1
|
Wu X, Yang Z, Zou J, Gao H, Shao Z, Li C, Lei P. Protein kinases in neurodegenerative diseases: current understandings and implications for drug discovery. Signal Transduct Target Ther 2025; 10:146. [PMID: 40328798 PMCID: PMC12056177 DOI: 10.1038/s41392-025-02179-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/03/2025] [Accepted: 02/12/2025] [Indexed: 05/08/2025] Open
Abstract
Neurodegenerative diseases (e.g., Alzheimer's, Parkinson's, Huntington's disease, and Amyotrophic Lateral Sclerosis) are major health threats for the aging population and their prevalences continue to rise with the increasing of life expectancy. Although progress has been made, there is still a lack of effective cures to date, and an in-depth understanding of the molecular and cellular mechanisms of these neurodegenerative diseases is imperative for drug development. Protein phosphorylation, regulated by protein kinases and protein phosphatases, participates in most cellular events, whereas aberrant phosphorylation manifests as a main cause of diseases. As evidenced by pharmacological and pathological studies, protein kinases are proven to be promising therapeutic targets for various diseases, such as cancers, central nervous system disorders, and cardiovascular diseases. The mechanisms of protein phosphatases in pathophysiology have been extensively reviewed, but a systematic summary of the role of protein kinases in the nervous system is lacking. Here, we focus on the involvement of protein kinases in neurodegenerative diseases, by summarizing the current knowledge on the major kinases and related regulatory signal transduction pathways implicated in diseases. We further discuss the role and complexity of kinase-kinase networks in the pathogenesis of neurodegenerative diseases, illustrate the advances of clinical applications of protein kinase inhibitors or novel kinase-targeted therapeutic strategies (such as antisense oligonucleotides and gene therapy) for effective prevention and early intervention.
Collapse
Affiliation(s)
- Xiaolei Wu
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhangzhong Yang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinjun Zou
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Querfurth HW, Lemere C, Ciola J, Havas D, Xia W, Lee HK. Target Validation Studies of PS48, a PDK-1 Allosteric Agonist, for the Treatment of Alzheimer's Disease Phenotype in APP/PS1 Transgenic Mice. Int J Mol Sci 2025; 26:3473. [PMID: 40331945 PMCID: PMC12027031 DOI: 10.3390/ijms26083473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
The Alzheimer's disease (AD)-affected brain is known to be deficient in the utilization of glucose, its main energy substrate, and systemic diabetes is a significant risk factor for AD. In the course of biochemical and molecular investigations into this puzzling relationship, it has been shown that resistance to insulin action is a prominent feature of early stages of AD in the brain, thereby contributing to an energy failure state and a decline in synaptic function. In one AD-like cellular model, we found that β-amyloid (Aβ) accumulation inhibited insulin signaling and cell viability through an alteration of the PI3K/PDK-1/Akt signal pathway, an effect overcome by mTORC2 stimulation. A PDK-1 allosteric agonist, PS48, as well as newly synthesized analogs, were also found to reverse the metabolic defects caused by intracellular Aβ42 accumulation. In vivo, we previously showed that oral dosing of PS48 significantly improves learning and memory in APP/PS1 transgenic mice. Herein, we present evidence using unbiased immunohistological quantification and Western blot analyses demonstrating that ingested PS48 crosses into brain tissue where it targeted Akt and GSK3-β activities. Beneficial effects on neuronal number and Tau phosphorylation were found. Not unexpectedly, Aβ levels remained unchanged. These results support a path toward a future therapeutic trial of this untested strategy and agent in humans.
Collapse
Affiliation(s)
- Henry W. Querfurth
- Tufts Medical Center, Department of Neurology and Tufts University School of Medicine, Department of Neuroscience, 800 Washington St., and 136 Harrison Ave., Boston, MA 02111, USA
| | - Cynthia Lemere
- Brigham and Women’s Hospital, ARCND, 60 Fenwood Rd., Hale Bldg. for Transformative Medicine, Boston, MA 02115, USA; (C.L.); (J.C.)
| | - Jason Ciola
- Brigham and Women’s Hospital, ARCND, 60 Fenwood Rd., Hale Bldg. for Transformative Medicine, Boston, MA 02115, USA; (C.L.); (J.C.)
| | - Daniel Havas
- Psychogenics Inc., 215 College Rd., Paramus, NJ 07652, USA;
| | - Weiming Xia
- Chobanian and Avedisian School of Medicine, Department of Pharmacology, Physiology and Biophysics, Boston University, 72 E. Concord St., Boston, MA 02118, USA;
| | - Han Kyu Lee
- Tufts Medical Center, Department of Neurology, 800 Washington St., Boston, MA 02111, USA;
| |
Collapse
|
3
|
Coschiera A, Yoshihara M, Lauter G, Ezer S, Pucci M, Li H, Kavšek A, Riedel CG, Kere J, Swoboda P. Primary cilia promote the differentiation of human neurons through the WNT signaling pathway. BMC Biol 2024; 22:48. [PMID: 38413974 PMCID: PMC10900739 DOI: 10.1186/s12915-024-01845-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: 07/18/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Primary cilia emanate from most human cell types, including neurons. Cilia are important for communicating with the cell's immediate environment: signal reception and transduction to/from the ciliated cell. Deregulation of ciliary signaling can lead to ciliopathies and certain neurodevelopmental disorders. In the developing brain cilia play well-documented roles for the expansion of the neural progenitor cell pool, while information about the roles of cilia during post-mitotic neuron differentiation and maturation is scarce. RESULTS We employed ciliated Lund Human Mesencephalic (LUHMES) cells in time course experiments to assess the impact of ciliary signaling on neuron differentiation. By comparing ciliated and non-ciliated neuronal precursor cells and neurons in wild type and in RFX2 -/- mutant neurons with altered cilia, we discovered an early-differentiation "ciliary time window" during which transient cilia promote axon outgrowth, branching and arborization. Experiments in neurons with IFT88 and IFT172 ciliary gene knockdowns, leading to shorter cilia, confirm these results. Cilia promote neuron differentiation by tipping WNT signaling toward the non-canonical pathway, in turn activating WNT pathway output genes implicated in cyto-architectural changes. CONCLUSIONS We provide a mechanistic entry point into when and how ciliary signaling coordinates, promotes and translates into anatomical changes. We hypothesize that ciliary alterations causing neuron differentiation defects may result in "mild" impairments of brain development, possibly underpinning certain aspects of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Andrea Coschiera
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Masahito Yoshihara
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba, Japan
- Chiba University, Chiba, Japan
| | - Gilbert Lauter
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden
- Uppsala University, Uppsala, Sweden
| | - Sini Ezer
- University of Helsinki, Stem Cells and Metabolism Research Program, and Folkhälsan Research Center, Helsinki, Finland
| | - Mariangela Pucci
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
- Department of Bioscience and Technology for Food, Agriculture and Environment, Teramo, Italy
- University of Teramo, Teramo, Italy
| | - Haonan Li
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Alan Kavšek
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Christian G Riedel
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
- University of Helsinki, Stem Cells and Metabolism Research Program, and Folkhälsan Research Center, Helsinki, Finland
| | - Peter Swoboda
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden.
