disease-modifying therapy for AD.
Collapse
Affiliation(s)
- Yunseon Yang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
| | - Min-Jong Seok
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
| | - Ye Eun Kim
- Department of Neurology, Hanyang University Guri Hospital, Hangyang University College of Medicine, Guri, Republic of Korea
- Graduate School of Translational Medicine, Hanyang University, Seoul, Republic of Korea
| | - Yunjung Choi
- Convergence Research Center for Brain Science, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jae-Jin Song
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
| | - Yanuar Alan Sulistio
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
| | - Seong-Hoon Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
| | - Mi-Yoon Chang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yun Kyung Kim
- Convergence Research Center for Brain Science, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Med, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Tae-Gyun Kim
- Innopeutics Corporation, Seoul, Republic of Korea
| | - Heh-In Im
- Convergence Research Center for Brain Science, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
- Division of Bio-Med, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea.
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, Hangyang University College of Medicine, Guri, Republic of Korea.
| | - Sang-Hun Lee
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea.
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea.
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Liu G, Yu Q, Zhu H, Tan B, Yu H, Li X, Lu Y, Li H. Amyloid-β mediates intestinal dysfunction and enteric neurons loss in Alzheimer's disease transgenic mouse. Cell Mol Life Sci 2023; 80:351. [PMID: 37930455 PMCID: PMC11072809 DOI: 10.1007/s00018-023-04948-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 06/02/2023] [Accepted: 06/21/2023] [Indexed: 11/07/2023]
Abstract
Alzheimer's disease (AD) is traditionally considered as a brain disorder featured by amyloid-β (Aβ) deposition. The current study on whether pathological changes of AD extend to the enteric nervous system (ENS) is still in its infancy. In this study, we found enteric Aβ deposition, intestinal dysfunction, and colonic inflammation in the young APP/PS1 mice. Moreover, these mice exhibited cholinergic and nitrergic signaling pathways damages and enteric neuronal loss. Our data show that Aβ42 treatment remarkably affected the gene expression of cultured myenteric neurons and the spontaneous contraction of intestinal smooth muscles. The intra-colon administration of Aβ42 induced ENS dysfunction, brain gliosis, and β-amyloidosis-like changes in the wild-type mice. Our results suggest that ENS mirrors the neuropathology observed in AD brains, and intestinal pathological changes may represent the prodromal events, which contribute to brain pathology in AD. In summary, our findings provide new opportunities for AD early diagnosis and prevention.
Collapse
Affiliation(s)
- Guoqiang Liu
- Medical College, Hubei University for Nationalities, Enshi, 445000, Hubei, China
| | - Quntao Yu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Houze Zhu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bo Tan
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongyan Yu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xinyan Li
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Youming Lu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Hao Li
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
23
|
Stallings NR, O'Neal MA, Hu J, Shen ZJ, Malter JS. Long-term normalization of calcineurin activity in model mice rescues Pin1 and attenuates Alzheimer's phenotypes without blocking peripheral T cell IL-2 response. Alzheimers Res Ther 2023; 15:179. [PMID: 37849016 PMCID: PMC10580561 DOI: 10.1186/s13195-023-01323-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Current treatments for Alzheimer's disease (AD) have largely failed to yield significant therapeutic benefits. Novel approaches are desperately needed to help address this immense public health issue. Data suggests that early intervention at the first stages of mild cognitive impairment may have a greater chance for success. The calcineurin (CN)-Pin1 signaling cascade can be selectively targeted with tacrolimus (FK506), a highly specific, FDA-approved CN inhibitor used safely for > 20 years in solid organ transplant recipients. AD prevalence was significantly reduced in solid organ recipients treated with FK506. METHODS Time release pellets were used to deliver constant FK506 dosage to APP/PS1 mice without deleterious manipulation or handling. Immunofluorescence, histology, molecular biology, and behavior were used to evaluate changes in AD pathology. RESULTS FK506 can be safely and consistently delivered into juvenile APP/PS1 mice via time-release pellets to levels roughly seen in transplant patients, leading to the normalization of CN activity and reduction or elimination of AD pathologies including synapse loss, neuroinflammation, and cognitive impairment. Pin1 activity and function were rescued despite the continuing presence of high levels of transgenic Aβ42. Indicators of neuroinflammation including Iba1 positivity and IL-6 production were also reduced to normal levels. Peripheral blood mononuclear cells (PBMC) obtained during treatment or splenocytes isolated at euthanasia activated normally after mitogens. CONCLUSIONS Low-dose, constant FK506 can normalize CNS CN and Pin1 activity, suppress neuroinflammation, and attenuate AD-associated pathology without blocking peripheral IL-2 responses making repurposed FK506 a viable option for early, therapeutic intervention in AD.
Collapse
Affiliation(s)
- Nancy R Stallings
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines, Dallas, TX, 75390, USA
| | - Melissa A O'Neal
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines, Dallas, TX, 75390, USA
| | - Jie Hu
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines, Dallas, TX, 75390, USA
| | - Zhong-Jian Shen
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines, Dallas, TX, 75390, USA
| | - James S Malter
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines, Dallas, TX, 75390, USA.
| |
Collapse
|
24
|
Garcia M, Paulus A, Vázquez-Reyes S, Klementieva O, Gouras G, Bachiller S, Deierborg T. Maternal separation differentially modulates early pathology by sex in 5xFAD Alzheimer's disease-transgenic mice. Brain Behav Immun Health 2023; 32:100663. [PMID: 37503358 PMCID: PMC10369403 DOI: 10.1016/j.bbih.2023.100663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/15/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. Most cases of AD are considered idiopathic and likely due to a combination of genetic, environmental, and lifestyle-related risk factors. Despite occurring decades before the typical age of an AD diagnosis, early-life stress (ELS) has been suggested to have long-lasting effects that may contribute to AD risk and pathogenesis. Still, the mechanisms that underlie the role of ELS on AD risk remain largely unknown. Here, we used 5xFAD transgenic mice to study relatively short-term alterations related to ELS in an AD-like susceptible mouse model at 6 weeks of age. To model ELS, we separated pups from their dams for 3 h per day from postnatal day 2-14. Around 6 weeks of age, we found that maternally separated (MS) 5xFAD mice, particularly female mice, displayed increased amyloid-β-immunoreactivity in the anterior cingulate cortex (ACC) and basolateral amygdala (BLA). In anterior cingulate cortex, we also noted significantly increased intraneuronal amyloid-β-immunoreactivity associated with MS but only in female mice. Moreover, IBA1-positive DAPI density was significantly increased in relation to MS in ACC and BLA, and microglia in BLA of MS mice had significantly different morphology compared to microglia in non-MS 5xFAD mice. Cytokine analysis showed that male MS mice, specifically, had increased levels of neuroinflammatory markers CXCL1 and IL-10 in hippocampal extracts compared to non-MS counterparts. Additionally, hippocampal extracts from both male and female MS 5xFAD mice had decreased levels of synapse- and activity-related markers Bdnf, 5htr6, Cox2, and Syp in hippocampus. Lastly, we performed behavioral tests to evaluate anxiety- and depressive-like behavior and working memory but could not detect any significant differences between groups. Overall, we detected several sex-specific molecular and cellular alterations in 6-week-old adolescent 5xFAD mice associated with MS that may help explain the connection between ELS and AD risk.
