1
|
Xiao Q, Wang D, Li D, Huang J, Ma F, Zhang H, Sheng Y, Zhang C, Ha X. Protein kinase C: A potential therapeutic target for endothelial dysfunction in diabetes. J Diabetes Complications 2023; 37:108565. [PMID: 37540984 DOI: 10.1016/j.jdiacomp.2023.108565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/13/2023] [Accepted: 07/22/2023] [Indexed: 08/06/2023]
Abstract
Protein kinase C (PKC) is a family of serine/threonine protein kinases that play an important role in many organs and systems and whose activation contributes significantly to endothelial dysfunction in diabetes. The increase in diacylglycerol (DAG) under high glucose conditions mediates PKC activation and synthesis, which stimulates oxidative stress and inflammation, resulting in impaired endothelial cell function. This article reviews the contribution of PKC to the development of diabetes-related endothelial dysfunction and summarizes the drugs that inhibit PKC activation, with the aim of exploring therapeutic modalities that may alleviate endothelial dysfunction in diabetic patients.
Collapse
Affiliation(s)
- Qian Xiao
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Dan Wang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Danyang Li
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Jing Huang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Feifei Ma
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, Gansu, China
| | - Haocheng Zhang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; The Second School of Clinical Medicine, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Yingda Sheng
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Caimei Zhang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Xiaoqin Ha
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China.
| |
Collapse
|
2
|
Lordén G, Newton A. Conventional protein kinase C in the brain: repurposing cancer drugs for neurodegenerative treatment? Neuronal Signal 2021; 5:NS20210036. [PMID: 34737895 PMCID: PMC8536831 DOI: 10.1042/ns20210036] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
Protein Kinase C (PKC) isozymes are tightly regulated kinases that transduce a myriad of signals from receptor-mediated hydrolysis of membrane phospholipids. They play an important role in brain physiology, and dysregulation of PKC activity is associated with neurodegeneration. Gain-of-function mutations in PKCα are associated with Alzheimer's disease (AD) and mutations in PKCγ cause spinocerebellar ataxia (SCA) type 14 (SCA14). This article presents an overview of the role of the conventional PKCα and PKCγ in neurodegeneration and proposes repurposing PKC inhibitors, which failed in clinical trials for cancer, for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Gema Lordén
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92037, U.S.A
| | - Alexandra C. Newton
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92037, U.S.A
| |
Collapse
|
3
|
Cheng XS, Shi FX, Zhao KP, Lin W, Li XY, Zhang J, Bu YY, Zhu R, Li XH, Duan DX, Ji XY, Wei JS, Wang JZ, Du J, Zhou XW. Nmnat2 attenuates amyloidogenesis and up-regulates ADAM10 in AMPK activity-dependent manner. Aging (Albany NY) 2021; 13:23620-23636. [PMID: 34644262 PMCID: PMC8580354 DOI: 10.18632/aging.203634] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/03/2021] [Indexed: 12/16/2022]
Abstract
Amyloid-β (Aβ) accumulating is considered as a causative factor for formation of senile plaque in Alzheimer’s disease (AD), but its mechanism is still elusive. The Nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2), a key redox cofactor for energy metabolism, is reduced in AD. Accumulative evidence has shown that the decrease of α-secretase activity, a disintegrin and metalloprotease domain 10 (ADAM10), is responsible for the increase of Aβ productions in AD patient’s brain. Here, we observe that the activity of α-secretase ADAM10 and levels of Nmnat2 are significantly decreased, meanwhile there is a simultaneous elevation of Aβ in Tg2576 mice. Over-expression of Nmnat2 increases the mRNA expression of α-secretase ADAM10 and its activity and inhibits Aβ production in N2a/APPswe cells, which can be abolished by Compound C, an AMPK antagonist, suggesting that AMPK is involved in over-expression of Nmnat2 against Aβ production. The further assays demonstrate that Nmnat2 activates AMPK by up-regulating the ratio of NAD+/NADH, moreover AMPK agonist AICAR can also increase ADAM10 activity and reduces Aβ1-40/1-42. Taken together, Nmnat2 suppresses Aβ production and up-regulates ADAM10 in AMPK activity-dependent manner, suggesting that Nmnat2 may serve as a new potential target in arresting AD.
Collapse
Affiliation(s)
- Xiang-Shu Cheng
- Department of Neurology, Translational Medicine Center, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China.,Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Brain Research Laboratory, Henan University, Kaifeng 475004, Henan, China
| | - Fang-Xiao Shi
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Kun-Peng Zhao
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Department of Psychiatry, Henan Key Lab of Biological Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, Henan, China
| | - Wang Lin
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xiao-Ying Li
- Department of Neurology, Translational Medicine Center, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China
| | - Jun Zhang
- Department of Neurology, Translational Medicine Center, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China
| | - Yao-Yao Bu
- Department of Neurology, Translational Medicine Center, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China
| | - Rui Zhu
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xiao-Hong Li
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Dong-Xiao Duan
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Department of Physiology, Basic Medical College, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xin-Ying Ji
- Department of Microbiology, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan, China
| | - Jian-She Wei
- Brain Research Laboratory, Henan University, Kaifeng 475004, Henan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jin Du
- Department of Neurology, Translational Medicine Center, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China.,Department of Respiratory, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China.,Brain Research Laboratory, Henan University, Kaifeng 475004, Henan, China
| | - Xin-Wen Zhou
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| |
Collapse
|
4
|
Cong W, Meng X, Li J, Zhang Q, Chen F, Liu W, Wang Y, Cheng S, Yao X, Yan J, Kim S, Saykin AJ, Liang H, Shen L. Genome-wide network-based pathway analysis of CSF t-tau/Aβ1-42 ratio in the ADNI cohort. BMC Genomics 2017; 18:421. [PMID: 28558704 PMCID: PMC5450240 DOI: 10.1186/s12864-017-3798-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 05/16/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The cerebrospinal fluid (CSF) levels of total tau (t-tau) and Aβ1-42 are potential early diagnostic markers for probable Alzheimer's disease (AD). The influence of genetic variation on these CSF biomarkers has been investigated in candidate or genome-wide association studies (GWAS). However, the investigation of statistically modest associations in GWAS in the context of biological networks is still an under-explored topic in AD studies. The main objective of this study is to gain further biological insights via the integration of statistical gene associations in AD with physical protein interaction networks. RESULTS The CSF and genotyping data of 843 study subjects (199 CN, 85 SMC, 239 EMCI, 207 LMCI, 113 AD) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were analyzed. PLINK was used to perform GWAS on the t-tau/Aβ1-42 ratio using quality controlled genotype data, including 563,980 single nucleotide polymorphisms (SNPs), with age, sex and diagnosis as covariates. Gene-level p-values were obtained by VEGAS2. Genes with p-value ≤ 0.05 were mapped on to a protein-protein interaction (PPI) network (9,617 nodes, 39,240 edges, from the HPRD Database). We integrated a consensus model strategy into the iPINBPA network analysis framework, and named it as CM-iPINBPA. Four consensus modules (CMs) were discovered by CM-iPINBPA, and were functionally annotated using the pathway analysis tool Enrichr. The intersection of four CMs forms a common subnetwork of 29 genes, including those related to tau phosphorylation (GSK3B, SUMO1, AKAP5, CALM1 and DLG4), amyloid beta production (CASP8, PIK3R1, PPA1, PARP1, CSNK2A1, NGFR, and RHOA), and AD (BCL3, CFLAR, SMAD1, and HIF1A). CONCLUSIONS This study coupled a consensus module (CM) strategy with the iPINBPA network analysis framework, and applied it to the GWAS of CSF t-tau/Aβ1-42 ratio in an AD study. The genome-wide network analysis yielded 4 enriched CMs that share not only genes related to tau phosphorylation or amyloid beta production but also multiple genes enriching several KEGG pathways such as Alzheimer's disease, colorectal cancer, gliomas, renal cell carcinoma, Huntington's disease, and others. This study demonstrated that integration of gene-level associations with CMs could yield statistically significant findings to offer valuable biological insights (e.g., functional interaction among the protein products of these genes) and suggest high confidence candidates for subsequent analyses.