| |
Collapse
|
4
|
Alexander C, Parsaee A, Vasefi M. Polyherbal and Multimodal Treatments: Kaempferol- and Quercetin-Rich Herbs Alleviate Symptoms of Alzheimer's Disease. BIOLOGY 2023; 12:1453. [PMID: 37998052 PMCID: PMC10669725 DOI: 10.3390/biology12111453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder impairing cognition and memory in the elderly. This disorder has a complex etiology, including senile plaque and neurofibrillary tangle formation, neuroinflammation, oxidative stress, and damaged neuroplasticity. Current treatment options are limited, so alternative treatments such as herbal medicine could suppress symptoms while slowing cognitive decline. We followed PRISMA guidelines to identify potential herbal treatments, their associated medicinal phytochemicals, and the potential mechanisms of these treatments. Common herbs, including Ginkgo biloba, Camellia sinensis, Glycyrrhiza uralensis, Cyperus rotundus, and Buplerum falcatum, produced promising pre-clinical results. These herbs are rich in kaempferol and quercetin, flavonoids with a polyphenolic structure that facilitate multiple mechanisms of action. These mechanisms include the inhibition of Aβ plaque formation, a reduction in tau hyperphosphorylation, the suppression of oxidative stress, and the modulation of BDNF and PI3K/AKT pathways. Using pre-clinical findings from quercetin research and the comparatively limited data on kaempferol, we proposed that kaempferol ameliorates the neuroinflammatory state, maintains proper cellular function, and restores pro-neuroplastic signaling. In this review, we discuss the anti-AD mechanisms of quercetin and kaempferol and their limitations, and we suggest a potential alternative treatment for AD. Our findings lead us to conclude that a polyherbal kaempferol- and quercetin-rich cocktail could treat AD-related brain damage.
Collapse
Affiliation(s)
- Claire Alexander
- Department of Biology, Lamar University, Beaumont, TX 77705, USA
| | - Ali Parsaee
- Biological Science, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Maryam Vasefi
- Department of Biology, Lamar University, Beaumont, TX 77705, USA
| |
Collapse
|
5
|
Abuelezz SA, Hendawy N. Spotlight on Coenzyme Q10 in scopolamine-induced Alzheimer's disease: oxidative stress/PI3K/AKT/GSK 3ß/CREB/BDNF/TrKB. J Pharm Pharmacol 2023:rgad048. [PMID: 37315215 DOI: 10.1093/jpp/rgad048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/28/2023] [Indexed: 06/16/2023]
Abstract
OBJECTIVES Excess amyloid beta (Aβ) and oxidative stress (OS) are inextricable hallmarks of the neuronal damage associated Alzheimer's disease. Aβ-induced cognitive and memory dysfunctions are mediated through different signalling pathways as phosphatidylinositol-3-kinase (PI3K) and their downstream intermediates including protein-kinase-B, known as Akt, glycogen-synthase-kinase-3β (GSK-3β), cAMP-response-element-binding-protein (CREB), brain-derived-neurotrophic factor (BDNF) and tropomyosin-related-kinase receptor-B (TrKB). The current work aims to investigate the protective potentials of CoQ10 against scopolamine (Scop)-induced cognitive disability and the contribution of PI3K/Akt/GSK-3β/CREB/BDNF/TrKB in the neuroprotection effects. METHODS The chronic co-administration of CQ10 (50, 100 and 200 mg/kg/day i.p.) with Scop in Wistar rats for 6 weeks were assayed both behaviourally and biochemically. KEY FINDINGS CoQ10 ameliorated the Scop-induced cognitive and memory defects by restoring alterations in novel object recognition and Morris water maze behavioural tests. CoQ10 favourably changed the Scop-induced deleterious effects in hippocampal malondialdehyde, 8-hydroxy-2' deoxyguanosine, antioxidants and PI3K/Akt/GSK-3β/CREB/BDNF/TrKB levels. CONCLUSIONS These results exhibited the neuroprotective effects of CoQ10 on Scop-induced AD and revealed its ability to inhibit oxidative stress, amyloid deposition and to modulate PI3K/Akt/GSK-3β/CREB/BDNF/TrKB pathway.
