1
|
Qin Q, Xia X, Qu J, Guan Z, Yin Y, Chang J, Yu C, Zhang T, Tang Y. Blood biomarkers of amyloid and tau pathologies, brain degeneration, inflammation, and oxidative stress in early- and late-onset Alzheimer's disease. J Alzheimers Dis 2025:13872877251340955. [PMID: 40336292 DOI: 10.1177/13872877251340955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
BackgroundNumerous blood biomarkers have emerged as promising biomarkers for Alzheimer's disease (AD) and cognitive decline, but limited knowledge exists concerning the difference of blood biomarkers between early-onset and late-onset cases.ObjectiveInvestigate blood biomarkers associated with amyloid and tau pathologies, brain degeneration, inflammation, and oxidative stress in individuals afflicted with both early-onset and late-onset AD, as well as in age-matched healthy controls.MethodsA total of 125 participants were enrolled. We assessed levels of 18 distinct blood biomarkers and their associations with cerebrospinal fluid biomarkers, neuropsychological test scores, APOE ε4 carrier status, and neuroimaging markers. The diagnostic potential of blood biomarkers was investigated.ResultsIn early-onset AD patients, levels of blood Interleukin (IL)-4, IL-6, and Tumor necrosis factor-alpha (TNF-α) were notably lower comparing to late-onset patients. AD patients exhibited higher blood levels of phosphorylated-tau181 (p-tau181), neurofilament light chain (NfL), and glial fibrillary acidic protein (GFAP), as well as lower levels of amyloid-β (Aβ)42 and IL-12p70. Oxidative stress markers, including malondialdehyde, total antioxidant capacity, and superoxide dismutase, exhibited a progressive trend across the continuum of AD. Inflammatory markers demonstrating correlations with neuroimaging markers. Blood levels of Aβ42, p-tau181, NfL, and GFAP associated with neuropsychological scores and effectively discriminated AD, with GFAP exhibiting particular relevance in early-onset cases.ConclusionsInflammatory markers exhibited differences between patients with early- and late-onset AD, associated with alterations in brain structure and function. With the progression of disease continuum, a decrement in antioxidant capacity was observed. Blood Aβ42, p-tau181, NfL, and GFAP showed promise in detecting cognitive decline and AD.
Collapse
Affiliation(s)
- Qi Qin
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xinyi Xia
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Junda Qu
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Zhongtian Guan
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Yunsi Yin
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Chang
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chaoji Yu
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tongtong Zhang
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yi Tang
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
- Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing, China
| |
Collapse
|
2
|
Badman J, Parracino A, Kumar R, Tambaro S. Insights into the intramembrane protease SPPL2b and its substrates: Functions and disease implications. Sci Signal 2025; 18:eadt2272. [PMID: 40327750 DOI: 10.1126/scisignal.adt2272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/21/2025] [Indexed: 05/08/2025]
Abstract
Specialized intramembrane proteases, known as iCLiPs, regulate the processing of transmembrane proteins by releasing intracellular domains, which can function as transcriptional regulators. The signal peptide peptidase-like (SPPL) family of iCLiPs, particularly SPPL2b, has roles in immune regulation, neuronal function, and disease pathogenesis. In the brain, SPPL2b localizes mainly in the plasma membrane of neurons and microglia and is abundant in the cortex and hippocampus. Its known substrates regulate neuronal growth, inflammation, and synaptic function, and increased amounts of SPPL2b have been found in postmortem brain tissue from patients with Alzheimer's disease. In this review, we discuss the currently known roles of SPPL2b, its substrates, and its disease implications. Understanding the downstream effects of SPPL2b-cleaved substrates will provide clearer insights into the impact of SPPL2b on cellular homeostasis and disease, potentially leading to new therapeutic strategies.
Collapse
Affiliation(s)
- Jack Badman
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna 171 64, Sweden
| | - Antonietta Parracino
- Department of Chemistry-BMC, Uppsala University, BMC Box 576, S-751 23 Uppsala, Sweden
| | - Rajnish Kumar
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna 171 64, Sweden
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005 Uttar Pradesh, India
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna 171 64, Sweden
| |
Collapse
|
3
|
Wang Y, Li S, Chen M, Zeng M, Zhou L, Yao R, Pang B, Xu Y, Cao S, Guo S, Cui X. Shenyu ningshen tablet reduced neuronal damage in the hippocampus of chronic restraint stress model rat by inhibiting A1-reactive astrocytes. Heliyon 2024; 10:e28916. [PMID: 38655362 PMCID: PMC11035944 DOI: 10.1016/j.heliyon.2024.e28916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024] Open
Abstract
Context Shenyu Ningshen (SYNS) tablet is the first pure Chinese medicinal small compound preparation approved for clinical trials for the treatment of depression in China. Clinical experiments confirmed that the formulation had a significant Improvement effect against depression due to the deficiency of both qi and yin. It has been shown to exhibit noticeable anti-inflammatory effect in an animal model of depression. Our previous study showed that SYNS could effectively inhibit the inflammatory response in a depression model. Aim of the study The purpose of this study was to investigate the protective effects of SYNS on neurons and explore whether the underlying mechanism was associated with A1s. Materials and methods The depression model of solitary raising-chronic restraint stress (CRS) rats was established; body weight examination, sugar water preference test, open field test, and histological analysis were performed to preliminarily verify the efficacy of the formulation. Subsequently, neuronal nucleus (NeuN) and synaptic-associated proteins (MAP2 and PSD95) were labeled, and the protective effect of SYNS on hippocampal neurons was observed based on the fluorescence intensity of the above indicators. Western blotting, histological examination, and immunofluorescence were used to evaluate the inhibitory effects of SYNS on neuroinflammation and activation of A1s in CRS depression model. Results SYNS improved behavioral indicators such as weight loss, pleasure loss, and reduced exercise volume in CRS rat model. SYNS restored the CRS-induced histopathological changes in the hippocampus. SYNS showed a certain degree of protective effect on synapses. Further, SYNS inhibited the activation of A1s by inhibiting neuroinflammatory factors in the hippocampus. Conclusion Our results showed that SYNS had a certain degree of neuroprotective effect, which might be related to its inhibition of the inflammatory response and A1s.
Collapse
Affiliation(s)
- Yaxin Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuran Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mengping Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Meihua Zeng
- Guangdong Si Ji Pharmaceutical Co., LTD, China
| | - Lirun Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rongmei Yao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bo Pang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingli Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shan Cao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shanshan Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolan Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Yun JH, Hong Y, Hong MH, Kim G, Lee JS, Woo RS, Lee J, Yang EJ, Kim IS. Anti-inflammatory effects of neuregulin-1 in HaCaT keratinocytes and atopic dermatitis-like mice stimulated with Der p 38. Cytokine 2024; 174:156439. [PMID: 38134557 DOI: 10.1016/j.cyto.2023.156439] [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: 07/25/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 12/24/2023]
Abstract
Neuregulin (NRG)-1 plays fundamental roles in several organ systems after binding to its receptors, ErbB2 and ErbB4. This study examines the role of NRG-1 in atopic dermatitis (AD), a chronic skin disease that causes dryness, pruritus, and inflammation. In mice administered Der p 38, the skin presents AD-like symptoms including filaggrin downregulation and infiltration of neutrophils and eosinophils. Noticeably, there is an increased expression of NRG-1, ErbB2, and ErbB4 in the skin. Upregulation of these proteins is significantly correlated to the clinical skin severity score. In human keratinocyte HaCaT cells, exposure to Der p 38 decreased filaggrin expression, and NRG-1 alone had no effect on the expression. However, co-treatment of Der p 38 with NRG-1 enhanced the filaggrin expression decreased by Der p 38. Pre-treatment with AG879 (an ErbB2 inhibitor) or ErbB4 siRNA blocked the recovery of filaggrin expression in the cells after co-treatment with Der p 38 and NRG-1. Der p 38 treatment enhanced the secretion of interleukin-6 (IL-6), IL-8, and monocyte chemoattractant protein-1 (MCP-1). Co-treatment of Der p 38 with NRG-1 lowered the cytokine secretion increased by Der p 38, although NRG-1 alone was not effective on cytokine alteration. Neutrophil apoptosis was not altered by NRG-1 or supernatants of cells treated with NRG-1, but the cell supernatants co-treated with Der p 38 and NRG-1 blocked the anti-apoptotic effects of Der p 38-treated supernatants on neutrophils, which was involved in the activation of caspase 9 and caspase 3. Taken together, we determined that NRG-1 has anti-inflammatory effects in AD triggered by Der p 38. These results will pave the way to understanding the functions of NRG-1 and in the future development of AD treatment.