Collapse
Affiliation(s)
- Wang Cong
- College of Automation, Harbin Engineering University, 145 Nantong Street, BLDG 61-5029, Harbin, 150001 China
| | - Xianglian Meng
- College of Automation, Harbin Engineering University, 145 Nantong Street, BLDG 61-5029, Harbin, 150001 China
- Harbin Huade University, No.288 Xue Yuan Rd. Limin Development Zone, Harbin, 150025 China
| | - Jin Li
- College of Automation, Harbin Engineering University, 145 Nantong Street, BLDG 61-5029, Harbin, 150001 China
| | - Qiushi Zhang
- College of Automation, Harbin Engineering University, 145 Nantong Street, BLDG 61-5029, Harbin, 150001 China
- College of Information Engineering, Northeast Dianli University, 169 Changchun Street, Jilin City, Jilin 132012 China
| | - Feng Chen
- College of Automation, Harbin Engineering University, 145 Nantong Street, BLDG 61-5029, Harbin, 150001 China
| | - Wenjie Liu
- College of Automation, Harbin Engineering University, 145 Nantong Street, BLDG 61-5029, Harbin, 150001 China
| | - Ying Wang
- College of Automation, Harbin Engineering University, 145 Nantong Street, BLDG 61-5029, Harbin, 150001 China
| | - Sipu Cheng
- College of Automation, Harbin Engineering University, 145 Nantong Street, BLDG 61-5029, Harbin, 150001 China
| | - Xiaohui Yao
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St, Suite 4100, Indianapolis, IN 46202 USA
- School of Informatics and Computing, Indiana University, 719 Indiana Avenue, Indianapolis, IN 46202 USA
| | - Jingwen Yan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St, Suite 4100, Indianapolis, IN 46202 USA
- School of Informatics and Computing, Indiana University, 719 Indiana Avenue, Indianapolis, IN 46202 USA
- Indiana University Network Science Institute, Bloomington, IN 47405 USA
| | - Sungeun Kim
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St, Suite 4100, Indianapolis, IN 46202 USA
- Indiana University Network Science Institute, Bloomington, IN 47405 USA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St, Suite 4100, Indianapolis, IN 46202 USA
- Indiana University Network Science Institute, Bloomington, IN 47405 USA
| | - Hong Liang
- College of Automation, Harbin Engineering University, 145 Nantong Street, BLDG 61-5029, Harbin, 150001 China
| | - Li Shen
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St, Suite 4100, Indianapolis, IN 46202 USA
- School of Informatics and Computing, Indiana University, 719 Indiana Avenue, Indianapolis, IN 46202 USA
| | - for the Alzheimer’s Disease Neuroimaging Initiative
- College of Automation, Harbin Engineering University, 145 Nantong Street, BLDG 61-5029, Harbin, 150001 China
- Harbin Huade University, No.288 Xue Yuan Rd. Limin Development Zone, Harbin, 150025 China
- College of Information Engineering, Northeast Dianli University, 169 Changchun Street, Jilin City, Jilin 132012 China
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St, Suite 4100, Indianapolis, IN 46202 USA
- School of Informatics and Computing, Indiana University, 719 Indiana Avenue, Indianapolis, IN 46202 USA
- Indiana University Network Science Institute, Bloomington, IN 47405 USA
| |
Collapse
|
5
|
Tam JHK, Cobb MR, Seah C, Pasternak SH. Tyrosine Binding Protein Sites Regulate the Intracellular Trafficking and Processing of Amyloid Precursor Protein through a Novel Lysosome-Directed Pathway. PLoS One 2016; 11:e0161445. [PMID: 27776132 PMCID: PMC5077117 DOI: 10.1371/journal.pone.0161445] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 08/07/2016] [Indexed: 01/18/2023] Open
Abstract
The amyloid hypothesis posits that the production of β-amyloid (Aβ) aggregates leads to neurodegeneration and cognitive decline associated with AD. Aβ is produced by sequential cleavage of the amyloid precursor protein (APP) by β- and γ-secretase. While nascent APP is well known to transit to the endosomal/ lysosomal system via the cell surface, we have recently shown that APP can also traffic to lysosomes intracellularly via its interaction with AP-3. Because AP-3 interacts with cargo protein via interaction with tyrosine motifs, we mutated the three tyrosines motif in the cytoplasmic tail of APP. Here, we show that the YTSI motif interacts with AP-3, and phosphorylation of the serine in this motif disrupts the interaction and decreases APP trafficking to lysosomes. Furthermore, we show that phosphorylation at this motif can decrease the production of neurotoxic Aβ 42. This demonstrates that reducing APP trafficking to lysosomes may be a strategy to reduce Aβ 42 in Alzheimer’s disease.
Collapse
Affiliation(s)
- Joshua H. K. Tam
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, Western University, London Ontario, Canada, N6A 5B7
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada, N6A 5B7
| | - M. Rebecca Cobb
- Program in Neuroscience, Western University, London, Ontario, Canada, N6A 5B7
| | - Claudia Seah
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, Western University, London Ontario, Canada, N6A 5B7
| | - Stephen H. Pasternak
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, Western University, London Ontario, Canada, N6A 5B7
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada, N6A 5B7
- Program in Neuroscience, Western University, London, Ontario, Canada, N6A 5B7
- Department of Clinical Neurological Sciences, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada, N6A 5B7
- * E-mail:
| |
Collapse
|
6
|
Lovestone S. Fleshing out the amyloid cascade hypothesis: the molecular biology of Alzheimer's disease. DIALOGUES IN CLINICAL NEUROSCIENCE 2012. [PMID: 22033981 PMCID: PMC3181594 DOI: 10.31887/dcns.2000.2.2/slovestone] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Alzheimer's disease (AD) is a disorder of two pathologies- plaques and tangles. The former have as a key constituent amyloid protein and the latter the microtubule-associaied protein tau. Genetics has demonstrated that changes in either protein are sufficient to cause dementia. The amyloid cascade hypothesis proposes that plaque-related changes precede tangle-related changes and positions amyloid as central to the degeneration of AD. All the evidence suggests this is correct, including evidence that presenil ins alter the processing of the amyloid precursor protein and evidence that disrupting the normal properties of tau underlies the related froniotemporal dementias. The amyloid cascade hypothesis has provided the basis for nearly a decade of intensive basic science - the skeleton of that hypothesis can now be fleshed out, and confidence is growing that this will result in useful disease-modifying therapies in the future.
Collapse
Affiliation(s)
- S Lovestone
- Author affiliations: Institute of Psychiatry, De Crespigny Park, London, UK
| |
Collapse
|
7
|
Haberman Y, Alon LT, Eliyahu E, Shalgi R. Receptor for activated C kinase (RACK) and protein kinase C (PKC) in egg activation. Theriogenology 2011; 75:80-9. [DOI: 10.1016/j.theriogenology.2010.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Revised: 07/18/2010] [Accepted: 07/18/2010] [Indexed: 11/24/2022]
|
8
|
Luciano M, Hansell NK, Lahti J, Davies G, Medland SE, Räikkönen K, Tenesa A, Widen E, McGhee KA, Palotie A, Liewald D, Porteous DJ, Starr JM, Montgomery GW, Martin NG, Eriksson JG, Wright MJ, Deary IJ. Whole genome association scan for genetic polymorphisms influencing information processing speed. Biol Psychol 2010; 86:193-202. [PMID: 21130836 DOI: 10.1016/j.biopsycho.2010.11.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 11/19/2010] [Accepted: 11/25/2010] [Indexed: 12/22/2022]
Abstract
Processing speed is an important cognitive function that is compromised in psychiatric illness (e.g., schizophrenia, depression) and old age; it shares genetic background with complex cognition (e.g., working memory, reasoning). To find genes influencing speed we performed a genome-wide association scan in up to three cohorts: Brisbane (mean age 16 years; N = 1659); LBC1936 (mean age 70 years, N = 992); LBC1921 (mean age 82 years, N = 307), and; HBCS (mean age 64 years, N =1080). Meta-analysis of the common measures highlighted various suggestively significant (p < 1.21 × 10⁻⁵) SNPs and plausible candidate genes (e.g., TRIB3). A biological pathways analysis of the speed factor identified two common pathways from the KEGG database (cell junction, focal adhesion) in two cohorts, while a pathway analysis linked to the GO database revealed common pathways across pairs of speed measures (e.g., receptor binding, cellular metabolic process). These highlighted genes and pathways will be able to inform future research, including results for psychiatric disease.