Collapse
Affiliation(s)
- M.G. Garcia
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - A. Paulus
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
- Medical Microspectroscopy, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - S. Vázquez-Reyes
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - O. Klementieva
- Medical Microspectroscopy, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - G.K. Gouras
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - S. Bachiller
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
- Institute of Biomedicine of Seville, IBiS/Virgen del Rocío University Hospital/CSIC/University of Seville/Seville, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - T. Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| |
Collapse
|
25
|
Kuhn MK, Fleeman RM, Beidler LM, Snyder AM, Chan DC, Proctor EA. Amyloid-β Pathology-Specific Cytokine Secretion Suppresses Neuronal Mitochondrial Metabolism. Cell Mol Bioeng 2023; 16:405-421. [PMID: 37811007 PMCID: PMC10550897 DOI: 10.1007/s12195-023-00782-y] [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: 02/17/2023] [Accepted: 08/28/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction Neuroinflammation and metabolic dysfunction are early alterations in Alzheimer's disease (AD) brain that are thought to contribute to disease onset and progression. Glial activation due to protein deposition results in cytokine secretion and shifts in brain metabolism, which have been observed in AD patients. However, the mechanism by which this immunometabolic feedback loop can injure neurons and cause neurodegeneration remains unclear. Methods We used Luminex XMAP technology to quantify hippocampal cytokine concentrations in the 5xFAD mouse model of AD at milestone timepoints in disease development. We used partial least squares regression to build cytokine signatures predictive of disease progression, as compared to healthy aging in wild-type littermates. We applied the disease-defining cytokine signature to wild-type primary neuron cultures and measured downstream changes in gene expression using the NanoString nCounter system and mitochondrial function using the Seahorse Extracellular Flux live-cell analyzer. Results We identified a pattern of up-regulated IFNγ, IP-10/CXCL10, and IL-9 as predictive of advanced disease. When healthy neurons were exposed to these cytokines in proportions found in diseased brain, gene expression of mitochondrial electron transport chain complexes, including ATP synthase, was suppressed. In live cells, basal and maximal mitochondrial respiration were impaired following cytokine stimulation. Conclusions We identify a pattern of cytokine secretion predictive of progressing amyloid-β pathology in the 5xFAD mouse model of AD that reduces expression of mitochondrial electron transport complexes and impairs mitochondrial respiration in healthy neurons. We establish a mechanistic link between disease-specific immune cues and impaired neuronal metabolism, potentially causing neuronal vulnerability and susceptibility to degeneration in AD. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00782-y.
Collapse
Affiliation(s)
- Madison K. Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA USA
| | - Rebecca M. Fleeman
- Department of Neurosurgery, Penn State College of Medicine, Hershey, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA
| | - Lynne M. Beidler
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA USA
| | - Amanda M. Snyder
- Department of Neurology, Penn State College of Medicine, Hershey, PA USA
| | - Dennis C. Chan
- Department of Neurosurgery, Penn State College of Medicine, Hershey, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA USA
| | - Elizabeth A. Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA USA
| |
Collapse
|
26
|
Cheng GWY, Ma IWT, Huang J, Yeung SHS, Ho P, Chen Z, Mak HKF, Herrup K, Chan KWY, Tse KH. Cuprizone drives divergent neuropathological changes in different mouse models of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.547147. [PMID: 37546935 PMCID: PMC10402084 DOI: 10.1101/2023.07.24.547147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Myelin degradation is a normal feature of brain aging that accelerates in Alzheimer's disease (AD). To date, however, the underlying biological basis of this correlation remains elusive. The amyloid cascade hypothesis predicts that demyelination is caused by increased levels of the β-amyloid (Aβ) peptide. Here we report on work supporting the alternative hypothesis that early demyelination is upstream of amyloid. We challenged two different mouse models of AD (R1.40 and APP/PS1) using cuprizone-induced demyelination and tracked the responses with both neuroimaging and neuropathology. In oppose to amyloid cascade hypothesis, R1.40 mice, carrying only a single human mutant APP (Swedish; APP SWE ) transgene, showed a more abnormal changes of magnetization transfer ratio and diffusivity than in APP/PS1 mice, which carry both APP SWE and a second PSEN1 transgene (delta exon 9; PSEN1 dE9 ). Although cuprizone targets oligodendrocytes (OL), magnetic resonance spectroscopy and targeted RNA-seq data in R1.40 mice suggested a possible metabolic alternation in axons. In support of alternative hypotheses, cuprizone induced significant intraneuronal amyloid deposition in young APP/PS1, but not in R1.40 mice, and it suggested the presence of PSEN deficiencies, may accelerate Aβ deposition upon demyelination. In APP/PS1, mature OL is highly vulnerable to cuprizone with significant DNA double strand breaks (53BP1 + ) formation. Despite these major changes in myelin, OLs, and Aβ immunoreactivity, no cognitive impairment or hippocampal pathology was detected in APP/PS1 mice after cuprizone treatment. Together, our data supports the hypothesis that myelin loss can be the cause, but not the consequence, of AD pathology. SIGNIFICANCE STATEMENT The causal relationship between early myelin loss and the progression of Alzheimer's disease remains unclear. Using two different AD mouse models, R1.40 and APP/PS1, our study supports the hypothesis that myelin abnormalities are upstream of amyloid production and deposition. We find that acute demyelination initiates intraneuronal amyloid deposition in the frontal cortex. Further, the loss of oligodendrocytes, coupled with the accelerated intraneuronal amyloid deposition, interferes with myelin tract diffusivity at a stage before any hippocampus pathology or cognitive impairments occur. We propose that myelin loss could be the cause, not the consequence, of amyloid pathology during the early stages of Alzheimer's disease.
Collapse
|
27
|
Zhang W, Xiao D, Mao Q, Xia H. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther 2023; 8:267. [PMID: 37433768 PMCID: PMC10336149 DOI: 10.1038/s41392-023-01486-5] [Citation(s) in RCA: 410] [Impact Index Per Article: 205.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 07/13/2023] Open
Abstract
Studies in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and Amyotrophic lateral sclerosis, Huntington's disease, and so on, have suggested that inflammation is not only a result of neurodegeneration but also a crucial player in this process. Protein aggregates which are very common pathological phenomenon in neurodegeneration can induce neuroinflammation which further aggravates protein aggregation and neurodegeneration. Actually, inflammation even happens earlier than protein aggregation. Neuroinflammation induced by genetic variations in CNS cells or by peripheral immune cells may induce protein deposition in some susceptible population. Numerous signaling pathways and a range of CNS cells have been suggested to be involved in the pathogenesis of neurodegeneration, although they are still far from being completely understood. Due to the limited success of traditional treatment methods, blocking or enhancing inflammatory signaling pathways involved in neurodegeneration are considered to be promising strategies for the therapy of neurodegenerative diseases, and many of them have got exciting results in animal models or clinical trials. Some of them, although very few, have been approved by FDA for clinical usage. Here we comprehensively review the factors affecting neuroinflammation and the major inflammatory signaling pathways involved in the pathogenicity of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic lateral sclerosis. We also summarize the current strategies, both in animal models and in the clinic, for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Weifeng Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China
| | - Dan Xiao
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, P.R. China
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Qinwen Mao
- Department of Pathology, University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China.