Collapse
Affiliation(s)
- Jeong Hee Yun
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Uijeongbu 11759, Republic of Korea
| | - Yujin Hong
- Department of Senior Healthcare, Graduate School, Eulji University, Uijeongbu 11759, Republic of Korea
| | - Min Hwa Hong
- Department of Senior Healthcare, Graduate School, Eulji University, Uijeongbu 11759, Republic of Korea
| | - Geunyeong Kim
- Department of Senior Healthcare, Graduate School, Eulji University, Uijeongbu 11759, Republic of Korea
| | - Ji-Sook Lee
- Department of Clinical Laboratory Science, Wonkwang Health Science University, Iksan 54538, Republic of Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, Eulji University School of Medicine, Daejeon 34824, Republic of Korea
| | - Juram Lee
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Uijeongbu 11759, Republic of Korea
| | - Eun Ju Yang
- Department of Biomedical Laboratory Science, Daegu Haany University, Gyeongsan 38610, Republic of Korea.
| | - In Sik Kim
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Uijeongbu 11759, Republic of Korea; Department of Senior Healthcare, Graduate School, Eulji University, Uijeongbu 11759, Republic of Korea.
| |
Collapse
|
5
|
Liu A, Fernandes BS, Citu C, Zhao Z. Unraveling the intercellular communication disruption and key pathways in Alzheimer's disease: an integrative study of single-nucleus transcriptomes and genetic association. Alzheimers Res Ther 2024; 16:3. [PMID: 38167548 PMCID: PMC10762817 DOI: 10.1186/s13195-023-01372-w] [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: 09/07/2023] [Accepted: 12/17/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Recently, single-nucleus RNA-seq (snRNA-seq) analyses have revealed important cellular and functional features of Alzheimer's disease (AD), a prevalent neurodegenerative disease. However, our knowledge regarding intercellular communication mediated by dysregulated ligand-receptor (LR) interactions remains very limited in AD brains. METHODS We systematically assessed the intercellular communication networks by using a discovery snRNA-seq dataset comprising 69,499 nuclei from 48 human postmortem prefrontal cortex (PFC) samples. We replicated the findings using an independent snRNA-seq dataset of 56,440 nuclei from 18 PFC samples. By integrating genetic signals from AD genome-wide association studies (GWAS) summary statistics and whole genome sequencing (WGS) data, we prioritized AD-associated Gene Ontology (GO) terms containing dysregulated LR interactions. We further explored drug repurposing for the prioritized LR pairs using the Therapeutic Targets Database. RESULTS We identified 190 dysregulated LR interactions across six major cell types in AD PFC, of which 107 pairs were replicated. Among the replicated LR signals, we found globally downregulated communications in the astrocytes-to-neurons signaling axis, characterized, for instance, by the downregulation of APOE-related and Calmodulin (CALM)-related LR interactions and their potential regulatory connections to target genes. Pathway analyses revealed 44 GO terms significantly linked to AD, highlighting Biological Processes such as 'amyloid precursor protein processing' and 'ion transmembrane transport,' among others. We prioritized several drug repurposing candidates, such as cromoglicate, targeting the identified dysregulated LR pairs. CONCLUSIONS Our integrative analysis identified key dysregulated LR interactions in a cell type-specific manner and the associated GO terms in AD, offering novel insights into potential therapeutic targets involved in disrupted cell-cell communication in AD.
Collapse
Affiliation(s)
- Andi Liu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St., Suite 600, Houston, TX, 77030, USA
| | - Brisa S Fernandes
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St., Suite 600, Houston, TX, 77030, USA
| | - Citu Citu
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St., Suite 600, Houston, TX, 77030, USA
| | - Zhongming Zhao
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St., Suite 600, Houston, TX, 77030, USA.
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, 37203, USA.
| |
Collapse
|
6
|
Liu J, Geraghty JR, Schram S, Cropper HC, Lei J, Loeb JA, Song F. Prevention of Alzheimer Pathology by Blocking Neuregulin Signaling on Microglia. eNeuro 2023; 10:ENEURO.0422-23.2023. [PMID: 37903620 PMCID: PMC10644371 DOI: 10.1523/eneuro.0422-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/01/2023] Open
Abstract
Plaque formation, microglial activation, and synaptic loss are pathologic hallmarks of Alzheimer's disease; however, removing plaques has had little clinical benefit. Here, we show that neuregulin-1, a glial growth factor, induces inflammatory cytokines and promotes phagocytic activity in vitro and augments microglial activation and plaque formation in 5XFAD Alzheimer's mice. Brain-specific targeting of neuregulin-1 by intraventricular delivery of a novel neuregulin-1 fusion protein antagonist, GlyB4, significantly alters microglial morphology and function to a nonpathogenic morphology in early-stage 5XFAD mice and prevents plaques from forming. Once plaques have already formed, GlyB4 reduces new plaque formation and prevents synaptic loss. Selective, targeted disruption of neuregulin-1 signaling on brain microglia with GlyB4 could be a novel "upstream" approach to slow or stop disease progression in Alzheimer's disease.
Collapse
Affiliation(s)
- Jianguo Liu
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Joseph R Geraghty
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Sarah Schram
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Haley C Cropper
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Justin Lei
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Jeffrey A Loeb
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Fei Song
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| |
Collapse
|
7
|
Liu A, Fernandes BS, Citu C, Zhao Z. Unraveling the intercellular communication disruption and key pathways in Alzheimer's disease: An integrative study of single-nucleus transcriptomes and genetic association. RESEARCH SQUARE 2023:rs.3.rs-3335643. [PMID: 37790454 PMCID: PMC10543294 DOI: 10.21203/rs.3.rs-3335643/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Background Recently, single-nucleus RNA-seq (snRNA-seq) analyses have revealed important cellular and functional features of Alzheimer's disease (AD), a prevalent neurodegenerative disease. However, our knowledge regarding intercellular communication mediated by dysregulated ligand-receptor (LR) interactions remains very limited in AD brains. Methods We systematically assessed the intercellular communication networks by using a discovery snRNA-seq dataset comprising 69,499 nuclei from 48 human postmortem prefrontal cortex (PFC) samples. We replicated the findings using an independent snRNA-seq dataset of 56,440 nuclei from 18 PFC samples. By integrating genetic signals from AD genome-wide association studies (GWAS) summary statistics and whole genome sequencing (WGS) data, we prioritized AD-associated Gene Ontology (GO) terms containing dysregulated LR interactions. We further explored drug repurposing for the prioritized LR pairs using the Therapeutic Targets Database. Results We identified 316 dysregulated LR interactions across six major cell types in AD PFC, of which 210 pairs were replicated. Among the replicated LR signals, we found globally downregulated communications in astrocytes-to-neurons signaling axis, characterized, for instance, by the downregulation of APOE-related and Calmodulin (CALM)-related LR interactions and their potential regulatory connections to target genes. Pathway analyses revealed 60 GO terms significantly linked to AD, highlighting Biological Processes such as 'amyloid precursor protein processing' and 'ion transmembrane transport', among others. We prioritized several drug repurposing candidates, such as cromoglicate, targeting the identified dysregulated LR pairs. Conclusions Our integrative analysis identified key dysregulated LR interactions in a cell type-specific manner and the associated GO terms in AD, offering novel insights into potential therapeutic targets involved in disrupted cell-cell communication in AD.
Collapse
Affiliation(s)
- Andi Liu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston
| | - Brisa S Fernandes
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston
| | - Citu Citu
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston
| |
Collapse
|
8
|
Vincent B, Maitra S. BACE1-dependent metabolism of neuregulin 1: Bridging the gap in explaining the occurrence of schizophrenia-like symptoms in Alzheimer's disease with psychosis? Ageing Res Rev 2023; 89:101988. [PMID: 37331479 DOI: 10.1016/j.arr.2023.101988] [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: 03/08/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
Alzheimer's disease is a neurodegenerative disease mainly characterized by cortico-neuronal atrophy, impaired memory and other cognitive declines. On the other hand, schizophrenia is a neuro-developmental disorder with an overtly active central nervous system pruning system resulting into abrupt connections with common symptoms including disorganised thoughts, hallucination and delusion. Nevertheless, the fronto-temporal anomaly presents itself as a common denominator for the two pathologies. There is even a strong presumption of increased risk of developing co-morbid dementia for schizophrenic individuals and psychosis for Alzheimer's disease patients, overall leading to a further deteriorated quality of life. However, convincing proofs of how these two disorders, although very distant from each other when considering their aetiology, develop coexisting symptoms is yet to be resolved. At the molecular level, the two primarily neuronal proteins β-amyloid precursor protein and neuregulin 1 have been considered in this relevant context, although the conclusions are for the moment only hypotheses. In order to propose a model for explaining the psychotic schizophrenia-like symptoms that sometimes accompany AD-associated dementia, this review projects out on the similar sensitivity shared by these two proteins regarding their metabolism by the β-site APP cleaving enzyme 1.