Collapse
Affiliation(s)
- Michelle Luciano
- Centre for Cognitive Aging and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Scotland, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Wang X, Chen Y, Wang X, Lu L. Genetic Regulatory Network Analysis forAppBased on Genetical Genomics Approach. Exp Aging Res 2010; 36:79-93. [DOI: 10.1080/03610730903418729] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
10
|
Weinreb O, Amit T, Mandel S, Youdim MBH. Neuroprotective molecular mechanisms of (-)-epigallocatechin-3-gallate: a reflective outcome of its antioxidant, iron chelating and neuritogenic properties. GENES AND NUTRITION 2009; 4:283-96. [PMID: 19756809 DOI: 10.1007/s12263-009-0143-4] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2009] [Accepted: 08/03/2009] [Indexed: 01/08/2023]
Abstract
Tea, the major source of dietary flavonoids, particularly the epicatechins, signifies the second most frequently consumed beverage worldwide, which varies its status from a simple ancient cultural drink to a nutrient component, endowed possible beneficial neuro-pharmacological actions. Accumulating evidence suggests that oxidative stress, resulting in reactive oxygen species generation, plays a pivotal role in neurodegenerative diseases, supporting the implementation of radical scavengers and metal chelating agents, such as natural tea polyphenols, for therapy. Vast epidemiology data indicate a correlation between occurrence of neurodegenerative disorders, such as Parkinson's and Alzheimer's diseases, and green tea consumption. In particular, recent literature strengthens the perception that diverse molecular signaling pathways, participating in the neuroprotective activity of the major green tea polyphenol, (-)-epigallocatechin-3-gallate (EGCG), renders this natural compound as potential agent to reduce the risk of various neurodegenerative diseases. In the current review, we discuss the studies concerning the mechanisms of action implicated in EGCG-induced neuroprotection and discuss the vision to translate these findings into a lifestyle arena.
Collapse
Affiliation(s)
- Orly Weinreb
- Eve Topf and USA National Parkinson Foundation Centers of Excellence for Neurodegenerative Diseases Research, Technion-Faculty of Medicine, Haifa, Israel.
| | | | | | | |
Collapse
|
11
|
ADAM-10-mediated N-cadherin cleavage is protein kinase C-alpha dependent and promotes glioblastoma cell migration. J Neurosci 2009; 29:4605-15. [PMID: 19357285 DOI: 10.1523/jneurosci.5126-08.2009] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
MMPs (matrix metalloproteinases) and the related "a disintegrin and metalloproteinases" (ADAMs) promote tumorigenesis by cleaving extracellular matrix and protein substrates, including N-cadherin. Although N-cadherin is thought to regulate cell adhesion, migration, and invasion, its role has not been characterized in glioblastomas (GBMs). In this study, we investigated the expression and function of posttranslational N-cadherin cleavage in GBM cells as well as its regulation by protein kinase C (PKC). N-Cadherin cleavage occurred at a higher level in glioblastoma cells than in non-neoplastic astrocytes. Treatment with the PKC activator phorbol 12-myristate 13-acetate (PMA) increased N-cadherin cleavage, which was reduced by pharmacological inhibitors and short interfering RNA (siRNA) specific for ADAM-10 or PKC-alpha. Furthermore, treatment of GBM cells with PMA induced the translocation of ADAM-10 to the cell membrane, the site at which N-cadherin was cleaved, and this translocation was significantly reduced by the PKC-alpha inhibitor Gö6976 [12-(2-cyanoethyl)-6,7,12,13-tetrahydro-13-methyl-5-oxo-5H-indolo[2,3-a]pyrrolo[3,4-c]carbazole] or PKC-alpha short hairpin RNA. In functional studies, N-cadherin cleavage was required for GBM cell migration, as depletion of N-cadherin cleavage by N-cadherin siRNA, ADAM-10 siRNA, or a cleavage-site mutant N-cadherin, decreased GBM cell migration. Together, these results suggest that N-cadherin cleavage is regulated by a PKC-alpha-ADAM-10 cascade in GBM cells and may be involved in mediating GBM cell migration.
Collapse
|
12
|
da Cruz e Silva OAB, Rebelo S, Vieira SI, Gandy S, da Cruz e Silva EF, Greengard P. Enhanced generation of Alzheimer's amyloid-beta following chronic exposure to phorbol ester correlates with differential effects on alpha and epsilon isozymes of protein kinase C. J Neurochem 2008; 108:319-30. [PMID: 19012746 DOI: 10.1111/j.1471-4159.2008.05770.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Alzheimer's amyloid precursor protein (APP) sorting and processing are modulated through signal transduction mechanisms regulated by protein phosphorylation. Notably, protein kinase C (PKC) appears to be an important component in signaling pathways that control APP metabolism. PKCs exist in at least 11 conventional and unconventional isoforms, and PKCalpha and PKCepsilon isoforms have been specifically implicated in controlling the generation of soluble APP and amyloid-beta (Abeta) fragments of APP, although identification of the PKC substrate phospho-state-sensitive effector proteins remains challenging. In the current study, we present evidence that chronic application of phorbol esters to cultured cells in serum-free medium is associated with several phenomena, namely: (i) PKCalpha down-regulation; (ii) PKCepsilon up-regulation; (iii) accumulation of APP and/or APP carboxyl-terminal fragments in the trans Golgi network; (iv) disappearance of fluorescence from cytoplasmic vesicles bearing a green fluorescent protein tagged form of APP; (v) insensitivity of soluble APP release following acute additional phorbol application; and (vi) elevated cellular APP mRNA levels and holoprotein, and secreted Abeta. These data indicate that, unlike acute phorbol ester application, which is accompanied by lowered Abeta generation, chronic phorbol ester treatment causes differential regulation of PKC isozymes and increased Abeta generation. These data have implications for the design of amyloid-lowering strategies based on modulating PKC activity.
Collapse
|
13
|
Rajagopal S, Fang H, Patanavanich S, Sando JJ, Kamatchi GL. Protein kinase C isozyme-specific potentiation of expressed Ca v 2.3 currents by acetyl-beta-methylcholine and phorbol-12-myristate, 13-acetate. Brain Res 2008; 1210:1-10. [PMID: 18420182 DOI: 10.1016/j.brainres.2008.03.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Accepted: 03/07/2008] [Indexed: 11/18/2022]
Abstract
Protein kinase C (PKC) is implicated in the potentiation of Ca v 2.3 currents by acetyl-beta-methylcholine (MCh), a muscarinic M1 receptor agonist or phorbol-12-myristate, 13-acetate (PMA). The PKC isozymes responsible for the action of MCh and PMA were investigated using translocation as a measure of activation and with isozyme-selective antagonists and siRNA. Ca v channels were expressed with alpha1 2.3, beta1b and alpha2delta subunits and muscarinic M1 receptors in the Xenopus oocytes and the expressed currents (I Ba) were studied using Ba2+ as the charge carrier. Translocation of PKC isozymes to the membrane studied by Western blot revealed that all eleven known PKC isozymes are present in the Xenopus oocytes. Exposure of the oocytes to MCh led to the translocation of PKC alpha whereas PMA activated PKC betaII and epsilon isozymes. The action of MCh was inhibited by Go 6976, an inhibitor of cPKC isozymes or PKC alpha siRNA. PMA-induced potentiation of Ca v 2.3 currents was inhibited by CG533 53, a PKC betaII antagonist, betaIIV5.3, a peptide translocation inhibitor of PKC betaII or PKC betaII siRNA. Similarly, epsilonV1.2, a peptide translocation inhibitor of PKC epsilon or PKC epsilon siRNA inhibited PMA action. The inhibitors of PKC increased the basal I Ba slightly. It is possible that some PKC isozymes have negative control over the I Ba. Our results implicate PKC alpha in the potentiation of Ca v 2.3 currents by MCh and PKC betaII and epsilon in the potentiation of Ca v 2.3 currents by PMA.
Collapse
Affiliation(s)
- Senthilkumar Rajagopal
- Department of Anesthesiology, P.O. Box 800710, University of Virginia Health Science Systems, 1766 Lane Road, Charlottesville, VA 22908-0710, USA
| | | | | | | | | |
Collapse
|
14
|
Yang HQ, Pan J, Ba MW, Sun ZK, Ma GZ, Lu GQ, Xiao Q, Chen SD. New protein kinase C activator regulates amyloid precursor protein processing in vitro by increasing alpha-secretase activity. Eur J Neurosci 2007; 26:381-91. [PMID: 17650113 DOI: 10.1111/j.1460-9568.2007.05648.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The beta amyloid (Abeta) cascade has been at the forefront of the hypothesis used to describe the pathogenesis of Alzheimer's disease (AD). It is generally accepted that drugs that can regulate the processing of the amyloid precursor protein (APP) toward the non-amyloidogenic pathway may have a therapeutic potential. Previous studies have shown that protein kinase C (PKC) hypofunction has an important role in AD pathophysiology. Therefore, the effects of a new PKC activator, alpha-APP modulator [(2S,5S)-(E,E)-8-(5-(4-(trifluoromethyl)phenyl)-2,4-pentadienoylamino)benzolactam (TPPB)], on APP processing were investigated. Using PC12 cells and SH-SY5Y(APP695) cells, it was found that TPPB promoted the secretion of sAPPalpha without affecting full-length expression of APP. The increase in sAPPalpha by TPPB was blocked by inhibitors of PKC, extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and tyrosine kinase, suggesting the involvement of these signal transduction pathways. TPPB increased alpha-secretase activity [a disintegrin and metalloproteinase (ADAM)10 and 17], as shown by direct fluorescence activity detection and Western blot analysis. TPPB-induced sAPPalpha release was blocked by the metalloproteinase inhibitor TAPI-2, furin inhibitor CMK and by the protein-trafficking inhibitor brefeldin. The results also showed that TPPB decreased beta-secretase activity, Abeta40 release and beta site APP-cleaving enzyme 1 (BACE1) expression, but did not significantly affect neprilysin (NEP) and insulin-degrading enzyme (IDE) expression. Our data indicate that TPPB could direct APP processing towards the non-amyloidogenic pathway by increasing alpha-secretase activity, and suggest its therapeutic potential in AD.
