1
|
Schaible P, Henschel J, Erny D. How the gut microbiota impacts neurodegenerative diseases by modulating CNS immune cells. J Neuroinflammation 2025; 22:60. [PMID: 40033338 PMCID: PMC11877772 DOI: 10.1186/s12974-025-03371-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide. Amyloid-β (Aβ) accumulation and neurofibrillary tangles are two key histological features resulting in progressive and irreversible neuronal loss and cognitive decline. The macrophages of the central nervous system (CNS) belong to the innate immune system and comprise parenchymal microglia and CNS-associated macrophages (CAMs) at the CNS interfaces (leptomeninges, perivascular space and choroid plexus). Microglia and CAMs have received attention as they may play a key role in disease onset and progression e. g., by clearing amyloid beta (Aβ) through phagocytosis. Genome-wide association studies (GWAS) have revealed that human microglia and CAMs express numerous risk genes for AD, further highlighting their potentially critical role in AD pathogenesis. Microglia and CAMs are tightly controlled by environmental factors, such as the host microbiota. Notably, it was further reported that the composition of the gut microbiota differed between AD patients and healthy individuals. Hence, emerging studies have analyzed the impact of gut bacteria in different preclinical mouse models for AD as well as in clinical studies, potentially enabling promising new therapeutic options.
Collapse
Affiliation(s)
- Philipp Schaible
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Julia Henschel
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Daniel Erny
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany.
| |
Collapse
|
2
|
Liu H, You M, Yang H, Wu X, Zhang S, Huang S, Gao H, Xie L. Exploring the molecular characterization of PANoptosis-related genes with features of immune dysregulation in Alzheimer's disease based on bulk and single-nuclei RNA sequencing. Metab Brain Dis 2025; 40:109. [PMID: 39841296 DOI: 10.1007/s11011-025-01540-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 01/15/2025] [Indexed: 01/23/2025]
Abstract
The immune system has emerged as a major factor in the pathogenesis of Alzheimer's disease (AD). PANoptosis is a newly defined programmed cell death mechanism related to many inflammatory diseases. This study aimed to identify the differentially expressed (DE) PANoptosis-related genes with characteristics of immune dysregulation (PRGIDs) in AD using bioinformatics analysis of bulk RNA-seq and single-nuclei RNA sequencing (snRNA-seq) data. To improve the robustness of gene selection, we integrated 3 microarray and 6 snRNA-seq datasets from the Gene Expression Omnibus (GEO), which allowed us to not only examine overall gene expression patterns but also assess the cellular specificity of gene expression at the single-cell level. This approach helped to identify cell-type-specific gene alterations that may be masked in bulk RNA-seq analyses. Relevant PANoptosis, immune dysregulation, and AD-related genes were obtained from the Genecards database. The AlzData database was also used in this study. Expression validation, the least absolute shrinkage and selection operator (LASSO) regression model, and CytoHubba algorithms were applied for key DE-PRGIDs selection. LASSO, Logistic, and Cox regressions were used to construct prognostic models. The receiver operating characteristic (ROC) curve and correlation analyses were conducted on key DE-PRGIDs. The Seurat package in R software was employed for performing snRNA-seq data processing. Uniform manifold approximation and projection (UMAP) was utilized for cell type annotation and PRGID cell visualization. The violin plot was applied for displaying expression levels of PRGIDs. High-dimensional consensus weighted gene co-expression network analysis (hdWGCNA) was conducted on microglia to identify gene modules and hub genes. Venn diagram analysis identified 250 PRGIDs and 39 DE-PRGIDs. NFKBIA was identified as the key gene. Prognostic models based on the expression level of NFKBIA were obtained. ROC curve analysis revealed its area under the curve (AUC) value: 0.661 in training set and 0.836 in validation set. The heatmap displayed the result of correlation analysis. SnRNA-seq data analysis identified 7 cell types. The UMAP and violin plots revealed highly expressed PRGIDs in microglia with remarkable differences between healthy controls and AD. hdWGCNA identified PVT1 and APOE as hub genes associated with microglia. In conclusion, our findings provide evidence that PANoptosis may play a role in the immune dysregulation observed in AD. PVT1 has been implicated in AD pathogenesis, potentially exerting its effects through the miR-488-3p/FOXD3/SCN2A axis.
Collapse
Affiliation(s)
- Hanjie Liu
- School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Haidian District, 100084, Beijing, P.R. China
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China
| | - Maochun You
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China
| | - Hui Yang
- Chengdu Shuangliu Hospital of Traditional Chinese Medicine, Chengdu, 610200, Sichuan, P.R. China
| | - Xiao Wu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China
| | - Siyu Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China
| | - Sihan Huang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China
| | - Huijuan Gao
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Shijingshan District, 100040, Beijing, P.R. China.
| | - Lushuang Xie
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China.
| |
Collapse
|
3
|
Ahn EH, Park JB. Molecular Mechanisms of Alzheimer's Disease Induced by Amyloid-β and Tau Phosphorylation Along with RhoA Activity: Perspective of RhoA/Rho-Associated Protein Kinase Inhibitors for Neuronal Therapy. Cells 2025; 14:89. [PMID: 39851517 PMCID: PMC11764136 DOI: 10.3390/cells14020089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
Amyloid-β peptide (Aβ) is a critical cause of Alzheimer's disease (AD). It is generated from amyloid precursor protein (APP) through cleavages by β-secretase and γ-secretase. γ-Secretase, which includes presenilin, is regulated by several stimuli. Tau protein has also been identified as a significant factor in AD. In particular, Tau phosphorylation is crucial for neuronal impairment, as phosphorylated Tau detaches from microtubules, leading to the formation of neurofibrillary tangles and the destabilization of the microtubule structure. This instability in microtubules damages axons and dendrites, resulting in neuronal impairment. Notably, Aβ is linked to Tau phosphorylation. Another crucial factor in AD is neuroinflammation, primarily occurring in the microglia. Microglia possess several receptors that bind with Aβ, triggering the expression and release of an inflammatory factor, although their main physiological function is to phagocytose debris and pathogens in the brain. NF-κB activation plays a major role in neuroinflammation. Additionally, the production of reactive oxygen species (ROS) in the microglia contributes to this neuroinflammation. In microglia, superoxide is produced through NADPH oxidase, specifically NOX2. Rho GTPases play an essential role in regulating various cellular processes, including cytoskeletal rearrangement, morphology changes, migration, and transcription. The typical function of Rho GTPases involves regulating actin filament formation. Neurons, with their complex processes and synapse connections, rely on cytoskeletal dynamics for structural support. Other brain cells, such as astrocytes, microglia, and oligodendrocytes, also depend on specific cytoskeletal structures to maintain their unique cellular architectures. Thus, the aberrant regulation of Rho GTPases activity can disrupt actin filaments, leading to altered cell morphology, including changes in neuronal processes and synapses, and potentially contributing to brain diseases such as AD.
Collapse
Affiliation(s)
- Eun Hee Ahn
- Department of Physiology, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea;
- Department of Neurology, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
- ELMED Co., Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
| |
Collapse
|
4
|
Sabrie Z, Temiz-Resitoglu M, Kalkan T, Kilic B, Tunctan B, Malik KU, Sahan-Firat S. Protection by selective mTORC2 inhibition of Zymosan-induced hypotension and systemic inflammation mediated via IKKα/IκB-α/NF-κB activation. Prostaglandins Other Lipid Mediat 2024; 175:106918. [PMID: 39461547 DOI: 10.1016/j.prostaglandins.2024.106918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Non-septic shock is a serious condition leading to multiple organ dysfunction. Although targeting the mammalian target of the rapamycin complex 1 (mTORC1) signaling pathway exerts potent anti-inflammatory activity, little is known about mTORC2's contribution to non-septic shock. Thus, our research aims to investigate mTORC2's contribution and associated changes of IκB kinase (IKKα)/inhibitor κB (IκB-α)/nuclear factor-ĸB (NF-κB) pathway on Zymosan (ZYM)-induced non-septic rat model using the novel mTORC2 selective inhibitor JR-AB2-011. Rats were given saline (4 ml/kg), dimethylsulfoxide (DMSO) (4 ml/kg), ZYM (500 mg/kg), and (or) JR-AB2-011 (1 mg/kg). Mean arterial pressure (MAP) and heart rate (HR) of rats were recorded. JR-AB2-011 reversed both ZYM-induced reduction in MAP and increase in HR. Protein expression and/or phosphorylation of rictor, protein kinase B (Akt), IκB-α, IKKα, NF-κB p65, inducible nitric oxide synthase (iNOS), nitrotyrosine, cyclooxygenase 2 (COX-2), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, besides prostaglandin (PG) E2 levels were measured. The enhanced expression of the proteins mentioned above has been inhibited by JR-AB2-011. These data suggest mTORC2's promising role in ZYM-induced hypotension and systemic inflammation mediated via IKKα/IκB-α/NF-κB pathway.
Collapse
Affiliation(s)
- Zainab Sabrie
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | | | - Taskin Kalkan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Banu Kilic
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Kafait U Malik
- Department of Pharmacology, College of Medicine, University of Tennessee, Center for Health Sciences, Memphis, TN, USA
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey.
