101
|
Nillert N, Boonyarat C, Welbat JU, Bunreungthong K, Puthongking P, Pannangrong W. Clausena Harmandiana root extract attenuated cognitive impairments via reducing amyloid accumulation and neuroinflammation in Aβ 1-42-induced rats. BMC Complement Med Ther 2022; 22:108. [PMID: 35439990 PMCID: PMC9019931 DOI: 10.1186/s12906-022-03591-4] [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: 12/20/2021] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) pathogenesis is associated with amyloid-β (Aβ)-induced neuroinflammation. In AD, the activation of microglia caused by Aβ accumulation is followed by the synthesis and release of pro-inflammatory cytokines, including interleukin-1β (IL-1β) and tumor necrosis factor-α (TNFα), and ultimately leads to cognitive impairments. Clausena harmandiana (CH) is a medicinal plant in the Rutaceae family and has been used in folk medicine to relieve illnesses such as stomachache and headache, and as a health tonic. Interestingly, CH root extract (CHRE) has several anti-inflammatory and other pharmacological activities, but there are no studies in AD-like animal models. OBJECTIVES This study aims to evaluate the effects of CHRE on cognitive impairments, increased Aβ1-42 protein levels, and neuroinflammation in Aβ1-42-induced rats. METHODS Forty-eight adult male Sprague-Dawley rats (250-300 g) were randomly divided into 6 groups (n = 8) of the sham control, V + Aβ, CB + Aβ CHRE125 + Aβ, CHRE250 + Aβ, and CHRE500 + Aβ. Sodium carboxymethylcellulose, Celebrex (10 mg/kg BW) and CHRE (125, 250, and 500 mg/kg BW) were given orally or without any treatment for 35 days. On day 21, aggregated Aβ1-42 at a concentration of 1 μg/μl were injected into both lateral ventricles (1 μl/side) of all treated rats, while sterilized normal saline were injected to untreated rats. Ten days later, the novel object recognition test was performed to assess their recognition memory. At the end of the test period, an overdose of thiopental sodium (120 mg/kg BW) and transcardial perfusion with 0.9% normal saline solution were used to euthanize all rats. Then Aβ1-42 protein levels and the expression of inflammatory markers (CD11b-positive microglia, IL-1β, and TNFα) were investigated in the cerebral cortex and hippocampus. RESULTS Pretreatment with CHRE at all doses could attenuate short- and long-term impairments in recognition memory. Additionally, CHRE also inhibited the increase of Aβ1-42 protein levels and the expression of inflammatory markers in both brain regions as well as receiving Celebrex. CONCLUSIONS This suggests that preventive treatment of CHRE might be a potential therapy against cognitive impairments via reducing Aβ1-42 protein levels and neuroinflammation caused by Aβ1-42.
Collapse
Affiliation(s)
- Nutchareeporn Nillert
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Chantana Boonyarat
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Jariya Umka Welbat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Komsun Bunreungthong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Ploenthip Puthongking
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Wanassanun Pannangrong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
102
|
Nelson AR. Peripheral Pathways to Neurovascular Unit Dysfunction, Cognitive Impairment, and Alzheimer’s Disease. Front Aging Neurosci 2022; 14:858429. [PMID: 35517047 PMCID: PMC9062225 DOI: 10.3389/fnagi.2022.858429] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. It was first described more than a century ago, and scientists are acquiring new data and learning novel information about the disease every day. Although there are nuances and details continuously being unraveled, many key players were identified in the early 1900’s by Dr. Oskar Fischer and Dr. Alois Alzheimer, including amyloid-beta (Aβ), tau, vascular abnormalities, gliosis, and a possible role of infections. More recently, there has been growing interest in and appreciation for neurovascular unit dysfunction that occurs early in mild cognitive impairment (MCI) before and independent of Aβ and tau brain accumulation. In the last decade, evidence that Aβ and tau oligomers are antimicrobial peptides generated in response to infection has expanded our knowledge and challenged preconceived notions. The concept that pathogenic germs cause infections generating an innate immune response (e.g., Aβ and tau produced by peripheral organs) that is associated with incident dementia is worthwhile considering in the context of sporadic AD with an unknown root cause. Therefore, the peripheral amyloid hypothesis to cognitive impairment and AD is proposed and remains to be vetted by future research. Meanwhile, humans remain complex variable organisms with individual risk factors that define their immune status, neurovascular function, and neuronal plasticity. In this focused review, the idea that infections and organ dysfunction contribute to Alzheimer’s disease, through the generation of peripheral amyloids and/or neurovascular unit dysfunction will be explored and discussed. Ultimately, many questions remain to be answered and critical areas of future exploration are highlighted.