Collapse
Affiliation(s)
- Bruno Vincent
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560 Valbonne, France.
| | - Subhamita Maitra
- Department of Molecular Biology, Umeå University, Umeå 90736, Sweden
| |
Collapse
|
9
|
Song J, Ma Z, Zhang H, Liang T, Zhang J. Identification of novel biomarkers linking depressive disorder and Alzheimer's disease based on an integrative bioinformatics analysis. BMC Genom Data 2023; 24:22. [PMID: 37061663 PMCID: PMC10105463 DOI: 10.1186/s12863-023-01120-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 03/16/2023] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND Previous reports revealed that a history of major depressive disorder (MDD) increased the risk of Alzheimer's disease (AD). The immune disorder is associated with MDD and AD pathophysiology. We aimed to identify differentially expressed immune-related genes (DEIRGs) that are involved in the pathogenesis of MDD and AD. METHODS We downloaded mRNA expression profiles (GSE76826 and GSE5281) from the Gene Expression Omnibus (GEO) database. The R software was used to identify DEIRGs for the two diseases separately. Functional enrichment analysis and PPI network of DEIRGs were performed. Finally, the relationship between shared DEIRGs and immune infiltrates of AD and MDD were analyzed, respectively. RESULTS A total of 121 DEIRGs linking AD and MDD were identified. These genes were significantly enriched in immune-related pathways, such as the JAK-STAT signaling pathway, regulation of chemotaxis, chemotaxis, cytokine-cytokine receptor interaction, and primary immunodeficiency. Furthermore, three shared DEIRGs (IL1R1, CHGB, and NRG1) were identified. Correlation analysis between DEIRGs and immune cells revealed that IL1R1 and NRG1 had a negative or positive correlation with some immune cells both in AD and MDD. CONCLUSION Both DEIRGs and immune cell infiltrations play a vital role in the pathogenesis of AD and MDD. Our findings indicated that there are common genes and biological processes between MDD and AD, which provides a theoretical basis for the study of the comorbidity of MDD and AD.
Collapse
Affiliation(s)
- Jin Song
- Out-Patient Department, Wuhan Mental Health Center, Wuhan, 430012, Hubei Province, China
- Out-Patient Department, Wuhan Hospital for Psychotherapy, Wuhan, 430012, Hubei Province, China
| | - Zilong Ma
- Ward of Sleep Disorders, Wuhan Mental Health Center, Wuhan, 430012, Hubei Province, China
- Ward of Sleep Disorders, Wuhan Hospital for Psychotherapy, Wuhan, 430012, Hubei Province, China
| | - Huishi Zhang
- Out-Patient Department, Wuhan Mental Health Center, Wuhan, 430012, Hubei Province, China.
- Out-Patient Department, Wuhan Hospital for Psychotherapy, Wuhan, 430012, Hubei Province, China.
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei Province, 430012, China.
| | - Ting Liang
- National Medical Institution Conducting Clinical Trials Office, Wuhan Mental Health Center, Wuhan, 430012, Hubei Province, China
- National Medical Institution Conducting Clinical Trials Office, Wuhan Hospital for Psychotherapy, Wuhan, 430012, Hubei Province, China
| | - Jun Zhang
- Ward of Traditional Chinese Medicine, Wuhan Mental Health Center, Wuhan, 430012, Hubei Province, China
- Ward of Traditional Chinese Medicine, Wuhan Hospital for Psychotherapy, Wuhan, 430012, Hubei Province, China
| |
Collapse
|
10
|
Fronza MG, Sacramento M, Alves D, Praticò D, Savegnago L. QTC-4-MeOBnE Ameliorated Depressive-Like Behavior and Memory Impairment in 3xTg Mice. Mol Neurobiol 2023; 60:1733-1745. [PMID: 36567360 DOI: 10.1007/s12035-022-03159-w] [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: 08/08/2022] [Accepted: 12/01/2022] [Indexed: 12/27/2022]
Abstract
Growing evidence has associated major depressive disorder (MDD) as a risk factor or prodromal syndrome for the occurrence of Alzheimer's disease (AD). Although this dilemma remains open, it is widely shown that a lifetime history of MDD is correlated with faster progression of AD pathology. Therefore, antidepressant drugs with neuroprotective effects could be an interesting therapeutic conception to target this issue simultaneously. In this sense, 1-(7-chloroquinolin-4-yl)-N-(4-methoxybenzyl)-5-methyl-1H-1,2,3-triazole-4- carboxamide (QTC-4-MeOBnE) was initially conceived as a multi-target ligand with affinity to β-secretase (BACE), glycogen synthase kinase 3β (GSK3β), and acetylcholinesterase but has also shown secondary effects on pathways involved in neuroinflammation and neurogenesis in preclinical models of AD. Herein, we investigated the effect of QTC-4-MeOBnE (1 mg/kg) administration for 45 days on depressive-like behavior and memory impairment in 3xTg mice, before the pathology is completely established. The treatment with QTC-4-MeOBnE prevented memory impairment and depressive-like behavior assessed by the Y-Maze task and forced swimming test. This effect was associated with the modulation of plural pathways involved in the onset and progression of AD, in cerebral structures of the cortex and hippocampus. Among them, the reduction of amyloid beta (Aβ) production mediated by changes in amyloid precursor protein metabolism and hippocampal tau phosphorylation through the inhibition of kinases. Additionally, QTC-4-MeOBnE also exerted beneficial effects on neuroinflammation and synaptic integrity. Overall, our studies suggest that QTC-4-MeOBnE has a moderate effect in a transgenic model of AD, indicating that perhaps studies regarding the neuropsychiatric effects as a neuroprotective molecule are more prone to be feasible.
Collapse
Affiliation(s)
- Mariana G Fronza
- Neurobiotechnology Research Group (GPN) - Postgraduate Program of Biotechnology, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil
| | - Manoela Sacramento
- Laboratory of Clean Organic Synthesis (LASOL), Postgraduate Program of Chemistry, UFPel, Pelotas, RS, Brazil
| | - Diego Alves
- Laboratory of Clean Organic Synthesis (LASOL), Postgraduate Program of Chemistry, UFPel, Pelotas, RS, Brazil
| | - Domenico Praticò
- Alzheimer's Center at Temple - ACT, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Lucielli Savegnago
- Neurobiotechnology Research Group (GPN) - Postgraduate Program of Biotechnology, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil.
| |
Collapse
|
11
|
Zhou A, Wang L, Pi X, Fan C, Chen W, Wang Z, Rong S, Wang T. Effects of perfluorooctane sulfonate (PFOS) on cognitive behavior and autophagy of male mice. J Toxicol Sci 2023; 48:513-526. [PMID: 37661368 DOI: 10.2131/jts.48.513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Perfluorooctane sulfonate (PFOS), an emerging environmental pollutant, is reported to cause neurotoxicity in animals and humans, but its underlying mechanisms are still unclear. We used in vivo models to investigate the effects of PFOS on cognition-related behaviors and related mechanisms. After 45 days of intragastric administration of PFOS (2 mg/kg or 8 mg/kg) in 7-week-old C57BL/6 mice, muscle strength, cognitive function and anxiety-like behavior were evaluated by a series of behavioral tests. The underling mechanisms of PFOS on impaired behaviors were evaluated by HE/Nissl staining, electron microscopy observation and western blot analysis. The results indicated that PFOS-exposed mice exhibited significant cognitive impairment, anxiety, neuronal degeneration and the abnormities of synaptic ultrastructure in the cortex and hippocampus. Western blot analysis indicated that PFOS exposure increased microtubule-associated protein light chain 3 (LC3) and decreased p62 protein levels, which may be associated with activation of autophagy leading to neuron damage. In summary, our results suggest that chronic exposure to PFOS adversely affects cognitive-related behavior in mice. These findings provide new mechanistic insights into PFOS-induced neurotoxicity.
Collapse
Affiliation(s)
- Aojia Zhou
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Li Wang
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Xuejiao Pi
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Cheng Fan
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Wenwen Chen
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Ziping Wang
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Shuang Rong
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Ting Wang
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| |
Collapse
|
12
|
Ma Y, Fan P, Zhao R, Zhang Y, Wang X, Cui W. Neuregulin-1 regulates the conversion of M1/M2 microglia phenotype via ErbB4-dependent inhibition of the NF-κB pathway. Mol Biol Rep 2022; 49:3975-3986. [PMID: 35166983 DOI: 10.1007/s11033-022-07249-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/09/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND The inflammatory response caused by microglia in the central nervous system plays an important role in Alzheimer's disease. Neuregulin-1 (NRG1) is a member of the neuregulin family and has been demonstrated to have anti-inflammatory properties. The relationship between NRG1, microglia phenotype and neuroinflammation remains unclear. MATERIALS AND METHODS BV2 cells were used to examine the mechanism of NRG1 in regulating microglia polarization. Neuronal apoptosis, inflammatory factors TNF-α and iNOS, microglia polarization, ErbB4 and NF-κB p65 expression were assessed. RESULTS We found that exogenous NRG1 treatment or overexpression improved microglial activity and reduced the secretion of the inflammatory factors TNF-α and iNOS in vitro. The expression of Bax in SH-SY5Y neuron cells incubated with medium collected from the NRG1 treatment group decreased. Additionally, our study showed that NRG1 treatment reduced the levels of the M1 microglia markers CD120 and iNOS and increased the levels of the M2 microglia markers CD206 and Arg-1. Furthermore, we observed that NRG1 treatment attenuated Aβ-induced NF-κB activation and promoted the expression of p-ErbB4 and that knockdown of ErbB4 abrogated the effects of NRG1 on NF-κB, Bax levels and M2 microglial polarization. CONCLUSION NRG1 inhibits the release of inflammatory factors in microglia and regulates the switching of the M1/M2 microglia phenotype, most likely via ErbB4-dependent inhibition of the NF-κB pathway.