Collapse
Affiliation(s)
- Hong-Qi Yang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Liron T, Seraya CB, Ish-Shalom M, Souroujon MC, Neumann D. Overexpression of amyloid precursor protein reduces epsilon protein kinase C levels. Neuroscience 2007; 146:152-9. [PMID: 17321053 DOI: 10.1016/j.neuroscience.2007.01.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 12/12/2006] [Accepted: 01/07/2007] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is characterized by extracellular deposits of amyloid beta peptide (Abeta), a peptide that is generated upon proteolytic cleavage of amyloid precursor protein (APP). The events leading to the development of AD and their sequence are not yet fully understood. Protein kinase C (PKC) has been suggested to have a significant role in controlling neuronal degeneration and in the aberrant signal transduction taking place in AD. Several studies document a deficit in PKC levels and activity in brains of AD patients when compared with those of normal controls. Such a decrease in PKC could have serious implications since certain PKC isozymes were shown to drive the APP proteolytic cleavage into a non-amyloidogenic pathway. Reduced levels of distinct PKC isozymes could thus contribute to driving APP processing toward an amyloidogenic pathway. The direct cause for the down-regulation of PKC in AD brains is still unknown. In that respect, we tested in this study whether APP may play a role in PKC reduction. We show in three different cell lines (CHO, COS and BOSC) that overexpression of APP leads to decreased PKC levels. This decrease was found to be specific for the epsilon PKC isozyme whereas the levels of delta, alpha and conventional PKC remained unchanged. Furthermore, we observed this decrease for both active, membrane-associated and inactive, cytosolic epsilon PKC. APP-driven decrease in epsilon PKC is most likely mediated by a factor in the culture medium, since transfer of medium from cultured cells overexpressing APP to naïve, non-overexpressing cells, has also led to the selective decrease in epsilon PKC levels. Taken together, our results suggest that APP expression levels may play a role in the decrease of epsilon PKC levels in AD brains and could thus affect the responsiveness of AD brain tissues to growth factors and neurotransmitters.
Collapse
Affiliation(s)
- T Liron
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, 69978 Tel-Aviv, Israel
| | | | | | | | | |
Collapse
|
16
|
Yang HQ, Ba MW, Ren RJ, Zhang YH, Ma JF, Pan J, Lu GQ, Chen SD. Mitogen activated protein kinase and protein kinase C activation mediate promotion of sAPPα secretion by deprenyl. Neurochem Int 2007; 50:74-82. [PMID: 16973242 DOI: 10.1016/j.neuint.2006.07.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Revised: 06/29/2006] [Accepted: 07/10/2006] [Indexed: 12/14/2022]
Abstract
The beta amyloid cascade plays a crucial role in the pathogenesis of Alzheimer's disease (AD). Therefore, drugs that regulate amyloid precursor protein (APP) processing toward the nonamyloidgenic pathway may have therapeutic potential. Many anti-dementia drugs can regulate APP processing in addition to their pharmacological properties. Deprenyl is a neuroprotective agent used to treat some neurodegenerative diseases, including AD. In the present study, the effects of deprenyl on APP processing were investigated. Using SK-N-SH and PC12 cells, it was demonstrated that deprenyl stimulated the release of the nonamyloidogenic alpha-secretase form of soluble APP (sAPPalpha) in a dose-dependent manner without affecting cellular APP expression. The increase of sAPPalpha secretion by deprenyl was blocked by the mitogen activated protein (MAP) kinase inhibitor U0126 and PD98059, and by the protein kinase C (PKC) inhibitor GF109203X and staurosporine, suggesting the involvement of these signal transduction pathways. Deprenyl induced phosphorylation of p42/44 MAP kinase, which was abolished by specific inhibitors of MAP kinase and PKC. Deprenyl also phosphorylated PKC and its major substrate, and myristoylated alanine-rich C kinase (MARCKS) at specific amino acid residues. The data also indicated that 10microM deprenyl successfully induced two PKC isoforms involved in the pathogenesis of AD, PKCalpha and PKCepsilon, to translocate from the cytosolic to the membrane fraction. This phenomenon was substantiated by immunocytochemistry staining. These data suggest a novel pharmacological mechanism in which deprenyl regulates the processing of APP via activation of the MAP kinase and PKC pathways, and that this mechanism may underlie the clinical efficacy of the drug in some AD patients.
Collapse
Affiliation(s)
- Hong-Qi Yang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Zhao B. The Health Effects of Tea Polyphenols and Their Antioxidant Mechanism. J Clin Biochem Nutr 2006. [DOI: 10.3164/jcbn.38.59] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
18
|
Mandel S, Weinreb O, Amit T, Youdim MBH. Mechanism of neuroprotective action of the anti-Parkinson drug rasagiline and its derivatives. ACTA ACUST UNITED AC 2005; 48:379-87. [PMID: 15850677 DOI: 10.1016/j.brainresrev.2004.12.027] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2004] [Accepted: 12/09/2004] [Indexed: 01/01/2023]
Abstract
The mitochondria are directly involved in cell survival and death. Drugs that protect mitochondria viability and prevent apoptotic cascade mechanisms involved in mitochondrial permeability transition pore (MPTp) will be cytoprotective. Rasagiline (N-propargyl-1R-aminoindan) is a novel, highly potent irreversible monoamine oxidase (MAO) B inhibitor, anti-Parkinson drug. Unlike selegiline, rasagiline is not derived from amphetamine, is not metabolized to neurotoxic l-methamphetamine derivative, nor does it have sympathomimetic activity. Rasagiline is effective as monotherapy or adjunct to L-dopa for patients with early and late Parkinson's disease (PD), and adverse events do not occur with greater frequency in subjects receiving rasagiline than those on placebo. Controlled studies indicate that it might have a disease-modifying effect in PD that may be related to neuroprotection. Its S-isomer, TVP1022, is a relatively inactive MAO inhibitor. However, both drugs have similar neuroprotective activities in neuronal cell cultures in response to various neurotoxins and in vivo (global ischemia, neurotrauma, head injury, anoxia, etc.), indicating that MAO inhibition is not a pre-requisite for neuroprotection. Structure activity studies have shown that the neuroprotective activity is associated with the propargyl moiety of rasagiline which protects mitochondrial viability and MPTp by activating Bcl-2 and protein kinase C (PKC), and down regulating pro-apoptotic FAS and Bax. Rasagiline and its derivatives also process amyloid precursor protein (APP) to the neuroprotective-neurotrophic soluble APP alpha (sAPPalpha) by PKC and MAP kinase-dependent activation of alpha-secretase. The neuroprotective activity of propargylamine has led us to develop novel bifunctional neuroprotective iron-chelating MAO-inhibiting drugs possessing propargyl moiety for the treatment of other neurodegenerative diseases.