| |
Collapse
|
28
|
Kuhn MK, Fleeman RM, Beidler LM, Snyder AM, Chan DC, Proctor EA. Alzheimer's disease-specific cytokine secretion suppresses neuronal mitochondrial metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.07.536014. [PMID: 37066287 PMCID: PMC10104145 DOI: 10.1101/2023.04.07.536014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Introduction Neuroinflammation and metabolic dysfunction are early alterations in Alzheimer's disease brain that are thought to contribute to disease onset and progression. Glial activation due to protein deposition results in cytokine secretion and shifts in brain metabolism, which have been observed in Alzheimer's disease patients. However, the mechanism by which this immunometabolic feedback loop can injure neurons and cause neurodegeneration remains unclear. Methods We used Luminex XMAP technology to quantify hippocampal cytokine concentrations in the 5xFAD mouse model of Alzheimer's disease at milestone timepoints in disease development. We used partial least squares regression to build cytokine signatures predictive of disease progression, as compared to healthy aging in wild-type littermates. We applied the disease-defining cytokine signature to wild-type primary neuron cultures and measured downstream changes in gene expression using the NanoString nCounter system and mitochondrial function using the Seahorse Extracellular Flux live-cell analyzer. Results We identified a pattern of up-regulated IFNγ, IP-10, and IL-9 as predictive of advanced disease. When healthy neurons were exposed to these cytokines in proportions found in diseased brain, gene expression of mitochondrial electron transport chain complexes, including ATP synthase, was suppressed. In live cells, basal and maximal mitochondrial respiration were impaired following cytokine stimulation. Conclusions An Alzheimer's disease-specific pattern of cytokine secretion reduces expression of mitochondrial electron transport complexes and impairs mitochondrial respiration in healthy neurons. We establish a mechanistic link between disease-specific immune cues and impaired neuronal metabolism, potentially causing neuronal vulnerability and susceptibility to degeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Madison K. Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Rebecca M. Fleeman
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Lynne M. Beidler
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA, USA
| | - Amanda M. Snyder
- Department of Neurology, Penn State College of Medicine, Hershey, PA, USA
| | - Dennis C. Chan
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Elizabeth A. Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
29
|
Zhang RL, Lei BX, Wu GY, Wang YY, Huang QH. Protective effects of berberine against β-amyloid-induced neurotoxicity in HT22 cells via the Nrf2/HO-1 pathway. Bioorg Chem 2023; 133:106210. [PMID: 36724611 DOI: 10.1016/j.bioorg.2022.106210] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 09/21/2022] [Accepted: 10/11/2022] [Indexed: 02/03/2023]
Abstract
Neuronal apoptosis has been found to have a pivotal role in the course of Alzheimer's disease (AD). Berberine (BBR), a potent antioxidant, occurs in plants such as Berberis, Phellodendron chinense, and Hydrastis canadensis. In this study, a neuronal apoptotic model was established in vitro using HT22 cells induced by Aβ25-35 to explore whether BBR contributes to protecting neurons against Aβ25-35-induced neurotoxicity, as well as its potential mechanisms. BBR was applied to HT22 cells for 1 h prior to exposing the cells to Aβ25-35 for 24 h. A CCK-8 assay was utilized to assess cell viability, and Annexin V - fluorescein isothiocyanate (FITC)/propidium iodide and Hoechst 33342 fluorescence staining were used to measure the rate of cell apoptosis. Existing scientific literature was also reviewed to further determine the effects of BBR on ROS production and mitochondrial function in HT22 cells. Furthermore, the expressions of proteins, including cytochrome C, cleaved caspase-3, p-p65, p65, and Nrf2/HO-1 antioxidant axis were assessed by Western blotting. The data indicated that BBR markedly improved cell viability, inhibited apoptosis and intracellular ROS levels, improved mitochondrial membrane potentials, decreased the rate of p-p65/p65, cytochrome C, and cleaved caspase-3, and intensified the activity of Nrf2/HO-1 antioxidants in HT22 cells. Overall, the findings indicated that BBR provides a certain level of neuroprotectiveness in HT22 cells exposed to Aβ25-35 via relieving oxidative stress, as well as by restraining the mitochondrial pathway of cellular apoptosis. In addition, the restraint of NF-κB activity and sensitization of the Nrf2/HO-1 antioxidant axis, which together are intimately involved in the neuroprotection of BBR, may be possible mechanisms accounting for its effectiveness against Aβ25-35in vitro.
Collapse
Affiliation(s)
- Ru-Lan Zhang
- Department of Chinese Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China
| | - Bing-Xi Lei
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Guo-Yong Wu
- Department of Thoracic Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China
| | - Yuan-Yuan Wang
- Department of Chinese Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China
| | - Qi-Hui Huang
- Department of Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China.
| |
Collapse
|
30
|
Mehta RI, Mehta RI. The Vascular-Immune Hypothesis of Alzheimer's Disease. Biomedicines 2023; 11:408. [PMID: 36830944 PMCID: PMC9953491 DOI: 10.3390/biomedicines11020408] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating and irreversible neurodegenerative disorder with unknown etiology. While its cause is unclear, a number of theories have been proposed to explain the pathogenesis of AD. In large part, these have centered around potential causes for intracerebral accumulation of beta-amyloid (βA) and tau aggregates. Yet, persons with AD dementia often exhibit autopsy evidence of mixed brain pathologies including a myriad of vascular changes, vascular brain injuries, complex brain inflammation, and mixed protein inclusions in addition to hallmark neuropathologic lesions of AD, namely insoluble βA plaques and neurofibrillary tangles (NFTs). Epidemiological data demonstrate that overlapping lesions diminish the βA plaque and NFT threshold necessary to precipitate clinical dementia. Moreover, a subset of persons who exhibit AD pathology remain resilient to disease while other persons with clinically-defined AD dementia do not exhibit AD-defining neuropathologic lesions. It is increasingly recognized that AD is a pathologically heterogeneous and biologically multifactorial disease with uncharacterized biologic phenomena involved in its genesis and progression. Here, we review the literature with regard to neuropathologic criteria and incipient AD changes, and discuss converging concepts regarding vascular and immune factors in AD.
Collapse
Affiliation(s)
- Rashi I. Mehta
- Department of Neuroradiology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Rupal I. Mehta
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
31
|
The Role of Dietary Lipids in Cognitive Health: Implications for Neurodegenerative Disease. Biomedicines 2022; 10:biomedicines10123250. [PMID: 36552006 PMCID: PMC9775642 DOI: 10.3390/biomedicines10123250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Neurodegenerative diseases are a group of disorders characterised by progressive loss of brain function. The most common of these is Alzheimer's disease, a form of dementia. Intake of macro- and micro-nutrients impacts brain function, including memory, learning, mood, and behaviour. Lipids, particularly phospholipids and sphingolipids, are crucial structural components of neural tissues and significantly affect cognitive function. The importance of functional foods in preventing cardiovascular disease is well-documented in the current literature. However, the significance of such foods for central nervous system health and neurodegenerative diseases is less recognized. Gut microbiome composition affects cognitive health and function, and dietary lipids are known to influence gut health. Thus, this review will discuss different sources of dietary lipids and their effect on cognitive functioning and their interaction with the gut microbiome in the context of neurodegenerative disease.
Collapse
|
32
|
Ge X, Wang L, Cui Q, Yan H, Wang Z, Ye S, Zhang Q, Fei A. Electroacupuncture improves cognitive impairment in diabetic cognitive dysfunction rats by regulating the mitochondrial autophagy pathway. J Physiol Sci 2022; 72:29. [PMID: 36418941 PMCID: PMC10717526 DOI: 10.1186/s12576-022-00854-0] [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: 05/22/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Diabetes-associated cognitive dysfunction has become a major public health concern. However, the mechanisms driving this disease are elusive. Herein, we explored how electroacupuncture improves learning and memory function in diabetic rats. METHODS The diabetic model was established by intraperitoneal injection of streptozotocin (STZ) in adult Sprague-Dawley rats. Rats were fed on high-fat and high-sugar diets. Learning and memory functions were assessed using behavioral tests. The hematoxylin and eosin (H&E) staining, Western blotting, real-time PCR, ELISA, immunohistochemistry, and transmission electronic microscopy (TEM) was performed to test related indicators. RESULTS High-fat and high-sugar diets impaired learning and memory function in rats, while electroacupuncture treatment reversed these changes. The model group presented highly prolonged escape latency compared to the control group, indicating impaired learning and memory functions. The TEM examination showed that electroacupuncture enhanced Aβ clearance and mitochondrial autophagy in hippocampal neuronal cells by increasing DISC1 expression. CONCLUSIONS Electroacupuncture improves learning and memory function in diabetic rats by increasing DISC1 expression to promote mitophagy. This enhanced Aβ clearance, alleviating cytotoxicity in hippocampal neuronal cells.