| |
Collapse
|
5
|
Macías F, Ulloa M, Clapp C, Martínez de la Escalera G, Arnold E. Prolactin protects hippocampal neurons against H2O2-induced neurotoxicity by suppressing BAX and NOX4 via the NF-κB signaling pathway. PLoS One 2024; 19:e0313328. [PMID: 39499702 PMCID: PMC11537405 DOI: 10.1371/journal.pone.0313328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/22/2024] [Indexed: 11/07/2024] Open
Abstract
Reactive oxygen species (ROS) are physiological byproducts of neuronal metabolism. However, an imbalance between ROS generation and antioxidant capacity, often driven by dysregulated pro-oxidant enzymes like nicotinamide adenine dinucleotide phosphate oxidases (NOX), can result in deleterious oxidative stress. This oxidative stress is a critical factor in the pathogenesis of neurodegenerative diseases. While interventions with broad-spectrum antioxidants have demonstrated limited efficacy, the modulation of endogenous antioxidant mechanisms presents a promising therapeutic avenue. Here, we investigated the potential of the neuroprotective hormone prolactin to mitigate oxidative stress and subsequent neuronal cell death. Prolactin protected primary mouse hippocampal neurons from hydrogen peroxide (H2O2)-induced oxidative damage. Prolactin reduced ROS levels, lipid peroxidation, and apoptosis, and its effects were occluded by a specific prolactin receptor antagonist (G129R-hPRL). Mechanistically, prolactin suppressed H2O2-induced mRNA upregulation of pro-oxidative Nox4 and pro-apoptotic Bax. Moreover, prolactin induced nuclear factor kappa B (NF-κB) nuclear translocation, and the inhibition of the NF-κB signaling pathway abolished the neuroprotective and transcriptional effects of prolactin, indicating its central role in prolactin-mediated protection. Our findings indicate that prolactin exerts potent antioxidant and neuroprotective effects by modulating the expression of Nox4 and Bax, thereby reducing ROS generation and neuronal apoptosis. This study underscores the therapeutic potential of prolactin in attenuating oxidative stress and suggests a possible role in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Fernando Macías
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, Querétaro, México
| | - Miriam Ulloa
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, Querétaro, México
| | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, Querétaro, México
| | - Gonzalo Martínez de la Escalera
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, Querétaro, México
| | - Edith Arnold
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, Querétaro, México
- CONAHCYT–Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, Querétaro, México
| |
Collapse
|
6
|
Kaltschmidt B, Czaniera NJ, Schulten W, Kaltschmidt C. NF-κB in Alzheimer's Disease: Friend or Foe? Opposite Functions in Neurons and Glial Cells. Int J Mol Sci 2024; 25:11353. [PMID: 39518906 PMCID: PMC11545113 DOI: 10.3390/ijms252111353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is a devasting neurodegenerative disease afflicting mainly glutamatergic neurons together with a massive neuroinflammation mediated by the transcription factor NF-κB. A 65%-plus increase in Alzheimer's patients by 2050 might be a major threat to society. Hallmarks of AD are neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau and amyloid beta (Aβ) plaques. Here, we review the potential involvement of transcription factor NF-κB by hereditary mutations of the tumor necrosis factor pathway in AD patients. One of the greatest genetic risk factors is APOE4. Recently, it was shown that the APOE4 allele functions as a null allele in human astrocytes not repressing NF-κB anymore. Moreover, NF-κB seems to be involved in the repair of DNA double-strand breaks during healthy learning and memory, a function blunted in AD. NF-κB could be a friend to healthy neurons by repressing apoptosis and necroptosis. But a loss of neuronal NF-κB and activation of glial NF-κB in AD makes it a foe of neuronal survival. Hopeful therapies include TNFR2 receptor bodies relieving the activation of glial NF-κB by TNFα.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, 33615 Bielefeld, Germany
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), 33615 Bielefeld, Germany
| | - Nele Johanne Czaniera
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
| | - Wiebke Schulten
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), 33615 Bielefeld, Germany
| |
Collapse
|
7
|
Qiao N, Shao H. Identification of neutrophil extracellular trap-related genes in Alzheimer's disease based on comprehensive bioinformatics analysis. Comput Methods Biomech Biomed Engin 2024:1-14. [PMID: 39314024 DOI: 10.1080/10255842.2024.2399029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease. There are currently no effective interventions to slow down or prevent the occurrence and progression of AD. Neutrophil extracellular traps (NETs) have been proven to be tightly linked to AD. This project attempted to identify hub genes for AD based on NETs. Gene expression profiles of the training set and validation set were downloaded from the Gene Expression Omnibus (GEO) database, including non-demented (ND) controls and AD samples. NET-related genes (NETRGs) were collected from the literature. Differential analysis identified 21 AD differentially expressed NETRGs (AD-DE-NETRGs) majorly linked to functions such as defense response to bacterium as well as pathways including IL-17 signaling pathway, as evidenced by enrichment analyses of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Protein-protein interaction (PPI) network, Minutia Cylinder-Code (MCC) algorithm, and molecular complex detection (MCODE) algorithm in the CytoHubba plug-in were employed to identify five hub genes (NFKBIA, SOCS3, CCL2, TIMP1, ACTB). Their diagnostic ability was validated in the validation set using receiver operating characteristic (ROC) curves and gene differential expression analysis. A total of 16 miRNAs and 132 lncRNAs were predicted through the mirDIP and ENCORI databases, and a lncRNA-miRNA-mRNA regulatory network was constructed using Cytoscape software. Small molecular compounds such as Benzo(a)pyrene and Copper Sulfate were predicted to target hub genes using the CTD database. This project successfully identified five hub genes, which may serve as potential biomarkers for AD, proffering clues for new therapeutic targets.
Collapse
Affiliation(s)
- Nana Qiao
- Department of Neurology, Xianyang Hospital of Yan'an University, Xianyang, China
| | - He Shao
- Department of Neurology, Xianyang Hospital of Yan'an University, Xianyang, China
| |
Collapse
|
8
|
Esmaealzadeh N, Miri MS, Mavaddat H, Peyrovinasab A, Ghasemi Zargar S, Sirous Kabiri S, Razavi SM, Abdolghaffari AH. The regulating effect of curcumin on NF-κB pathway in neurodegenerative diseases: a review of the underlying mechanisms. Inflammopharmacology 2024; 32:2125-2151. [PMID: 38769198 DOI: 10.1007/s10787-024-01492-1] [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: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Neurodegenerative diseases are part of the central nervous system (CNS) disorders that indicate their presence with neuronal loss, neuroinflammation, and increased oxidative stress. Several pathophysiological factors and biomarkers are involved in this inflammatory process causing these neurological disorders. The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is an inflammation element, which induced transcription and appears to be one of the important players in physiological procedures, especially nervous disorders. NF-κB can impact upon series of intracellular actions and induce or inhibit many inflammation-related pathways. Multiple reports have focused on the modification of NF-κB activity, controlling its expression, translocation, and signaling pathway in neurodegenerative disorders and injuries like Alzheimer's disease (AD), spinal cord injuries (SCI), and Parkinson's disease (PD). Curcumin has been noted to be a popular anti-oxidant and anti-inflammatory substance and is the foremost natural compound produced by turmeric. According to various studies, when playing an anti-inflammatory role, it interacts with several modulating proteins of long-standing disease signaling pathways and has an unprovocative consequence on pro-inflammatory cytokines. This review article determined to figure out curcumin's role in limiting the promotion of neurodegenerative disease via influencing the NF-κB signaling route. Preclinical studies were gathered from plenty of scientific platforms including PubMed, Scopus, Cochrane, and Google Scholar to evaluate this hypothesis. Extracted findings from the literature review explained the repressing impact of Curcumin on the NF-κB signaling pathway and, occasionally down-regulating the cytokine expression. Yet, there is an essential need for further analysis and specific clinical experiments to fully understand this subject.
Collapse
Affiliation(s)
- Niusha Esmaealzadeh
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Traditional Persian Medicine and Complementary Medicine (PerCoMed) Student Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahdis Sadat Miri
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., P. O. Box: 19419-33111, Tehran, Iran
| | - Helia Mavaddat
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., P. O. Box: 19419-33111, Tehran, Iran
| | - Amirreza Peyrovinasab
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., P. O. Box: 19419-33111, Tehran, Iran
| | - Sara Ghasemi Zargar
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., P. O. Box: 19419-33111, Tehran, Iran
| | - Shirin Sirous Kabiri
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., P. O. Box: 19419-33111, Tehran, Iran
| | - Seyed Mehrad Razavi
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., P. O. Box: 19419-33111, Tehran, Iran.
| | - Amir Hossein Abdolghaffari
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., P. O. Box: 19419-33111, Tehran, Iran.
| |
Collapse
|
9
|
Dai Y, Lin J, Chen X, Ren J, Wu C, Shen H, Li X, Yu J, Jiang B, Yu L. NAMPT/NAD +/PARP1 Pathway Regulates CFA-Induced Inflammatory Pain via NF-κB Signaling in Rodents. Adv Biol (Weinh) 2024; 8:e2400028. [PMID: 38463014 DOI: 10.1002/adbi.202400028] [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: 01/16/2024] [Revised: 02/26/2024] [Indexed: 03/12/2024]
Abstract
Emerging evidence has implicated nicotinamide adenine dinucleotide (NAD+) metabolism in various inflammatory diseases. In the study, the role of NAD+ metabolism in Complete Freund's Adjuvant (CFA)-evoked inflammatory pain and the underlying mechanisms are investigated. The study demonstrated that CFA induced upregulation of nicotinamide phosphoribosyltransferase (NAMPT) in dorsal root ganglia (DRG) without significant changes in the spinal cord. Inhibition of NAMPT expression by intrathecal injection of NAMPT siRNA alleviated CFA-induced pain-like behavior, decreased NAD+ contents in DRG, and lowered poly-(ADP-ribose) polymerase 1 (PARP1) activity levels. These effects are all reversed by the supplement of nicotinamide mononucleotide (NMN). Inhibition of PARP1 expression by intrathecal injection of PARP1 siRNA alleviated CFA-induced pain-like behavior, while elevated NAD+ levels of DRG. The analgesic effect of inhibiting NAMPT/NAD+/PARP1 axis can be attributed to the downregulation of the NF-κB/IL-1β inflammatory pathway. Double immunofluorescence staining showed that the expression of NAMPT/NAD+/PARP1 axis is restricted to DRG neurons. In conclusion, PARP1 activation in response to CFA stimulation, fueled by NAMPT-derived NAD+, mediates CFA-induced inflammatory pain through NF-κB/IL-1β inflammatory pathway.
Collapse
Affiliation(s)
- Yi Dai
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jiaqi Lin
- East Hospital Affiliated to Tongji University, Shanghai, 200000, China
| | - Xiangde Chen
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jinxuan Ren
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Chengwei Wu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Lishui Municipal Central Hospital, Lishui, 323000, China
| | - Huihui Shen
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Xue Li
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jing Yu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Baochun Jiang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Lina Yu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
10
|
Basheer N, Buee L, Brion JP, Smolek T, Muhammadi MK, Hritz J, Hromadka T, Dewachter I, Wegmann S, Landrieu I, Novak P, Mudher A, Zilka N. Shaping the future of preclinical development of successful disease-modifying drugs against Alzheimer's disease: a systematic review of tau propagation models. Acta Neuropathol Commun 2024; 12:52. [PMID: 38576010 PMCID: PMC10993623 DOI: 10.1186/s40478-024-01748-5] [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: 01/11/2024] [Accepted: 02/21/2024] [Indexed: 04/06/2024] Open
Abstract
The transcellular propagation of the aberrantly modified protein tau along the functional brain network is a key hallmark of Alzheimer's disease and related tauopathies. Inoculation-based tau propagation models can recapitulate the stereotypical spread of tau and reproduce various types of tau inclusions linked to specific tauopathy, albeit with varying degrees of fidelity. With this systematic review, we underscore the significance of judicious selection and meticulous functional, biochemical, and biophysical characterization of various tau inocula. Furthermore, we highlight the necessity of choosing suitable animal models and inoculation sites, along with the critical need for validation of fibrillary pathology using confirmatory staining, to accurately recapitulate disease-specific inclusions. As a practical guide, we put forth a framework for establishing a benchmark of inoculation-based tau propagation models that holds promise for use in preclinical testing of disease-modifying drugs.
Collapse
Affiliation(s)
- Neha Basheer
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Luc Buee
- Inserm, CHU Lille, CNRS, LilNCog - Lille Neuroscience & Cognition, University of Lille, 59000, Lille, France.
| | - Jean-Pierre Brion
- Faculty of Medicine, Laboratory of Histology, Alzheimer and Other Tauopathies Research Group (CP 620), ULB Neuroscience Institute (UNI), Université Libre de Bruxelles, 808, Route de Lennik, 1070, Brussels, Belgium
| | - Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Muhammad Khalid Muhammadi
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Jozef Hritz
- CEITEC Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 62500, Brno, Czech Republic
| | - Tomas Hromadka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Ilse Dewachter
- Biomedical Research Institute, BIOMED, Hasselt University, 3500, Hasselt, Belgium
| | - Susanne Wegmann
- German Center for Neurodegenerative Diseases, Charitéplatz 1, 10117, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Isabelle Landrieu
- CNRS EMR9002 - BSI - Integrative Structural Biology, 59000, Lille, France
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, 59000, Lille, France
| | - Petr Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Amritpal Mudher
- School of Biological Sciences, Faculty of Environment and Life Sciences, University of Southampton, Highfield Campus, Southampton, SO17 1BJ, UK
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia.
- AXON Neuroscience R&D Services SE, Dubravska Cesta 9, 845 10, Bratislava, Slovakia.
| |
Collapse
|
11
|
de la Fuente AG, Pelucchi S, Mertens J, Di Luca M, Mauceri D, Marcello E. Novel therapeutic approaches to target neurodegeneration. Br J Pharmacol 2023; 180:1651-1673. [PMID: 36965025 PMCID: PMC10952850 DOI: 10.1111/bph.16078] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 03/27/2023] Open
Abstract
Ageing is the main risk factor common to most primary neurodegenerative disorders. Indeed, age-related brain alterations have been long considered to predispose to neurodegeneration. Although protein misfolding and the accumulation of toxic protein aggregates have been considered as causative events in neurodegeneration, several other biological pathways affected by brain ageing also contribute to pathogenesis. Here, we discuss the evidence showing the involvement of the mechanisms controlling neuronal structure, gene expression, autophagy, cell metabolism and neuroinflammation in the onset and progression of neurodegenerative disorders. Furthermore, we review the therapeutic strategies currently under development or as future approaches designed to normalize these pathways, which may then increase brain resilience to cope with toxic protein species. In addition to therapies targeting the insoluble protein aggregates specifically associated with each neurodegenerative disorder, these novel pharmacological approaches may be part of combined therapies designed to rescue brain function.