Collapse
Affiliation(s)
- Silvia Mandel
- Eve Topf and NPF Centers of Excellence for Neurodegenerative Diseases Research, Israel
| | | | | | | |
Collapse
|
19
|
Eliyahu E, Tsaadon A, Shtraizent N, Shalgi R. The involvement of protein kinase C and actin filaments in cortical granule exocytosis in the rat. Reproduction 2005; 129:161-70. [PMID: 15695610 DOI: 10.1530/rep.1.00424] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mammalian sperm–egg fusion results in cortical granule exocytosis (CGE) and resumption of meiosis. Studies of various exocytotic cells suggest that filamentous actin (F-actin) blocks exocytosis by excluding secretory vesicles from the plasma membrane. However, the exact function of these microfilaments, in mammalian egg CGE, is still elusive. In the present study we investigated the role of actin in the process of CGE, and the possible interaction between actin and protein kinase C (PKC), by using coimmunoprecipitation, immunohistochemistry and confocal microscopy. We identified an interaction between actin and the PKC alpha isoenzyme in non-activated metaphase II (MII) eggs and in eggs activated by phorbol ester 12-O-tetradecanoyl phorbol-13-acetate (TPA). F-actin was evenly distributed throughout the egg’s cytosol with a marked concentration at the cortex and at the plasma membrane. A decrease in the fluorescence signal of F-actin, which represents its depolymerization/reorganization, was detected upon fertilization and upon parthenogenetic activation. Exposing the eggs to drugs that cause either polymerization or depolymerization of actin (jasplakinolide (JAS) and cytochalasin D (CD) respectively) did not induce or prevent CGE. However, CD, but not JAS, followed by a low dose of TPA doubled the percentage of eggs undergoing complete CGE, as compared with TPA alone. We further demonstrated that myristoylated alanin-rich C kinase substrate (MARCKS), a protein known to cross-link F-actin in other cell types, is expressed in rat eggs and is colocalized with actin. In view of our results, we suggest that the cytoskeletal cortex is not a mere physical barrier that blocks CGE, but rather a dynamic network that can be maneuvered towards allowing CGE by activated actin-associated proteins and/or by activated PKC.
Collapse
Affiliation(s)
- E Eliyahu
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | | | | | | |
Collapse
|
20
|
Weinreb O, Mandel S, Amit T, Youdim MBH. Neurological mechanisms of green tea polyphenols in Alzheimer's and Parkinson's diseases. J Nutr Biochem 2004; 15:506-16. [PMID: 15350981 DOI: 10.1016/j.jnutbio.2004.05.002] [Citation(s) in RCA: 296] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Revised: 05/10/2004] [Accepted: 05/26/2004] [Indexed: 01/04/2023]
Abstract
Tea consumption is varying its status from a mere ancient beverage and a lifestyle habit, to a nutrient endowed with possible prospective neurobiological-pharmacological actions beneficial to human health. Accumulating evidence suggest that oxidative stress resulting in reactive oxygen species generation and inflammation play a pivotal role in neurodegenerative diseases, supporting the implementation of radical scavengers, transition metal (e.g., iron and copper) chelators, and nonvitamin natural antioxidant polyphenols in the clinic. These observations are in line with the current view that polyphenolic dietary supplementation may have an impact on cognitive deficits in individuals of advanced age. As a consequence, green tea polyphenols are now being considered as therapeutic agents in well controlled epidemiological studies, aimed to alter brain aging processes and to serve as possible neuroprotective agents in progressive neurodegenerative disorders such as Parkinson's and Alzheimer's diseases. In particular, literature on the putative novel neuroprotective mechanism of the major green tea polyphenol, (-)-epigallocatechin-3-gallate, are examined and discussed in this review.
Collapse
Affiliation(s)
- Orly Weinreb
- Eve Topf and USA National Parkinson Foundation Centers of Excellence for Neurodegenerative Diseases Research and Department of Pharmacology, Rappaport Family Research Institute, Technion-Faculty of Medicine, 31096 Haifa, Israel
| | | | | | | |
Collapse
|
21
|
Lanni C, Mazzucchelli M, Porrello E, Govoni S, Racchi M. Differential involvement of protein kinase C alpha and epsilon in the regulated secretion of soluble amyloid precursor protein. ACTA ACUST UNITED AC 2004; 271:3068-75. [PMID: 15233804 DOI: 10.1111/j.1432-1033.2004.04240.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We investigated the differential role of protein kinase C (PKC) isoforms in the regulated proteolytic release of soluble amyloid precursor protein (sAPPalpha) in SH-SY5Y neuroblastoma cells. We used cells stably transfected with cDNAs encoding either PKCalpha or PKCepsilon in the antisense orientation, producing a reduction of the expression of PKCalpha and PKCepsilon, respectively. Reduced expression of PKCalpha and/or PKCepsilon did not modify the response of the kinase to phorbol ester stimulation, demonstrating translocation of the respective isoforms from the cytosolic fraction to specific intracellular compartments with an interesting differential localization of PKCalpha to the plasma membrane and PKCepsilon to Golgi-like structures. Reduced expression of PKCalpha significantly impaired the secretion of sAPPalpha induced by treatment with phorbol esters. Treatment of PKCalpha-deficient cells with carbachol induced a significant release of sAPPalpha. These results suggest that the involvement of PKCalpha in carbachol-induced sAPPalpha release is negligible. The response to carbachol is instead completely blocked in PKCepsilon-deficient cells suggesting the importance of PKCepsilon in coupling cholinergic receptors with APP metabolism.
Collapse
Affiliation(s)
- Cristina Lanni
- Department of Experimental and Applied Pharmacology, Centre of Excellence in Applied Biology and School of Pharmacy, University of Pavia, Viale Taramelli 14, 27100 Pavia, Italy
| | | | | | | | | |
Collapse
|
22
|
Zhang HY, Yan H, Tang XC. Huperzine A enhances the level of secretory amyloid precursor protein and protein kinase C-alpha in intracerebroventricular beta-amyloid-(1-40) infused rats and human embryonic kidney 293 Swedish mutant cells. Neurosci Lett 2004; 360:21-4. [PMID: 15082169 DOI: 10.1016/j.neulet.2004.01.055] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2003] [Revised: 01/01/2004] [Accepted: 01/10/2004] [Indexed: 10/26/2022]
Abstract
We examined whether huperzine A (HupA), a promising therapeutic agent for Alzheimer's disease, could alter the processing of amyloid precursor protein (APP) in rats with beta-amyloid protein-(1-40) (Abeta(1-40)) infusion into the cerebral ventricle and in human embryonic kidney 293 (HEK293sw) cells. Daily intraperitoneal administration of HupA for 12 consecutive days produced significant reversals of the Abeta(1-40)-induced down-regulation of secretory APP (APPs) and protein kinase C (PKC) in rats. In the HEK293sw cells, the level of APPs was increased significantly with HupA treatment, and there was a similar change in PKCalpha level under the same condition. However, no significant alternations in the levels of PKCdelta and PKC were found after HupA treatment. These findings suggest that HupA may affect the processing of APP by up-regulating PKC, especially PKCalpha.
Collapse
Affiliation(s)
- Hai Yan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | | | | |
Collapse
|
23
|
Mandel S, Weinreb O, Amit T, Youdim MBH. Cell signaling pathways in the neuroprotective actions of the green tea polyphenol (-)-epigallocatechin-3-gallate: implications for neurodegenerative diseases. J Neurochem 2004; 88:1555-69. [PMID: 15009657 DOI: 10.1046/j.1471-4159.2003.02291.x] [Citation(s) in RCA: 249] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Accumulating evidence supports the hypothesis that brain iron misregulation and oxidative stress (OS), resulting in reactive oxygen species (ROS) generation from H2O2 and inflammatory processes, trigger a cascade of events leading to apoptotic/necrotic cell death in neurodegenerative disorders, such as Parkinson's (PD), Alzheimer's (AD) and Huntington's diseases, and amyotrophic lateral sclerosis (ALS). Thus, novel therapeutic approaches aimed at neutralization of OS-induced neurotoxicity, support the application of ROS scavengers, transition metals (e.g. iron and copper) chelators and non-vitamin natural antioxidant polyphenols, in monotherapy, or as part of antioxidant cocktail formulation for these diseases. Both experimental and epidemiological evidence demonstrate that flavonoid polyphenols, particularly from green tea and blueberries, improve age-related cognitive decline and are neuroprotective in models of PD, AD and cerebral ischemia/reperfusion injuries. However, recent studies indicate that the radical scavenger property of green tea polyphenols is unlikely to be the sole explanation for their neuroprotective capacity and in fact, a wide spectrum of cellular signaling events may well account for their biological actions. In this article, the currently established mechanisms involved in the beneficial health action and emerging studies concerning the putative novel molecular neuroprotective activity of green tea and its major polyphenol (-)-epigallocatechin-3-gallate (EGCG), will be reviewed and discussed.