Collapse
Affiliation(s)
- Xia Ge
- Department of Endocrinology, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230001, China
| | - Ling Wang
- College of Second Clinical Medical, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Qianqian Cui
- Department of Emergency, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230001, China
| | - Hongli Yan
- Department of Acupuncture-Moxibustion and Rehabilitation, Mingguang Hospital of Traditional Chinese Medicine, Chuzhou, 239499, China
| | - Zhongbao Wang
- College of Third Clinical Medical, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Shandong Ye
- Department of Endocrinology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230002, China.
| | - Qingping Zhang
- College of Acupuncture-Moxibustion and Tuina, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Aihua Fei
- Department of Endocrinology, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230001, China.
| |
Collapse
|
33
|
Faridar A, Vasquez M, Thome AD, Yin Z, Xuan H, Wang JH, Wen S, Li X, Thonhoff JR, Zhao W, Zhao H, Beers DR, Wong STC, Masdeu JC, Appel SH. Ex vivo expanded human regulatory T cells modify neuroinflammation in a preclinical model of Alzheimer's disease. Acta Neuropathol Commun 2022; 10:144. [PMID: 36180898 PMCID: PMC9524037 DOI: 10.1186/s40478-022-01447-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Background Regulatory T cells (Tregs) play a neuroprotective role by suppressing microglia and macrophage-mediated inflammation and modulating adaptive immune reactions. We previously documented that Treg immunomodulatory mechanisms are compromised in Alzheimer’s disease (AD). Ex vivo expansion of Tregs restores and amplifies their immunosuppressive functions in vitro. A key question is whether adoptive transfer of ex vivo expanded human Tregs can suppress neuroinflammation and amyloid pathology in a preclinical mouse model. Methods An immunodeficient mouse model of AD was generated by backcrossing the 5xFAD onto Rag2 knockout mice (5xFAD-Rag2KO). Human Tregs were expanded ex vivo for 24 days and administered to 5xFAD-Rag2KO. Changes in amyloid burden, microglia characteristics and reactive astrocytes were evaluated using ELISA and confocal microscopy. NanoString Mouse AD multiplex gene expression analysis was applied to explore the impact of ex vivo expanded Tregs on the neuroinflammation transcriptome. Results Elimination of mature B and T lymphocytes and natural killer cells in 5xFAD-Rag2KO mice was associated with upregulation of 95 inflammation genes and amplified number of reactive microglia within the dentate gyrus. Administration of ex vivo expanded Tregs reduced amyloid burden and reactive glial cells in the dentate gyrus and frontal cortex of 5xFAD-Rag2KO mice. Interrogation of inflammation gene expression documented down-regulation of pro-inflammatory cytokines (IL1A&B, IL6), complement cascade (C1qa, C1qb, C1qc, C4a/b), toll-like receptors (Tlr3, Tlr4 and Tlr7) and microglial activations markers (CD14, Tyrobp,Trem2) following Treg administration. Conclusions Ex vivo expanded Tregs with amplified immunomodulatory function, suppressed neuroinflammation and alleviated AD pathology in vivo. Our results provide preclinical evidences for Treg cell therapy as a potential treatment strategy in AD. Supplementary Information The online version contains supplementary material available at 10.1186/s40478-022-01447-z.
Collapse
Affiliation(s)
- Alireza Faridar
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Matthew Vasquez
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - Aaron D Thome
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Zheng Yin
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - Hui Xuan
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Jing Hong Wang
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Shixiang Wen
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Xuping Li
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston, TX, USA
| | - Jason R Thonhoff
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Weihua Zhao
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Hong Zhao
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - David R Beers
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Stephen T C Wong
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - Joseph C Masdeu
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Stanley H Appel
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA.
| |
Collapse
|
34
|
Luo M, Lee LKC, Peng B, Choi CHJ, Tong WY, Voelcker NH. Delivering the Promise of Gene Therapy with Nanomedicines in Treating Central Nervous System Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201740. [PMID: 35851766 PMCID: PMC9475540 DOI: 10.1002/advs.202201740] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/19/2022] [Indexed: 06/01/2023]
Abstract
Central Nervous System (CNS) diseases, such as Alzheimer's diseases (AD), Parkinson's Diseases (PD), brain tumors, Huntington's disease (HD), and stroke, still remain difficult to treat by the conventional molecular drugs. In recent years, various gene therapies have come into the spotlight as versatile therapeutics providing the potential to prevent and treat these diseases. Despite the significant progress that has undoubtedly been achieved in terms of the design and modification of genetic modulators with desired potency and minimized unwanted immune responses, the efficient and safe in vivo delivery of gene therapies still poses major translational challenges. Various non-viral nanomedicines have been recently explored to circumvent this limitation. In this review, an overview of gene therapies for CNS diseases is provided and describes recent advances in the development of nanomedicines, including their unique characteristics, chemical modifications, bioconjugations, and the specific applications that those nanomedicines are harnessed to deliver gene therapies.
Collapse
Affiliation(s)
- Meihua Luo
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Australian Institute for Bioengineering and Nanotechnologythe University of QueenslandSt LuciaQLD4072Australia
| | - Leo Kit Cheung Lee
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Bo Peng
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Frontiers Science Center for Flexible ElectronicsXi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical materials & EngineeringNorthwestern Polytechnical UniversityXi'an710072China
| | - Chung Hang Jonathan Choi
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Wing Yin Tong
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO)ClaytonVIC3168Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClaytonVIC3168Australia
- Materials Science and EngineeringMonash University14 Alliance LaneClaytonVIC3800Australia
| |
Collapse
|
35
|
Goel P, Chakrabarti S, Goel K, Bhutani K, Chopra T, Bali S. Neuronal cell death mechanisms in Alzheimer's disease: An insight. Front Mol Neurosci 2022; 15:937133. [PMID: 36090249 PMCID: PMC9454331 DOI: 10.3389/fnmol.2022.937133] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Regulated cell death (RCD) is an ordered and tightly orchestrated set of changes/signaling events in both gene expression and protein activity and is responsible for normal development as well as maintenance of tissue homeostasis. Aberrant activation of this pathway results in cell death by various mechanisms including apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death. Such pathological changes in neurons alone or in combination have been observed in the pathogenesis of various neurodegenerative diseases including Alzheimer's disease (AD). Pathological hallmarks of AD focus primarily on the accumulation of two main protein markers: amyloid β peptides and abnormally phosphorylated tau proteins. These protein aggregates result in the formation of A-β plaques and neuro-fibrillary tangles (NFTs) and induce neuroinflammation and neurodegeneration over years to decades leading to a multitude of cognitive and behavioral deficits. Autopsy findings of AD reveal massive neuronal death manifested in the form of cortical volume shrinkage, reduction in sizes of gyri to up to 50% and an increase in the sizes of sulci. Multiple forms of cell death have been recorded in neurons from different studies conducted so far. However, understanding the mechanism/s of neuronal cell death in AD patients remains a mystery as the trigger that results in aberrant activation of RCD is unknown and because of the limited availability of dying neurons. This review attempts to elucidate the process of Regulated cell death, how it gets unregulated in response to different intra and extracellular stressors, various forms of unregulated cell death, their interplay and their role in pathogenesis of Alzheimer's Disease in both human and experimental models of AD. Further we plan to explore the correlation of both amyloid-beta and Tau with neuronal loss as seen in AD.
Collapse
Affiliation(s)
- Parul Goel
- Department of Biochemistry, Shri Atal Bihari Vajpayee Government Medical College Chhainsa, Faridabad, India
| | - Sasanka Chakrabarti
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Kapil Goel
- Department of Community Medicine and School of Public Health, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Karanpreet Bhutani
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Tanya Chopra
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Sharadendu Bali
- Department of Surgery, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| |
Collapse
|
36
|
Flores-Aguilar L, Hall H, Orciani C, Foret MK, Kovecses O, Ducatenzeiler A, Cuello AC. Early loss of locus coeruleus innervation promotes cognitive and neuropathological changes before amyloid plaque deposition in a transgenic rat model of Alzheimer's disease. Neuropathol Appl Neurobiol 2022; 48:e12835. [PMID: 35822518 DOI: 10.1111/nan.12835] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/31/2022] [Accepted: 07/06/2022] [Indexed: 11/29/2022]
Abstract
AIMS The locus coeruleus (LC) is the main source of noradrenaline (NA) in the mammalian brain and has been found to degenerate during the initial stages of Alzheimer's disease (AD). Recent studies indicate that at late stages of the amyloid pathology, LC-pathological alterations accelerate AD-like pathology progression by interfering with the neuromodulatory and anti-inflammatory properties of NA. However, the impact of LC degeneration at the earliest stages of amyloidosis on the AD-like pathology is not well understood. METHODS The LC was lesioned in wild-type (wt) and McGill-R-Thy1-APP transgenic rats (APP tg) by administering N-(2-chloroethyl)-N-ethyl-bromo-benzylamine (DSP4) before amyloid plaque deposition. Cognitive deficits and AD-like neuropathological changes were measured after the LC lesion. RESULTS Four months post-treatment, rats displayed a decrease in brain noradrenergic innervation. The LC lesion in APP tg-treated rats enhanced cognitive deficits and decreased hippocampal cholinergic innervation and neurotrophin expression. In addition, the APP tg-treated rats displayed an increased microglial and astroglial cell number in close vicinity to hippocampal amyloid-beta burdened neurons. The recruited microglia showed cellular alterations indicative of an intermediate activation state. CONCLUSIONS Our results indicate that early LC demise aggravates the early neuroinflammatory process, cognitive impairments, cholinergic deficits and neurotrophin deregulation at the earliest stages of the human-like brain amyloidosis.