Collapse
Affiliation(s)
- Alerie G. de la Fuente
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
- Instituto de Neurociencias CSIC‐UMHAlicanteSpain
- Wellcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
- Institute of Molecular BiologyLeopold‐Franzens‐Universität InnsbruckInnsbruckAustria
| | - Jerome Mertens
- Institute of Molecular BiologyLeopold‐Franzens‐Universität InnsbruckInnsbruckAustria
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Daniela Mauceri
- Institute of Anatomy and Cell BiologyDepartment of Molecular and Cellular Neuroscience, University of MarburgMarburgGermany
- Department of NeurobiologyInterdisciplinary Centre for Neurosciences (IZN), Heidelberg UniversityHeidelbergGermany
| | - Elena Marcello
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| |
Collapse
|
12
|
Waigi EW, Webb RC, Moss MA, Uline MJ, McCarthy CG, Wenceslau CF. Soluble and insoluble protein aggregates, endoplasmic reticulum stress, and vascular dysfunction in Alzheimer's disease and cardiovascular diseases. GeroScience 2023; 45:1411-1438. [PMID: 36823398 PMCID: PMC10400528 DOI: 10.1007/s11357-023-00748-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/28/2023] [Indexed: 02/25/2023] Open
Abstract
Dementia refers to a particular group of symptoms characterized by difficulties with memory, language, problem-solving, and other thinking skills that affect a person's ability to perform everyday activities. Alzheimer's disease (AD) is the most common form of dementia, affecting about 6.2 million Americans aged 65 years and older. Likewise, cardiovascular diseases (CVDs) are a major cause of disability and premature death, impacting 126.9 million adults in the USA, a number that increases with age. Consequently, CVDs and cardiovascular risk factors are associated with an increased risk of AD and cognitive impairment. They share important age-related cardiometabolic and lifestyle risk factors, that make them among the leading causes of death. Additionally, there are several premises and hypotheses about the mechanisms underlying the association between AD and CVD. Although AD and CVD may be considered deleterious to health, the study of their combination constitutes a clinical challenge, and investigations to understand the mechanistic pathways for the cause-effect and/or shared pathology between these two disease constellations remains an active area of research. AD pathology is propagated by the amyloid β (Aβ) peptides. These peptides give rise to small, toxic, and soluble Aβ oligomers (SPOs) that are nonfibrillar, and it is their levels that show a robust correlation with the extent of cognitive impairment. This review will elucidate the interplay between the effects of accumulating SPOs in AD and CVDs, the resulting ER stress response, and their role in vascular dysfunction. We will also address the potential underlying mechanisms, including the possibility that SPOs are among the causes of vascular injury in CVD associated with cognitive decline. By revealing common mechanistic underpinnings of AD and CVD, we hope that novel experimental therapeutics can be designed to reduce the burden of these devastating diseases. Graphical abstract Alzheimer's disease (AD) pathology leads to the release of Aβ peptides, and their accumulation in the peripheral organs has varying effects on various components of the cardiovascular system including endoplasmic reticulum (ER) stress and vascular damage. Image created with BioRender.com.
Collapse
Affiliation(s)
- Emily W Waigi
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - R Clinton Webb
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
| | - Melissa A Moss
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, USA
| | - Mark J Uline
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, USA
| | - Cameron G McCarthy
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
| | - Camilla Ferreira Wenceslau
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA.
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA.
| |
Collapse
|
13
|
Schnöder L, Quan W, Yu Y, Tomic I, Luo Q, Hao W, Peng G, Li D, Fassbender K, Liu Y. Deficiency of IKKβ in neurons ameliorates Alzheimer's disease pathology in APP- and tau-transgenic mice. FASEB J 2023; 37:e22778. [PMID: 36688823 DOI: 10.1096/fj.202201512r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/21/2022] [Accepted: 01/06/2023] [Indexed: 01/24/2023]
Abstract
In Alzheimer's disease (AD) brain, inflammatory activation regulates protein levels of amyloid-β-peptide (Aβ) and phosphorylated tau (p-tau), as well as neurodegeneration; however, the regulatory mechanisms remain unclear. We constructed APP- and tau-transgenic AD mice with deletion of IKKβ specifically in neurons, and observed that IKKβ deficiency reduced cerebral Aβ and p-tau, and modified inflammatory activation in both AD mice. However, neuronal deficiency of IKKβ decreased apoptosis and maintained synaptic proteins (e.g., PSD-95 and Munc18-1) in the brain and improved cognitive function only in APP-transgenic mice, but not in tau-transgenic mice. Additionally, IKKβ deficiency decreased BACE1 protein and activity in APP-transgenic mouse brain and cultured SH-SY5Y cells. IKKβ deficiency increased expression of PP2A catalytic subunit isoform A, an enzyme dephosphorylating cerebral p-tau, in the brain of tau-transgenic mice. Interestingly, deficiency of IKKβ in neurons enhanced autophagy as indicated by the increased ratio of LC3B-II/I in brains of both APP- and tau-transgenic mice. Thus, IKKβ deficiency in neurons ameliorates AD-associated pathology in APP- and tau-transgenic mice, perhaps by decreasing Aβ production, increasing p-tau dephosphorylation, and promoting autophagy-mediated degradation of BACE1 and p-tau aggregates in the brain. However, IKKβ deficiency differently protects neurons in APP- and tau-transgenic mice. Further studies are needed, particularly in the context of interaction between Aβ and p-tau, before IKKβ/NF-κB can be targeted for AD therapies.
Collapse
Affiliation(s)
- Laura Schnöder
- Department of Neurology, Saarland University, Homburg, Germany
| | - Wenqiang Quan
- Department of Neurology, Saarland University, Homburg, Germany
- Department of Clinical Laboratory, Tongji Hospital, Tongji University Medical School, Shanghai, China
| | - Ye Yu
- Department of Neurology, Saarland University, Homburg, Germany
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Inge Tomic
- Department of Neurology, Saarland University, Homburg, Germany
| | - Qinghua Luo
- Department of Neurology, Saarland University, Homburg, Germany
| | - Wenlin Hao
- Department of Neurology, Saarland University, Homburg, Germany
| | - Guoping Peng
- Department of Neurology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dong Li
- Department of Clinical Laboratory, Tongji Hospital, Tongji University Medical School, Shanghai, China
| | | | - Yang Liu
- Department of Neurology, Saarland University, Homburg, Germany
- Department of Clinical Laboratory, Tongji Hospital, Tongji University Medical School, Shanghai, China
| |
Collapse
|
14
|
Kaltschmidt B, Helweg LP, Greiner JFW, Kaltschmidt C. NF-κB in neurodegenerative diseases: Recent evidence from human genetics. Front Mol Neurosci 2022; 15:954541. [PMID: 35983068 PMCID: PMC9380593 DOI: 10.3389/fnmol.2022.954541] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
The transcription factor NF-κB is commonly known to drive inflammation and cancer progression, but is also a crucial regulator of a broad range of cellular processes within the mammalian nervous system. In the present review, we provide an overview on the role of NF-κB in the nervous system particularly including its constitutive activity within cortical and hippocampal regions, neuroprotection as well as learning and memory. Our discussion further emphasizes the increasing role of human genetics in neurodegenerative disorders, namely, germline mutations leading to defects in NF-κB-signaling. In particular, we propose that loss of function mutations upstream of NF-κB such as ADAM17, SHARPIN, HOIL, or OTULIN affect NF-κB-activity in Alzheimer’s disease (AD) patients, in turn driving anatomical defects such as shrinkage of entorhinal cortex and the limbic system in early AD. Similarly, E3 type ubiquitin ligase PARKIN is positively involved in NF-κB signaling. PARKIN loss of function mutations are most frequently observed in Parkinson’s disease patients. In contrast to AD, relying on germline mutations of week alleles and a disease development over decades, somatic mutations affecting NF-κB activation are commonly observed in cells derived from glioblastoma multiforme (GBM), the most common malignant primary brain tumor. Here, our present review particularly sheds light on the mutual exclusion of either the deletion of NFKBIA or amplification of epidermal growth factor receptor (EGFR) in GBM, both resulting in constitutive NF-κB-activity driving tumorigenesis. We also discuss emerging roles of long non-coding RNAs such as HOTAIR in suppressing phosphorylation of IκBα in the context of GBM. In summary, the recent progress in the genetic analysis of patients, particularly those suffering from AD, harbors the potential to open up new vistas for research and therapy based on TNFα/NF-κB pathway and neuroprotection.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Department of Molecular Neurobiology, Bielefeld University, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), Bielefeld, Germany
- Department of Cell Biology, Biological Faculty, University of Bielefeld, Bielefeld, Germany
- *Correspondence: Barbara Kaltschmidt,
| | - Laureen P. Helweg
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), Bielefeld, Germany
- Department of Cell Biology, Biological Faculty, University of Bielefeld, Bielefeld, Germany
| | - Johannes F. W. Greiner
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), Bielefeld, Germany
- Department of Cell Biology, Biological Faculty, University of Bielefeld, Bielefeld, Germany
| | - Christian Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), Bielefeld, Germany
- Department of Cell Biology, Biological Faculty, University of Bielefeld, Bielefeld, Germany
| |
Collapse
|
15
|
Zhang Q, Li J, Weng L. Identification and Validation of Aging-Related Genes in Alzheimer’s Disease. Front Neurosci 2022; 16:905722. [PMID: 35615282 PMCID: PMC9124812 DOI: 10.3389/fnins.2022.905722] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 04/13/2022] [Indexed: 12/17/2022] Open
Abstract
Aging is recognized as the key risk factor for Alzheimer’s disease (AD). This study aimed to identify and verify potential aging-related genes associated with AD using bioinformatics analysis. Aging-related differential expression genes (ARDEGs) were determined by the intersection of limma test, weighted correlation network analysis (WGCNA), and 1153 aging and senescence-associated genes. Potential biological functions and pathways of ARDEGs were determined by GO, KEGG, GSEA, and GSVA. Then, LASSO algorithm was used to identify the hub genes and the diagnostic ability of the five ARDEGs in discriminating AD from the healthy control samples. Further, the correlation between hub ARDEGs and clinical characteristics was explored. Finally, the expression level of the five ARDEGs was validated using other four GEO datasets and blood samples of patients with AD and healthy individuals. Five ARDEGs (GFAP, PDGFRB, PLOD1, MAP4K4, and NFKBIA) were obtained. For biological function analysis, aging, cellular senescence, and Ras protein signal transduction regulation were enriched. Diagnostic ability of the five ARDEGs in discriminating AD from the control samples demonstrated a favorable diagnostic value. Eventually, quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) validation test revealed that compared with healthy controls, the mRNA expression level of PDGFRB, PLOD1, MAP4K4, and NFKBIA were elevated in AD patients. In conclusion, this study identified four ARDEGs (PDGFRB, PLOD1, MAP4K4, and NFKBIA) associated with AD. They provide an insight into potential novel biomarkers for diagnosing AD and monitoring progression.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hydrocephalus Center, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Weng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- *Correspondence: Ling Weng,
| |
Collapse
|
16
|
Microglial NF-κB drives tau spreading and toxicity in a mouse model of tauopathy. Nat Commun 2022; 13:1969. [PMID: 35413950 PMCID: PMC9005658 DOI: 10.1038/s41467-022-29552-6] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/21/2022] [Indexed: 01/31/2023] Open
Abstract
Activation of microglia is a prominent pathological feature in tauopathies, including Alzheimer's disease. How microglia activation contributes to tau toxicity remains largely unknown. Here we show that nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, activated by tau, drives microglial-mediated tau propagation and toxicity. Constitutive activation of microglial NF-κB exacerbated, while inactivation diminished, tau seeding and spreading in young PS19 mice. Inhibition of NF-κB activation enhanced the retention while reduced the release of internalized pathogenic tau fibrils from primary microglia and rescued microglial autophagy deficits. Inhibition of microglial NF-κB in aged PS19 mice rescued tau-mediated learning and memory deficits, restored overall transcriptomic changes while increasing neuronal tau inclusions. Single cell RNA-seq revealed that tau-associated disease states in microglia were diminished by NF-κB inactivation and further transformed by constitutive NF-κB activation. Our study establishes a role for microglial NF-κB signaling in mediating tau spreading and toxicity in tauopathy.