Collapse
Affiliation(s)
- Sally A Abuelezz
- Clinical Pharmacology Department, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Nevien Hendawy
- Clinical Pharmacology Department, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
- Basic Medical Sciences Department, Faculty of Medicine, Faculty of Medicine, Galala University, Suez, Egypt
| |
Collapse
|
6
|
Gamit N, Dharmarajan A, Sethi G, Warrier S. Want of Wnt in Parkinson's disease: Could sFRP disrupt interplay between Nurr1 and Wnt signaling? Biochem Pharmacol 2023; 212:115566. [PMID: 37088155 DOI: 10.1016/j.bcp.2023.115566] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Nuclear receptor related 1 (Nurr1) is a transcription factor known to regulate the development and maintenance of midbrain dopaminergic (mDA) neurons. Reports have confirmed that defect or obliteration of Nurr1 results in neurodegeneration and motor function impairment leading to Parkinson's disease (PD). Studies have also indicated that Nurr1 regulates the expression of alpha-synuclein (α-SYN) and mutations in Nurr1 cause α-SYN overexpression, thereby increasing the risk of PD. Nurr1 is modulated via various pathways including Wnt signaling pathway which is known to play an important role in neurogenesis and deregulation of it contributes to PD pathogenesis. Both Wnt/β-catenin dependent and independent pathways are implicated in the activation of Nurr1 and subsequent downregulation of α-SYN. This review highlights the interaction between Nurr1 and Wnt signaling pathways in mDA neuronal development. We further hypothesize how modulation of Wnt signaling pathway by its antagonist, secreted frizzled related proteins (sFRPs) could be a potential route to treat PD.
Collapse
Affiliation(s)
- Naisarg Gamit
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600 116, India; School of Pharmacy and Biomedical Sciences, Curtin Medical School, Curtin University, Perth, Western Australia 6102, Australia; Curtin Health and Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia; School of Human Sciences, Faculty of Life and Physical Sciences, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore 117 600, Singapore
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India; Cuor Stem Cellutions Pvt Ltd, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India.
| |
Collapse
|
7
|
He C, Zhao X, Lei Y, Nie J, Lu X, Song J, Wang L, Li H, Liu F, Zhang Y, Niu Q. Whole-transcriptome analysis of aluminum-exposed rat hippocampus and identification of ceRNA networks to investigate neurotoxicity of Al. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:1401-1417. [PMID: 34900398 PMCID: PMC8636738 DOI: 10.1016/j.omtn.2021.11.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022]
Abstract
Aluminum is a known neurotoxin that can induce Aβ deposition and abnormal phosphorylation of tau protein, leading to Alzheimer disease (AD)-like damages such as neuronal damage and decreased learning and memory functions. In this study, we constructed a rat model of subchronic aluminum maltol exposure, and the whole-transcriptome sequencing was performed on the hippocampus of the control group and the middle-dose group. A total of 167 miRNAs, 37 lncRNAs, 256 mRNAs, and 64 circRNAs expression changed. The Kyoto Encyclopedia of Genes and Genomes showed that PI3K/AKT pathway was the most enriched pathway of DEGs, and IRS1 was the core molecule in the PPI network. circRNA/lncRNA-miRNA-mRNA networks of all DEGs, DEGs in the PI3K/AKT pathway, and IRS1 were constructed by Cytoscape. Molecular experiment results showed that aluminum inhibited the IRS1/PI3K/AKT pathway and increased the content of Aβ and tau. In addition, we also constructed an AAV intervention rat model, proving that inhibition of miR-96-5p expression might resist aluminum-induced injury by upregulating expression of IRS1. In general, these results suggest that the ceRNA networks are involved in the neurotoxic process of aluminum, providing a new strategy for studying the toxicity mechanism of aluminum and finding biological targets for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Chanting He
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi 030001, China
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan, Shanxi 030001, China
- Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiaoyan Zhao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yang Lei
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jisheng Nie
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiaoting Lu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jing Song
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Linping Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Huan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Fangqu Liu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yidan Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi 030001, China
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| |
Collapse
|
8
|
Villavicencio Tejo F, Quintanilla RA. Contribution of the Nrf2 Pathway on Oxidative Damage and Mitochondrial Failure in Parkinson and Alzheimer's Disease. Antioxidants (Basel) 2021; 10:1069. [PMID: 34356302 PMCID: PMC8301100 DOI: 10.3390/antiox10071069] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 12/17/2022] Open
Abstract
The increase in human life expectancy has become a challenge to reduce the deleterious consequences of aging. Nowadays, an increasing number of the population suffer from age-associated neurodegenerative diseases including Parkinson's disease (PD) and Alzheimer's disease (AD). These disorders present different signs of neurodegeneration such as mitochondrial dysfunction, inflammation, and oxidative stress. Accumulative evidence suggests that the transcriptional factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) plays a vital defensive role orchestrating the antioxidant response in the brain. Nrf2 activation promotes the expression of several antioxidant enzymes that exert cytoprotective effects against oxidative damage and mitochondrial impairment. In this context, several studies have proposed a role of Nrf2 in the pathogenesis of PD and AD. Thus, we consider it important to summarize the ongoing literature related to the effects of the Nrf2 pathway in the context of these diseases. Therefore, in this review, we discuss the mechanisms involved in Nrf2 activity and its connection with mitochondria, energy supply, and antioxidant response in the brain. Furthermore, we will lead our discussion to identify the participation of the Nrf2 pathway in mitochondrial impairment and neurodegeneration present in PD and AD. Finally, we will discuss the therapeutic effects that the Nrf2 pathway activation could have on the cognitive impairment, neurodegeneration, and mitochondrial failure present in PD and AD.