Collapse
Affiliation(s)
- Lisi Flores-Aguilar
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.,Current affiliation: Department of Pathology and Laboratory Medicine, University of California, Irvine, United States of America
| | - Hélène Hall
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Chiara Orciani
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Morgan K Foret
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Olivia Kovecses
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | | | - A Claudio Cuello
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Visiting Professor, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
37
|
Zhang L, Liu Y, Wang X, Wang D, Wu H, Chen H, Chen J, Liu Y. Treadmill exercise improve recognition memory by TREM2 pathway to inhibit hippocampal microglial activation and neuroinflammation in Alzheimer's disease model. Physiol Behav 2022; 251:113820. [PMID: 35452628 DOI: 10.1016/j.physbeh.2022.113820] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/30/2022] [Accepted: 04/16/2022] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease-related cognition impairment is correlated with increased neuroinflammation. Studies show that physical exercises improve cognitive function and regulate neuroinflammation. However, no sufficient studies have been performed to directly observe the mechanism of exercise-related effects on microglia and neuroinflammation, in association with memory function under Alzheimer's disease. This study aims to explore the relationship of TREM2, microglia activation and neuroinflammation in the development of Alzheimer's disease, followed by investigating why physical exercises improve cognition in the Alzheimer's disease model by means of the adeno-associated virus (AAV) injection. We found that: 1) Recognition memory impairment in Aβ-induced Alzheimer's disease model was associated with the reduction in TREM2 which induced microglial activation and neuroinflammation; 2) Exercise activated the TREM2 pathway, which was necessary for inhibiting microglial activation and neuroinflammation, leading to improved recognition memory in the Alzheimer's disease model. Together, the improvement of AD-associated recognition memory by exercises is associated with up-regulation of the TREM2 pathway which promotes the phenotypic conversion of microglia and decreases the level of neuroinflammation.
Collapse
Affiliation(s)
- Linlin Zhang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, 350007, China; Comprehensive Key Laboratory of Sports Ability Evaluation and Research of the General Administration of Sport of China, Beijing Key Laboratory of Sports Function Assessment and Technical Analysis, Capital University of Physical Education and Sports, Beijing, 100191, China.
| | - Yanzhong Liu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, 350007, China
| | - Xin Wang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, 350007, China
| | - Dan Wang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, 350007, China
| | - Hao Wu
- Comprehensive Key Laboratory of Sports Ability Evaluation and Research of the General Administration of Sport of China, Beijing Key Laboratory of Sports Function Assessment and Technical Analysis, Capital University of Physical Education and Sports, Beijing, 100191, China
| | - Haichun Chen
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, 350007, China
| | - Jiaxin Chen
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, 350007, China
| | - Yiping Liu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, 350007, China.
| |
Collapse
|
38
|
Welch G, Tsai LH. Mechanisms of DNA damage-mediated neurotoxicity in neurodegenerative disease. EMBO Rep 2022; 23:e54217. [PMID: 35499251 PMCID: PMC9171412 DOI: 10.15252/embr.202154217] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/18/2022] [Accepted: 04/19/2022] [Indexed: 12/26/2022] Open
Abstract
Neurons are highly susceptible to DNA damage accumulation due to their large energy requirements, elevated transcriptional activity, and long lifespan. While newer research has shown that DNA breaks and mutations may facilitate neuron diversity during development and neuronal function throughout life, a wealth of evidence indicates deficient DNA damage repair underlies many neurological disorders, especially age-associated neurodegenerative diseases. Recently, efforts to clarify the molecular link between DNA damage and neurodegeneration have improved our understanding of how the genomic location of DNA damage and defunct repair proteins impact neuron health. Additionally, work establishing a role for senescence in the aging and diseased brain reveals DNA damage may play a central role in neuroinflammation associated with neurodegenerative disease.
Collapse
Affiliation(s)
- Gwyneth Welch
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
39
|
Ring J, Tadic J, Ristic S, Poglitsch M, Bergmann M, Radic N, Mossmann D, Liang Y, Maglione M, Jerkovic A, Hajiraissi R, Hanke M, Küttner V, Wolinski H, Zimmermann A, Domuz Trifunović L, Mikolasch L, Moretti DN, Broeskamp F, Westermayer J, Abraham C, Schauer S, Dammbrueck C, Hofer SJ, Abdellatif M, Grundmeier G, Kroemer G, Braun RJ, Hansen N, Sommer C, Ninkovic M, Seba S, Rockenfeller P, Vögtle F, Dengjel J, Meisinger C, Keller A, Sigrist SJ, Eisenberg T, Madeo F. The HSP40 chaperone Ydj1 drives amyloid beta 42 toxicity. EMBO Mol Med 2022; 14:e13952. [PMID: 35373908 PMCID: PMC9081910 DOI: 10.15252/emmm.202113952] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 01/22/2023] Open
Abstract
Amyloid beta 42 (Abeta42) is the principal trigger of neurodegeneration during Alzheimer's disease (AD). However, the etiology of its noxious cellular effects remains elusive. In a combinatory genetic and proteomic approach using a yeast model to study aspects of intracellular Abeta42 toxicity, we here identify the HSP40 family member Ydj1, the yeast orthologue of human DnaJA1, as a crucial factor in Abeta42-mediated cell death. We demonstrate that Ydj1/DnaJA1 physically interacts with Abeta42 (in yeast and mouse), stabilizes Abeta42 oligomers, and mediates their translocation to mitochondria. Consequently, deletion of YDJ1 strongly reduces co-purification of Abeta42 with mitochondria and prevents Abeta42-induced mitochondria-dependent cell death. Consistently, purified DnaJ chaperone delays Abeta42 fibrillization in vitro, and heterologous expression of human DnaJA1 induces formation of Abeta42 oligomers and their deleterious translocation to mitochondria in vivo. Finally, downregulation of the Ydj1 fly homologue, Droj2, improves stress resistance, mitochondrial morphology, and memory performance in a Drosophila melanogaster AD model. These data reveal an unexpected and detrimental role for specific HSP40s in promoting hallmarks of Abeta42 toxicity.