Collapse
|
17
|
Sajad M, Kumar R, Thakur SC. History in Perspective: The Prime Pathological Players and Role of Phytochemicals in Alzheimer’s Disease. IBRO Neurosci Rep 2022; 12:377-389. [PMID: 35586776 PMCID: PMC9108734 DOI: 10.1016/j.ibneur.2022.04.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/25/2022] [Indexed: 11/01/2022] Open
Abstract
Alzheimer's disease is a steadily progressive, irreversible neurological disorder that is most frequently categorized under the umbrella term "neurodegeneration". Several attempts are underway to clarify the pathogenic mechanisms, identify the aetiologies, and determine a pathway by which the therapeutic steps can be implemented. Oxidative stress is one of the pathogenic processes, which is commonly believed to be associated with neurodegenerative diseases. Accumulation of extracellular amyloid-β protein (Aβ), hyperphosphorylation of tau, initiation of neurometabolic reactions characterized by the loss of neuronal function and synaptic failure, and decreased or lost learning capability and memory function are the most central neuropathological characteristics of AD. According to the amyloid cascade hypothesis, the enhanced deposition of Aβ deposits and neurofibrillary tangles due to hyperphosphorylation of Tau activates the cascade reactions in the brain. These reactions affect the synaptic activity and activation of microglia, which results in neuroinflammation due to enhanced immune function. Plant-based phytochemicals have also been used long ago against several diseases. Phytoconstituents play a significant neuroprotective property by preventing the pathophysiology of the disease. In this review, we have discussed the formation and crosstalk between amyloid and tau pathologies as well as the effect of neuroinflammation on the progression of AD. We have specifically focused on the formation of NFT, β-amyloids, inflammation, and pathophysiology of AD and the role of phytochemicals in the prevention of AD. AD is an insidious, slowly progressive, and neurodegenerative disorder. Common symptoms are memory loss, difficulty in recalling, and understanding. β-amyloids and Neurofibrillary tangles are the main factors in AD pathogenesis. Activated microglia and oxidative stress have different effects on AD progression. Phytochemicals show a key role against AD by inhibiting several pathways.
Collapse
|
18
|
Jurcau A, Simion A. Oxidative Stress in the Pathogenesis of Alzheimer's Disease and Cerebrovascular Disease with Therapeutic Implications. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 19:94-108. [PMID: 32124703 DOI: 10.2174/1871527319666200303121016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 01/18/2020] [Accepted: 01/28/2020] [Indexed: 12/15/2022]
Abstract
The significant gain in life expectancy led to an increase in the incidence and prevalence of dementia. Although vascular risk factors have long and repeatedly been shown to increase the risk of Alzheimer's Disease (AD), translating these findings into effective preventive measures has failed. In addition, the finding that incident ischemic stroke approximately doubles the risk of a patient to develop AD has been recently reinforced. Current knowledge and pathogenetic hypotheses of AD are discussed. The implication of oxidative stress in the development of AD is reviewed, with special emphasis on its sudden burst in the setting of acute ischemic stroke and the possible link between this increase in oxidative stress and consequent cognitive impairment. Current knowledge and future directions in the prevention and treatment of AD are discussed outlining the hypothesis of a possible beneficial effect of antioxidant treatment in acute ischemic stroke in delaying the onset/progression of dementia.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Faculty of Medicine and Pharmacy, University of Oradea, 410154 Oradea, Romania.,Clinical Municipal Hospital "Dr. G Curteanu", Neurology Ward, Oradea, Romania
| | - Aurel Simion
- Faculty of Medicine and Pharmacy, University of Oradea, 410154 Oradea, Romania.,Clinical Municipal Hospital "Dr. G Curteanu", Neurological Rehabilitation Ward, Oradea, Romania
| |
Collapse
|
19
|
Yakupova EI, Bobyleva LG, Shumeyko SA, Vikhlyantsev IM, Bobylev AG. Amyloids: The History of Toxicity and Functionality. BIOLOGY 2021; 10:biology10050394. [PMID: 34062910 PMCID: PMC8147320 DOI: 10.3390/biology10050394] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022]
Abstract
Proteins can perform their specific function due to their molecular structure. Partial or complete unfolding of the polypeptide chain may lead to the misfolding and aggregation of proteins in turn, resulting in the formation of different structures such as amyloid aggregates. Amyloids are rigid protein aggregates with the cross-β structure, resistant to most solvents and proteases. Because of their resistance to proteolysis, amyloid aggregates formed in the organism accumulate in tissues, promoting the development of various diseases called amyloidosis, for instance Alzheimer's diseases (AD). According to the main hypothesis, it is considered that the cause of AD is the formation and accumulation of amyloid plaques of Aβ. That is why Aβ-amyloid is the most studied representative of amyloids. Therefore, in this review, special attention is paid to the history of Aβ-amyloid toxicity. We note the main problems with anti-amyloid therapy and write about new views on amyloids that can play positive roles in the different organisms including humans.
Collapse
Affiliation(s)
- Elmira I. Yakupova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +7-(985)687-77-27
| | - Liya G. Bobyleva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| | - Sergey A. Shumeyko
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| | - Ivan M. Vikhlyantsev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| | - Alexander G. Bobylev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| |
Collapse
|
20
|
Romero-Nava R, Alarcón-Aguilar FJ, Giacoman-Martínez A, Blancas-Flores G, Aguayo-Cerón KA, Ballinas-Verdugo MA, Sánchez-Muñoz F, Huang F, Villafaña-Rauda S, Almanza-Pérez JC. Glycine is a competitive antagonist of the TNF receptor mediating the expression of inflammatory cytokines in 3T3-L1 adipocytes. Inflamm Res 2021; 70:605-618. [PMID: 33877377 DOI: 10.1007/s00011-021-01462-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/16/2021] [Accepted: 04/05/2021] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE To determine the involvement of TNF-α and glycine receptors in the inhibition of pro-inflammatory adipokines in 3T3-L1 cells. METHODS RT-PCR evidenced glycine receptors in 3T3-L1 adipocytes. 3T3-L1 cells were transfected with siRNA for the glycine (Glrb) and TNF1a (Tnfrsf1a) receptors and confirmed by confocal microscopy. Transfected cells were treated with glycine (10 mM). The expressions of TNF-α and IL-6 mRNA were measured by qRT-PCR, while concentrations were quantified by ELISA. RESULTS Glycine decreased the expression and concentration of TNF-α and IL-6; this effect did not occur in the absence of TNF-α receptor due to siRNA. In contrast, glycine produced only slight changes in the expression of TNF-α and IL-6 in the absence of the glycine receptor due to siRNA. A docking analysis confirmed the possibility of binding glycine to the TNF-α1a receptor. CONCLUSION These findings support the idea that glycine could partially inhibit the binding of TNF-α to its receptor and provide clues about the mechanisms by which glycine inhibits the secretion of pro-inflammatory adipokines in adipocytes through the TNF-α receptor.
Collapse
MESH Headings
- 3T3-L1 Cells
- Adipocytes/metabolism
- Adiponectin/genetics
- Animals
- Cytokines/genetics
- Cytokines/metabolism
- Gene Expression
- Glycine/pharmacology
- Mice
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/metabolism
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- Receptors, Glycine/genetics
- Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type II/genetics
Collapse
Affiliation(s)
- Rodrigo Romero-Nava
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C.P. 09340, Mexico City, Mexico
- Departamento de Farmacología y Toxicología, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
- Sección de Posgrado, Laboratorio de Señalización Intracelular, Escuela Superior de Medicina del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Francisco J Alarcón-Aguilar
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C.P. 09340, Mexico City, Mexico
| | - Abraham Giacoman-Martínez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C.P. 09340, Mexico City, Mexico
| | - Gerardo Blancas-Flores
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C.P. 09340, Mexico City, Mexico
| | - Karla A Aguayo-Cerón
- Sección de Posgrado, Laboratorio de Señalización Intracelular, Escuela Superior de Medicina del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Martha A Ballinas-Verdugo
- Departamento de Inmunología, Instituto Nacional de Cardiología (Ignacio Chávez), Mexico City, Mexico
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología (Ignacio Chávez), Mexico City, Mexico
| | - Fengyang Huang
- Departamento de Farmacología y Toxicología, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Santiago Villafaña-Rauda
- Sección de Posgrado, Laboratorio de Señalización Intracelular, Escuela Superior de Medicina del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Julio C Almanza-Pérez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C.P. 09340, Mexico City, Mexico.
| |
Collapse
|
21
|
Bedada FB, Ntekim OE, Nwulia EO, Fungwe TV, Nadarajah SR, Obisesan TO. Exercise Training-Increased FBXO32 and FOXO1 in a Gender-Dependent Manner in Mild Cognitively Impaired African Americans: GEMS-1 Study. Front Aging Neurosci 2021; 13:641758. [PMID: 33935685 PMCID: PMC8079639 DOI: 10.3389/fnagi.2021.641758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/18/2021] [Indexed: 01/29/2023] Open
Abstract
The ubiquitin proteasome system (UPS) and FOXOs transcription factors play a pivotal role in cellular clearance and minimizing the accumulation of Aβ in neurodegeneration (ND). In African Americans (AAs) with mild cognitive impairment (MCI), the role of components of UPS and FOXOs; and whether they are amenable to exercise effects is unknown. We hypothesized that exercise can enhance cellular clearance systems during aging and ND by increasing expressions of FBXO32 and FOXO1. To test this hypothesis, we used TaqMan gene expression analysis in peripheral blood (PB) to investigate the component of UPS and FOXOs; and provide mechanistic insight at baseline, during exercise, and in both genders. At baseline, levels of FBXO32 were higher in women than in men. In our attempt to discern gender-specific exercise-related changes, we observed that levels of FBXO32 increased in men but not in women. Similarly, levels of FOXO1 increased in men only. These data suggest that a graded dose of FBXO32 and FOXO1 may be beneficial when PB cells carrying FBXO32 and FOXO1 summon into the brain in response to Alzheimer's disease (AD) perturbation (docking station PB cells). Our observation is consistent with emerging studies that exercise allows the trafficking of blood factors. Given the significance of FBXO32 and FOXO1 to ND and associated muscle integrity, our findings may explain, at least in part, the benefits of exercise on memory, associated gait, and balance perturbation acknowledged to herald the emergence of MCI.