Collapse
Affiliation(s)
| | - Rodrigo A Quintanilla
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile;
| |
Collapse
|
9
|
Gupta R, Sahu M, Srivastava D, Tiwari S, Ambasta RK, Kumar P. Post-translational modifications: Regulators of neurodegenerative proteinopathies. Ageing Res Rev 2021; 68:101336. [PMID: 33775891 DOI: 10.1016/j.arr.2021.101336] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/10/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022]
Abstract
One of the hallmark features in the neurodegenerative disorders (NDDs) is the accumulation of aggregated and/or non-functional protein in the cellular milieu. Post-translational modifications (PTMs) are an essential regulator of non-functional protein aggregation in the pathogenesis of NDDs. Any alteration in the post-translational mechanism and the protein quality control system, for instance, molecular chaperone, ubiquitin-proteasome system, autophagy-lysosomal degradation pathway, enhances the accumulation of misfolded protein, which causes neuronal dysfunction. Post-translational modification plays many roles in protein turnover rate, accumulation of aggregate and can also help in the degradation of disease-causing toxic metabolites. PTMs such as acetylation, glycosylation, phosphorylation, ubiquitination, palmitoylation, SUMOylation, nitration, oxidation, and many others regulate protein homeostasis, which includes protein structure, functions and aggregation propensity. Different studies demonstrated the involvement of PTMs in the regulation of signaling cascades such as PI3K/Akt/GSK3β, MAPK cascade, AMPK pathway, and Wnt signaling pathway in the pathogenesis of NDDs. Further, mounting evidence suggests that targeting different PTMs with small chemical molecules, which acts as an inhibitor or activator, reverse misfolded protein accumulation and thus enhances the neuroprotection. Herein, we briefly discuss the protein aggregation and various domain structures of different proteins involved in the NDDs, indicating critical amino acid residues where PTMs occur. We also describe the implementation and involvement of various PTMs on signaling cascade and cellular processes in NDDs. Lastly, we implement our current understanding of the therapeutic importance of PTMs in neurodegeneration, along with emerging techniques targeting various PTMs.
Collapse
|
10
|
Changes in 24 h Rhythmicity of Spontaneous Locomotor Activity in the Triple Transgenic Mouse for Alzheimer's Disease (3xTg-AD) in a Jet Lag Protocol: Correlations with Retinal Sensitivity. J Circadian Rhythms 2021; 19:7. [PMID: 34163535 PMCID: PMC8194968 DOI: 10.5334/jcr.214] [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] [Indexed: 12/01/2022] Open
Abstract
The progression of amyloid plaques and neurofibrillary tangles in different brain areas is associated with the effects of Alzheimer’s disease (AD). In addition to cognitive impairment, circadian alterations in locomotor activity have also been detected, but they have not been characterized in a jet lag protocol. Therefore, the present study aimed to compare 3xTg-AD and non-transgenic mice in changes of 24 h cycles of spontaneous locomotor activity in a jet lag protocol, in an environment without a running wheel, at 3 different states of neuronal damage: early, intermediate and advanced (3, 8 and 13 months, respectively). The 3xTg-AD mice at 3 months presented differences in phase angle and acrophase, and differentially increased activity after advances more than after delays. At 13 months, a shortening of the free-running period in constant darkness was also noted. 3xTg-AD mice showed a significant increase (123%) in global activity at 8 to 13 months and in nighttime activity (153%) at 13 months. In the advance protocol (ADV), 3xTg-AD mice displayed a significant increase in global activity (171%) at 8 and 13 months. The differences in masking effect were evident at 8 months. To assess a possible retinal dysfunction that could interfere with photic entrainment as part of the neurodegenerative process, we compared electroretinogram recordings. The results showed early deterioration in the retinal response to light flashes in mesopic conditions, observed in the B-wave latency and amplitude. Thus, our study presents new behavioral and pathological characteristics of 3xTg-AD mice and reveals the usefulness of non-invasive tools in early diagnosis.
Collapse
|
11
|
Wang J, Chen X, Bai W, Wang Z, Xiao W, Zhu J. Study on Mechanism of Ginkgo biloba L. Leaves for the Treatment of Neurodegenerative Diseases Based on Network Pharmacology. Neurochem Res 2021; 46:1881-1894. [PMID: 33988813 DOI: 10.1007/s11064-021-03315-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 01/20/2023]
Abstract
Ginkgo biloba L. leaves (GBLs), as widely used plant extract sources, significantly improve cognitive, learning and memory function in patients with dementia. However, few studies have been conducted on the specific mechanism of Neurodegenerative diseases (NDs). In this study, network pharmacology was employed to elucidate potential mechanism of GBLs in the treatment of NDs. Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) was used to obtain the chemical components in accordance with the screening principles of oral availability and drug-like property. Potential targets of GBLs were integrated with disease targets, and intersection targets were exactly the potential action targets of GBLs for treating NDs; these key targets were enriched and analyzed by the protein protein interaction (PPI) analysis and molecular docking verification. Key genes were ultimately used to find the biological pathway and explain the therapeutic mechanism by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Twenty-seven active components of GBLs may affect biological processes such as oxidative reactions and activate transcription factor activities. These components may also affect 120 metabolic pathways, such as the PI3K/AKT pathway, by regulating 147 targets, including AKT1, ALB, HSP90AA1, PTGS2, MMP9, EGFR and APP. By using the software iGEMDOCK, the main target proteins were found to bind well to the main active components of GBLs. GBLs have the characteristics of multi-component and multi-target synergistic effect on the treatment of NDs, which preliminarily predicted its possible molecular mechanism of action, and provided the basis for the follow-up study.