Collapse
|
40
|
Liu J, Jiao L, Zhong X, Yao W, Du K, Lu S, Wu Y, Ma T, Tong J, Xu M, Jiang W, Wang Y, He M, Xin W, Liu M. Platelet Activating Factor Receptor Exaggerates Microglia-Mediated Microenvironment by IL10-STAT3 Signaling: A Novel Potential Biomarker and Target for Diagnosis and Treatment of Alzheimer's Disease. Front Aging Neurosci 2022; 14:856628. [PMID: 35572136 PMCID: PMC9096237 DOI: 10.3389/fnagi.2022.856628] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022] Open
Abstract
Background Early diagnosis and effective intervention are the keys to delaying the progression of Alzheimer's Disease (AD). Therefore, we aimed to identify new biomarkers for the early diagnosis of AD through bioinformatic analysis and elucidate the possible underlying mechanisms. Methods and Results GSE1297, GSE63063, and GSE110226 datasets from the GEO database were used to screen the highly differentially expressed genes. We identified a potential biomarker, Platelet activating factor receptor (PTAFR), significantly upregulated in the brain tissue, peripheral blood, and cerebrospinal fluid of AD patients. Furthermore, PTAFR levels in the plasma and brain tissues of APP/PS1 mice were significantly elevated. Simultaneously, PTAFR could mediate the inflammatory responses to exaggerate the microenvironment, particularly mediated by the microglia through the IL10-STAT3 pathway. In addition, PTAFR was a putative target of anti-AD compounds, including EGCG, donepezil, curcumin, memantine, and Huperzine A. Conclusion PTAFR was a potential biomarker for early AD diagnosis and treatment which correlated with the microglia-mediated microenvironment. It is an important putative target for the development of a novel strategy for clinical treatment and drug discovery for AD.
Collapse
Affiliation(s)
- Junxiu Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Linchi Jiao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Xin Zhong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Weifan Yao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Senxu Lu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Yuqiang Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Tianxin Ma
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Junhui Tong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Mingyue Xu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Wenjuan Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Yubao Wang
- Liaoning Medical Diagnosis and Treatment Center, Shenyang, China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China,Miao He,
| | - Wei Xin
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,The First Affiliated Hospital of China Medical University, Shenyang, China,Wei Xin,
| | - Mingyan Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China,*Correspondence: Mingyan Liu,
| |
Collapse
|
41
|
Gallego Villarejo L, Bachmann L, Marks D, Brachthäuser M, Geidies A, Müller T. Role of Intracellular Amyloid β as Pathway Modulator, Biomarker, and Therapy Target. Int J Mol Sci 2022; 23:ijms23094656. [PMID: 35563046 PMCID: PMC9103247 DOI: 10.3390/ijms23094656] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
The β- and γ-secretase-driven cleavage of the amyloid precursor protein (APP) gives rise to the amyloid β peptide, which is believed to be the main driver of neurodegeneration in Alzheimer’s disease (AD). As it is prominently detectable in extracellular plaques in post-mortem AD brain samples, research in recent decades focused on the pathological role of extracellular amyloid β aggregation, widely neglecting the potential meaning of very early generation of amyloid β inside the cell. In the last few years, the importance of intracellular amyloid β (iAβ) as a strong player in neurodegeneration has been indicated by a rising number of studies. In this review, iAβ is highlighted as a crucial APP cleavage fragment, able to manipulate intracellular pathways and foster neurodegeneration. We demonstrate its relevance as a pathological marker and shed light on initial studies aiming to modulate iAβ through pharmacological treatment, which has been shown to have beneficial effects on cognitive properties in animal models. Finally, we display the relevance of viral infections on iAβ generation and point out future directions urgently needed to manifest the potential relevance of iAβ in Alzheimer’s disease.
Collapse
Affiliation(s)
- Lucia Gallego Villarejo
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Lisa Bachmann
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - David Marks
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Maite Brachthäuser
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Alexander Geidies
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Thorsten Müller
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, 80336 Munich, Germany
- Correspondence:
| |
Collapse
|
42
|
Benavides GA, Mueller T, Darley-Usmar V, Zhang J. Optimization of measurement of mitochondrial electron transport activity in postmortem human brain samples and measurement of susceptibility to rotenone and 4-hydroxynonenal inhibition. Redox Biol 2022; 50:102241. [PMID: 35066289 PMCID: PMC8792425 DOI: 10.1016/j.redox.2022.102241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/25/2022] Open
Abstract
Mitochondrial function is required to meet the energetic and metabolic requirements of the brain. Abnormalities in mitochondrial function, due to genetic or developmental factors, mitochondrial toxins, aging or insufficient mitochondrial quality control contribute to neurological and psychiatric diseases. Studying bioenergetics from postmortem human tissues has been challenging due to the diverse range of human genetics, health conditions, sex, age, and postmortem interval. Furthermore, fresh tissues that were in the past required for assessment of mitochondrial respiratory function were rarely available. Recent studies established protocols to use in bioenergetic analyses from frozen tissues using animal models and cell cultures. In this study we optimized these methods to determine the activities of mitochondrial electron transport in postmortem human brain. Further we demonstrate how these samples can be used to assess the susceptibility to the mitochondrial toxin rotenone and exposure to the reactive lipid species 4-hydroxynonenal. The establishment of such an approach will significantly impact translational studies of human diseases by allowing measurement of mitochondrial function in human tissue repositories.
Collapse
Affiliation(s)
- Gloria A Benavides
- Department of Pathology, Mitochondrial Medicine Laboratory, Birmingham, AL, 35294, USA
| | - Toni Mueller
- Department of Pathology, Mitochondrial Medicine Laboratory, Birmingham, AL, 35294, USA
| | - Victor Darley-Usmar
- Department of Pathology, Mitochondrial Medicine Laboratory, Birmingham, AL, 35294, USA
| | - Jianhua Zhang
- Department of Pathology, Mitochondrial Medicine Laboratory, Birmingham, AL, 35294, USA; Birmingham VA Medical Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
43
|
Lin Y, Ma HY, Wang Y, He J, Liu HJ. Identification of Potential Core Genes for the Rupture of Intracranial Aneurysms by a Bioinformatics Analysis. Front Genet 2022; 13:875007. [PMID: 35432454 PMCID: PMC9006073 DOI: 10.3389/fgene.2022.875007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Previous studies, using autopsy and angiography, have shown that 3.6–6% of the population have intracranial aneurysms, and the rupture of aneurysm can lead to brain dysfunction or even death in patients. Methods: To explore potential preventional target genes for the ruptured of aneurysm, we analyze three gene expression datasets (GSE13353, GSE15629 and GSE54083) derived from the GEO database. We confirm DEGs associated with the unrupture of aneurysms by R package. DAVID version provides functional classification and annotation analyses of associated genes, including GO and KEGG pathway. PPI of these DEGs is analyzed based on the string database and visualized by Cytoscape software. DEGs are verified by qRT-PCR using samples isolated from the patients. Results: 249 overlapping DEGs, including 96 up-regulated genes and 153 down-regulated genes are screened using the Venn diagram webtool. The GO term and KEGG pathways analysis results indicate that these DEGs are mainly enriched in protein phosphorylation, apoptotic process and inflammatory response in the BP term and focal adhesion, thyroid hormone signaling pathway, ErbB signaling pathway, cytokine-cytokine receptor interaction and some disease processes in the KEGG pathways. 6 candidates are confirmed by Cytoscape software and qRT-PCR, including APP, JUN, GSK3B, ErbB2, PPBP and THBS1. Conclusions: Our data and previous studies show that ErbB2 and THBS1 are crucial to prevent aneurysm rupture, while APP, JUN, GSK3B and PPBP performs the opposite role, and further experiments are needed to verify these findings.
Collapse
|
44
|
Li C, Liu M, Xia J, Mei L, Yang Q, Shi F, Zhang H, Shen D. Predicting Brain Amyloid-β PET Grades with Graph Convolutional Networks Based on Functional MRI and Multi-Level Functional Connectivity. J Alzheimers Dis 2022; 86:1679-1693. [PMID: 35213377 DOI: 10.3233/jad-215497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The detection of amyloid-β (Aβ) deposition in the brain provides crucial evidence in the clinical diagnosis of Alzheimer's disease (AD). However, the current positron emission tomography (PET)-based brain Aβ examination suffers from the problems of coarse visual inspection (in many cases, with 2-class stratification) and high scanning cost. OBJECTIVE 1) To characterize the non-binary Aβ deposition levels in the AD continuum based on clustering of PET data, and 2) to explore the feasibility of predicting individual Aβ deposition grades with non-invasive functional magnetic resonance imaging (fMRI). METHODS 1) Individual whole-brain Aβ-PET images from the OASIS-3 dataset (N = 258) were grouped into three clusters (grades) with t-SNE and k-means. The demographical data as well as global and regional standard uptake value ratios (SUVRs) were compared among the three clusters with Chi-square tests or ANOVA tests. 2) From resting-state fMRI, both conventional functional connectivity (FC) and high-order FC networks were constructed and the topological architectures of the two networks were jointly learned with graph convolutional networks (GCNs) to predict the Aβ-PET grades for each individual. RESULTS We found three clearly separated clusters, indicating three Aβ-PET grades. There were significant differences in gender, age, cognitive ability, APOE type, as well as global and regional SUVRs among the three grades we found. The prediction of Aβ-PET grades with GCNs on FC for the 258 participants in the AD continuum reached a satisfactory averaged accuracy (78.8%) in the two-class classification tasks. CONCLUSION The results demonstrated the feasibility of using deep learning on a non-invasive brain functional imaging technique to approximate PET-based Aβ deposition grading.