Collapse
|
103
|
Johansen M, Joensen S, Restorff M, Stórá T, Christy D, Gustavsson EK, Bian J, Guo Y, Farrer MJ, Petersen MS. Polygenic risk of Alzheimer's disease in the Faroe Islands. Eur J Neurol 2022; 29:2192-2200. [PMID: 35384166 DOI: 10.1111/ene.15351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/01/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The Faroe Islands are a geographically isolated population in the North Atlantic with a similar prevalence Alzheimer's disease (AD) and all cause dementia as other European nations. However, the genetic risk underlying Alzheimer's disease and other dementia susceptibility has yet to be elucidated. METHODS Forty-nine single nucleotide polymorphisms (SNPs) were genotyped in 174 patients with AD and other dementias and 159 healthy controls. Single variant and polygenic risk score (PRS) associations, with/without APOE variability, were assessed by logistic regression. Performance was examined using receiver operating characteristics 'area under the curve' analysis (ROC AUC). RESULTS APOE rs429358 was associated with AD in the Faroese cohort after correction for multiple testing (OR=6.32, CI[3.98-10.05], p=6.31e-15 ), with suggestive evidence for three other variants: NECTIN2 rs41289512 (OR 2.05, CI[1.20-3.51], p=0.01), HLA-DRB1 rs6931277 (OR 0.67, CI[0.48-0.94], p=0.02), and APOE rs7412 [ε2] (OR 0.28, CI[0.11-0.73], p=0.01). PRS were associated with AD with or without the inclusion of APOE (PRS+APOE OR=4.5. CI[2.90-5.85, p=4.56e-15 and PRS-APOE OR=1.53, CI[1.21-1.98], p=6.82e-4 ). AD ROC AUC analyses demonstrated a PRS+APOE AUC=80.3% and PRS-APOE AUC=63.4%. However, PRS+APOE was also significantly associated with all cause dementia (OR=3.39, CI[2.51-4.71], p= 2.50e-14 ) with an AUC=76.9%, i.e. all cause dementia did show similar results albeit less significant. DISCUSSION In the Faroe Islands, SNP analyses highlighted APOE and immunogenomic variability in AD and dementia risk. PRS+APOE , based on 25 SNPs/loci, had excellent sensitivity and specificity for Alzheimer's disease with AUC of 80.3%. High PRS were also associated with an earlier onset of late-onset AD.
Collapse
Affiliation(s)
- Malan Johansen
- Center of Health Science, University of the Faroe Islands, Vestarabryggja 15, Tórshavn, Faroe Islands.,Department of Occupational Medicine and Public Health, The Faroese Hospital System, Sigmundargøta 5, Tórshavn, Faroe Islands
| | - Sofus Joensen
- The Dementia Clinic, Psychiatric Center, National Hospital of the Faroe Islands, J. C. Svabos gøta 41-49, Tórshavn, Faroe Islands
| | - Marjun Restorff
- The Dementia Clinic, Psychiatric Center, National Hospital of the Faroe Islands, J. C. Svabos gøta 41-49, Tórshavn, Faroe Islands
| | - Tórmóður Stórá
- The Dementia Clinic, Psychiatric Center, National Hospital of the Faroe Islands, J. C. Svabos gøta 41-49, Tórshavn, Faroe Islands
| | - Darren Christy
- Centre for Applied Neurogenetics, University of British Columbia, C201-4500 Oak Street, Vancouver, BC, Canada
| | - Emil K Gustavsson
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, UK.,Department of Neurodegenerative Disease, Institute of Neurology, University College London, Queen Square, London, UK
| | - Jiang Bian
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, 2004 Mowry RD, Gainesville, FL, USA
| | - Yi Guo
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, 2004 Mowry RD, Gainesville, FL, USA
| | - Matthew J Farrer
- Centre for Applied Neurogenetics, University of British Columbia, C201-4500 Oak Street, Vancouver, BC, Canada.,McKnight Brain Institute, Department of Neurology, University of Florida, 1149 Newell Drive, Gainesville, FL, USA
| | - Maria Skaalum Petersen
- Center of Health Science, University of the Faroe Islands, Vestarabryggja 15, Tórshavn, Faroe Islands.,Department of Occupational Medicine and Public Health, The Faroese Hospital System, Sigmundargøta 5, Tórshavn, Faroe Islands
| |
Collapse
|
104
|
Hur JY. γ-Secretase in Alzheimer's disease. Exp Mol Med 2022; 54:433-446. [PMID: 35396575 PMCID: PMC9076685 DOI: 10.1038/s12276-022-00754-8] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/05/2022] [Accepted: 01/20/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is caused by synaptic and neuronal loss in the brain. One of the characteristic hallmarks of AD is senile plaques containing amyloid β-peptide (Aβ). Aβ is produced from amyloid precursor protein (APP) by sequential proteolytic cleavages by β-secretase and γ-secretase, and the polymerization of Aβ into amyloid plaques is thought to be a key pathogenic event in AD. Since γ-secretase mediates the final cleavage that liberates Aβ, γ-secretase has been widely studied as a potential drug target for the treatment of AD. γ-Secretase is a transmembrane protein complex containing presenilin, nicastrin, Aph-1, and Pen-2, which are sufficient for γ-secretase activity. γ-Secretase cleaves >140 substrates, including APP and Notch. Previously, γ-secretase inhibitors (GSIs) were shown to cause side effects in clinical trials due to the inhibition of Notch signaling. Therefore, more specific regulation or modulation of γ-secretase is needed. In recent years, γ-secretase modulators (GSMs) have been developed. To modulate γ-secretase and to understand its complex biology, finding the binding sites of GSIs and GSMs on γ-secretase as well as identifying transiently binding γ-secretase modulatory proteins have been of great interest. In this review, decades of findings on γ-secretase in AD are discussed.