Collapse
Affiliation(s)
- Jing Wang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, Liaoning, People's Republic of China.,Institute of Chemistry and Applications of Plant Resources, Dalian Polytechnic University, Dalian, 116034, Liaoning, People's Republic of China.,Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222000, Jiangsu, People's Republic of China.,State Key Laboratory of Pharmaceutical New-tech for Chinese Medicine, Lianyungang, 222000, Jiangsu, People's Republic of China
| | - Xialin Chen
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222000, Jiangsu, People's Republic of China.,State Key Laboratory of Pharmaceutical New-tech for Chinese Medicine, Lianyungang, 222000, Jiangsu, People's Republic of China
| | - Weirong Bai
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222000, Jiangsu, People's Republic of China.,State Key Laboratory of Pharmaceutical New-tech for Chinese Medicine, Lianyungang, 222000, Jiangsu, People's Republic of China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222000, Jiangsu, People's Republic of China.,State Key Laboratory of Pharmaceutical New-tech for Chinese Medicine, Lianyungang, 222000, Jiangsu, People's Republic of China
| | - Wei Xiao
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222000, Jiangsu, People's Republic of China. .,State Key Laboratory of Pharmaceutical New-tech for Chinese Medicine, Lianyungang, 222000, Jiangsu, People's Republic of China.
| | - Jingbo Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, Liaoning, People's Republic of China. .,Institute of Chemistry and Applications of Plant Resources, Dalian Polytechnic University, Dalian, 116034, Liaoning, People's Republic of China.
| |
Collapse
|
12
|
Henriques AD, Machado-Silva W, Leite RE, Suemoto CK, Leite KR, Srougi M, Pereira AC, Jacob-Filho W, Nóbrega OT. Genome-wide profiling and predicted significance of post-mortem brain microRNA in Alzheimer’s disease. Mech Ageing Dev 2020; 191:111352. [DOI: 10.1016/j.mad.2020.111352] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022]
|
13
|
Knockout of p75 neurotrophin receptor attenuates the hyperphosphorylation of Tau in pR5 mouse model. Aging (Albany NY) 2019; 11:6762-6791. [PMID: 31479419 PMCID: PMC6756909 DOI: 10.18632/aging.102202] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 08/12/2019] [Indexed: 02/02/2023]
Abstract
p75 neurotrophin receptor (p75NTR) has been implicated in Alzheimer's disease (AD). However, whether p75NTR is involved in Tau hyperphosphorylation, one of the pathologies observed in AD, remains unclear. In our previous study, the extracellular domain of p75NTR blocked amyloid beta (Aβ) toxicity and attenuated Aβ-induced Tau hyperphosphorylation. Here we show that, in the absence of Aβ, p75NTR regulates Tau phosphorylation in the transgenic mice with the P301L human Tau mutation (pR5). The knockout of p75NTR in pR5 mice attenuated the phosphorylation of human Tau. In addition, the elevated activity of kinases responsible for Tau phosphorylation including glycogen synthase kinase 3 beta; cyclin-dependent-kinase 5; and Rho-associated protein kinase was also inhibited when p75NTR is knocked out in pR5 mice at 9 months of age. The increased caspase-3 activity observed in pR5 mice was also abolished in the absence of p75NTR. Our study also showed that p75NTR is required for Aβ- and pro-brain derived neurotrophin factor (proBDNF)-induced Tau phosphorylation, in vitro. Overall, our data indicate that p75NTR is required for Tau phosphorylation, a key event in the formation of neurofibrillary tangles, another hallmark of AD. Thus, targeting p75NTR could reduce or prevent the pathologic hyperphosphorylation of Tau.
Collapse
|
14
|
Shaping the Nrf2-ARE-related pathways in Alzheimer's and Parkinson's diseases. Ageing Res Rev 2019; 54:100942. [PMID: 31415806 DOI: 10.1016/j.arr.2019.100942] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 08/02/2019] [Accepted: 08/09/2019] [Indexed: 12/20/2022]
Abstract
A failure in redox homeostasis is a common hallmark of Alzheimer's Disease (AD) and Parkinson's Disease (PD), two age-dependent neurodegenerative disorders (NDD), causing increased oxidative stress, oxidized/damaged biomolecules, altered neuronal function and consequent cell death. Activation of nuclear factor erythroid 2-related factor 2 (Nrf2), a redox-regulated transcription factor, results in upregulation of cytoprotective and antioxidant enzymes/proteins, protecting against oxidative stress. Nrf2 regulation is achieved by various proteins and pathways, at both cytoplasmatic and nuclear level; however, the elaborate network of mechanisms involved in Nrf2 regulation may restrain Nrf2 pathway normal activity. Indeed, altered Nrf2 activity is involved in aging and NDD, such as AD and PD. Therefore, understanding the diversity of Nrf2 control mechanisms and regulatory proteins is of high interest, since more effective NDD therapeutics can be identified. In this review, we first introduce Keap1-Nrf2-ARE structure, function and regulation, with a special focus on the several pathways involved in Nrf2 positive and negative modulation, namely p62, PKC, PI3K/Akt/GSK-3β, NF-kB and p38 MAPK. We then briefly describe the evidences for oxidative stress and Nrf2 pathway deregulation in different stages of NDDs. Finally, we discuss the potential of Nrf2-related pathways as potential therapeutic targets to possibly prevent or slowdown NDD progression.
Collapse
|
15
|
Park JS, Lee J, Jung ES, Kim MH, Kim IB, Son H, Kim S, Kim S, Park YM, Mook-Jung I, Yu SJ, Lee JH. Brain somatic mutations observed in Alzheimer's disease associated with aging and dysregulation of tau phosphorylation. Nat Commun 2019; 10:3090. [PMID: 31300647 PMCID: PMC6626023 DOI: 10.1038/s41467-019-11000-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 06/14/2019] [Indexed: 01/24/2023] Open
Abstract
The role of brain somatic mutations in Alzheimer’s disease (AD) is not well understood. Here, we perform deep whole-exome sequencing (average read depth 584×) in 111 postmortem hippocampal formation and matched blood samples from 52 patients with AD and 11 individuals not affected by AD. The number of somatic single nucleotide variations (SNVs) in AD brain specimens increases significantly with aging, and the rate of mutation accumulation in the brain is 4.8-fold slower than that in AD blood. The putatively pathogenic brain somatic mutations identified in 26.9% (14 of 52) of AD individuals are enriched in PI3K-AKT, MAPK, and AMPK pathway genes known to contribute to hyperphosphorylation of tau. We show that a pathogenic brain somatic mutation in PIN1 leads to a loss-of-function mutation. In vitro mimicking of haploinsufficiency of PIN1 aberrantly increases tau phosphorylation and aggregation. This study provides new insights into the genetic architecture underlying the pathogenesis of AD. The role of brain somatic mutations in neurodegenerative diseases such as Alzheimer’s disease (AD) is not well understood. Here the authors carry out high-depth exome sequencing ~500× on brain tissue from patients with AD and controls, and identify mutations in a number of genes that are known to contribute to phosphorylation and aggregation of tau, including PIN1.