Collapse
Affiliation(s)
- Fikru B. Bedada
- Department of Clinical Laboratory Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Oyonumo E. Ntekim
- Department of Nutritional Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Evaristus O. Nwulia
- Department of Psychiatry, Howard University Hospital, Washington, DC, United States
| | - Thomas V. Fungwe
- Department of Nutritional Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Sheeba Raaj Nadarajah
- Department of Nursing, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Thomas O. Obisesan
- Division of Geriatrics, Department of Medicine, Howard University Hospital, Washington, DC, United States
| |
Collapse
|
22
|
Liu X, Wang K, Wei X, Xie T, Lv B, Zhou Q, Wang X. Interaction of NF-κB and Wnt/β-catenin Signaling Pathways in Alzheimer's Disease and Potential Active Drug Treatments. Neurochem Res 2021; 46:711-731. [PMID: 33523396 DOI: 10.1007/s11064-021-03227-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/24/2020] [Accepted: 01/02/2021] [Indexed: 12/25/2022]
Abstract
The most important neuropathological features of Alzheimer's disease (AD) are extracellular amyloid-β protein (Aβ) deposition, tau protein hyperphosphorylation and activation of neurometabolic reaction in the brain accompanied by neuronal and synaptic damage, and impaired learning and memory function. According to the amyloid cascade hypothesis, increased Aβ deposits in the brain to form the core of the senile plaques that initiate cascade reactions, affecting the synapses and stimulating activation of microglia, resulting in neuroinflammation. A growing number of studies has shown that NF-κB and Wnt/β-catenin pathways play important roles in neurodegenerative diseases, especially AD. In this review, we briefly introduce the connection between neuroinflammation-mediated synaptic dysfunction in AD and elaborated on the mechanism of these two signaling pathways in AD-related pathological changes, as well as their interaction. Based on our interest in natural compounds, we also briefly introduce and conduct preliminary screening of potential therapeutics for AD.
Collapse
Affiliation(s)
- Xiao Liu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Kaiyue Wang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xing Wei
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tian Xie
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bin Lv
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qian Zhou
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishiku, Kitakyushu, 807-8555, Japan
| | - Xiaoying Wang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China. .,College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
23
|
Snow WM, Cadonic C, Cortes-Perez C, Adlimoghaddam A, Roy Chowdhury SK, Thomson E, Anozie A, Bernstein MJ, Gough K, Fernyhough P, Suh M, Albensi BC. Sex-Specific Effects of Chronic Creatine Supplementation on Hippocampal-Mediated Spatial Cognition in the 3xTg Mouse Model of Alzheimer's Disease. Nutrients 2020; 12:nu12113589. [PMID: 33238473 PMCID: PMC7700653 DOI: 10.3390/nu12113589] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
The creatine (Cr) energy system has been implicated in Alzheimer’s disease (AD), including reductions in brain phosphoCr and Cr kinase, yet no studies have examined the neurobehavioral effects of Cr supplementation in AD, including the 3xTg mouse model. This studied investigated the effects of Cr supplementation on spatial cognition, plasticity- and disease-related protein levels, and mitochondrial function in the 3xTg hippocampus. Here, 3xTg mice were fed a control or Cr-supplemented (3% Cr (w/w)) diet for 8–9 weeks and tested in the Morris water maze. Mitochondrial oxygen consumption (Seahorse) and protein levels (Western blots) were measured in the hippocampus in subsets of mice. Overall, 3xTg females exhibited impaired memory as compared to males. In females, Cr supplementation decreased escape latency and was associated with increased spatial search strategy use. In males, Cr supplementation decreased the use of spatial search strategies. Pilot data indicated mitochondrial enhancements with Cr supplementation in both sexes. In females, Cr supplementation increased CREB phosphorylation and levels of IκB (NF-κB suppressor), CaMKII, PSD-95, and high-molecular-weight amyloid β (Aβ) species, whereas Aβ trimers were reduced. These data suggest a beneficial preventative effect of Cr supplementation in females and warrant caution against Cr supplementation in males in the AD-like brain.
Collapse
Affiliation(s)
- Wanda M. Snow
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Centre for the Advancement of Teaching and Learning, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Correspondence: or (W.M.S); (B.C.A.); Tel.: +1-204-235-3942 (B.C.A.)
| | - Chris Cadonic
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Claudia Cortes-Perez
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Aida Adlimoghaddam
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Subir K. Roy Chowdhury
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Research Institute in Oncology, CancerCare Manitoba, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Ella Thomson
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Adama Anozie
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Michael J. Bernstein
- Department of Psychological and Social Sciences, Pennsylvania State University Abington, Abington, PA 19001, USA;
| | - Kathleen Gough
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada;
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Miyoung Suh
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Benedict C. Albensi
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Correspondence: or (W.M.S); (B.C.A.); Tel.: +1-204-235-3942 (B.C.A.)
| |
Collapse
|
24
|
da Silva Mesquita R, Kyrylchuk A, Costa de Oliveira R, Costa Sá IS, Coutinho Borges Camargo G, Soares Pontes G, Moura Araújo da Silva F, Saraiva Nunomura RDC, Grafov A. Alkaloids of Abuta panurensis Eichler: In silico and in vitro study of acetylcholinesterase inhibition, cytotoxic and immunomodulatory activities. PLoS One 2020; 15:e0239364. [PMID: 32991579 PMCID: PMC7523975 DOI: 10.1371/journal.pone.0239364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
Natural products obtained from species of the genus Abuta (Menispermaceae) are known as ethnobotanicals that are attracting increasing attention due to a wide range of their pharmacological properties. In this study, the alkaloids stepharine and 5-N-methylmaytenine were first isolated from branches of Abuta panurensis Eichler, an endemic species from the Amazonian rainforest. Structure of the compounds was elucidated by a combination of 1D and 2D NMR spectroscopic and MS and HRMS spectrometric techniques. Interaction of the above-mentioned alkaloids with acetylcholinesterase enzyme and interleukins IL-6 and IL-8 was investigated in silico by molecular docking. The molecules under investigation were able to bind effectively with the active sites of the AChE enzyme, IL-6, and IL-8 showing affinity towards the proteins. Along with the theoretical study, acetylcholinesterase enzyme inhibition, cytotoxic, and immunomodulatory activity of the compounds were assessed by in vitro assays. The data obtained in silico corroborate the results of AChE enzyme inhibition, the IC50 values of 61.24μM for stepharine and 19.55μM for 5-N-methylmaytenine were found. The compounds showed cytotoxic activity against two tumor cell lines (K562 and U937) with IC50 values ranging from 11.77 μM to 28.48 μM. The in vitro assays revealed that both alkaloids were non-toxic to Vero and human PBMC cells. As for the immunomodulatory activity, both compounds inhibited the production of IL-6 at similar levels. Stepharine inhibited considerably the production of IL-8 in comparison to 5-N-methylmaytenine, which showed a dose dependent action (inhibitory at the IC50 dose, and stimulatory at the twofold IC50 one). Such a behavior may possibly be explained by different binding modes of the alkaloids to the interleukin structural fragments. Occurrence of the polyamine alkaloid 5-N-methylmaytenine was reported for the first time for the Menispermaceae family, as well as the presence of stepharine in A. panurensis.
Collapse
Affiliation(s)
| | - Andrii Kyrylchuk
- Institute of Organic Chemistry, National Academy of Sciences (NAS), Kyiv, Ukraine
| | - Regiane Costa de Oliveira
- Post-Graduate Program in Hematology, University of the State of Amazonas (UEA), Manaus, Amazonas, Brazil
| | | | | | - Gemilson Soares Pontes
- Post-Graduate Program in Hematology, University of the State of Amazonas (UEA), Manaus, Amazonas, Brazil
- Laboratory of Virology, National Institute of Amazonian Research (INPA), Manaus, Amazonas, Brazil
| | - Felipe Moura Araújo da Silva
- Analytical Center –Multidisciplinary Support Center (CAM), Federal University of Amazonas (UFAM), Manaus, Amazonas, Brazil
| | | | - Andriy Grafov
- Department of Chemistry, University of Helsinki, Helsinki, Finland
| |
Collapse
|
25
|
Aluminum-induced "mixed" cell death in mice cerebral tissue and potential intervention. Neurotox Res 2019; 37:835-846. [PMID: 31721047 DOI: 10.1007/s12640-019-00123-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/10/2019] [Accepted: 10/04/2019] [Indexed: 01/14/2023]
Abstract
The brain is one of organs vulnerable to aluminum insult. Aluminum toxicity is involved in neurobehavioral deficit, neuronal cell dysfunction, and death. The aim of this study are as follows: (1) to evaluate the repairing efficiency of Necrostatin-1 (Nec-1), a cell death inhibitor, and Z-VAD-FMK, a pan-caspase inhibitor, on Al-induced neurobehavioral deficit and neuronal cell death, in order to evidence the cell death inducing ability of aluminum, and (2) to primarily explore the possibility of treating neuronal cell loss-related disease, such as Alzheimer's disease, with Nec-1 and Z-VAD in Al-induced dementia animal model. We found Nec-1 and Z-VAD-FMK alone or in combination could reduce aluminum-induced learning and memory impairment in mice. Pathohistological results indicated that Nec-1 and Z-VAD-FMK can decrease Al-induced neuronal death cell. In addition, some cell death-associated proteins in cell death signal pathway were inhibited by Nec-1 and Z-VAD-FMK in Al-exposed mice. In conclusions, Nec-1 and Z-VAD-FMK can repair the injury of learning and memory induced by aluminum in mice. Furthermore, Nec-1 was more obvious to repair the injury of learning and memory function compared with Z-VAD-FMK. Nec-1 and Z-VAD-FMK can repair the Al-induced morphological injury of cell and reduce the amounts of dead cell, and repairing effects were more significant at higher doses. The effect of Nec-1 was stronger than Z-VAD-FMK, though their mechanism was different. The combination of them had the strongest effect. Our study evidenced Al-induced neuronal necroptosis and apoptosis existing in animal model and suggested potential therapeutic effects of Nec-1 and Z-VAD-FMK on neuronal cell death in neurodegenerative diseases.
Collapse
|
26
|
Liu N, Yu Z, Xun Y, Shu P, Yue Y, Yuan S, Jiang Y, Huang Z, Yang X, Feng X, Xiang S, Wang X. Amyloid-β25-35 Upregulates Endogenous Neuroprotectant Neuroglobin via NFκB Activation in vitro. J Alzheimers Dis 2019; 64:1163-1174. [PMID: 30010125 DOI: 10.3233/jad-180163] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Neuroglobin (Ngb) has been reported to be increased in early and moderately advanced Alzheimer's disease (AD) stages but declined in the severe stage. However, its regulatory mechanisms and pathophysiological roles in the disease remain to be defined. In this study, we found that Ngb expression was significantly upregulated by low dose Aβ25-35, the neurotoxic fragment of Aβ1 - 40 and Aβ1 - 42, but was not further increased by a higher dose of Aβ25-35. Mutation analysis and supershift assay demonstrated that transcription factor Nuclear Factor κB (NFκB), κB2 and κB3 sites located in mouse Ngb promoter region were involved in dynamic regulation of Ngb expression in response to different doses of Aβ25-35 stimulation. In addition, we found that suppression of endogenous Ngb expression exacerbated Aβ25-35-induced neuronal cell death and mitochondrial dysfunction. Our results indicate that endogenous Ngb expression may be upregulated by low dose Aβ25-35, which is responsible for protecting against Aβ25-35-mediated neurotoxicity. These experimental findings suggest that upregulation of endogenous Ngb expression might be an effective intervention approach for AD.