Collapse
Affiliation(s)
- Yuqi Ma
- Department of Human Anatomy and Histoembryology, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, China
| | - Peixia Fan
- Department of Human Anatomy and Histoembryology, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, China
| | - Rui Zhao
- Department of Human Anatomy and Histoembryology, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, China
| | - Yinghua Zhang
- Department of Human Anatomy and Histoembryology, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, China
| | - Xianwei Wang
- Department of Human Anatomy and Histoembryology, Xinxiang Medical University, Xinxiang, China. .,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China.
| | - Weigang Cui
- Department of Human Anatomy and Histoembryology, Xinxiang Medical University, Xinxiang, China. .,Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
13
|
Yuan J, Wang H, Wang Y, Wang Z, Huo Q, Dai X, Zhang J, Sun Y. Rapid Identification of 3,6'-Disinapoyl Sucrose Metabolites in Alzheimer's Disease Model Mice Using UHPLC-Orbitrap Mass Spectrometry. Molecules 2021; 27:114. [PMID: 35011346 PMCID: PMC8746568 DOI: 10.3390/molecules27010114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system characterized by the progressive impairment of neural activity. Studies have shown that 3,6'-disinapoyl sucrose (DISS) can alleviate the pathological symptoms of AD through the activation of the cAMP/CREB/BDNF signaling pathway. However, the exact biochemical mechanisms of action of DISS are not clear. This study explores metabolism of DISS in an AD mouse model, induced by the microinjection of a lentiviral expression plasmid of the APPswe695 gene into CA1 of the hippocampus. After gavage administration of DISS (200 mg/kg), the kidneys, livers, brains, plasma, urine, and feces were collected for UHPLC-Orbitrap mass spectrometry analysis. Twenty metabolites, including the prototype drug of DISS, were positively or tentatively identified based on accurate mass measurements, characteristic fragmentation behaviors, and retention times. Thus, the metabolic pathways of DISS in AD mice were preliminarily elucidated through the identification of metabolites, such as ester bond cleavage, demethoxylation, demethylation, and sinapic acid-related products. Furthermore, differences in the in vivo distribution of several metabolites were observed between the model and sham control groups. These findings can provide a valuable reference for the pharmacological mechanisms and biosafety of DISS.
Collapse
Affiliation(s)
- Jiaqi Yuan
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| | - Han Wang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| | - Yunting Wang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| | - Zijian Wang
- Beijing Research Institution of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China;
| | - Qing Huo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| | - Jiayu Zhang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China;
| | - Yaxuan Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| |
Collapse
|
14
|
Martínez-Iglesias O, Naidoo V, Cacabelos N, Cacabelos R. Epigenetic Biomarkers as Diagnostic Tools for Neurodegenerative Disorders. Int J Mol Sci 2021; 23:13. [PMID: 35008438 PMCID: PMC8745005 DOI: 10.3390/ijms23010013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/03/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
Epigenetics is the study of heritable changes in gene expression that occur without alterations to the DNA sequence, linking the genome to its surroundings. The accumulation of epigenetic alterations over the lifespan may contribute to neurodegeneration. The aim of the present study was to identify epigenetic biomarkers for improving diagnostic efficacy in patients with neurodegenerative diseases. We analyzed global DNA methylation, chromatin remodeling/histone modifications, sirtuin (SIRT) expression and activity, and the expression of several important neurodegeneration-related genes. DNA methylation, SIRT expression and activity and neuregulin 1 (NRG1), microtubule-associated protein tau (MAPT) and brain-derived neurotrophic factor (BDNF) expression were reduced in buffy coat samples from patients with neurodegenerative disorders. Our data suggest that these epigenetic biomarkers may be useful in clinical practical for the diagnosis, surveillance, and prognosis of disease activity in patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Olaia Martínez-Iglesias
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, 15165 Corunna, Spain; (V.N.); (N.C.); (R.C.)
| | | | | | | |
Collapse
|
15
|
Schramm S, Jokisch M, Jöckel KH, Herring A, Keyvani K. Is kallikrein-8 a blood biomarker for detecting amnestic mild cognitive impairment? Results of the population-based Heinz Nixdorf Recall study. Alzheimers Res Ther 2021; 13:202. [PMID: 34930454 PMCID: PMC8690879 DOI: 10.1186/s13195-021-00945-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/06/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Kallikrein-8 (KLK8) might be an early blood-biomarker of Alzheimer's disease (AD). We examined whether blood KLK8 is elevated in persons with amnestic mild cognitive impairment (aMCI) which is a precursor of AD, compared to cognitively unimpaired (CU) controls. METHODS Forty cases and 80 controls, matched by sex and age (± 3years), were participants of the longitudinal population-based Heinz Nixdorf Recall study (baseline: 2000-2003). Standardized cognitive performance was assessed 5 (T1) and 10 years after baseline (T2). Cases were CU at T1 and had incidental aMCI at T2. Controls were CU at T1 and T2. Blood KLK8 was measured at T2. Using multiple logistic regression the association between KLK8 in cases vs. controls was investigated by estimating odds ratios (OR) and 95% confidence intervals (95%CI), adjusted for inter-assay variability and freezing duration. Using receiver operating characteristic (ROC) analysis, the diagnostic accuracy of KLK8 was determined by estimating the area under the curve (AUC) and 95%CI (adjusted for inter-assay variability, freezing duration, age, sex). RESULTS Thirty-seven participants with aMCI vs. 72 CU (36.7%women, 71.0±8.0 (mean±SD) years) had valid KLK8 measurements. Mean KLK8 was higher in cases than in controls (911.6±619.8 pg/ml vs.783.1±633.0 pg/ml). Fully adjusted, a KLK8 increase of 500pg/ml was associated with a 2.68 (1.05-6.84) higher chance of having aMCI compared to being CU. With an AUC of 0.92 (0.86-0.97), blood KLK8 was a strong discriminator for aMCI and CU. CONCLUSION This is the first population-based study to demonstrate the potential clinical utility of blood KLK8 as a biomarker for incipient AD.
Collapse
Affiliation(s)
- Sara Schramm
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany.
| | - Martha Jokisch
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Karl-Heinz Jöckel
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Arne Herring
- Institute of Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
16
|
Untargeted Metabolomic Profiling of Cuprizone-Induced Demyelination in Mouse Corpus Callosum by UPLC-Orbitrap/MS Reveals Potential Metabolic Biomarkers of CNS Demyelination Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7093844. [PMID: 34567412 PMCID: PMC8457991 DOI: 10.1155/2021/7093844] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/13/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is a neurodegenerative disorder characterized by periodic neuronal demyelination, which leads to a range of symptoms and eventually to disability. The goal of this research was to use UPLC-Orbitrap/MS to identify validated biomarkers and explore the metabolic mechanisms of MS in mice. Thirty-two C57BL/6 male mice were randomized into two groups that were fed either normal food or 0.2% CPZ for 11 weeks. The mouse demyelination model was assessed by LFB and the expression of MBP by immunofluorescence and immunohistochemistry. The metabolites of the corpus callosum were quantified using UPLC-Orbitrap/MS. The mouse pole climbing experiment was used to assess coordination ability. Multivariate statistical analysis was adopted for screening differential metabolites, and the ingenuity pathway analysis (IPA) was used to reveal the metabolite interaction network. We successfully established the demyelination model. The CPZ group slowly lost weight and showed an increased pole climbing time during feeding compared to the CON group. A total of 81 metabolites (VIP > 1 and P < 0.05) were determined to be enriched in 24 metabolic pathways; 41 metabolites were markedly increased, while 40 metabolites were markedly decreased in the CPZ group. The IPA results revealed that these 81 biomarker metabolites were associated with neuregulin signaling, PI3K-AKT signaling, mTOR signaling, and ERK/MAPK signaling. KEGG pathway analysis showed that two significantly different metabolic pathways were enriched, namely, the glycerophospholipid and sphingolipid metabolic pathways, comprising a total of nine biomarkers. Receiver operating characteristic analysis showed that the metabolites (e.g., PE (16 : 0/22 : 6(4Z, 7Z, 10Z, 13Z, 16Z, 19Z)), PC (18 : 0/22 : 4(7Z, 10Z, 13Z, 16Z)), cytidine 5′-diphosphocholine, PS (18 : 0/22 : 6(4Z, 7Z, 10Z, 13Z, 16Z, 19Z)), glycerol 3-phosphate, SM (d18 : 0/16 : 1(9Z)), Cer (d18:1/18 : 0), galabiosylceramide (d18:1/18 : 0), and GlcCer (d18:1/18 : 0)) have good discrimination ability for the CPZ group. In conclusion, the differential metabolites have great potential to serve as biomarkers of demyelinating diseases. In addition, we identified metabolic pathways associated with CPZ-induced demyelination pathogenesis, which provided a new perspective for understanding the relationship between metabolites and CNS demyelination pathogenesis.