Collapse
Affiliation(s)
- Ji-Yeun Hur
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
105
|
Filipello F, Goldsbury C, You SF, Locca A, Karch CM, Piccio L. Soluble TREM2: Innocent bystander or active player in neurological diseases? Neurobiol Dis 2022; 165:105630. [PMID: 35041990 PMCID: PMC10108835 DOI: 10.1016/j.nbd.2022.105630] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/14/2022] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is an innate immune receptor expressed by macrophages and microglia in the central nervous system (CNS). TREM2 has attracted a lot of interest in the past decade for its critical role in modulating microglia functions under homeostatic conditions and in neurodegenerative diseases. Genetic variation in TREM2 is sufficient to cause Nasu-Hakola disease, a rare pre-senile dementia with bone cysts, and to increase risk for Alzheimer's disease, frontotemporal dementia, and other neurodegenerative disorders. Beyond the role played by TREM2 genetic variants in these diseases, TREM2 engagement is a key step in microglia activation in response to different types of tissue injury (e.g. β-Amyloid deposition, demyelination, apoptotic cell death) leading to enhanced microglia metabolism, phagocytosis, proliferation and survival. TREM2 also exists as a soluble form (sTREM2), generated from receptor shedding or alternative splicing, which is detectable in plasma and cerebrospinal fluid (CSF). Genetic variation, physiological conditions and disease status impact CSF sTREM2 levels. Clinical and preclinical studies suggest that targeting and/or monitoring sTREM2 could have clinical and therapeutic implications. Despite the critical role of sTREM2 in neurologic disease, its function remains poorly understood. Here, we review the current literature on sTREM2 regarding its origin, genetic variation, and possible functions as a biomarker in neurological disorders and as a potential active player in CNS diseases and target for therapies.
Collapse
Affiliation(s)
- Fabia Filipello
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Claire Goldsbury
- Brain and Mind Centre and Charles Perkins Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia
| | - Shih Feng You
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Alberto Locca
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laura Piccio
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Brain and Mind Centre and Charles Perkins Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
106
|
Shen L, Zhang H, Lin J, Gao Y, Chen M, Khan NU, Tang X, Hong Q, Feng C, Zhao Y, Cao X. A Combined Proteomics and Metabolomics Profiling to Investigate the Genetic Heterogeneity of Autistic Children. Mol Neurobiol 2022; 59:3529-3545. [PMID: 35348996 DOI: 10.1007/s12035-022-02801-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/16/2022] [Indexed: 11/30/2022]
Abstract
Autism spectrum disorder (ASD) has become one of the most common neurological developmental disorders in children. However, the study of ASD diagnostic markers faces significant challenges due to the existence of heterogeneity. In this study, genetic testing was performed on children who were clinically diagnosed with ASD. Children with ASD susceptibility genes and healthy controls were studied. The proteomics of plasma and peripheral blood mononuclear cells (PBMCs) as well as plasma metabolomics were carried out. The results showed that although there was genetic heterogeneity in children with ASD, the differentially expressed proteins (DEPs) in plasma, peripheral blood mononuclear cells, and differential metabolites in plasma could still effectively distinguish autistic children from controls. The mechanism associated with them focuses on several common and previously reported mechanisms of ASD. The biomarkers for ASD diagnosis could be found by taking differentially expressed proteins and differential metabolites into consideration. Integrating omics data, glycerophospholipid metabolism and N-glycan biosynthesis might play a critical role in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.,Brain Disease and Big Data Research Institute, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Jing Lin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, People's Republic of China
| | - Yan Gao
- Maternal and Child Health Hospital of Baoan, Shenzhen, 518100, People's Republic of China
| | - Margy Chen
- Department of Psychology, Emory University, Atlanta, GA, 30322, USA
| | - Naseer Ullah Khan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Qi Hong
- Maternal and Child Health Hospital of Baoan, Shenzhen, 518100, People's Republic of China
| | - Chengyun Feng
- Maternal and Child Health Hospital of Baoan, Shenzhen, 518100, People's Republic of China
| | - Yuxi Zhao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.