Collapse
Affiliation(s)
- Silvia Mandel
- Eve Topf and USA National Parkinson Foundation Centers of Excellence for Neurodegenerative Diseases Research and Department of Pharmacology, Technion-Faculty of Medicine, Haifa, Israel
| | | | | | | |
Collapse
|
24
|
Fährmann M, Kaufhold M, Pfeiffer AF, Seidler U. Protein kinase C-alpha attenuates cholinergically stimulated gastric acid secretion of rabbit parietal cells. Br J Pharmacol 2003; 139:545-54. [PMID: 12788814 PMCID: PMC1573865 DOI: 10.1038/sj.bjp.0705211] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
(1) The phorbolester 12-O-tetradecanoyl phorbol-13-acetate (TPA), an activator of protein kinase C (PKC), inhibits cholinergic stimulation of gastric acid secretion. We observed that this effect strongly correlated with the inhibition of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activity in rabbit parietal cells. (2) The aim of this study was to specify the function of PKC-alpha in cholinergically stimulated H(+) secretion. PKC-alpha represents the only calcium-dependent PKC isoenzyme that has been detected in rabbit parietal cells. (3) Gö 6976, an inhibitor of calcium-dependent PKC, concentration-dependently antagonized the inhibitory effect of TPA, and, therefore, revealed the action of PKC-alpha on carbachol-induced acid secretion in rabbit parietal cells. (4) TPA exerted no additive inhibition of carbachol-stimulated acid secretion if acid secretion was partially inhibited by the potent CaMKII inhibitor 1-[N,O-bis(5-isoquinolinsulfonyl)-N-methyl-L-tyrosyl]-4-phenyl-piperazine (KN-62). (5) Since both kinase modulators, TPA and KN-62, affected no divergent signal transduction pathways in the parietal cell, an in vitro model has been used to study if PKC directly targets CaMKII. CaMKII purified from parietal cell-containing gastric mucosa of pig, was transphosphorylated by purified cPKC containing PKC-alpha up to 1.8 mol P(i) per mol CaMKII in vitro. The autonomy site of CaMKII was not transphosphorylated by PKC. (6) The phosphotransferase activity of the purified CaMKII was in vitro inhibited after transphosphorylation by PKC if calmodulin was absent during transphosphorylation. Attenuation of CaMKII activity by PKC showed strong similarity to the downregulation of CaMKII by basal autophosphorylation. (7) Our results suggest that PKC-alpha and CaMKII are closely functionally linked in a cholinergically induced signalling pathway in rabbit parietal cells. We assume that in cholinergically stimulated parietal cells PKC-alpha transinhibits CaMKII activity, resulting in an attenuation of acid secretion.
Collapse
Affiliation(s)
- Michael Fährmann
- Institut für Zoophysiologie der Westfälischen Wilhelms-Universität Münster, Hindenburgplatz 55, D-48143 Münster, Germany.
| | | | | | | |
Collapse
|
25
|
Benussi L, Ghidoni R, Steinhoff T, Alberici A, Villa A, Mazzoli F, Nicosia F, Barbiero L, Broglio L, Feudatari E, Signorini S, Finckh U, Nitsch RM, Binetti G. Alzheimer disease-associated cystatin C variant undergoes impaired secretion. Neurobiol Dis 2003; 13:15-21. [PMID: 12758063 DOI: 10.1016/s0969-9961(03)00012-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
CST3 is the coding gene for cystatin C (CysC). CST3 B/B homozygosity is associated with an increased risk of developing Alzheimer disease. We performed CysC analysis on human primary skin fibroblasts obtained from donors carrying A/A, A/B, and B/B CST3. Pulse-chase experiments demonstrated that the release of the B variant of CysC has a different temporal pattern compared to that of the A one. Fibroblasts B/B homozygous displayed a reduced secretion of CysC due to a less efficient cleavage of the signal peptide, as suggested by high-resolution Western blot analysis and by in vitro assay. In the brain, the reduced level of CysC may represent the molecular factor responsible for the increased risk of Alzheimer disease.
Collapse
Affiliation(s)
- Luisa Benussi
- Neurobiology Lab, IRCCS Centro San Giovanni di Dio, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Levites Y, Amit T, Mandel S, Youdim MBH. Neuroprotection and neurorescue against Abeta toxicity and PKC-dependent release of nonamyloidogenic soluble precursor protein by green tea polyphenol (-)-epigallocatechin-3-gallate. FASEB J 2003; 17:952-4. [PMID: 12670874 DOI: 10.1096/fj.02-0881fje] [Citation(s) in RCA: 286] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Green tea extract and its main polyphenol constituent (-)-epigallocatechin-3-gallate (EGCG) possess potent neuroprotective activity in cell culture and mice model of Parkinson's disease. The central hypothesis guiding this study is that EGCG may play an important role in amyloid precursor protein (APP) secretion and protection against toxicity induced by beta-amyloid (Abeta). The present study shows that EGCG enhances (approximately 6-fold) the release of the non-amyloidogenic soluble form of the amyloid precursor protein (sAPPalpha) into the conditioned media of human SH-SY5Y neuroblastoma and rat pheochromocytoma PC12 cells. sAPPalpha release was blocked by the hydroxamic acid-based metalloprotease inhibitor Ro31-9790, which indicated mediation via alpha-secretase activity. Inhibition of protein kinase C (PKC) with the inhibitor GF109203X, or by down-regulation of PKC, blocked the EGCG-induced sAPPalpha secretion, suggesting the involvement of PKC. Indeed, EGCG induced the phosphorylation of PKC, thus identifying a novel PKC-dependent mechanism of EGCG action by activation of the non-amyloidogenic pathway. EGCG is not only able to protect, but it can rescue PC12 cells against the beta-amyloid (Abeta) toxicity in a dose-dependent manner. In addition, administration of EGCG (2 mg/kg) to mice for 7 or 14 days significantly decreased membrane-bound holoprotein APP levels, with a concomitant increase in sAPPalpha levels in the hippocampus. Consistently, EGCG markedly increased PKCalpha and PKC in the membrane and the cytosolic fractions of mice hippocampus. Thus, EGCG has protective effects against Abeta-induced neurotoxicity and regulates secretory processing of non-amyloidogenic APP via PKC pathway.
Collapse
Affiliation(s)
- Yona Levites
- Eve Topf and USA National Parkinson Foundation, Centers of Excellence for Neurodegenerative Diseases Research, Technion Faculty of Medicine, Haifa, Israel
| | | | | | | |
Collapse
|
27
|
Racchi M, Mazzucchelli M, Pascale A, Sironi M, Govoni S. Role of protein kinase Calpha in the regulated secretion of the amyloid precursor protein. Mol Psychiatry 2003; 8:209-16. [PMID: 12610653 DOI: 10.1038/sj.mp.4001204] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Protein kinase C (PKC) has a key role in the signal transduction machinery involved in the regulation of amyloid precursor protein (APP) metabolism. Direct and indirect receptor-mediated activation of PKC has been shown to increase the release of soluble APP (sAPPalpha) and reduce the secretion of beta-amyloid peptides. Experimental evidence suggests that specific isoforms of PKC, such as PKCalpha and PKC epsilon, are involved in the regulation of APP metabolism. In this study, we characterized the role of PKCalpha in the regulated secretion of APP using wild-type SH-SY5Y neuroblastoma cells and cells transfected with a plasmid expressing PKCalpha antisense cDNA. Cells expressing antisense PKCalpha secrete less sAPPalpha in response to phorbol esters. In contrast, carbachol increases the secretion of sAPPalpha to similar levels in wild-type cells and in cells transfected with antisense PKCalpha by acting on APP metabolism through an indirect pathway partially involving the activation of PKC. These results suggest that the direct PKC-dependent activation of the APP secretory pathway is compromised by reduced PKCalpha expression and a specific role of this isoform in these mechanisms. On the other hand, indirect pathways that are also partially dependent on the mitogen-activated protein kinase signal transduction mechanism remain unaffected and constitute a redundant, compensatory mechanism within the APP secretory pathway.
Collapse
Affiliation(s)
- M Racchi
- Department of Experimental and Applied Pharmacology, University of Pavia, Viale Taramelli 14, Pavia 27100, Italy
| | | | | | | | | |
Collapse
|
28
|
Pakaski M, Balaspiri L, Checler F, Kasa P. Human amyloid-beta causes changes in the levels of endothelial protein kinase C and its alpha isoform in vitro. Neurochem Int 2002; 41:409-14. [PMID: 12213228 DOI: 10.1016/s0197-0186(02)00026-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Amyloid-beta (A(beta)) deposits and neurofibrillary pathology are characteristic features of Alzheimer's disease (AD). The association of A(beta) with cerebral vessels is an intriguing feature of AD. While there is considerable evidence of altered activities of the major isoforms of protein kinase C (PKC) in the vasculature and neurons of AD brains, little is known about the relationship between the Abeta toxicity and the altered PKC levels in cerebral endothelial cells. In this study, cultured brain endothelial cells exposed to A(beta)1-40 revealed a translocation of PKC from the membrane fraction to the cytosol. The content of the isoform PKC(alpha), involved in the regulation of amyloid precursor protein (APP) secretion, was decreased in the membrane-bound fraction of rat endothelial cells and increased in the cytosol after A(beta)1-40 treatment. These data suggest that the accumulation of A(beta) peptide in the cerebral vasculature may play a significant role in the down-regulation of PKC seen in the AD cerebral vasculature.