Collapse
Affiliation(s)
- Chaolin Li
- School of Education, Guangzhou University, Guangzhou, China.,School of Biomedical Engineering, Shanghai Tech University, Shanghai, China
| | - Mianxin Liu
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, China
| | - Jing Xia
- Institute of Brain-Intelligence Technology, Zhangjiang Lab, Shanghai, China
| | - Lang Mei
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, China
| | - Qing Yang
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, China
| | - Feng Shi
- Department of Research and Development, United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Han Zhang
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, China
| | - Dinggang Shen
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, China.,Department of Research and Development, United Imaging Intelligence Co., Ltd., Shanghai, China
| |
Collapse
|
45
|
Bartley SC, Proctor MT, Xia H, Ho E, Kang DS, Schuster K, Bicca MA, Seckler HS, Viola KL, Patrie SM, Kelleher NL, De Mello FG, Klein WL. An Essential Role for Alzheimer’s-Linked Amyloid Beta Oligomers in Neurodevelopment: Transient Expression of Multiple Proteoforms during Retina Histogenesis. Int J Mol Sci 2022; 23:ijms23042208. [PMID: 35216328 PMCID: PMC8875314 DOI: 10.3390/ijms23042208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/04/2022] Open
Abstract
Human amyloid beta peptide (Aβ) is a brain catabolite that at nanomolar concentrations can form neurotoxic oligomers (AβOs), which are known to accumulate in Alzheimer’s disease. Because a predisposition to form neurotoxins seems surprising, we have investigated whether circumstances might exist where AβO accumulation may in fact be beneficial. Our investigation focused on the embryonic chick retina, which expresses the same Aβ as humans. Using conformation-selective antibodies, immunoblots, mass spectrometry, and fluorescence microscopy, we discovered that AβOs are indeed present in the developing retina, where multiple proteoforms are expressed in a highly regulated cell-specific manner. The expression of the AβO proteoforms was selectively associated with transiently expressed phosphorylated Tau (pTau) proteoforms that, like AβOs, are linked to Alzheimer’s disease (AD). To test whether the AβOs were functional in development, embryos were cultured ex ovo and then injected intravitreally with either a beta-site APP-cleaving enzyme 1 (BACE-1) inhibitor or an AβO-selective antibody to prematurely lower the levels of AβOs. The consequence was disrupted histogenesis resulting in dysplasia resembling that seen in various retina pathologies. We suggest the hypothesis that embryonic AβOs are a new type of short-lived peptidergic hormone with a role in neural development. Such a role could help explain why a peptide that manifests deleterious gain-of-function activity when it oligomerizes in the aging brain has been evolutionarily conserved.
Collapse
Affiliation(s)
- Samuel C. Bartley
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Madison T. Proctor
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Hongjie Xia
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Evelyn Ho
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Dong S. Kang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Kristen Schuster
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Maíra A. Bicca
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Henrique S. Seckler
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA; (H.S.S.); (S.M.P.)
| | - Kirsten L. Viola
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Steven M. Patrie
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA; (H.S.S.); (S.M.P.)
| | - Neil L. Kelleher
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA;
| | - Fernando G. De Mello
- Instituto de Biofísica Carlos Chagas Filho (IBCCF), Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - William L. Klein
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Chicago, IL 60611, USA
- Correspondence: ; Tel.: +1-847-591-5510
| |
Collapse
|
46
|
Austad SN, Ballinger S, Buford TW, Carter CS, Smith DL, Darley-Usmar V, Zhang J. Targeting whole body metabolism and mitochondrial bioenergetics in the drug development for Alzheimer's disease. Acta Pharm Sin B 2022; 12:511-531. [PMID: 35256932 PMCID: PMC8897048 DOI: 10.1016/j.apsb.2021.06.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/26/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Aging is by far the most prominent risk factor for Alzheimer's disease (AD), and both aging and AD are associated with apparent metabolic alterations. As developing effective therapeutic interventions to treat AD is clearly in urgent need, the impact of modulating whole-body and intracellular metabolism in preclinical models and in human patients, on disease pathogenesis, have been explored. There is also an increasing awareness of differential risk and potential targeting strategies related to biological sex, microbiome, and circadian regulation. As a major part of intracellular metabolism, mitochondrial bioenergetics, mitochondrial quality-control mechanisms, and mitochondria-linked inflammatory responses have been considered for AD therapeutic interventions. This review summarizes and highlights these efforts.
Collapse
Key Words
- ACE2, angiotensin I converting enzyme (peptidyl-dipeptidase A) 2
- AD, Alzheimer's disease
- ADP, adenosine diphosphate
- ADRD, AD-related dementias
- Aβ, amyloid β
- CSF, cerebrospinal fluid
- Circadian regulation
- DAMPs
- DAMPs, damage-associated molecular patterns
- Diabetes
- ER, estrogen receptor
- ETC, electron transport chain
- FCCP, trifluoromethoxy carbonylcyanide phenylhydrazone
- FPR-1, formyl peptide receptor 1
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide-1
- HBP, hexoamine biosynthesis pathway
- HTRA, high temperature requirement A
- Hexokinase biosynthesis pathway
- I3A, indole-3-carboxaldehyde
- IRF-3, interferon regulatory factor 3
- LC3, microtubule associated protein light chain 3
- LPS, lipopolysaccharide
- LRR, leucine-rich repeat
- MAVS, mitochondrial anti-viral signaling
- MCI, mild cognitive impairment
- MRI, magnetic resonance imaging
- MRS, magnetic resonance spectroscopy
- Mdivi-1, mitochondrial division inhibitor 1
- Microbiome
- Mitochondrial DNA
- Mitochondrial electron transport chain
- Mitochondrial quality control
- NLRP3, leucine-rich repeat (LRR)-containing protein (NLR)-like receptor family pyrin domain containing 3
- NOD, nucleotide-binding oligomerization domain
- NeuN, neuronal nuclear protein
- PET, fluorodeoxyglucose (FDG)-positron emission tomography
- PKA, protein kinase A
- POLβ, the base-excision repair enzyme DNA polymerase β
- ROS, reactive oxygen species
- Reactive species
- SAMP8, senescence-accelerated mice
- SCFAs, short-chain fatty acids
- SIRT3, NAD-dependent deacetylase sirtuin-3
- STING, stimulator of interferon genes
- STZ, streptozotocin
- SkQ1, plastoquinonyldecyltriphenylphosphonium
- T2D, type 2 diabetes
- TCA, Tricarboxylic acid
- TLR9, toll-like receptor 9
- TMAO, trimethylamine N-oxide
- TP, tricyclic pyrone
- TRF, time-restricted feeding
- cAMP, cyclic adenosine monophosphate
- cGAS, cyclic GMP/AMP synthase
- hAPP, human amyloid precursor protein
- hPREP, human presequence protease
- i.p., intraperitoneal
- mTOR, mechanistic target of rapamycin
- mtDNA, mitochondrial DNA
- αkG, alpha-ketoglutarate
Collapse
Affiliation(s)
- Steven N. Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Thomas W. Buford
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christy S. Carter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
47
|
Heysieattalab S, Sadeghi L. Ecballium elaterium attenuates neuroinflammation in an animal model of Alzheimer's disease through modulation of nuclear factor κB pathway. AVICENNA JOURNAL OF PHYTOMEDICINE 2022; 12:89-100. [PMID: 35145898 PMCID: PMC8801219 DOI: 10.22038/ajp.2021.18881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Sustained inflammation, which could be promoted by Aβ aggregation and tau hyperphosphorylation, is a critical player in Alzheimer's disease (AD) pathogenesis. In the first phase, this study was designed to evaluate the anti-inflammatory properties of Ecballium elaterium (EE), as a Mediterranean therapeutic plant, and its effects on biochemical and behavioral signs of nucleus basalis of Meynert lesioned (NBML) rats, as an approved model of AD. In the second phase, we investigated the effect of EE on nuclear factor (NF)-κB pathway which is responsible for encoding proteins involved in the inflammatory cascade. MATERIALS AND METHODS Animals were divided randomly into four groups as following: control, NBML rats (AD), AD rats that were treated by high- and low-dose EE. Prostaglandins (PGs) levels were measured by enzyme-linked immunosorbent assay (ELISA) kits. Cyclooxygenase-2 (COX-2) and acetylcholinesterase (AChE) levels were assessed by fluorometric kit and Elman method, respectively. Behavioral signs were evaluated by Morris Water Maze (MWM) test and inflammatory proteins content was analyzed by immunoblotting method. RESULTS According to the results, treatment of NBML rats with EE fruit juice reduced PGs and cytokines more than 2-fold in comparison with AD rats through inhibition of COX-2 enzyme. Attenuation of inflammatory response in NBML rats was accompanied by reduced AChE activity (about 3-fold) and improved learning ability. Interestingly, EE reduced NF-κB expression for about 3-fold which resulted in a more than 10-fold increase in IκBα/P-IκBα ratio. CONCLUSION Our results confirmed the TNF-α/cytokines/NF-κB/COX-2 pathway involves as the main inflammatory response in NBML rats. We also provided biochemical and behavioral evidence which introduces EE as an anti-inflammatory adjuvant to improve pathophysiological signs in patients suffering from AD and related dementia.