| |
Collapse
|
107
|
Srinivasan G, Brafman DA. The Emergence of Model Systems to Investigate the Link Between Traumatic Brain Injury and Alzheimer's Disease. Front Aging Neurosci 2022; 13:813544. [PMID: 35211003 PMCID: PMC8862182 DOI: 10.3389/fnagi.2021.813544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous epidemiological studies have demonstrated that individuals who have sustained a traumatic brain injury (TBI) have an elevated risk for developing Alzheimer's disease and Alzheimer's-related dementias (AD/ADRD). Despite these connections, the underlying mechanisms by which TBI induces AD-related pathology, neuronal dysfunction, and cognitive decline have yet to be elucidated. In this review, we will discuss the various in vivo and in vitro models that are being employed to provide more definite mechanistic relationships between TBI-induced mechanical injury and AD-related phenotypes. In particular, we will highlight the strengths and weaknesses of each of these model systems as it relates to advancing the understanding of the mechanisms that lead to TBI-induced AD onset and progression as well as providing platforms to evaluate potential therapies. Finally, we will discuss how emerging methods including the use of human induced pluripotent stem cell (hiPSC)-derived cultures and genome engineering technologies can be employed to generate better models of TBI-induced AD.
Collapse
Affiliation(s)
| | - David A. Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
108
|
TREM2 Ameliorates Lipopolysaccharide-Induced Oxidative Stress Response and Neuroinflammation by Promoting Sirtuin3 in BV2 Cells. Neurotox Res 2022; 40:56-65. [PMID: 35013907 DOI: 10.1007/s12640-021-00459-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 10/19/2022]
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) plays a crucial role in modulating microglial-mediated neuroinflammation. The NAD-dependent deacetylase protein Sirtuin 3 (SIRT3) regulates mitochondrial oxidative stress response and neuroinflammation. TREM2 deficiency impairs the denovo synthesis pathway of NAD+. Therefore, the aim of this study was to investigate the potential role of TREM2 and SIRT3 in LPS-induced oxidative stress and neuroinflammation in BV2 cells. Lentivirus vector-mediated TREM2 overexpression (TREM2-OE) and corresponding negative control vector (TREM2-NC) were synthesized. BV2 cells were treated with LPS and/or TREM2-OE. 3-TYP, a selective SIRT3 inhibitor, was applied to determine the role of SIRT3 in the anti-oxidant and anti-inflammatory effects of TREM2. TREM2, SIRT3, NLRP3 inflammasome, caspase-1, postsynaptic density-95 (PSD-95), and brain derived neurotrophic factor (BDNF) were measured by Western blot analysis. Superoxide dismutase (SOD) was tested by SOD Assay Kit. Reactive oxygen species (ROS) expression was examined by immunofluorescence. Interleukin 1β (IL-1β) was determined by ELISA. Contents of NAD+ and NADH were detected by WST-8 method. LPS (1ug/ml for 24 h) significantly decreased TREM2 expression at both RNA and protein levels (p < 0.01 and p < 0.05, respectively). Lower levels of SIRT3 protein and NAD+ were also detected following LPS stimulation (p < 0.05 and p < 0.05, respectively). LPS significantly enhanced ROS, NLRP3, caspase-1, and IL-1β expression (p < 0.01, p < 0.05, p < 0.05, and p < 0.01, respectively). PSD-95 and BDNF expression were decreased triggered by LPS (p < 0.05 and p < 0.05, respectively). TREM2 overexpression enhanced NAD+ and SIRT3 protein expression following LPS challenge in BV2 cells (p < 0.01 and p < 0.05, respectively). TREM2 alleviated LPS-induced oxidative stress and neuroinflammation (p < 0.01 and p < 0.05, respectively). Similarly, TREM2 overexpression upregulated PSD-95 and BDNF expression (p < 0.05 and p < 0.05, respectively). The anti-oxidant and anti-inflammatory effects of TREM2 were partially abrogated by SIRT3 antagonist 3-TYP (p < 0.05 and p < 0.05, respectively). Similarly, selective SIRT3 inhibition also partially abrogated TREM2-induced BDNF protein upregulation (p < 0.05) but failed to influence PSD-95 protein expression following LPS stimulation. LPS induces oxidative stress and neuroinflammation in BV2 cells, which may be mediated in part by the downregulation of TREM2 and SIRT3. TREM2 overexpression ameliorates LPS-induced oxidative stress and neuroinflammation through enhancing SIRT3 function via NAD+.