Collapse
Affiliation(s)
- Jun Sung Park
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Junehawk Lee
- Center for Supercomputing Applications, Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Republic of Korea
| | - Eun Sun Jung
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.,Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Myeong-Heui Kim
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Il Bin Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hyeonju Son
- Department of Biomedical Systems Informatics, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Sanghyeon Kim
- Laboratory of Brain Research, Stanley Medical Research Institute (SMRI), 9800 Medical Center Drive, Suite C-050, Rockville, MD, 20850, USA
| | - Young Mok Park
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.,Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seok Jong Yu
- Center for Supercomputing Applications, Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Republic of Korea.
| | - Jeong Ho Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea. .,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
16
|
Song Z, Han X, Shen L, Zou H, Zhang B, Liu J, Gong A. PTEN silencing enhances neuronal proliferation and differentiation by activating PI3K/Akt/GSK3β pathway in vitro. Exp Cell Res 2018; 363:179-187. [PMID: 29305963 DOI: 10.1016/j.yexcr.2018.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/03/2017] [Accepted: 01/02/2018] [Indexed: 01/09/2023]
Abstract
The failure of neuronal proliferation and differentiation is a major obstacle for neural repair and regeneration after traumatic central nervous system (CNS) injury. PTEN acts as an intrinsic brake on the neuronal cells, but its roles and mechanism still remain to be clarified. Herein, for the first time we confirmed that PTEN had a dual effect on the neuronal cells in vitro. Firstly, we found that PTEN knockdown significantly promoted cell proliferation and differentiation. Then, PTEN knockdown activated PI3K/Akt and Wnt/β-catenin pathways in vitro. Further evidence revealed that GSK3β as a key node involved in PTEN controlling cell proliferation and differentiation in PC12 cells. In addition, we identified that PTEN-GSK3β pathway modulated neuronal proliferation via β-catenin. Taken together, these results suggest that PTEN silencing enhances neuronal proliferation and differentiation by activating PI3K/Akt/GSK3β pathway that it may be a promising therapeutic approach for CNS injury.
Collapse
Affiliation(s)
- Zhiwen Song
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xiu Han
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Liming Shen
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Hongjun Zou
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining 272000, China
| | - Jinbo Liu
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| | - Aihua Gong
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
17
|
Li H, Wang R. Blocking SIRT1 inhibits cell proliferation and promotes aging through the PI3K/AKT pathway. Life Sci 2017; 190:84-90. [DOI: 10.1016/j.lfs.2017.09.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/18/2017] [Accepted: 09/25/2017] [Indexed: 12/20/2022]
|
18
|
Caspase-3-dependent cleavage of Akt modulates tau phosphorylation via GSK3β kinase: implications for Alzheimer's disease. Mol Psychiatry 2017; 22:1002-1008. [PMID: 28138159 DOI: 10.1038/mp.2016.214] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/26/2016] [Accepted: 09/14/2016] [Indexed: 01/23/2023]
Abstract
The pathological hallmark of Alzheimer's disease (AD) is accumulation of misfolded amyloid-β peptides and hyperphosphorylated tau protein in the brain. Increasing evidence suggests that serine-aspartyl proteases-caspases are activated in the AD brain. Previous studies identified a caspase-3 cleavage site within the amyloid-β precursor protein, and a caspase-3 cleavage of tau as the mechanisms involved in the development of Aβ and tau neuropathology, respectively. However, the potential role that caspase-3 could have on tau metabolism remains unknown. In the current studies, we provide experimental evidence that caspase-3 directly and specifically regulates tau phosphorylation, and demonstrate that this effect is mediated by the GSK3β kinase pathway via a caspase-3-dependent cleavage of the protein kinase B (also known as Akt). In addition, we confirm these results in vivo by using a transgenic mouse model of AD. Collectively, our findings demonstrate a new role for caspase-3 in the neurobiology of tau, and suggest that therapeutic strategies aimed at inhibiting this protease-dependent cleavage of Akt may prove beneficial in preventing tau hyperphosphorylation and subsequent neuropathology in AD and related tauopathies.