Collapse
Affiliation(s)
- Ning Liu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China.,Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhanyang Yu
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yu Xun
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Pan Shu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yiwei Yue
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, China.,Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shishan Yuan
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Yinghua Jiang
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Zixuan Huang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Xing Feng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Shuanglin Xiang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiaoying Wang
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Jha NK, Jha SK, Kar R, Nand P, Swati K, Goswami VK. Nuclear factor-kappa β as a therapeutic target for Alzheimer's disease. J Neurochem 2019; 150:113-137. [PMID: 30802950 DOI: 10.1111/jnc.14687] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/16/2019] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a typical progressive, chronic neurodegenerative disorder with worldwide prevalence. Its clinical manifestation involves the presence of extracellular plaques and intracellular neurofibrillary tangles (NFTs). NFTs occur in brain tissues as a result of both Aβ agglomeration and Tau phosphorylation. Although there is no known cure for AD, research into possible cures and treatment options continues using cell-cultures and model animals/organisms. The nuclear factor-kappa β (NF-κβ) plays an active role in the progression of AD. Impairment to this signaling module triggers undesirable phenotypic changes such as neuroinflammation, activation of microglia, oxidative stress related complications, and apoptotic cell death. These imbalances further lead to homeostatic abnormalities in the brain or in initial stages of AD essentially pushing normal neurons toward the degeneration process. Interestingly, the role of NF-κβ signaling associated receptor-interacting protein kinase is currently observed in apoptotic and necrotic cell death, and has been reported in brains. Conversely, the NF-κβ signaling pathway has also been reported to be involved in normal brain functioning. This pathway plays a crucial role in maintaining synaptic plasticity and balancing between learning and memory. Since any impairment in the pathways associated with NF-κβ signaling causes altered neuronal dynamics, neurotherapeutics using compounds including, antioxidants, bioflavonoids, and non-steroidal anti-inflammatory drugs against such abnormalities offer possibilities to rectify aberrant excitatory neuronal activity in AD. In this review, we have provided an extensive overview of the crucial role of NF-κβ signaling in normal brain homeostasis. We have also thoroughly outlined several established pathomechanisms associated with NF-κβ pathways in AD, along with their respective therapeutic approaches.
Collapse
Affiliation(s)
- Niraj Kumar Jha
- Department of Biotechnology, Noida Institute of Engineering & Technology (NIET), Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Rohan Kar
- Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Parma Nand
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Kumari Swati
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Vineet Kumar Goswami
- Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| |
Collapse
|
28
|
Specific alterations in the circulating levels of the SIRT1, TLR4, and IL7 proteins in patients with dementia. Exp Gerontol 2018; 111:203-209. [DOI: 10.1016/j.exger.2018.07.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/20/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022]
|
29
|
Chen Z, Xiang Y, Bao B, Wu X, Xia Z, You J, Nie H. Simvastatin improves cerebrovascular injury caused by ischemia‑reperfusion through NF‑κB‑mediated apoptosis via MyD88/TRIF signaling. Mol Med Rep 2018; 18:3177-3184. [PMID: 30066928 PMCID: PMC6102662 DOI: 10.3892/mmr.2018.9337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 05/02/2018] [Indexed: 01/26/2023] Open
Abstract
Cerebrovascular injury is the most prevalent human cerebrovascular disease and frequently results in ischemic stroke. Simvastatin may be a potential therapeutic agent for the treatment of patients with cerebrovascular injury. The present study aimed to investigate the efficacy of and the potential mechanisms regulated by simvastatin in a rat model of ischemia-reperfusion (I/R)-induced cerebrovascular injury. Cerebrovascular injury model rats were established and were subsequently treated with simvastatin or a vehicle control following I/R injury. Cell damage, neurological functions and neuronal apoptosis were examined, as well as the nuclear factor (NF)-κB-mediated myeloid differentiation primary response protein 88 (MyD88)/toll-interleukin-1 receptor domain-containing adapter molecule 1 (TRIF) signaling pathway following simvastatin treatment. The results of the present study demonstrated that simvastatin treatment led to a reduction in cell damage, improvement of neurological functions and decreased neuronal apoptosis compared with vehicle-treated I/R model rats, 14 days post-treatment. In addition, simvastatin treatment reduced cerebral water content and blood-brain barrier disruption in cerebrovascular injury induced by I/R. The results also revealed that simvastatin treatment inhibited neuronal apoptosis via the NF-κB-mediated MyD88/TRIF signaling pathway. In conclusion, simvastatin treatment may reduce I/R-induced neuronal apoptosis via inhibition of the NF-κB-mediated MyD88/TRIF signaling pathway.
Collapse
Affiliation(s)
- Zhiying Chen
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Yuanyuan Xiang
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Bing Bao
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Xiangbin Wu
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Zhongbin Xia
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Jianyou You
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Hongbing Nie
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| |
Collapse
|
30
|
Hadem IKH, Majaw T, Kharbuli B, Sharma R. Beneficial effects of dietary restriction in aging brain. J Chem Neuroanat 2017; 95:123-133. [PMID: 29031555 DOI: 10.1016/j.jchemneu.2017.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/14/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022]
Abstract
Aging is a multifactorial complex process that leads to the deterioration of biological functions wherein its underlying mechanism is not fully elucidated. It affects the organism at the molecular and cellular level that contributes to the deterioration of structural integrity of the organs. The central nervous system is the most vulnerable organ affected by aging and its effect is highly heterogeneous. Aging causes alteration in the structure, metabolism and physiology of the brain leading to impaired cognitive and motor-neural functions. Dietary restriction (DR), a robust mechanism that extends lifespan in various organisms, ameliorates brain aging by reducing oxidative stress, improving mitochondrial function, activating anti-inflammatory responses, promoting neurogenesis and increasing synaptic plasticity. It also protects and prevents age-related structural changes. DR alleviates many age-associated diseases including neurodegeneration and improves cognitive functions. DR inhibits/activates nutrient signaling cascades such as insulin/IGF-1, mTOR, AMPK and sirtuins. Because of its sensitivity to energy status and hormones, AMPK is considered as the global nutrient sensor. This review will present an elucidative potential role of dietary restriction in the prevention of phenotypic features during aging in brain and its diverse mechanisms.
Collapse
Affiliation(s)
| | - Teikur Majaw
- Department of Biochemistry, North-Eastern Hill University, Shillong 793022, Meghalaya, India
| | - Babiangshisha Kharbuli
- Department of Biochemistry, North-Eastern Hill University, Shillong 793022, Meghalaya, India
| | - Ramesh Sharma
- Department of Biochemistry, North-Eastern Hill University, Shillong 793022, Meghalaya, India.
| |
Collapse
|
31
|
Sakamoto K, Okuwaki T, Ushikubo H, Mori A, Nakahara T, Ishii K. Activation inhibitors of nuclear factor kappa B protect neurons against the NMDA-induced damage in the rat retina. J Pharmacol Sci 2017; 135:S1347-8613(17)30162-7. [PMID: 29110956 DOI: 10.1016/j.jphs.2017.09.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/13/2017] [Indexed: 01/08/2023] Open
Abstract
We reported that high-mobility group Box-1 (HMGB1) was involved in excitoneurotoxicity in the retina. HMGB1 is known to activate nuclear factor kappa B (NF-κB). However, the role of NF-κB in excitotoxicity is still controversial. Here, we demonstrated that NF-κB activation induced by NMDA led to the retinal neurotoxicity. Male Sprague-Dawley rats were used, and NMDA (200 nmol/eye) and bovine HMGB1 (15 μg/eye) were intravitreally injected. Triptolide (500 pmol/eye), BAY 11-7082 (500 pmol/eye), and IMD-0354 (7.5 nmol/eye), NF-κB inhibitors, were co-injected with NMDA or HMGB1. Retinal sections were obtained seven days after intravitreal injection. Cell loss in the ganglion cell layer was observed in the HMGB1- and the NMDA-treated retina. All of the NF-κB inhibitors used in this study reduced the damage. BAY 11-7082 reduced the expression of phosphorylated NF-κB 12 h after NMDA injection, upregulation of GFAP immunoreactivity induced by NMDA 12 and 48 h after NMDA injection, and the number of TUNEL-positive cells 48 h after NMDA injection. The results suggest that NF-κB activation is one of the mechanisms of the retinal neuronal death that occurs 48 h after NMDA injection or later. Prevention of NF-kB activation is a candidate for the treatment of retinal neurodegeneration associated with excitotoxicity.
Collapse
Affiliation(s)
- Kenji Sakamoto
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan.
| | - Tatsuya Okuwaki
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Hiroko Ushikubo
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Kunio Ishii
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| |
Collapse
|
32
|
Uchida S, Shumyatsky GP. Synaptically Localized Transcriptional Regulators in Memory Formation. Neuroscience 2017; 370:4-13. [PMID: 28733211 DOI: 10.1016/j.neuroscience.2017.07.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 01/07/2023]
Abstract
At the neuronal cell level, long-term memory formation emerges from interactions between initial activity-dependent molecular changes at the synapse and subsequent regulation of gene transcription in the nucleus. This in turn leads to strengthening of the connections back at the synapse that received the initial signal. However, the mechanisms through which this synapse-to-nucleus molecular exchange occurs remain poorly understood. Here we discuss recent studies that delineate nucleocytoplasmic transport of a special class of synaptically localized transcriptional regulators that upon receiving initial external signal by the synapse move to the nucleus to modulate gene transcription.
Collapse
Affiliation(s)
- Shusaku Uchida
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Gleb P Shumyatsky
- Department of Genetics, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA.
| |
Collapse
|
33
|
Chen YC. Impact of a discordant helix on β-amyloid structure, aggregation ability and toxicity. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:681-687. [DOI: 10.1007/s00249-017-1235-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/26/2017] [Accepted: 06/26/2017] [Indexed: 11/24/2022]
|
34
|
Transcriptional regulation of RACK1 and modulation of its expression: Role of steroid hormones and significance in health and aging. Cell Signal 2017; 35:264-271. [DOI: 10.1016/j.cellsig.2017.02.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/07/2017] [Accepted: 02/08/2017] [Indexed: 12/27/2022]
|
35
|
Amanullah A, Upadhyay A, Chhangani D, Joshi V, Mishra R, Yamanaka K, Mishra A. Proteasomal Dysfunction Induced By Diclofenac Engenders Apoptosis Through Mitochondrial Pathway. J Cell Biochem 2017; 118:1014-1027. [DOI: 10.1002/jcb.25666] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/01/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Ayeman Amanullah
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Deepak Chhangani
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Vibhuti Joshi
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Ribhav Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology Research Institute of Environmental Medicine; Nagoya University Furo-cho; Chikusa-ku Nagoya 464-8601 Japan
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| |
Collapse
|
36
|
Snow WM, Albensi BC. Neuronal Gene Targets of NF-κB and Their Dysregulation in Alzheimer's Disease. Front Mol Neurosci 2016; 9:118. [PMID: 27881951 PMCID: PMC5101203 DOI: 10.3389/fnmol.2016.00118] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/25/2016] [Indexed: 11/21/2022] Open
Abstract
Although, better known for its role in inflammation, the transcription factor nuclear factor kappa B (NF-κB) has more recently been implicated in synaptic plasticity, learning, and memory. This has been, in part, to the discovery of its localization not just in glia, cells that are integral to mediating the inflammatory process in the brain, but also neurons. Several effectors of neuronal NF-κB have been identified, including calcium, inflammatory cytokines (i.e., tumor necrosis factor alpha), and the induction of experimental paradigms thought to reflect learning and memory at the cellular level (i.e., long-term potentiation). NF-κB is also activated after learning and memory formation in vivo. In turn, activation of NF-κB can elicit either suppression or activation of other genes. Studies are only beginning to elucidate the multitude of neuronal gene targets of NF-κB in the normal brain, but research to date has confirmed targets involved in a wide array of cellular processes, including cell signaling and growth, neurotransmission, redox signaling, and gene regulation. Further, several lines of research confirm dysregulation of NF-κB in Alzheimer's disease (AD), a disorder characterized clinically by a profound deficit in the ability to form new memories. AD-related neuropathology includes the characteristic amyloid beta plaque formation and neurofibrillary tangles. Although, such neuropathological findings have been hypothesized to contribute to memory deficits in AD, research has identified perturbations at the cellular and synaptic level that occur even prior to more gross pathologies, including transcriptional dysregulation. Indeed, synaptic disturbances appear to be a significant correlate of cognitive deficits in AD. Given the more recently identified role for NF-κB in memory and synaptic transmission in the normal brain, the expansive network of gene targets of NF-κB, and its dysregulation in AD, a thorough understanding of NF-κB-related signaling in AD is warranted and may have important implications for uncovering treatments for the disease. This review aims to provide a comprehensive view of our current understanding of the gene targets of this transcription factor in neurons in the intact brain and provide an overview of studies investigating NF-κB signaling, including its downstream targets, in the AD brain as a means of uncovering the basic physiological mechanisms by which memory becomes fragile in the disease.