Collapse
|
109
|
Ferrer I. Alzheimer's disease is an inherent, natural part of human brain aging: an integrated perspective. FREE NEUROPATHOLOGY 2022; 3:17. [PMID: 37284149 PMCID: PMC10209894 DOI: 10.17879/freeneuropathology-2022-3806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/21/2022] [Indexed: 06/08/2023]
Abstract
Alzheimer disease is one of the most challenging demons in our society due to its very high prevalence and its clinical manifestations which cause deterioration of cognition, intelligence, and emotions - the very capacities that distinguish Homo sapiens from other animal species. Besides the personal, social, and economical costs, late stages of AD are vivid experiences for the family, relatives, friends, and general observers of the progressive ruin of an individual who turns into a being with lower mental and physical capacities than less evolved species. A human brain with healthy cognition, conscience, and emotions can succeed in dealing with most difficulties that life may pose. Without these capacities, the same person probably cannot. Due, in part, to this emotional impact, the absorbing study of AD has generated, over the years, a fascinating and complex story of theories, hypotheses, controversies, fashion swings, and passionate clashes, together with tremendous efforts and achievements geared to improve understanding of the pathogenesis and treatment of the disorder. Familal AD is rare and linked to altered genetic information associated with three genes. Sporadic AD (sAD) is much more common and multifactorial. A major point of clinical discussion has been, and still is, establishing the differences between brain aging and sAD. This is not a trivial question, as the neuropathological and molecular characteristics of normal brain aging and the first appearance of early stages of sAD-related pathology are not easily distinguishable in most individuals. Another important point is confidence in assigning responsibility for the beginning of sAD to a few triggering molecules, without considering the wide number of alterations that converge in the pathogenesis of aging and sAD. Genetic risk factors covering multiple molecular signals are increasing in number. In the same line, molecular pathways are altered at early stages of sAD pathology, currently grouped under the aegis of normal brain aging, only to increase massively at advanced stages of the process. Sporadic AD is here considered an inherent, natural part of human brain aging, which is prevalent in all humans, and variably present or not in a few individuals in other species. The progression of the process has devastating effects in a relatively low percentage of human beings eventually evolving to dementia. The continuum of brain aging and sAD implies the search for a different approach in the study of human brain aging at the first stages of the biological process, and advances in the use of new technologies aimed at slowing down the molecular defects underlying human brain aging and sAD at the outset, and transfering information and tasks to AI and coordinated devices.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Emeritus Researcher of the Bellvitge Institute of Biomedical Research (IDIBELL); Biomedical Research Network of Neurodegenerative Diseases (CIBERNED); Institute of Neurosciences, University of Barcelona; Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
110
|
Targeting PKC in microglia to promote remyelination and repair in the CNS. Curr Opin Pharmacol 2021; 62:103-108. [PMID: 34965482 DOI: 10.1016/j.coph.2021.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/19/2021] [Indexed: 01/28/2023]
Abstract
Microglia and CNS-infiltrating macrophages play significant roles in the pathogenesis of neuroinflammatory and neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Prolonged and dysregulated inflammatory responses by these innate immune cells can have deleterious effects on the surrounding CNS microenvironment, which can worsen neurodegeneration and demyelination. However, although chronic activation of pro-inflammatory microglia is maladaptive, other functional microglial subtypes play beneficial roles during CNS repair and regeneration. Therefore, there is a tremendous interest in understanding the underlying mechanism of the activation of these reparative/regenerative microglia. In this review, we focus on the potential role of PKC, a downstream signaling molecule of TREM2 and PLCγ2, and PKC modulators in promoting the activation of reparative/regenerative microglial subtypes as a novel therapy for neuroinflammatory and neurodegenerative diseases.