Collapse
|
17
|
Espeso-Gil S, Holik AZ, Bonnin S, Jhanwar S, Chandrasekaran S, Pique-Regi R, Albaigès-Ràfols J, Maher M, Permanyer J, Irimia M, Friedländer MR, Pons-Espinal M, Akbarian S, Dierssen M, Maass PG, Hor CN, Ossowski S. Environmental Enrichment Induces Epigenomic and Genome Organization Changes Relevant for Cognition. Front Mol Neurosci 2021; 14:664912. [PMID: 34025350 PMCID: PMC8131874 DOI: 10.3389/fnmol.2021.664912] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/09/2021] [Indexed: 01/11/2023] Open
Abstract
In early development, the environment triggers mnemonic epigenomic programs resulting in memory and learning experiences to confer cognitive phenotypes into adulthood. To uncover how environmental stimulation impacts the epigenome and genome organization, we used the paradigm of environmental enrichment (EE) in young mice constantly receiving novel stimulation. We profiled epigenome and chromatin architecture in whole cortex and sorted neurons by deep-sequencing techniques. Specifically, we studied chromatin accessibility, gene and protein regulation, and 3D genome conformation, combined with predicted enhancer and chromatin interactions. We identified increased chromatin accessibility, transcription factor binding including CTCF-mediated insulation, differential occupancy of H3K36me3 and H3K79me2, and changes in transcriptional programs required for neuronal development. EE stimuli led to local genome re-organization by inducing increased contacts between chromosomes 7 and 17 (inter-chromosomal). Our findings support the notion that EE-induced learning and memory processes are directly associated with the epigenome and genome organization.
Collapse
Affiliation(s)
- Sergio Espeso-Gil
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Genetics and Genome Biology Program, SickKids Research Institute, Toronto, ON, Canada
| | - Aliaksei Z. Holik
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Sarah Bonnin
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Shalu Jhanwar
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Sandhya Chandrasekaran
- MD/PhD Program in the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Júlia Albaigès-Ràfols
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Michael Maher
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jon Permanyer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Manuel Irimia
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Pg. Lluis Companys 23, Barcelona, Spain
| | - Marc R. Friedländer
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Meritxell Pons-Espinal
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Schahram Akbarian
- Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Philipp G. Maass
- Genetics and Genome Biology Program, SickKids Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Charlotte N. Hor
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Stephan Ossowski
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| |
Collapse
|
18
|
Herring A, Kurapati NK, Krebs S, Grammon N, Scholz LM, Voss G, Miah MR, Budny V, Mairinger F, Haase K, Teuber-Hanselmann S, Dobersalske C, Schramm S, Jöckel KH, Münster Y, Keyvani K. Genetic knockdown of Klk8 has sex-specific multi-targeted therapeutic effects on Alzheimer's pathology in mice. Neuropathol Appl Neurobiol 2021; 47:611-624. [PMID: 33341972 DOI: 10.1111/nan.12687] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/23/2020] [Accepted: 12/14/2020] [Indexed: 01/22/2023]
Abstract
AIMS Previous work in our lab has identified the protease kallikrein-8 (KLK8) as a potential upstream mover in the pathogenesis of Alzheimer's disease (AD). We showed pathologically elevated levels of KLK8 in the cerebrospinal fluid and blood of patients with mild cognitive impairment or dementia due to AD, and in brains of patients and transgenic CRND8 (TgCRND8) mice in incipient stages of the disease. Furthermore, short-term antibody-mediated KLK8 inhibition in moderate stage disease alleviated AD pathology in female mice. However, it remains to be shown whether long-term reversal of KLK8 overexpression can also counteract AD. Therefore, the effects of genetic Klk8-knockdown were determined in TgCRND8 mice. METHODS The effects of heterozygous ablation of murine Klk8 (mKlk8) gene on AD pathology of both sexes were examined by crossbreeding TgCRND8 [hAPP+/-] with mKlk8-knockdown [mKlk8+/-] mice resulting in animals with or without AD pathology which revealed pathologically elevated or normal KLK8 levels. RESULTS mKlk8-knockdown had negligible effects on wildtype animals but led to significant decline of amyloid beta (Aβ) and tau pathology as well as an improvement of structural neuroplasticity in a sex-specific manner in transgenics. These changes were mediated by a shift to non-amyloidogenic cleavage of the human amyloid precursor protein (APP), recovery of the neurovascular unit and maintaining microglial metabolic fitness. Mechanistically, Klk8-knockdown improved Aβ phagocytosis in primary glia and Aβ resistance in primary neurons. Most importantly, transgenic mice revealed less anxiety and a better memory performance. CONCLUSIONS These results reinforce the potential of KLK8 as a therapeutic target in AD.
Collapse
Affiliation(s)
- Arne Herring
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Nirup K Kurapati
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Sofia Krebs
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Nils Grammon
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Luisa M Scholz
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Gerrit Voss
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Muhammad R Miah
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Vanessa Budny
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Fabian Mairinger
- Institute of Pathology, University of Duisburg-Essen, Essen, Germany
| | - Katharina Haase
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | | | - Celia Dobersalske
- DKFZ-Division of Translational Neurooncology, West German Cancer Center, German Cancer Consortium (DKTK) Partner Site, University Hospital Essen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sara Schramm
- Institute of Medical Informatics, Biometry and Epidemiology, University of Duisburg-Essen, Essen, Germany
| | - Karl-Heinz Jöckel
- Institute of Medical Informatics, Biometry and Epidemiology, University of Duisburg-Essen, Essen, Germany
| | - Yvonne Münster
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
19
|
Seo HJ, Park JE, Choi SM, Kim T, Cho SH, Lee KH, Song WK, Song J, Jeong HS, Kim DH, Kim BC. Inhibitory Neural Network's Impairments at Hippocampal CA1 LTP in an Aged Transgenic Mouse Model of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22020698. [PMID: 33445678 PMCID: PMC7828160 DOI: 10.3390/ijms22020698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/03/2021] [Accepted: 01/08/2021] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by a rapid accumulation of amyloid β (Aβ) protein in the hippocampus, which impairs synaptic structures and neuronal signal transmission, induces neuronal loss, and diminishes memory and cognitive functions. The present study investigated the impact of neuregulin 1 (NRG1)-ErbB4 signaling on the impairment of neural networks underlying hippocampal long-term potentiation (LTP) in 5xFAD mice, a model of AD with greater symptom severity than that of TG2576 mice. Specifically, we observed parvalbumin (PV)-containing hippocampal interneurons, the effect of NRG1 on hippocampal LTP, and the functioning of learning and memory. We found a significant decrease in the number of PV interneurons in 11-month-old 5xFAD mice. Moreover, synaptic transmission in the 5xFAD mice decreased at 6 months of age. The 11-month-old transgenic AD mice showed fewer inhibitory PV neurons and impaired NRG1-ErbB4 signaling than did wild-type mice, indicating that the former exhibit the impairment of neuronal networks underlying LTP in the hippocampal Schaffer-collateral pathway. In conclusion, this study confirmed the impaired LTP in 5xFAD mice and its association with aberrant NRG1-ErbB signaling in the neuronal network.
Collapse
Affiliation(s)
- Hyeon Jeong Seo
- Department of Biomedical Sciences, Graduate School, Chonnam National University, Gwangju 61186, Korea;
| | - Jung Eun Park
- Department of Biomedical Science, College of Natural Sciences, Chosun University, Gwangju 61452, Korea;
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Korea
| | - Seong-Min Choi
- Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Korea; (S.-M.C.); (S.H.C.)
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
| | - Taekyoung Kim
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
| | - Soo Hyun Cho
- Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Korea; (S.-M.C.); (S.H.C.)
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Medical School & Hwasun Hospital, Hwasun 58128, Korea;
| | - Woo Keun Song
- Cell Logistics and Silver Health Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Dong Hyun Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49236, Korea
- Correspondence: (D.H.K.); (B.C.K.); Tel.: +82-51-200-7583 (D.H.K.); +82-62-220-6123 (B.C.K.)
| | - Byeong C. Kim
- Department of Biomedical Sciences, Graduate School, Chonnam National University, Gwangju 61186, Korea;
- Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Korea; (S.-M.C.); (S.H.C.)
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
- Correspondence: (D.H.K.); (B.C.K.); Tel.: +82-51-200-7583 (D.H.K.); +82-62-220-6123 (B.C.K.)
| |
Collapse
|
20
|
Ou GY, Lin WW, Zhao WJ. Neuregulins in Neurodegenerative Diseases. Front Aging Neurosci 2021; 13:662474. [PMID: 33897409 PMCID: PMC8064692 DOI: 10.3389/fnagi.2021.662474] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/16/2021] [Indexed: 02/05/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), are typically characterized by progressive neuronal loss and neurological dysfunctions in the nervous system, affecting both memory and motor functions. Neuregulins (NRGs) belong to the epidermal growth factor (EGF)-like family of extracellular ligands and they play an important role in the development, maintenance, and repair of both the central nervous system (CNS) and peripheral nervous system (PNS) through the ErbB signaling pathway. They also regulate multiple intercellular signal transduction and participate in a wide range of biological processes, such as differentiation, migration, and myelination. In this review article, we summarized research on the changes and roles of NRGs in neurodegenerative diseases, especially in AD. We elaborated on the structural features of each NRG subtype and roles of NRG/ErbB signaling networks in neurodegenerative diseases. We also discussed the therapeutic potential of NRGs in the symptom remission of neurodegenerative diseases, which may offer hope for advancing related treatment.