Collapse
Affiliation(s)
- Magdolna Pakaski
- Department of Psychiatry, Alzheimer's Disease Research Centre, University of Szeged, Somogyi 4, H-6720 Szeged, Hungary.
| | | | | | | |
Collapse
|
29
|
Abstract
Upon sperm-egg interaction, an increase in intracellular calcium concentration ([Ca(2+)](i)) is observed. Several studies reported that cortical reaction (CR) can be triggered not only by a [Ca(2+)](i) rise but also by protein kinase C (PKC) activation. Because the CR is regarded as a Ca(2+)-dependent exocytotic process and because the calcium-dependent conventional PKCs (cPKC) alpha and beta II are considered as exocytosis mediators in various cell systems, we chose to study activation of the cPKC in the rat egg during in vivo fertilization and parthenogenetic activation. By using immunohistochemistry and confocal microscopy techniques, we demonstrated, for the first time, the activation of the cPKC alpha, beta I, and beta II during in vivo fertilization. All three isozymes examined presented translocation to the egg's plasma membrane as early as the sperm-binding stage. However, the kinetics of their translocation was not identical. Activation of cPKC alpha was obtained by the phorbol ester 12-O-tetradecanoyl phorbol-13-acetate (TPA) or by 1-oleoyl-2-acetylglycerol (OAG) but not by the calcium ionophore ionomycin. PKC alpha translocation was first detected 5-10 min after exposure to TPA and reached a maximum at 20 min, whereas in eggs activated by OAG, translocation of PKC alpha was observed almost immediately and reached a maximum within 5 min. These results suggest that, although [Ca(2+)](i) elevation on its own does not activate PKC alpha, it may accelerate OAG-induced PKC alpha activation. We also demonstrate a successful inhibition of the CR by a myristoylated PKC pseudosubstrate (myrPKCPsi), a specific PKC inhibitor. Our study suggests that exocytosis can be triggered independently either by a [Ca(2+)](i) rise or by PKC.
Collapse
Affiliation(s)
- Efrat Eliyahu
- Department of Embryology and Teratology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | | |
Collapse
|
30
|
Xu KP, Zoukhri D, Zieske JD, Dartt DA, Sergheraert C, Loing E, Yu FS. A role for MAP kinase in regulating ectodomain shedding of APLP2 in corneal epithelial cells. Am J Physiol Cell Physiol 2001; 281:C603-14. [PMID: 11443060 DOI: 10.1152/ajpcell.2001.281.2.c603] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported an increased secretion of amyloid precursor-like protein 2 (APLP2) in the healing corneal epithelium. The present study sought to investigate signal transduction pathways involved in APLP2 shedding in vitro. APLP2 was constitutively shed and released into culture medium in SV40-immortalized human corneal epithelial cells as assessed by Western blotting, flow cytometry, and indirect immunofluorescence. Activation of protein kinase C (PKC) by phorbol 12-myristate 13-acetate (PMA) caused significant increases in APLP2 shedding. This was inhibited by staurosporine and a PKC-epsilon-specific, N-myristoylated peptide inhibitor. Epidermal growth factor (EGF) also induced APLP2 accumulation in culture medium. Basal APLP2 shedding as well as that induced by PMA and EGF was blocked by a mitogen-activated protein kinase (MAPK) kinase inhibitor, U-0126. Our results suggest that MAPK activity accounts for basal as well as PKC- and EGF-induced APLP2 shedding. In addition, PKC-epsilon may be involved in the induction of APLP2 shedding in corneal epithelial cells.
Collapse
Affiliation(s)
- K P Xu
- Schepens Eye Research Institute and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Rossner S, Mendla K, Schliebs R, Bigl V. Protein kinase Calpha and beta1 isoforms are regulators of alpha-secretory proteolytic processing of amyloid precursor protein in vivo. Eur J Neurosci 2001; 13:1644-8. [PMID: 11328358 DOI: 10.1046/j.0953-816x.2001.01525.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have recently shown that in utero treatment of guinea pigs with the DNA methylating substance methylazoxymethanol acetate (MAM) results in neocortical microencephalopathy, increased protein kinase C (PKC) activity and altered processing of the amyloid precursor protein (APP) in neocortex of offspring. Here we show that PKCalpha and PKCbeta1 are the key regulators of alpha-secretory APP processing in guinea pig neocortex under these experimental conditions in vivo. This conclusion is based on the selective translocation of PKCalpha and PKCbeta1 isoforms to the cell membrane in MAM-treated guinea pigs, as revealed by Western blot analysis and by immunocytochemistry. Additionally, we observed that [3H]phorbol ester binding to protein kinase C increased by 38% and enhanced basal PKC activity by 58% in the neocortex of microencephalic guinea pigs. Inhibition of PKCalpha/PKCbeta1 by Gö6976 abolished this difference, suggesting that constitutive overactivation of these PKC isoforms accounts for the increase in total PKC activity. We also observed a strong positive correlation between levels of alpha-secretase-processed APP and PKC activity in the neocortex of individual animals, providing further evidence for a significant role of classical PKC isoforms in nonamyloidogenic APP processing.
Collapse
Affiliation(s)
- S Rossner
- Department of Neurochemistry, Paul Flechsig Institute for Brain Research, Jahnallee 59, 04109 Leipzig, Germany.
| | | | | | | |
Collapse
|
32
|
Roßner S, Mehlhorn G, Schliebs R, Bigl V. Increased neuronal and glial expression of protein kinase C isoforms in neocortex of transgenic Tg2576 mice with amyloid pathology. Eur J Neurosci 2001. [DOI: 10.1111/j.1460-9568.2001.01388.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Rossner S, Mehlhorn G, Schliebs R, Bigl V. Increased neuronal and glial expression of protein kinase C isoforms in neocortex of transgenic Tg2576 mice with amyloid pathology. Eur J Neurosci 2001. [DOI: 10.1046/j.1460-9568.2001.01388.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
34
|
Rossner S, Beck M, Stahl T, Mendla K, Schliebs R, Bigl V. Constitutive overactivation of protein kinase C in guinea pig brain increases alpha-secretory APP processing without decreasing beta-amyloid generation. Eur J Neurosci 2000; 12:3191-200. [PMID: 10998103 DOI: 10.1046/j.1460-9568.2000.00211.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Whilst it is generally accepted that the activation of protein kinase C (PKC) increases amyloid precursor protein (APP) secretion in vitro, the role of PKC in the regulation of APP processing and beta-amyloid generation in vivo is still not well understood. In order to address this question, we established the animal model of neocortical microencephalopathy in guinea pigs caused by in utero treatment with methylazoxymethanol acetate, a DNA-methylating substance that eliminates proliferating cells of neuroepithelial origin. The induction of this neocortical malformation is accompanied by constitutive overactivation of PKC in the neocortex of the offspring. In the cortical and hippocampal tissues of juvenile microencephalic guinea pigs (postnatal day 30), we observed significant increases in basal (by 58% and 74%, respectively,) and phorbol ester-stimulated PKC enzyme activity (by 47% and 71%) as compared to age-matched control animals. In the same cortical/hippocampal preparations of methylazoxymethanol-treated animals, there was increased alpha-secretion of APP by 35% and 30% as measured by Western blot analysis using the antibody 6E10, whilst total APP secretion as well as APP mRNA expression remained unaltered. This upregulation of APP alpha-secretion was limited to brain areas that displayed elevated PKC activity. However, constitutive overactivation of neocortical PKC did not affect the generation of beta-amyloid peptides 1-40 or 1-42 as measured by ELISA, suggesting that only the alpha-secretase pathway of APP processing is affected by chronic PKC overactivation in vivo.