Collapse
|
111
|
Hermans SJ, Nero TL, Morton CJ, Gooi JH, Crespi GAN, Hancock NC, Gao C, Ishii K, Markulić J, Parker MW. Structural biology of cell surface receptors implicated in Alzheimer’s disease. Biophys Rev 2021; 14:233-255. [DOI: 10.1007/s12551-021-00903-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023] Open
|
112
|
IKK2/NF-κB Activation in Astrocytes Reduces amyloid β Deposition: A Process Associated with Specific Microglia Polarization. Cells 2021; 10:cells10102669. [PMID: 34685649 PMCID: PMC8534251 DOI: 10.3390/cells10102669] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that is accompanied by pronounced neuroinflammatory responses mainly characterized by marked microgliosis and astrogliosis. However, it remains open as to how different aspects of astrocytic and microglial activation affect disease progression. Previously, we found that microglia expansion in the spinal cord, initiated by IKK2/NF-κB activation in astrocytes, exhibits stage-dependent beneficial effects on the progression of amyotrophic lateral sclerosis. Here, we investigated the impact of NF-κB-initiated neuroinflammation on AD pathogenesis using the APP23 mouse model of AD in combination with conditional activation of IKK2/NF-κB signaling in astrocytes. We show that NF-κB activation in astrocytes triggers a distinct neuroinflammatory response characterized by striking astrogliosis as well as prominent microglial reactivity. Immunohistochemistry and Congo red staining revealed an overall reduction in the size and number of amyloid plaques in the cerebral cortex and hippocampus. Interestingly, isolated primary astrocytes and microglia cells exhibit specific marker gene profiles which, in the case of microglia, point to an enhanced plaque clearance capacity. In contrast, direct IKK2/NF-κB activation in microglia results in a pro-inflammatory polarization program. Our findings suggest that IKK2/NF-κB signaling in astrocytes may activate paracrine mechanisms acting on microglia function but also on APP processing in neurons.
Collapse
|
113
|
Femminella GD, Pagano G, Liccardo D, Cannavo A. Editorial: Molecular Mechanisms Involved in Heart Failure, Parkinson's, and Alzheimer's Diseases. Front Mol Biosci 2021; 8:754987. [PMID: 34568433 PMCID: PMC8455875 DOI: 10.3389/fmolb.2021.754987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 08/25/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Gennaro Pagano
- Roche Pharmaceutical Research and Early Development, NRD Neuroscience and Rare Diseases, Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Daniela Liccardo
- Center for Translational Medical Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Alessandro Cannavo
- Department of Translational Medical Science, Federico II University of Naples, Naples, Italy
| |
Collapse
|
114
|
Salminen A, Kaarniranta K, Kauppinen A. Hypoxia/ischemia impairs CD33 (Siglec-3)/TREM2 signaling: Potential role in Alzheimer's pathogenesis. Neurochem Int 2021; 150:105186. [PMID: 34530055 DOI: 10.1016/j.neuint.2021.105186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/22/2022]
Abstract
Recent genetic and molecular studies have indicated that the innate immune system, especially microglia, have a crucial role in the accumulation of β-amyloid plaques in Alzheimer's disease (AD). In particular, the CD33 receptor, also called Siglec-3, inhibits the TREM2 receptor-induced phagocytic activity of microglia. CD33 receptors recognize the α2,3 and α2,6-linked sialic groups in tissue glycocalyx, especially sialylated gangliosides in human brain. The CD33 receptor triggers cell-type specific responses, e.g., in microglia, CD33 inhibits phagocytosis, whereas in natural killer cells, it inhibits the cytotoxic activity of the NKG2D receptor. Nonetheless, the regulation of the activity of CD33 receptor needs to be clarified. For example, it seems that hypoxia/ischemia, a potential cause of AD pathology, increases the expression of CD33 and its downstream target SHP-1, a tyrosine phosphatase which suppresses the phagocytosis driven by TREM2. Moreover, hypoxia/ischemia increases the deposition of sialylated gangliosides, e.g., GM1, GM2, GM3, and GD1, which are ligands for inhibitory CD33/Siglec-3 receptors. In addition, β-amyloid peptides bind to the sialylated gangliosides in raft-like clusters and subsequently these gangliosides act as seeds for the formation of β-amyloid plaques in AD pathology. It is known that senile plaques contain sialylated GM1, GM2, and GM3 gangliosides, i.e., the same species induced by hypoxia/ischemia treatment. Sialylated gangliosides in plaques might stimulate the CD33/Siglec-3 receptors of microglia and thus impede TREM2-driven phagocytosis. We propose that hypoxia/ischemia, e.g., via the accumulation of sialylated gangliosides, prevents the phagocytosis of β-amyloid deposits by inhibiting CD33/TREM2 signaling.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, FI-70029, KYS, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| |
Collapse
|
115
|
Abstract
Several genes in innate immunity have been implicated in Alzheimer's disease (AD). However, the effect of innate immunity on amyloid β (Aβ) production, which makes amyloid plaques in AD brains, was previously not known. Recently, the antiviral protein interferon-induced transmembrane protein 3 (IFITM3) has been identified as a novel γ-secretase modulatory protein for Aβ production. In this review, the mechanisms of how innate immunity modulates Aβ production via IFITM3-γ-secretase complexes and contributes to AD pathogenesis are discussed.