Collapse
Affiliation(s)
- Guan-yong Ou
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Wen-wen Lin
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Wei-jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- *Correspondence: Wei-jiang Zhao
| |
Collapse
|
21
|
Increased Levels of Serum Neuregulin 1 Associated with Cognitive Impairment in Vascular Dementia. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6683747. [PMID: 33274218 PMCID: PMC7676920 DOI: 10.1155/2020/6683747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/21/2022]
Abstract
Objective Neuregulin 1 (NRG 1) is a member of the epidermal growth factor (EGF) family and is believed to play an important role in neuroplasticity. However, the relationship between NRG 1 and vascular dementia (VaD) is poorly understood. The purpose of this study is to explore the correlation between neuregulin 1 and VaD. Patients and Methods. From October 2018 to September 2020, 93 VaD patients and 79 control populations who attended Liaocheng People's Hospital were included in the study. Baseline characteristics including age, gender, years of education, HDL, LDL, FBG, SBP, and DBP are collected. At the same time, peripheral blood was collected, and the concentration of serum NRG 1 was detected by enzyme-linked immunosorbent assay (ELISA). All research subjects received professional cognitive function assessment. Results A total of 93 VaD patients and 79 controls were enrolled. There was no significant difference in age, gender, years of education, HDL, LDL, FBG, SBP, and DBP between the two groups (p > 0.05). However, compared with the control group, VaD patients have lower MoCA and higher serum NRG 1 levels, and the difference is statistically significant (p < 0.001). The correlation analysis of MoCA and baseline characteristics showed that the MoCA score in VaD was significantly negatively correlated with serum NRG 1 (r = −0.374, p = 0.036). The results of multivariate regression showed that the MoCA score of VaD patients was only associated with NRG 1 (β = 0.258, p = 0.012). Conclusions The concentration of serum NRG 1 in VaD patients is significantly increased, which may be an independent risk factor for cognitive impairment in VaD patients.
Collapse
|
22
|
Paterson C, Cumming B, Law AJ. Temporal Dynamics of the Neuregulin-ErbB Network in the Murine Prefrontal Cortex across the Lifespan. Cereb Cortex 2020; 30:3325-3339. [PMID: 31897479 DOI: 10.1093/cercor/bhz312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Neuregulin-ErbB signaling is essential for numerous functions in the developing, adult, and aging brain, particularly in the prefrontal cortex (PFC). Mouse models with disrupted Nrg and/or ErbB genes are relevant to psychiatric, developmental, and age-related disorders, displaying a range of abnormalities stemming from cortical circuitry impairment. Many of these models display nonoverlapping phenotypes dependent upon the gene target and timing of perturbation, suggesting that cortical expression of the Nrg-ErbB network undergoes temporal regulation across the lifespan. Here, we report a comprehensive temporal expression mapping study of the Nrg-ErbB signaling network in the mouse PFC across postnatal development through aging. We find that Nrg and ErbB genes display distinct expression profiles; moreover, splice isoforms of these genes are differentially expressed across the murine lifespan. We additionally find a developmental switch in ErbB4 splice isoform expression potentially mediated through coregulation of the lncRNA Miat expression. Our results are the first to comprehensively and quantitatively map the expression patterns of the Nrg-ErbB network in the mouse PFC across the postnatal lifespan and may help disentangle the pathway's involvement in normal cortical sequences of events across the lifespan, as well as shedding light on the pathophysiological mechanisms of abnormal Nrg-ErbB signaling in neurological disease.
Collapse
Affiliation(s)
- Clare Paterson
- Department of Psychiatry, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brooke Cumming
- Department of Psychiatry, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Amanda J Law
- Department of Psychiatry, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA.,Department of Cell and Developmental Biology, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA.,Department of Medicine, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
23
|
Ferdousi F, Kondo S, Sasaki K, Uchida Y, Ohkohchi N, Zheng YW, Isoda H. Microarray analysis of verbenalin-treated human amniotic epithelial cells reveals therapeutic potential for Alzheimer's Disease. Aging (Albany NY) 2020; 12:5516-5538. [PMID: 32224504 PMCID: PMC7138585 DOI: 10.18632/aging.102985] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/24/2020] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) has become a major world health problem as the population ages. There is still no available treatment that can stop or reverse the progression of AD. Human amnion epithelial cells (hAECs), an alternative source for stem cells, have shown neuroprotective and neurorestorative potentials when transplanted in vivo. Besides, studies have suggested that stem cell priming with plant-derived bioactive compounds can enhance stem cell proliferation and differentiation and improve the disease-treating capability of stem cells. Verbenalin is an iridoid glucoside found in medicinal herbs of Verbenaceae family. In the present study, we have conducted microarray gene expression profiling of verbenalin-treated hAECs to explore its therapeutic potential for AD. Gene set enrichment analysis revealed verbenalin treatment significantly enriched AD-associated gene sets. Genes associated with lysosomal dysfunction, pathologic angiogenesis, pathologic protein aggregation, circadian rhythm, age-related neurometabolism, and neurogenesis were differentially expressed in the verbenalin-treated hAECs compared to control cells. Additionally, the neuroprotective effect of verbenalin was confirmed against amyloid beta-induced neurotoxicity in human neuroblastoma SH-SY5Y cells. Our present study is the first to report the therapeutic potential of verbenalin for AD; however, further in-depth research in the in vitro and in vivo models are required to confirm our preliminary findings.
Collapse
Affiliation(s)
- Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba 305-8577, Ibaraki, Japan
| | - Shinji Kondo
- R&D Center for Tailor-Made QOL, University of Tsukuba, Tsukuba 305-8550, Ibaraki, Japan
| | - Kazunori Sasaki
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba 305-8577, Ibaraki, Japan
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Ibaraki, Japan
| | - Yoshiaki Uchida
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Nobuhiro Ohkohchi
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Yun-Wen Zheng
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba 305-8577, Ibaraki, Japan
- R&D Center for Tailor-Made QOL, University of Tsukuba, Tsukuba 305-8550, Ibaraki, Japan
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Ibaraki, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| |
Collapse
|
24
|
Scapin C, Ferri C, Pettinato E, Zambroni D, Bianchi F, Del Carro U, Belin S, Caruso D, Mitro N, Pellegatta M, Taveggia C, Schwab MH, Nave KA, Feltri ML, Wrabetz L, D'Antonio M. Enhanced axonal neuregulin-1 type-III signaling ameliorates neurophysiology and hypomyelination in a Charcot-Marie-Tooth type 1B mouse model. Hum Mol Genet 2020; 28:992-1006. [PMID: 30481294 DOI: 10.1093/hmg/ddy411] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/30/2018] [Accepted: 11/22/2018] [Indexed: 12/11/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) neuropathies are a group of genetic disorders that affect the peripheral nervous system with heterogeneous pathogenesis and no available treatment. Axonal neuregulin 1 type III (Nrg1TIII) drives peripheral nerve myelination by activating downstream signaling pathways such as PI3K/Akt and MAPK/Erk that converge on master transcriptional regulators of myelin genes, such as Krox20. We reasoned that modulating Nrg1TIII activity may constitute a general therapeutic strategy to treat CMTs that are characterized by reduced levels of myelination. Here we show that genetic overexpression of Nrg1TIII ameliorates neurophysiological and morphological parameters in a mouse model of demyelinating CMT1B, without exacerbating the toxic gain-of-function that underlies the neuropathy. Intriguingly, the mechanism appears not to be related to Krox20 or myelin gene upregulation, but rather to a beneficial rebalancing in the stoichiometry of myelin lipids and proteins. Finally, we provide proof of principle that stimulating Nrg1TIII signaling, by pharmacological suppression of the Nrg1TIII inhibitor tumor necrosis factor-alpha-converting enzyme (TACE/ADAM17), also ameliorates the neuropathy. Thus, modulation of Nrg1TIII by TACE/ADAM17 inhibition may represent a general treatment for hypomyelinating neuropathies.