Collapse
Affiliation(s)
- Wanda M Snow
- Division of Neurodegenerative Disorders, St. Boniface Hospital ResearchWinnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of ManitobaWinnipeg, MB, Canada
| | - Benedict C Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital ResearchWinnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of ManitobaWinnipeg, MB, Canada
| |
Collapse
|
37
|
Dvoriantchikova G, Pappas S, Luo X, Ribeiro M, Danek D, Pelaez D, Park KK, Ivanov D. Virally delivered, constitutively active NFκB improves survival of injured retinal ganglion cells. Eur J Neurosci 2016; 44:2935-2943. [PMID: 27564592 DOI: 10.1111/ejn.13383] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 08/21/2016] [Accepted: 08/23/2016] [Indexed: 12/17/2022]
Abstract
As axon damage and retinal ganglion cell (RGC) loss lead to blindness, therapies that increase RGC survival and axon regrowth have direct clinical relevance. Given that NFκB signaling is critical for neuronal survival and may regulate neurite growth, we investigated the therapeutic potential of NFκB signaling in RGC survival and axon regeneration. Although both NFκB subunits (p65 and p50) are present in RGCs, p65 exists in an inactive (unphosphorylated) state when RGCs are subjected to neurotoxic conditions. In this study, we used a phosphomimetic approach to generate DNA coding for an activated (phosphorylated) p65 (p65mut), then employed an adeno-associated virus serotype 2 (AAV2) to deliver the DNA into RGCs. We tested whether constitutive p65mut expression prevents death and facilitates neurite outgrowth in RGCs subjected to transient retinal ischemia or optic nerve crush (ONC), two models of neurotoxicity. Our data indicate that RGCs treated with AAV2-p65mut displayed a significant increase in survival compared to controls in ONC model (77 ± 7% vs. 25 ± 3%, P-value = 0.0001). We also found protective effect of modified p65 in RGCs of ischemic retinas (55 ± 12% vs. 35 ± 6%), but not to a statistically significant degree (P-value = 0.14). We did not detect a difference in axon regeneration between experimental and control animals after ONC. These findings suggest that increased NFκB signaling in RGCs attenuates retinal damage in animal models of neurodegeneration, but insignificantly impacts axon regeneration.
Collapse
Affiliation(s)
- Galina Dvoriantchikova
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Steve Pappas
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Xueting Luo
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Marcio Ribeiro
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dagmara Danek
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Daniel Pelaez
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Kevin K Park
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dmitry Ivanov
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.,Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
38
|
Abdel-Aziz AK, Mantawy EM, Said RS, Helwa R. The tyrosine kinase inhibitor, sunitinib malate, induces cognitive impairment in vivo via dysregulating VEGFR signaling, apoptotic and autophagic machineries. Exp Neurol 2016; 283:129-41. [PMID: 27288242 DOI: 10.1016/j.expneurol.2016.06.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 05/14/2016] [Accepted: 06/06/2016] [Indexed: 01/29/2023]
Abstract
Chemobrain refers to a cluster of cognitive deficits which affects almost 4-75% of chemotherapy-treated cancer patients. Sunitinib, an FDA-approved multityrosine kinase inhibitor, is currently used in treating different types of tumors. Despite being regarded as targeted therapy which blunts sustained angiogenesis in cancer milieu through inhibiting vascular endothelial growth factor receptor 2 (VEGFR2) signaling, the latter has a cardinal role in cognition. Recent clinical reports warned that sunitinib adversely affected memory processing in cancer patients. Nevertheless, the underlying mechanisms have not been investigated yet. Hence, we explored the impact of a clinically relevant dose of sunitinib on memory processing in vivo and questioned the implication of VEGFR2 signaling, autophagy and apoptosis. Strikingly, sunitinib preferentially impaired spatial cognition as evidenced in Morris water maze, T-maze and passive avoidance task. Consistently, sunitinib degenerated cortical and hippocampal neurons as assessed by histopathological examination and toluidine blue staining. Ultrastructural examination also depicted chromatin condensation, mitochondrial damage and accumulated autophagosomes. Digging deeper, central VEGF/VEGFR2/mTOR signaling was robustly suppressed. Besides, sunitinib boosted cortical and hippocampal p53 and executioner caspase-3 and decreased nuclear factor kappa B and Bcl-2 levels promoting apoptotic cell death. It also profoundly impeded neuronal autophagic flux as shown by decreased beclin-1 and Atg5 and increased p62/SQTSM1 levels. To our knowledge, this is the first study to provide molecular insights into sunitinib-induced chemofog where impeded VEGFR2 signaling and autophagic and hyperactivated apoptotic machineries act in neurodegenerative concert. Importantly, our findings shed light on potential therapeutic strategies to be exploited in the management of sunitinib-induced chemobrain.
Collapse
Affiliation(s)
- Amal Kamal Abdel-Aziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Riham Soliman Said
- National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Reham Helwa
- Zoology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
39
|
RAGE Expression and ROS Generation in Neurons: Differentiation versus Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9348651. [PMID: 27313835 PMCID: PMC4897723 DOI: 10.1155/2016/9348651] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/03/2016] [Indexed: 12/27/2022]
Abstract
RAGE is a multiligand receptor able to bind advanced glycation end-products (AGEs), amphoterin, calgranulins, and amyloid-beta peptides, identified in many tissues and cells, including neurons. RAGE stimulation induces the generation of reactive oxygen species (ROS) mainly through the activity of NADPH oxidases. In neuronal cells, RAGE-induced ROS generation is able to favor cell survival and differentiation or to induce death through the imbalance of redox state. The dual nature of RAGE signaling in neurons depends not only on the intensity of RAGE activation but also on the ability of RAGE-bearing cells to adapt to ROS generation. In this review we highlight these aspects of RAGE signaling regulation in neuronal cells.
Collapse
|
40
|
Dietary phytochemicals and neuro-inflammaging: from mechanistic insights to translational challenges. IMMUNITY & AGEING 2016; 13:16. [PMID: 27081392 PMCID: PMC4831196 DOI: 10.1186/s12979-016-0070-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/30/2016] [Indexed: 12/20/2022]
Abstract
An extensive literature describes the positive impact of dietary phytochemicals on overall health and longevity. Dietary phytochemicals include a large group of non-nutrients compounds from a wide range of plant-derived foods and chemical classes. Over the last decade, remarkable progress has been made to realize that oxidative and nitrosative stress (O&NS) and chronic, low-grade inflammation are major risk factors underlying brain aging. Accumulated data strongly suggest that phytochemicals from fruits, vegetables, herbs, and spices may exert relevant negative immunoregulatory, and/or anti-O&NS activities in the context of brain aging. Despite the translational gap between basic and clinical research, the current understanding of the molecular interactions between phytochemicals and immune-inflammatory and O&NS (IO&NS) pathways could help in designing effective nutritional strategies to delay brain aging and improve cognitive function. This review attempts to summarise recent evidence indicating that specific phytochemicals may act as positive modulators of IO&NS pathways by attenuating pro-inflammatory pathways associated with the age-related redox imbalance that occurs in brain aging. We will also discuss the need to initiate long-term nutrition intervention studies in healthy subjects. Hence, we will highlight crucial aspects that require further study to determine effective physiological concentrations and explore the real impact of dietary phytochemicals in preserving brain health before the onset of symptoms leading to cognitive decline and inflammatory neurodegeneration.
Collapse
|
41
|
Xing C, Lo EH. Help-me signaling: Non-cell autonomous mechanisms of neuroprotection and neurorecovery. Prog Neurobiol 2016; 152:181-199. [PMID: 27079786 DOI: 10.1016/j.pneurobio.2016.04.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 04/06/2016] [Accepted: 04/09/2016] [Indexed: 12/11/2022]
Abstract
Self-preservation is required for life. At the cellular level, this fundamental principle is expressed in the form of molecular mechanisms for preconditioning and tolerance. When the cell is threatened, internal cascades of survival signaling become triggered to protect against cell death and defend against future insults. Recently, however, emerging findings suggest that this principle of self-preservation may involve not only intracellular signals; the release of extracellular signals may provide a way to recruit adjacent cells into an amplified protective program. In the central nervous system where multiple cell types co-exist, this mechanism would allow threatened neurons to "ask for help" from glial and vascular compartments. In this review, we describe this new concept of help-me signaling, wherein damaged or diseased neurons release signals that may shift glial and vascular cells into potentially beneficial phenotypes, and help remodel the neurovascular unit. Understanding and dissecting these non-cell autonomous mechanisms of self-preservation in the CNS may lead to novel opportunities for neuroprotection and neurorecovery.
Collapse
Affiliation(s)
- Changhong Xing
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| | - Eng H Lo
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
42
|
Kaltschmidt B, Kaltschmidt C. NF-KappaB in Long-Term Memory and Structural Plasticity in the Adult Mammalian Brain. Front Mol Neurosci 2015; 8:69. [PMID: 26635522 PMCID: PMC4656838 DOI: 10.3389/fnmol.2015.00069] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/30/2015] [Indexed: 11/13/2022] Open
Abstract
The transcription factor nuclear factor kappaB (NF-κB) is a well-known regulator of inflammation, stress, and immune responses as well as cell survival. In the nervous system, NF-κB is one of the crucial components in the molecular switch that converts short- to long-term memory-a process that requires de novo gene expression. Here, the researches published on NF-κB and downstream target genes in mammals will be reviewed, which are necessary for structural plasticity and long-term memory, both under normal and pathological conditions in the brain. Genetic evidence has revealed that NF-κB regulates neuroprotection, neuronal transmission, and long-term memory. In addition, after genetic ablation of all NF-κB subunits, a severe defect in hippocampal adult neurogenesis was observed during aging. Proliferation of neural precursors is increased; however, axon outgrowth, synaptogenesis, and tissue homeostasis of the dentate gyrus are hampered. In this process, the NF-κB target gene PKAcat and other downstream target genes such as Igf2 are critically involved. Therefore, NF-κB activity seems to be crucial in regulating structural plasticity and replenishment of granule cells within the hippocampus throughout the life. In addition to the function of NF-κB in neurons, we will discuss on a neuroinflammatory role of the transcription factor in glia. Finally, a model for NF-κB homeostasis on the molecular level is presented, in order to explain seemingly the contradictory, the friend or foe, role of NF-κB in the nervous system.
Collapse
|
43
|
Martire S, Mosca L, d'Erme M. PARP-1 involvement in neurodegeneration: A focus on Alzheimer's and Parkinson's diseases. Mech Ageing Dev 2015; 146-148:53-64. [PMID: 25881554 DOI: 10.1016/j.mad.2015.04.001] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/26/2015] [Accepted: 04/06/2015] [Indexed: 12/17/2022]
Abstract
DNA damage is the prime activator of the enzyme poly(ADP-ribose)polymerase1 (PARP-1) whose overactivation has been proven to be associated with the pathogenesis of numerous central nervous system disorders, such as ischemia, neuroinflammation, and neurodegenerative diseases. Under oxidative stress conditions PARP-1 activity increases, leading to an accumulation of ADP-ribose polymers and NAD(+) depletion, that induces energy crisis and finally cell death. This review aims to explain the contribution of PARP-1 in neurodegenerative diseases, focusing on Alzheimer's and Parkinson's disease, to stimulate further studies on this issue and thereby engage a new perspective regarding the design of possible therapeutic agents or the identification of biomarkers.