Collapse
Affiliation(s)
- S Rossner
- Department of Neurochemistry, Paul Flechsig Institute for Brain Research, Jahnallee 59, 04109 Leipzig, Germany.
| | | | | | | | | | | |
Collapse
|
35
|
Dodart JC, Mathis C, Ungerer A. The beta-amyloid precursor protein and its derivatives: from biology to learning and memory processes. Rev Neurosci 2000; 11:75-93. [PMID: 10718147 DOI: 10.1515/revneuro.2000.11.2-3.75] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Intensive investigation towards the understanding of the biology and physiological functions of the beta-amyloid precursor protein (APP) have been supported since it is known that a 39-43 amino acid fragment of APP, called the beta-amyloid protein (Abeta), accumulates in the brain parenchyma to form the typical lesions associated with Alzheimer's disease (AD). It emerges from extensive data that APP and its derivatives show a wide range of contrasting physiological properties and therefore might be involved in distinct physiological functions. Abeta has been shown to disrupt neuronal activity and to demonstrate neurotoxic properties in a wide range of experimental procedures. In contrast, both in vitro and in vivo studies suggest that APP and/or its secreted forms are important factors involved in the viability, growth and morphological and functional plasticity of nerve cells. Furthermore, several recent studies suggest that APP and its derivatives have an important role in learning and memory processes. Memory impairments can be induced in animals by intracerebral treatment with Abeta. Altered expression of the APP gene in aged animals or in genetically-modified animals also leads to memory deficits. By contrast, secreted forms of APP have recently been shown to facilitate learning and memory processes in mice. These interesting findings open novel perspectives to understand the involvement of APP in the development of cognitive deficits associated with AD. In this review, we summarize the current data concerning the biology and the behavioral effects of APP and its derivatives which may be relevant to the roles of these proteins in memory and in AD pathology.
Collapse
Affiliation(s)
- J C Dodart
- Laboratoire d'Ethologie et Neurobiologie, URA-CNRS 1295, ULP, Strasbourg, France.
| | | | | |
Collapse
|
36
|
Racchi M, Govoni S. Rationalizing a pharmacological intervention on the amyloid precursor protein metabolism. Trends Pharmacol Sci 1999; 20:418-23. [PMID: 10498955 DOI: 10.1016/s0165-6147(99)01380-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The treatment of Alzheimer's disease remains a major challenge because of our incomplete understanding of the triggering events that lead to the selective neurodegeneration characteristic of Alzheimer's brains. The rational design of a pharmacological intervention is therefore a great theoretical challenge. One approach involves the study of the pharmacological modulation of the amyloid precursor protein metabolism, in which the goal is to reduce the formation of beta-amyloid in the hope of reducing the formation of a potentially neurotoxic peptide. Such an approach has led to the identification of a complex intracellular mechanism that can be regulated by neurotransmitters and other ligands.
Collapse
Affiliation(s)
- M Racchi
- Institute of Pharmacology, University of Pavia, Viale Taramelli 14 27100 Pavia, Italy.
| | | |
Collapse
|
37
|
Racchi M, Solano DC, Sironi M, Govoni S. Activity of alpha-secretase as the common final effector of protein kinase C-dependent and -independent modulation of amyloid precursor protein metabolism. J Neurochem 1999; 72:2464-70. [PMID: 10349856 DOI: 10.1046/j.1471-4159.1999.0722464.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The metabolic fate of the amyloid precursor protein (APP) is one of the key factors in the pathogenesis of Alzheimer's disease (AD). A complex cellular mechanism regulates the rate at which the precursor is cleaved by alpha-secretase and released as soluble protein in the extracellular space. We show here that alpha-secretase constitutes the common final effector of several independent means of stimulation of soluble APP (sAPP) release. The release of sAPP by alpha-secretase resembles that of several other membrane-bound proteins with soluble counterparts, a process that is sensitive to matrix metalloprotease inhibitors. The hydroxamic acid-based compound KD-IX-73-4 inhibits phorbol ester-mediated sAPP release from COS cells with an IC50 of 8 microM, consistent with previous data for the same compound against leukocyte L-selectin shedding. Beyond direct protein kinase C (PKC) activation we show that KD-IX-73-4 inhibits also receptor-mediated sAPP release induced by carbachol in SH-SY5Y cells and by bradykinin in human skin fibroblasts, with the latter being a PKC-independent mechanism. Altogether these data suggest that all pharmacological means of stimulating sAPP release converge to a hydroxamic acid-based inhibitor-sensitive proteolytic enzyme. Moreover, because KD-IX-73-4 was effective in the inhibition of stimulated but not constitutive sAPP release, these data suggest the existence of different enzymes regulating the two metabolic pathways leading to sAPP secretion.
Collapse
Affiliation(s)
- M Racchi
- IRCCS Centro San Giovanni di Dio-FBF, Brescia, Italy
| | | | | | | |
Collapse
|
38
|
Goekjian PG, Wu GZ, Chen S, Zhou L, Jirousek MR, Gillig JR, Ballas LM, Dixon JT. Synthesis of Fluorinated Macrocyclic Bis(indolyl)maleimides as Potential 19F NMR Probes for Protein Kinase C. J Org Chem 1999. [DOI: 10.1021/jo9808876] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
39
|
Jung SS, Gauthier S, Cashman NR. Beta-amyloid precursor protein is detectable on monocytes and is increased in Alzheimer's disease. Neurobiol Aging 1999; 20:249-57. [PMID: 10588572 DOI: 10.1016/s0197-4580(99)00051-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Using the anti-beta-amyloid precursor protein (betaAPP) monoclonal antibodies 4G8, 6E10 and 22C11 and flow cytometry, we report that human circulating peripheral blood monocytes display surface immunoreactivity for betaAPP. In contrast, circulating lymphocytes do not possess cell surface betaAPP immunoreactivity, despite similar levels of betaAPP expression. Immunoblotting analysis showed that monocytes, but not lymphocytes, possess an 82 kDa C-terminal betaAPP fragment consistent with a processed transmembrane species. Monocyte surface betaAPP was upregulated approximately threefold by activation with lipopolysaccharide and interferon-gamma, activation did not produce detectable betaAPP on the cell surface of lymphocytes. Surface betaAPP immunoreactivity was reduced in a normal aged population compared to normal young controls (Young = 81.07 +/- 13.67 mean fluorescence units, Aged = 36.74 +/- 3.81, p < 0.01), but was significantly increased in AD subjects compared to age-matched healthy controls (AD = 60.31 +/- 7.42, p < 0.05). Our data suggest that a proportion of peripheral A beta may be derived from monocyte/macrophages, and that defects in brain cell processing of betaAPP in AD may be shared by this readily accessible peripheral cell.
Collapse
Affiliation(s)
- S S Jung
- Department of Microbiology & Immunology, Montréal Neurological Institute, McGill University, PQ, Canada
| | | | | |
Collapse
|
40
|
Abstract
Multiple lines of evidence suggest that increased production and/or deposition of the beta-amyloid peptide, derived from the amyloid precursor protein, contributes to Alzheimer's disease. A growing list of neurotransmitters, growth factors, cytokines, and hormones have been shown to regulate amyloid precursor protein processing. Although traditionally thought to be mediated by activation of protein kinase C, recent data have implicated other signaling mechanisms in the regulation of this process. Moreover, novel mechanisms of regulation involving cholesterol-, apolipoprotein E-, and stress-activated pathways have been identified. As the phenotypic changes associated with Alzheimer's disease encompass many of these signaling systems, it is relevant to determine how altered cell signaling may be contributing to increasing brain amyloid burden. We review the myriad ways in which first messengers regulate amyloid precursor protein catabolism as well as the signal transduction cascades that give rise to these effects.
Collapse
Affiliation(s)
- J Mills
- Kinsmen Laboratory of Neurological Research, Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
41
|
Biological activities of amyloid precursor protein. ACTA BIOLOGICA HUNGARICA 1998. [DOI: 10.1007/bf03542974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
42
|
Raz T, Eliyahu E, Yesodi V, Shalgi R. Profile of protein kinase C isozymes and their possible role in mammalian egg activation. FEBS Lett 1998; 431:415-8. [PMID: 9714554 DOI: 10.1016/s0014-5793(98)00806-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Western blot analysis was used to investigate protein kinase C (PKC) profile of rat eggs. The presence of eight PKC isozymes was demonstrated: conventional PKC alpha, beta and gamma; novel PKC delta, epsilon and mu; atypical PKC zeta and lambda. PKC alpha was detected by RT-PCR as well. PKC translocation from the cytosol to the plasma membrane served as a marker for enzyme activation. Immunofluorescence confocal microscopy demonstrated a relatively uniform distribution of PKC alpha, betaI, and betaII throughout the cytosol of metaphase II arrested eggs. PKC accumulation at the plasma membrane was detected 5 min after exposure to 12-O-tetradecanoyl phorbol-13-acetate and increased with time, thus demonstrating activation of these PKCs.
Collapse
Affiliation(s)
- T Raz
- Department of Embryology and Teratology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | | | | | | |
Collapse
|