Collapse
Affiliation(s)
| | | | | | | | - Francesca Bianchi
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Ubaldo Del Carro
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | | | - Donatella Caruso
- DiSFeB-Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nico Mitro
- DiSFeB-Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Marta Pellegatta
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Carla Taveggia
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Markus H Schwab
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany.,Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Klaus-Armin Nave
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - M Laura Feltri
- DIBIT, Divisions of Genetics and Cell Biology.,Hunter James Kelly Research Institute.,Department of Neurology.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Lawrence Wrabetz
- DIBIT, Divisions of Genetics and Cell Biology.,Hunter James Kelly Research Institute.,Department of Neurology.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | | |
Collapse
|
25
|
Jorba G, Aguirre-Plans J, Junet V, Segú-Vergés C, Ruiz JL, Pujol A, Fernández-Fuentes N, Mas JM, Oliva B. In-silico simulated prototype-patients using TPMS technology to study a potential adverse effect of sacubitril and valsartan. PLoS One 2020; 15:e0228926. [PMID: 32053711 PMCID: PMC7018085 DOI: 10.1371/journal.pone.0228926] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/26/2020] [Indexed: 12/11/2022] Open
Abstract
Unveiling the mechanism of action of a drug is key to understand the benefits and adverse reactions of a medication in an organism. However, in complex diseases such as heart diseases there is not a unique mechanism of action but a wide range of different responses depending on the patient. Exploring this collection of mechanisms is one of the clues for a future personalized medicine. The Therapeutic Performance Mapping System (TPMS) is a Systems Biology approach that generates multiple models of the mechanism of action of a drug. Each molecular mechanism generated could be associated to particular individuals, here defined as prototype-patients, hence the generation of models using TPMS technology may be used for detecting adverse effects to specific patients. TPMS operates by (1) modelling the responses in humans with an accurate description of a protein network and (2) applying a Multilayer Perceptron-like and sampling strategy to find all plausible solutions. In the present study, TPMS is applied to explore the diversity of mechanisms of action of the drug combination sacubitril/valsartan. We use TPMS to generate a wide range of models explaining the relationship between sacubitril/valsartan and heart failure (the indication), as well as evaluating their association with macular degeneration (a potential adverse effect). Among the models generated, we identify a set of mechanisms of action associated to a better response in terms of heart failure treatment, which could also be associated to macular degeneration development. Finally, a set of 30 potential biomarkers are proposed to identify mechanisms (or prototype-patients) more prone of suffering macular degeneration when presenting good heart failure response. All prototype-patients models generated are completely theoretical and therefore they do not necessarily involve clinical effects in real patients. Data and accession to software are available at http://sbi.upf.edu/data/tpms/.
Collapse
Affiliation(s)
- Guillem Jorba
- Anaxomics Biotech SL, Barcelona, Catalonia, Spain
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Science, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Joaquim Aguirre-Plans
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Science, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Valentin Junet
- Anaxomics Biotech SL, Barcelona, Catalonia, Spain
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Catalonia, Spain
| | | | | | - Albert Pujol
- Anaxomics Biotech SL, Barcelona, Catalonia, Spain
| | - Narcís Fernández-Fuentes
- Department of Biosciences, U Science Tech, Universitat de Vic-Universitat Central de Catalunya, Vic, Catalonia, Spain
| | | | - Baldo Oliva
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Science, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| |
Collapse
|
26
|
Arginase Inhibition Supports Survival and Differentiation of Neuronal Precursors in Adult Alzheimer's Disease Mice. Int J Mol Sci 2020; 21:ijms21031133. [PMID: 32046281 PMCID: PMC7037054 DOI: 10.3390/ijms21031133] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 01/23/2023] Open
Abstract
Adult neurogenesis is a complex physiological process, which plays a central role in maintaining cognitive functions, and consists of progenitor cell proliferation, newborn cell migration, and cell maturation. Adult neurogenesis is susceptible to alterations under various physiological and pathological conditions. A substantial decay of neurogenesis has been documented in Alzheimer’s disease (AD) patients and animal AD models; however, several treatment strategies can halt any further decline and even induce neurogenesis. Our previous results indicated a potential effect of arginase inhibition, with norvaline, on various aspects of neurogenesis in triple-transgenic mice. To better evaluate this effect, we chronically administered an arginase inhibitor, norvaline, to triple-transgenic and wild-type mice, and applied an advanced immunohistochemistry approach with several biomarkers and bright-field microscopy. Remarkably, we evidenced a significant reduction in the density of neuronal progenitors, which demonstrate a different phenotype in the hippocampi of triple-transgenic mice as compared to wild-type animals. However, norvaline showed no significant effect upon the progenitor cell number and constitution. We demonstrated that norvaline treatment leads to an escalation of the polysialylated neuronal cell adhesion molecule immunopositivity, which suggests an improvement in the newborn neuron survival rate. Additionally, we identified a significant increase in the hippocampal microtubule-associated protein 2 stain intensity. We also explore the molecular mechanisms underlying the effects of norvaline on adult mice neurogenesis and provide insights into their machinery.
Collapse
|
27
|
Teuber-Hanselmann S, Rekowski J, Vogelgsang J, von Arnim C, Reetz K, Stang A, Jöckel KH, Wiltfang J, Esselmann H, Otto M, Tumani H, Herring A, Keyvani K. CSF and blood Kallikrein-8: a promising early biomarker for Alzheimer's disease. J Neurol Neurosurg Psychiatry 2020; 91:40-48. [PMID: 31371645 PMCID: PMC6952834 DOI: 10.1136/jnnp-2019-321073] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 11/05/2022]
Abstract
OBJECTIVE There is still an urgent need for supportive minimally invasive and cost-effective biomarkers for early diagnosis of Alzheimer's disease (AD). Previous work in our lab has identified Kallikrein-8 (KLK8) as a potential candidate since it shows an excessive increase in human brain in preclinical disease stages. The aim of this study was to evaluate the diagnostic performance of cerebrospinal fluid (CSF) and blood KLK8 for AD and mild cognitive impairment (MCI) due to AD. METHODS In this multi-centre trans-sectional study, clinical and laboratory data as well as CSF and/or blood serum samples of 237 participants, including 98 patients with mild AD, 21 with MCI due to AD and 118 controls were collected. CSF and/or serum KLK8 levels were analysed by ELISA. The diagnostic accuracy of KLK8 in CSF and blood was determined using receiver operating characteristic (ROC) analyses and compared with that of CSF core biomarkers Aβ42, P-tau and T-tau. RESULTS The diagnostic accuracy of CSF KLK8 was as good as that of core CSF biomarkers for AD (area under the curve (AUC)=0.89) and in case of MCI (AUC=0.97) even superior to CSF Aβ42. Blood KLK8 was a similarly strong discriminator for MCI (AUC=0.94) but slightly weaker for AD (AUC=0.83). CONCLUSIONS This is the first study to demonstrate the potential clinical utility of blood and CSF KLK8 as a biomarker for incipient AD. Future prospective validation studies are warranted.
Collapse
Affiliation(s)
- Sarah Teuber-Hanselmann
- Institute of Neuropathology, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Jan Rekowski
- Institute of Medical Informatics, Biometry and Epidemiology, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Jonathan Vogelgsang
- Department of Psychiatry and Psychotherapy, University Medical Center, Gottingen, Germany
| | - Christine von Arnim
- Department of Neurology, University of Ulm, Ulm, Germany.,Clinic for Neurogeriatrics and Neurological Rehabilitation, RKU-University and Rehabilitation Hospital Ulm, Ulm, Germany
| | - Kathrin Reetz
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Andreas Stang
- Center of Clinical Epidemiology, c/o Institute of Medical Informatics, Biometry and Epidemiology, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Karl-Heinz Jöckel
- Institute of Medical Informatics, Biometry and Epidemiology, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center, Gottingen, Germany.,iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | - Herrmann Esselmann
- Department of Psychiatry and Psychotherapy, University Medical Center, Gottingen, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | - Arne Herring
- Institute of Neuropathology, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
28
|
Ledonne A, Mercuri NB. On the Modulatory Roles of Neuregulins/ErbB Signaling on Synaptic Plasticity. Int J Mol Sci 2019; 21:ijms21010275. [PMID: 31906113 PMCID: PMC6981567 DOI: 10.3390/ijms21010275] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/27/2019] [Accepted: 12/29/2019] [Indexed: 12/14/2022] Open
Abstract
Neuregulins (NRGs) are a family of epidermal growth factor-related proteins, acting on tyrosine kinase receptors of the ErbB family. NRGs play an essential role in the development of the nervous system, since they orchestrate vital functions such as cell differentiation, axonal growth, myelination, and synapse formation. They are also crucially involved in the functioning of adult brain, by directly modulating neuronal excitability, neurotransmission, and synaptic plasticity. Here, we provide a review of the literature documenting the roles of NRGs/ErbB signaling in the modulation of synaptic plasticity, focusing on evidence reported in the hippocampus and midbrain dopamine (DA) nuclei. The emerging picture shows multifaceted roles of NRGs/ErbB receptors, which critically modulate different forms of synaptic plasticity (LTP, LTD, and depotentiation) affecting glutamatergic, GABAergic, and DAergic synapses, by various mechanisms. Further, we discuss the relevance of NRGs/ErbB-dependent synaptic plasticity in the control of brain processes, like learning and memory and the known involvement of NRGs/ErbB signaling in the modulation of synaptic plasticity in brain’s pathological conditions. Current evidence points to a central role of NRGs/ErbB receptors in controlling glutamatergic LTP/LTD and GABAergic LTD at hippocampal CA3–CA1 synapses, as well as glutamatergic LTD in midbrain DA neurons, thus supporting that NRGs/ErbB signaling is essential for proper brain functions, cognitive processes, and complex behaviors. This suggests that dysregulated NRGs/ErbB-dependent synaptic plasticity might contribute to mechanisms underlying different neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Ada Ledonne
- Department of Experimental Neuroscience, Santa Lucia Foundation, Via del Fosso di Fiorano, no 64, 00143 Rome, Italy;
- Correspondence: ; Tel.: +3906-501703160; Fax: +3906-501703307
| | - Nicola B. Mercuri
- Department of Experimental Neuroscience, Santa Lucia Foundation, Via del Fosso di Fiorano, no 64, 00143 Rome, Italy;
- Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier no 1, 00133 Rome, Italy
| |
Collapse
|
29
|
Nrg1 Intracellular Signaling Is Neuroprotective upon Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3930186. [PMID: 31583038 PMCID: PMC6754950 DOI: 10.1155/2019/3930186] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/13/2019] [Accepted: 07/29/2019] [Indexed: 01/08/2023]
Abstract
The schizophrenia risk gene NRG1 controls the formation of excitatory and inhibitory synapses in cortical circuits. While the expression of different NRG1 isoforms occurs during development, adult neurons primarily express the CRD-NRG1 isoform characterized by a highly conserved intracellular domain (NRG1-ICD). We and others have demonstrated that Nrg1 intracellular signaling promotes dendrite elongation and excitatory connections during neuronal development. However, the role of Nrg1 intracellular signaling in adult neurons and pathological conditions remains largely unaddressed. Here, we investigated the role of Nrg1 intracellular signaling in neuroprotection and stroke. Our bioinformatic analysis revealed the evolutionary conservation of the NRG1-ICD and a decrease in NRG1 expression with age in the human frontal cortex. Hence, we first evaluated whether Nrg1 signaling may affect pathological hallmarks in an in vitro model of neuronal senescence; however, our data failed to reveal a role for Nrg1 in the activation of the stress-related pathway p38 MAPK and DNA damage. Previous studies demonstrated that the soluble EGF domain of Nrg1 alleviated brain ischemia, a pathological process involving the generation of free radicals, reactive oxygen species (ROS), and excitotoxicity. Hence, we tested the hypothesis that Nrg1 intracellular signaling could be neuroprotective in stroke. We discovered that Nrg1 expression significantly increased neuronal survival upon oxygen-glucose deprivation (OGD), an established in vitro model for stroke. Notably, the specific activation of Nrg1 intracellular signaling by expression of the Nrg1-ICD protected neurons from OGD. Additionally, time-lapse experiments confirmed that Nrg1 intracellular signaling increased the survival of neurons exposed to OGD. Finally, we investigated the relevance of Nrg1 intracellular signaling in stroke in vivo. Using viral vectors, we expressed the Nrg1-ICD in cortical neurons and subsequently challenged them by a focal hemorrhagic stroke; our data indicated that Nrg1 intracellular signaling improved neuronal survival in the infarcted area. Altogether, these data highlight Nrg1 intracellular signaling as neuroprotective upon ischemic lesion both in vitro and in vivo. Given the complexity of the neurotoxic effects of stroke and the involvement of various mechanisms, such as the generation of ROS, excitotoxicity, and inflammation, further studies are required to determine the molecular bases of the neuroprotective effect of Nrg1 intracellular signaling. In conclusion, our research highlights the stimulation of Nrg1 intracellular signaling as a promising target for cortical stroke treatment.