Collapse
Affiliation(s)
- Ji-Yeun Hur
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
116
|
Regnier-Golanov AS, Dündar F, Zumbo P, Betel D, Hernandez MS, Peterson LE, Lo EH, Golanov EV, Britz GW. Hippocampal Transcriptome Changes After Subarachnoid Hemorrhage in Mice. Front Neurol 2021; 12:691631. [PMID: 34354664 PMCID: PMC8329593 DOI: 10.3389/fneur.2021.691631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/11/2021] [Indexed: 11/13/2022] Open
Abstract
After subarachnoid hemorrhage (SAH), up to 95% of surviving patients suffer from post-SAH syndrome, which includes cognitive deficits with impaired memory, executive functions, and emotional disturbances. Although these long-term cognitive deficits are thought to result from damage to temporomesial-hippocampal areas, the underlying mechanisms remain unknown. To fill this gap in knowledge, we performed a systematic RNA sequencing screen of the hippocampus in a mouse model of SAH. SAH was induced by perforation of the circle of Willis in mice. Four days later, hippocampal RNA was obtained from SAH and control (sham perforation) mice. Next-generation RNA sequencing was used to determine differentially expressed genes in the whole bilateral hippocampi remote from the SAH bleeding site. Functional analyses and clustering tools were used to define molecular pathways. Differential gene expression analysis detected 642 upregulated and 398 downregulated genes (false discovery rate <0.10) in SAH compared to Control group. Functional analyses using IPA suite, Gene Ontology terms, REACTOME pathways, and MsigDB Hallmark gene set collections revealed suppression of oligodendrocytes/myelin related genes, and overexpression of genes related to complement system along with genes associated with innate and adaptive immunity, and extracellular matrix reorganization. Interferon regulatory factors, TGF-β1, and BMP were identified as major orchestrating elements in the hippocampal tissue response. The MEME-Suite identified binding motifs of Krüppel-like factors, zinc finger transcription factors, and interferon regulatory factors as overrepresented DNA promoter motifs. This study provides the first systematic gene and pathway database of the hippocampal response after SAH. Our findings suggest that damage of the entorhinal cortex by subarachnoid blood may remotely trigger specific hippocampal responses, which include suppression of oligodendrocyte function. Identification of these novel pathways may allow for development of new therapeutic approaches for post-SAH cognitive deficits.
Collapse
Affiliation(s)
- Angelique S. Regnier-Golanov
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Friederike Dündar
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
| | - Paul Zumbo
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States
| | - Magda S. Hernandez
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Leif E. Peterson
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX, United States
| | - Eng H. Lo
- Laboratory of Neuroprotection Research, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Eugene V. Golanov
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Gavin W. Britz
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
117
|
Shaw BC, Katsumata Y, Simpson JF, Fardo DW, Estus S. Analysis of Genetic Variants Associated with Levels of Immune Modulating Proteins for Impact on Alzheimer's Disease Risk Reveal a Potential Role for SIGLEC14. Genes (Basel) 2021; 12:genes12071008. [PMID: 34208838 PMCID: PMC8303736 DOI: 10.3390/genes12071008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 01/22/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified immune-related genes as risk factors for Alzheimer’s disease (AD), including TREM2 and CD33, frequently passing a stringent false-discovery rate. These genes either encode or signal through immunomodulatory tyrosine-phosphorylated inhibitory motifs (ITIMs) or activation motifs (ITAMs) and govern processes critical to AD pathology, such as inflammation and amyloid phagocytosis. To investigate whether additional ITIM and ITAM-containing family members may contribute to AD risk and be overlooked due to the stringent multiple testing in GWAS, we combined protein quantitative trait loci (pQTL) data from a recent plasma proteomics study with AD associations in a recent GWAS. We found that pQTLs for genes encoding ITIM/ITAM family members were more frequently associated with AD than those for non-ITIM/ITAM genes. Further testing of one family member, SIGLEC14 which encodes an ITAM, uncovered substantial copy number variations, identified an SNP as a proxy for gene deletion, and found that gene expression correlates significantly with gene deletion. We also found that SIGLEC14 deletion increases the expression of SIGLEC5, an ITIM. We conclude that many genes in this ITIM/ITAM family likely impact AD risk, and that complex genetics including copy number variation, opposing function of encoded proteins, and coupled gene expression may mask these AD risk associations at the genome-wide level.