Collapse
|
19
|
Yu HJ, Koh SH. The role of PI3K/AKT pathway and its therapeutic possibility in Alzheimer's disease. ACTA ACUST UNITED AC 2017. [DOI: 10.7599/hmr.2017.37.1.18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Hyun-Jung Yu
- Department of Neurology, Bundang Jesaeng Hospital, Gyeonggi, South Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, Gyeonggi, South Korea
| |
Collapse
|
20
|
Neuroprotective Effect of Fisetin Against Amyloid-Beta-Induced Cognitive/Synaptic Dysfunction, Neuroinflammation, and Neurodegeneration in Adult Mice. Mol Neurobiol 2016; 54:2269-2285. [DOI: 10.1007/s12035-016-9795-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/11/2016] [Indexed: 12/22/2022]
|
21
|
Shi Y, Shu B, Yang R, Xu Y, Xing B, Liu J, Chen L, Qi S, Liu X, Wang P, Tang J, Xie J. Wnt and Notch signaling pathway involved in wound healing by targeting c-Myc and Hes1 separately. Stem Cell Res Ther 2015; 6:120. [PMID: 26076648 PMCID: PMC4501079 DOI: 10.1186/s13287-015-0103-4] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 01/20/2015] [Accepted: 05/20/2015] [Indexed: 12/18/2022] Open
Abstract
Introduction Wnt and Notch signaling pathways are critically involved in relative cell fate decisions within the development of cutaneous tissues. Moreover, several studies identified the above two pathways as having a significant role during wound healing. However, their biological effects during cutaneous tissues repair are unclear. Methods We employed a self-controlled model (Sprague–Dawley rats with full-thickness skin wounds) to observe the action and effect of Wnt/β-catenin and Notch signalings in vivo. The quality of wound repair relevant to the gain/loss-of-function Wnt/β-catenin and Notch activation was estimated by hematoxylin-and-eosin and Masson staining. Immunofluorescence analysis and Western blot analysis were used to elucidate the underlying mechanism of the regulation of Wnt and Notch signaling pathways in wound healing. Meanwhile, epidermal stem cells (ESCs) were cultured in keratinocyte serum-free medium with Jaggedl or in DAPT (N-[(3,5-difluorophenyl)acetyl]-L-alanyl-2-phenyl]glycine-1,1-dimethylethyl) to investigate whether the interruption of Notch signaling contributes to the expression of Wnt/β-catenin signaling. Results The results showed that in vivo the gain-of-function Wnt/β-catenin and Notch activation extended the ability to promote wound closure. We further determined that activation or inhibition of Wnt signaling and Notch signaling can affect the proliferation of ESCs, the differentiation and migration of keratinocytes, and follicle regeneration by targeting c-Myc and Hes1, which ultimately lead to enhanced or delayed wound healing. Furthermore, Western blot analysis suggested that the two pathways might interact in vivo and in vitro. Conclusion These results suggest that Wnt and Notch signalings play important roles in cutaneous repair by targeting c-Myc and Hes1 separately. What’s more, interaction between the above two pathways might act as a vital role in regulation of wound healing.
Collapse
Affiliation(s)
- Yan Shi
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Bin Shu
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Ronghua Yang
- Department of Burns, Fo Shan Hospital of Sun Yat-Sen University, Lingnan avenue, Foshan, 528000, China.
| | - Yingbin Xu
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Bangrong Xing
- Department of Burns, Third Affiliated Hospital of Sun Yat-sen University, Tianhe road, Guangzhou, 510620, China.
| | - Jian Liu
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Lei Chen
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Shaohai Qi
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Xusheng Liu
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Peng Wang
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Jinming Tang
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Julin Xie
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| |
Collapse
|
22
|
Lauzon MA, Daviau A, Marcos B, Faucheux N. Growth factor treatment to overcome Alzheimer's dysfunctional signaling. Cell Signal 2015; 27:1025-38. [PMID: 25744541 DOI: 10.1016/j.cellsig.2015.02.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/16/2015] [Indexed: 10/23/2022]
Abstract
The number of people suffering from Alzheimer's disease (AD) will increase as the world population ages, creating a huge socio-economic burden. The three pathophysiological hallmarks of AD are the cholinergic system dysfunction, the β-amyloid peptide deposition and the Tau protein hyperphosphorylation. Current treatments have only transient effects and each tends to concentrate on a single pathophysiological aspect of AD. This review first provides an overall view of AD in terms of its pathophysiological symptoms and signaling dysfunction. We then examine the therapeutic potential of growth factors (GFs) by showing how they can overcome the dysfunctional cell signaling that occurs in AD. Finally, we discuss new alternatives to GFs that help overcome the problem of brain uptake, such as small peptides, with evidence from some of our unpublished data on human neuronal cell line.
Collapse
Affiliation(s)
- Marc-Antoine Lauzon
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Alex Daviau
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Bernard Marcos
- Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Nathalie Faucheux
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada.
| |
Collapse
|
23
|
Hui J, Zhang J, Kim H, Tong C, Ying Q, Li Z, Mao X, Shi G, Yan J, Zhang Z, Xi G. Fluoxetine regulates neurogenesis in vitro through modulation of GSK-3β/β-catenin signaling. Int J Neuropsychopharmacol 2015; 18:pyu099. [PMID: 25522429 PMCID: PMC4376550 DOI: 10.1093/ijnp/pyu099] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND It is generally accepted that chronic treatment with antidepressants increases hippocampal neurogenesis, but the molecular mechanisms underlying their effects are unknown. Recently, glycogen synthase kinase-3 beta (GSK-3β)/β-catenin signaling was shown to be involved in the mechanism of how antidepressants might influence hippocampal neurogenesis. METHODS The aim of this study was to determine whether GSK-3β/β-catenin signaling is involved in the alteration of neurogenesis as a result of treatment with fluoxetine, a selective serotonin reuptake inhibitor. The mechanisms involved in fluoxetine's regulation of GSK-3β/β-catenin signaling pathway were also examined. RESULTS Our results demonstrated that fluoxetine increased the proliferation of embryonic neural precursor cells (NPCs) by up-regulating the phosphorylation of Ser9 on GSK-3β and increasing the level of nuclear β-catenin. The overexpression of a stabilized β-catenin protein (ΔN89 β-catenin) significantly increased NPC proliferation, while inhibition of β-catenin expression in NPCs led to a significant decrease in the proliferation and reduced the proliferative effects induced by fluoxetine. The effects of fluoxetine-induced up-regulation of both phosphorylation of Ser9 on GSK-3β and nuclear β-catenin were significantly prevented by the 5-hydroxytryptamine-1A (5-HT1A) receptor antagonist WAY-100635. CONCLUSIONS The results demonstrate that fluoxetine may increase neurogenesis via the GSK-3β/β-catenin signaling pathway that links postsynaptic 5-HT1A receptor activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Guangjun Xi
- Department of Critical Care Medicine, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China (Drs Hui and Yan); Department of Neurology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China (Drs J Zhang, Li, Mao, Shi, and Xi); Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, University of Southern California, Los Angeles, CA (Drs Kim, Tong, and Ying); Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing, China (Dr Z Zhang).