Collapse
|
30
|
Polis B, Srikanth KD, Gurevich V, Gil-Henn H, Samson AO. L-Norvaline, a new therapeutic agent against Alzheimer's disease. Neural Regen Res 2019; 14:1562-1572. [PMID: 31089055 PMCID: PMC6557086 DOI: 10.4103/1673-5374.255980] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Growing evidence highlights the role of arginase activity in the manifestation of Alzheimer’s disease (AD). Upregulation of arginase was shown to contribute to neurodegeneration. Regulation of arginase activity appears to be a promising approach for interfering with the pathogenesis of AD. Therefore, the enzyme represents a novel therapeutic target. In this study, we administered an arginase inhibitor, L-norvaline (250 mg/L), for 2.5 months to a triple-transgenic model (3×Tg-AD) harboring PS1M146V, APPSwe, and tauP301L transgenes. Then, the neuroprotective effects of L-norvaline were evaluated using immunohistochemistry, proteomics, and quantitative polymerase chain reaction assays. Finally, we identified the biological pathways activated by the treatment. Remarkably, L-norvaline treatment reverses the cognitive decline in AD mice. The treatment is neuroprotective as indicated by reduced beta-amyloidosis, alleviated microgliosis, and reduced tumor necrosis factor transcription levels. Moreover, elevated levels of neuroplasticity related postsynaptic density protein 95 were detected in the hippocampi of mice treated with L-norvaline. Furthermore, we disclosed several biological pathways, which were involved in cell survival and neuroplasticity and were activated by the treatment. Through these modes of action, L-norvaline has the potential to improve the symptoms of AD and even interferes with its pathogenesis. As such, L-norvaline is a promising neuroprotective molecule that might be tailored for the treatment of a range of neurodegenerative disorders. The study was approved by the Bar-Ilan University Animal Care and Use Committee (approval No. 82-10-2017) on October 1, 2017.
Collapse
Affiliation(s)
- Baruh Polis
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine; Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Kolluru D Srikanth
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Vyacheslav Gurevich
- Laboratory of Cancer Personalized Medicine and Diagnostic Genomics, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Hava Gil-Henn
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Abraham O Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
31
|
Administration of Momordica charantia Enhances the Neuroprotection and Reduces the Side Effects of LiCl in the Treatment of Alzheimer's Disease. Nutrients 2018; 10:nu10121888. [PMID: 30513908 PMCID: PMC6316175 DOI: 10.3390/nu10121888] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/14/2018] [Accepted: 11/28/2018] [Indexed: 12/15/2022] Open
Abstract
Recently, the use of natural food supplements to reduce the side effects of chemical compounds used for the treatment of various diseases has become popular. Lithium chloride (LiCl) has some protective effects in neurological diseases, including Alzheimer’s disease (AD). However, its toxic effects on various systems and some relevant interactions with other drugs limit its broader use in clinical practice. In this study, we investigated the in vitro and in vivo pharmacological functions of LiCl combined with Momordica charantia (MC) in the treatment of AD. The in vitro results show that the order of the neuroprotective effect is MC5, MC3, MC2, and MC5523 under hyperglycemia or tau hyperphosphorylation. Therefore, MC5523 (80 mg/kg; oral gavage) and/or LiCl (141.3 mg/kg; intraperitoneal injection) were applied to ovariectomized (OVX) 3×Tg-AD female and C57BL/6J (B6) male mice that received intracerebroventricular injections of streptozotocin (icv-STZ, 3 mg/kg) for 28 days. We found that the combined treatment not only increased the survival rate by reducing hepatotoxicity but also increased neuroprotection associated with anti-gliosis in the icv-STZ OVX 3×Tg-AD mice. Furthermore, the cotreatment with MC5523 and LiCl prevented memory deficits associated with reduced neuronal loss, gliosis, oligomeric Aβ level, and tau hyperphosphorylation and increased the expression levels of synaptic-related protein and pS9-GSK3β (inactive form) in the icv-STZ B6 mice. Therefore, MC5523 combined with LiCl could be a potential strategy for the treatment of AD.
Collapse
|
32
|
Sasmita AO. Current viral-mediated gene transfer research for treatment of Alzheimer’s disease. Biotechnol Genet Eng Rev 2018; 35:26-45. [DOI: 10.1080/02648725.2018.1523521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Andrew Octavian Sasmita
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
33
|
Tamura H, Shiosaka S, Morikawa S. Trophic modulation of gamma oscillations: The key role of processing protease for Neuregulin-1 and BDNF precursors. Neurochem Int 2018; 119:2-10. [DOI: 10.1016/j.neuint.2017.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/11/2017] [Accepted: 12/08/2017] [Indexed: 12/26/2022]
|
34
|
Soluble epoxide hydrolase inhibition alleviated cognitive impairments via NRG1/ErbB4 signaling after chronic cerebral hypoperfusion induced by bilateral carotid artery stenosis in mice. Brain Res 2018; 1699:89-99. [PMID: 30343686 DOI: 10.1016/j.brainres.2018.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/01/2018] [Accepted: 07/02/2018] [Indexed: 12/19/2022]
Abstract
Cerebral ischemic stroke is associated with a high rate of incidence, prevalence and mortality globally. Carotid artery stenosis, which is mainly caused by atherosclerosis plaque, results in chronic cerebral hypoperfusion and predominantly increases the risk of ischemic stroke. In the present study, we used bilateral common carotid artery stenosis (BCAS) model by placing microcoils of 0.18 mm diameter encompassing both common carotid arteries respectively, to mimic the pathogenesis of carotid artery atherosclerosis and intensively explore the pathology. We found that BCAS injury for 1 month impaired spatial cognitive functions significantly, and inhibited synaptic plasticity, including hippocampal long-term potentiation (LTP) inhibition, dendritic spine density reduction and synaptic associative proteins disorder. BCAS-induced cerebral hypoperfused mice treated with 1-(1-propanoylpiperidin-4-yl)-3-[4-(trifluoromethoxy)phenyl]urea (TPPU), a potent soluble epoxide hydrolase (sEH) inhibitor, exhibited amelioration of cognitive dysfunction and improved synaptic plasticity. The neural protective effects of TPPU on BCAS-induced cerebral hypoperfusion might due to activation of neuregulin-1 (NRG1)/ErbB4 signaling, and triggered PI3K-Akt pathways subsequently. Our results suggested that sEH inhibition could exert multi-target protective effects and alleviate spatial cognitive dysfunctions after chronic cerebral hypoperfusion in mice.
Collapse
|