Collapse
Affiliation(s)
- Sara Martire
- Department of Biochemical Sciences, Sapienza University of Roma, Italy
| | - Luciana Mosca
- Department of Biochemical Sciences, Sapienza University of Roma, Italy
| | - Maria d'Erme
- Department of Biochemical Sciences, Sapienza University of Roma, Italy.
| |
Collapse
|
44
|
Minter MR, Main BS, Brody KM, Zhang M, Taylor JM, Crack PJ. Soluble amyloid triggers a myeloid differentiation factor 88 and interferon regulatory factor 7 dependent neuronal type-1 interferon response in vitro. J Neuroinflammation 2015; 12:71. [PMID: 25879763 PMCID: PMC4407532 DOI: 10.1186/s12974-015-0263-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/09/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Neuro-inflammation has long been implicated as a contributor to the progression of Alzheimer's disease in both humans and animal models. Type-1 interferons (IFNs) are pleiotropic cytokines critical in mediating the innate immune pro-inflammatory response. The production of type-1 IFNs following pathogen detection is, in part, through the activation of the toll-like receptors (TLRs) and subsequent signalling through myeloid differentiation factor-88 (Myd88) and interferon regulatory factors (IRFs). We have previously identified that neuronal type-1 IFN signalling, through the type-1 interferon alpha receptor-1 (IFNAR1), is detrimental in models of AD. Using an in vitro approach, this study investigated the TLR network as a potential production pathway for neuronal type-1 IFNs in response to Aβ. METHODS Wildtype and Myd88(-/-) primary cultured cortical and hippocampal neurons were treated with 2.5 μM Aβ1-42 for 24 to 72 h or 1 to 10 μM Aβ1-42 for 72 h. Human BE(2)M17 neuroblastoma cells stably expressing an IRF7 small hairpin RNA (shRNA) or negative control shRNA construct were subjected to 7.5 μM Aβ1-42/Aβ42-1 for 24 to 96 h, 2.5 to 15 μM Aβ1-42 for 96 h or 100 ng/ml LPS for 0.5 to 24 h. Q-PCR was used to analyse IFNα, IFNβ, IL-1β, IL-6 and TNFα mRNA transcript levels. Phosphorylation of STAT-3 was detected by Western blot analysis, and cell viability was assessed by MTS assay. RESULTS Reduced IFNα, IFNβ, IL-1β, IL-6 and TNFα expression was detected in Aβ1-42-treated Myd88(-/-) neurons compared to wildtype cells. This correlated with reduced phosphorylation of STAT-3, a downstream type-1 IFN signalling mediator. Significantly, Myd88(-/-) neuronal cultures were protected against Aβ1-42-induced neurotoxicity compared to wildtype as determined by MTS assay. Knockdown of IRF7 in M17 cells was sufficient in blocking IFNα, IFNβ and p-STAT-3 induction to both Aβ1-42 and the TLR4 agonist LPS. M17 IRF7 KD cells were also protected against Aβ1-42-induced cytotoxicity. CONCLUSIONS This study confirms that the neuronal type-1 IFN response to soluble amyloid is mediated primarily through TLRs. This production is dependent upon Myd88 and IRF7 signalling. This study suggests that targeting this pathway to modulate neuronal type-1 IFN levels may be beneficial in controlling Aβ-induced neurotoxicity.
Collapse
Affiliation(s)
- Myles Robert Minter
- Department of Pharmacology and Therapeutics, University of Melbourne, 8th floor, Medical building, Grattan St, Parkville, Melbourne, 3010, VIC, Australia.
| | - Bevan Scott Main
- Department of Pharmacology and Therapeutics, University of Melbourne, 8th floor, Medical building, Grattan St, Parkville, Melbourne, 3010, VIC, Australia.
| | - Kate Maree Brody
- Department of Pharmacology and Therapeutics, University of Melbourne, 8th floor, Medical building, Grattan St, Parkville, Melbourne, 3010, VIC, Australia.
| | - Moses Zhang
- Department of Pharmacology and Therapeutics, University of Melbourne, 8th floor, Medical building, Grattan St, Parkville, Melbourne, 3010, VIC, Australia.
| | - Juliet Marie Taylor
- Department of Pharmacology and Therapeutics, University of Melbourne, 8th floor, Medical building, Grattan St, Parkville, Melbourne, 3010, VIC, Australia.
| | - Peter John Crack
- Department of Pharmacology and Therapeutics, University of Melbourne, 8th floor, Medical building, Grattan St, Parkville, Melbourne, 3010, VIC, Australia.
| |
Collapse
|
45
|
Arepalli SK, Choi M, Jung JK, Lee H. Novel NF-κB inhibitors: a patent review (2011 – 2014). Expert Opin Ther Pat 2015; 25:319-34. [DOI: 10.1517/13543776.2014.998199] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
46
|
Marwarha G, Raza S, Meiers C, Ghribi O. Leptin attenuates BACE1 expression and amyloid-β genesis via the activation of SIRT1 signaling pathway. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:1587-95. [PMID: 24874077 PMCID: PMC4125440 DOI: 10.1016/j.bbadis.2014.05.015] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/13/2014] [Accepted: 05/16/2014] [Indexed: 02/07/2023]
Abstract
The aspartyl protease β-site AβPP-cleaving enzyme 1 (BACE1) catalyzes the rate-limiting step in Aβ production, a peptide at the nexus of neurodegenerative cascades in Alzheimer Disease (AD). The adipocytokine leptin has been demonstrated to reduce Aβ production and decrease BACE1 activity and expression levels. However, the signaling cascades involved in the leptin-induced mitigation in Aβ levels and BACE1 expression levels have not been elucidated. We have demonstrated that the transcription factor nuclear factor - kappa B (NF-κB) positively regulates BACE1 transcription. NF-κB activity is tightly regulated by the mammalian sirtuin SIRT1. Multiple studies have cogently evinced that leptin activates the metabolic master regulator SIRT1. In this study, we determined the extent to which SIRT1 expression and activity regulate the leptin-induced attenuation in BACE1 expression and Aβ levels in cultured human neuroblastoma SH-SY5Y cells. This study also elucidated and delineated the signal transduction pathways involved in the leptin induced mitigation in BACE1 expression. Our results demonstrate for the first time that leptin attenuates the activation and transcriptional activity of NF-κB by reducing the acetylation of the p65 subunit in a SIRT1-dependent manner. Furthermore, our data shows that leptin reduces the NF-κB-mediated transcription of BACE1 and consequently reduces Amyloid-β genesis. Our study provides a valuable insight and a novel mechanism by which leptin reduces BACE1 expression and Amyloid-β production and may help design potential therapeutic interventions.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Shaneabbas Raza
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Craig Meiers
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Othman Ghribi
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, USA.
| |
Collapse
|
47
|
Inhibiting IκB kinase-β downregulates inflammatory cytokines in injured discs and neuropeptides in dorsal root ganglia innervating injured discs in rats. Spine (Phila Pa 1976) 2014; 39:1171-7. [PMID: 24825147 DOI: 10.1097/brs.0000000000000374] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Quantitative and immunohistological analysis of the efficacy of an IκB kinase-β (IKKβ) inhibitor in an injured intervertebral disc (IVD) model. OBJECTIVE To elucidate the efficacy of an IKKβ inhibitor on inflammatory cytokine levels in injured IVDs or on neuropeptide levels in the dorsal root ganglia (DRG) neurons innervating injured IVDs in rats. SUMMARY OF BACKGROUND DATA Multiple studies have suggested that upregulation of inflammatory cytokines in damaged IVDs causes discogenic low back pain. The efficacy of blocking individual inflammatory cytokines is limited; however, inflammatory cytokine stimuli often require IKKβ to activate nuclear factor-k B. METHODS Sprague-Dawley rats were divided into 3 groups: sham, saline (disc-injury plus saline), and IKKβ (disc-injury plus anti-IKKβ). To induce injury, IVDs were repeatedly punctured.Experiment 1: Four, 7, and 14 days postinjury, coccygeal (Co) 5/6, Co6/7, and Co7/8 IVDs were resected and tumor necrosis factor-α, interleukin (IL)-1β, and IL-6 levels were quantified by enzyme-linked immunosorbent assay. Experiment 2: The neurotracer Fluoro-Gold was injected into injured L5-L6 IVDs and uninjured sham group IVDs to detect DRG neurons. One week postsurgery, L1-L6 DRGs were immunolabeled with the neuropeptide calcitonin gene-related peptide. The proportions of Fluoro-Gold-labeled calcitonin gene-related peptide-immunoreactive DRG neurons were assessed. RESULTS Experiment 1: IVD levels of tumor necrosis factor-α (through 2 wk), IL-1β (at 4 d), and IL-6 (at 4 d) were significantly higher in the saline group than in the sham group, and significantly lower in the IKKβ group than in the saline group (P < 0.05). Experiment 2: The percentage of calcitonin gene-related peptide-immunoreactive Fluoro-Gold-labeled DRG neurons was significantly higher in the saline group than in the sham group, and significantly lower in the IKKβ group than in the saline group (P < 0.05). CONCLUSION Injury-induced upregulation of inflammatory cytokines within IVDs and increased levels of neuropeptides within DRG neurons can be suppressed by inhibiting IKKβ. LEVEL OF EVIDENCE N/A.
Collapse
|
48
|
Complex regulation of acute and chronic neuroinflammatory responses in mouse models deficient for nuclear factor kappa B p50 subunit. Neurobiol Dis 2014; 64:16-29. [DOI: 10.1016/j.nbd.2013.12.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 11/11/2013] [Accepted: 12/04/2013] [Indexed: 12/29/2022] Open
|
49
|
Chen LJ, Sun BH, Qu JP, Xu S, Li S. Avermectin induced inflammation damage in king pigeon brain. CHEMOSPHERE 2013; 93:2528-2534. [PMID: 24134892 DOI: 10.1016/j.chemosphere.2013.09.058] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 09/03/2013] [Accepted: 09/10/2013] [Indexed: 06/02/2023]
Abstract
To determine the effect of Avermectin (AVM) on inflammation damage in king pigeon brain, eighty two-month-old American king pigeons were randomly divided into four groups, and were fed with either commercial diet or AVM-supplemented diet containing 20 mg kg(-1)diet, 40 mg kg(-1)diet, and 60 mg kg(-1)diet AVM for 30, 60 and 90 d, respectively. Then, the expression level of inflammatory factors (iNOS, PTGEs, NF-κB), histological damage, and ultra-structural damage were examined. It showed that AVM caused higher expressions (P<0.05) of iNOS, PTGEs, NF-κB with disorganized histological and ultra-structural structures in cerebrum, cerebellum, and optic lobe. Meanwhile, inflammatory and histopathological damage were induced by AVM in king pigeon brains. In addition, the main targeted organelle in nervous system was mitochondria, which indicated that mitochondria may be relevant to the process of inflammation induced by AVM. To our best knowledge, this is the first report to study the toxic effect of AVM on inflammatory damage in king pigeon. Thus, the information presented in this study is believed to be helpful in supplementing data for further AVM toxicity study.
Collapse
Affiliation(s)
- Li-Jie Chen
- Department of Veterinary Physiology, Northeast Agricultural University, Harbin 150030, PR China; Department of Neurology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, PR China
| | | | | | | | | |
Collapse
|
50
|
Stimulation of central β2-adrenoceptors suppresses NFκB activity in rat brain: A role for IκB. Neurochem Int 2013; 63:368-78. [DOI: 10.1016/j.neuint.2013.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 07/08/2013] [Accepted: 07/20/2013] [Indexed: 11/19/2022]
|