Collapse
Affiliation(s)
- Benjamin C. Shaw
- Department of Physiology, University of Kentucky, Lexington, KY 40506, USA; (B.C.S.); (J.F.S.)
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA;
| | - Yuriko Katsumata
- Department of Biostatistics, University of Kentucky, Lexington, KY 40506, USA;
| | - James F. Simpson
- Department of Physiology, University of Kentucky, Lexington, KY 40506, USA; (B.C.S.); (J.F.S.)
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA;
| | - David W. Fardo
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA;
- Department of Biostatistics, University of Kentucky, Lexington, KY 40506, USA;
| | - Steven Estus
- Department of Physiology, University of Kentucky, Lexington, KY 40506, USA; (B.C.S.); (J.F.S.)
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA;
- Correspondence: ; Tel.: +1-859-218-2388
| |
Collapse
|
118
|
Kurt Jellinger 90: his contribution to neuroimmunology. J Neural Transm (Vienna) 2021; 128:1545-1550. [PMID: 34110492 PMCID: PMC8528743 DOI: 10.1007/s00702-021-02358-4] [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: 04/22/2021] [Accepted: 05/27/2021] [Indexed: 11/05/2022]
Abstract
This review honors Kurt Jellinger on his 90th birthday as one of the most outstanding neuropathologists, who has contributed immensely to neuroscience due to his vast experience and collection of excellently documented autopsy cases. Two of his many insightful reports are highlighted here. One report focuses on the pathogenesis of inflammatory demyelinating diseases and investigates the neuropathology in autopsy tissue of a patient, who developed an MS-like disease after repeated treatment with lyophilized bovine brain cells in 1958. More than 60 years later, after reinvestigation of the historic samples in 2015 and subsequent mRNA isolation, next generation sequencing and reconstruction of the antibody, we succeeded in identifying myelin oligodendrocyte glycoprotein (MOG) as the target antigen and provided the missing element between the pathomechanisms in classic EAE animal models and transfer of this disease process into humans. A second significant example of Kurt Jellinger’s contribution to neuroscience was a report on the role of MS in the development of Alzheimer's disease (AD), which found that AD pathology is present to the same extent in demyelinated and non-demyelinated cortical areas in MS and the incidence for AD pathology in elderly MS patients is comparable to the normal-aging population. This indicates that chronic inflammation in the MS cortex alone does not significantly predispose to the development of cortical AD pathology. These and other findings were only possible due to the broad collection of extremely well-defined material established by Kurt Jellinger, which ultimately continues to contribute to translational neuroscience, even decades later.
Collapse
|
119
|
Editorial: Understanding factors that, beyond plaques and tangles, contribute to the heterogeneity of Alzheimer disease and implications for the development of biomarkers and design of interventions. Curr Opin Neurol 2021; 34:226-227. [PMID: 33664207 DOI: 10.1097/wco.0000000000000915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
120
|
Frautschy SA. CR3 ruffles FcγR's claim over phagocytic cups. J Biol Chem 2021; 296:100801. [PMID: 34019878 PMCID: PMC8191298 DOI: 10.1016/j.jbc.2021.100801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Phagocytosis plays diverse roles in biology, but our understanding of the purpose, interplay, and cell signaling mechanisms associated with different modes of phagocytosis is limited, without being able to capture and visualize each step in this rapid process from the beginning to end. A new study by Walbaum et al. uses stunning time-lapse 3D imaging of the engulfment of erythrocytes by macrophages via sinking, ruffling, and cup formation, unequivocally confirming a visionary 44-year-old theory derived from still electron microscopy photos that phagocytosis mediated by complement receptor CR3 occurs via a sinking mechanism and antibody-mediated phagocytosis occurs via phagocytic cup formation. The article also challenges the dogma, showing that phagocytic cup formation is not unique to antibody receptor phagocytosis, rather CR3 plays a complex role in different modes of phagocytosis. For example, inhibition of antibody-mediated phagocytosis leads to a compensatory upregulation of CR3-mediated sinking phagocytosis. These findings animate, in vivid colors, processes previously only captured as stills, exposing interactions between different phagocytic mechanisms and altering our basic understanding of this important process.
Collapse
Affiliation(s)
- S A Frautschy
- Geriatric Research Education and Clinical Center, Veterans Greater Los Angeles HealthCare System, Los Angeles, California, USA; Departments of Neurology and Medicine, University of California, Los Angeles, Los Angeles, California, USA.
| |
Collapse
|