Collapse
Affiliation(s)
| | - Leila Sadeghi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
48
|
Gabrielli M, Prada I, Joshi P, Falcicchia C, D’Arrigo G, Rutigliano G, Battocchio E, Zenatelli R, Tozzi F, Radeghieri A, Arancio O, Origlia N, Verderio C. OUP accepted manuscript. Brain 2022; 145:2849-2868. [PMID: 35254410 PMCID: PMC9420022 DOI: 10.1093/brain/awac083] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 01/17/2022] [Accepted: 02/13/2022] [Indexed: 11/27/2022] Open
Abstract
Synaptic dysfunction is an early mechanism in Alzheimer’s disease that involves progressively larger areas of the brain over time. However, how it starts and propagates is unknown. Here we show that amyloid-β released by microglia in association with large extracellular vesicles (Aβ-EVs) alters dendritic spine morphology in vitro, at the site of neuron interaction, and impairs synaptic plasticity both in vitro and in vivo in the entorhinal cortex–dentate gyrus circuitry. One hour after Aβ-EV injection into the mouse entorhinal cortex, long-term potentiation was impaired in the entorhinal cortex but not in the dentate gyrus, its main target region, while 24 h later it was also impaired in the dentate gyrus, revealing a spreading of long-term potentiation deficit between the two regions. Similar results were obtained upon injection of extracellular vesicles carrying Aβ naturally secreted by CHO7PA2 cells, while neither Aβ42 alone nor inflammatory extracellular vesicles devoid of Aβ were able to propagate long-term potentiation impairment. Using optical tweezers combined to time-lapse imaging to study Aβ-EV–neuron interaction, we show that Aβ-EVs move anterogradely at the axon surface and that their motion can be blocked through annexin-V coating. Importantly, when Aβ-EV motility was inhibited, no propagation of long-term potentiation deficit occurred along the entorhinal–hippocampal circuit, implicating large extracellular vesicle motion at the neuron surface in the spreading of long-term potentiation impairment. Our data indicate the involvement of large microglial extracellular vesicles in the rise and propagation of early synaptic dysfunction in Alzheimer’s disease and suggest a new mechanism controlling the diffusion of large extracellular vesicles and their pathogenic signals in the brain parenchyma, paving the way for novel therapeutic strategies to delay the disease.
Collapse
Affiliation(s)
| | - Ilaria Prada
- CNR Institute of Neuroscience, Vedano al Lambro, MB 20854, Italy
| | - Pooja Joshi
- CNR Institute of Neuroscience, Vedano al Lambro, MB 20854, Italy
| | | | - Giulia D’Arrigo
- CNR Institute of Neuroscience, Vedano al Lambro, MB 20854, Italy
| | - Grazia Rutigliano
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, Pisa 56127, Italy
- CNR Institute of Clinical Physiology, Pisa 56124, Italy
| | - Elisabetta Battocchio
- CNR Institute of Neuroscience, Vedano al Lambro, MB 20854, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
| | - Rossella Zenatelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Francesca Tozzi
- Bio@SNS Laboratory, Scuola Normale Superiore, Pisa, 56124, Italy
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
- Consorzio Sistemi a Grande Interfase (CSGI), Department of Chemistry, University of Florence, Sesto Fiorentino, FI 50019, Italy
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York 10032, NY, USA
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Nicola Origlia
- Correspondence may also be addressed to: Nicola Origlia CNR Institute of Neuroscience, via Moruzzi 1 Pisa, 56124, Italy E-mail:
| | - Claudia Verderio
- Correspondence to: Claudia Verderio CNR Institute of Neuroscience via Raoul Follereau 3, Vedano al Lambro MB, 20854, Italy E-mail:
| |
Collapse
|
49
|
Velasco-Bolom JL, Domínguez L. Mechanistic regulation of γ-secretase by their substrates. Phys Chem Chem Phys 2022; 24:19223-19232. [DOI: 10.1039/d2cp01714h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
γ-Secretase (GS) is a transmembrane (TM) enzyme that plays important roles in the processing of approximately 90 substrates.
Collapse
Affiliation(s)
- José-Luis Velasco-Bolom
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Laura Domínguez
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
50
|
Ginsberg SD, Joshi S, Sharma S, Guzman G, Wang T, Arancio O, Chiosis G. The penalty of stress - Epichaperomes negatively reshaping the brain in neurodegenerative disorders. J Neurochem 2021; 159:958-979. [PMID: 34657288 PMCID: PMC8688321 DOI: 10.1111/jnc.15525] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/22/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023]
Abstract
Adaptation to acute and chronic stress and/or persistent stressors is a subject of wide interest in central nervous system disorders. In this context, stress is an effector of change in organismal homeostasis and the response is generated when the brain perceives a potential threat. Herein, we discuss a nuanced and granular view whereby a wide variety of genotoxic and environmental stressors, including aging, genetic risk factors, environmental exposures, and age- and lifestyle-related changes, act as direct insults to cellular, as opposed to organismal, homeostasis. These two concepts of how stressors impact the central nervous system are not mutually exclusive. We discuss how maladaptive stressor-induced changes in protein connectivity through epichaperomes, disease-associated pathologic scaffolds composed of tightly bound chaperones, co-chaperones, and other factors, impact intracellular protein functionality altering phenotypes, that in turn disrupt and remodel brain networks ranging from intercellular to brain connectome levels. We provide an evidence-based view on how these maladaptive changes ranging from stressor to phenotype provide unique precision medicine opportunities for diagnostic and therapeutic development, especially in the context of neurodegenerative disorders including Alzheimer's disease where treatment options are currently limited.
Collapse
Affiliation(s)
- Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, USA
- Departments of Psychiatry, Neuroscience & Physiology, the NYU Neuroscience Institute, New York University Grossman School of Medicine, New York City, New York, USA
| | - Suhasini Joshi
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Gianny Guzman
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Tai Wang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Columbia University, New York City, New York, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, New York, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| |
Collapse
|