| |
Collapse
|
24
|
Bayod S, Felice P, Andrés P, Rosa P, Camins A, Pallàs M, Canudas AM. Downregulation of canonical Wnt signaling in hippocampus of SAMP8 mice. Neurobiol Aging 2014; 36:720-9. [PMID: 25443287 DOI: 10.1016/j.neurobiolaging.2014.09.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 09/14/2014] [Accepted: 09/17/2014] [Indexed: 01/09/2023]
Abstract
In the adult brain, canonical Wnt (Wnt/β-catenin) signaling modulates neuronal function, hippocampal neurogenesis, and synaptic plasticity. Indeed, growing evidence suggests that downregulation of Wnt signaling could be involved in the cognitive decline associated with aging and also with the physiopathology of Alzheimer's disease (AD). However, the molecular basis remains unknown. At present, SAMP8 is an experimental model that has been proposed for studying age-related neurodegenerative changes associated with aging and the pathogenesis of AD. Here, we examined Wnt signaling in the hippocampus of SAMP8 mice at 9 and 12 months of age, as well as in its control-strain SAMR1 mice. Our results showed increased Dickkopf-1 protein levels in SAMP8 with age, in addition to GSK-3 α/β activation and hyperphosphorylated tau. Consequently, higher β-catenin phosphorylation at Ser(33,37) and Thr(41), which promotes its degradation, along with a decrease in active β-catenin (ABC) in the nucleus, were observed in SAMP8, mainly at the age of 12 months. Moreover, nuclear levels of Dvl3 were lower in 9- and 12-month-old SAMP8 mice. Related to these findings, SAMP8 showed an increase in neuronal loss in the hippocampus that was associated with lower protein levels of the antiapoptotic protein and the Wnt target gene, Bcl-2, in addition to an increase in the proapototic protein Bax. Our results suggest a relationship between age-related downregulation of canonical Wnt signaling and neuronal loss observed in the hippocampus of SAMP8 mice. Thus, enhancing Wnt signaling may represent a novel neuroprotective strategy aimed at counteracting the cognitive decline that is associated not only with aging but also with AD.
Collapse
Affiliation(s)
- Sergi Bayod
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Paolo Felice
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Pol Andrés
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Paolo Rosa
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Antoni Camins
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Mercè Pallàs
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna-Maria Canudas
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
25
|
Kitagishi Y, Nakanishi A, Ogura Y, Matsuda S. Dietary regulation of PI3K/AKT/GSK-3β pathway in Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2014; 6:35. [PMID: 25031641 PMCID: PMC4075129 DOI: 10.1186/alzrt265] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Alzheimer’s disease (AD) is characterized by the formation of senile plaques and neurofibrillary tangles composed of phosphorylated Tau. Several findings suggest that correcting signal dysregulation for Tau phosphorylation in AD may offer a potential therapeutic approach. The PI3K/AKT/GSK-3β pathway has been shown to play a pivotal role in neuroprotection, enhancing cell survival by stimulating cell proliferation and inhibiting apoptosis. This pathway appears to be crucial in AD because it promotes protein hyper-phosphorylation in Tau. Understanding those regulations may provide a better efficacy of new therapeutic approaches. In this review, we summarize advances in the involvement of the PI3K/AKT/GSK-3β pathways in cell signaling of neuronal cells. We also review recent studies on the features of several diets and the signaling pathway involved in AD.
Collapse
Affiliation(s)
- Yasuko Kitagishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - Atsuko Nakanishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - Yasunori Ogura
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| |
Collapse
|
26
|
Short-term high-fat-and-fructose feeding produces insulin signaling alterations accompanied by neurite and synaptic reduction and astroglial activation in the rat hippocampus. J Cereb Blood Flow Metab 2014; 34:1001-8. [PMID: 24667917 PMCID: PMC4050245 DOI: 10.1038/jcbfm.2014.48] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 11/09/2022]
Abstract
Chronic consumption of high-fat-and-fructose diets (HFFD) is associated with the development of insulin resistance (InsRes) and obesity. Systemic insulin resistance resulting from long-term HFFD feeding has detrimental consequences on cognitive performance, neurogenesis, and long-term potentiation establishment, accompanied by neuronal alterations in the hippocampus. However, diet-induced hippocampal InsRes has not been reported. Therefore, we investigated whether short-term HFFD feeding produced hippocampal insulin signaling alterations associated with neuronal changes in the hippocampus. Rats were fed with a control diet or an HFFD consisting of 10% lard supplemented chow and 20% high-fructose syrup in the drinking water. Our results show that 7 days of HFFD feeding induce obesity and InsRes, associated with the following alterations in the hippocampus: (1) a decreased insulin signaling; (2) a decreased hippocampal weight; (3) a reduction in dendritic arborization in CA1 and microtubule-associated protein 2 (MAP-2) levels; (4) a decreased dendritic spine number in CA1 and synaptophysin content, along with an increase in tau phosphorylation; and finally, (5) an increase in reactive astrocyte associated with microglial changes. To our knowledge, this is the first report addressing hippocampal insulin signaling, as well as morphologic, structural, and functional modifications due to short-term HFFD feeding in the rat.
Collapse
|
27
|
Yu UY, Yoo BC, Ahn JH. Regulatory B Subunits of Protein Phosphatase 2A Are Involved in Site-specific Regulation of Tau Protein Phosphorylation. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:155-61. [PMID: 24757378 PMCID: PMC3994303 DOI: 10.4196/kjpp.2014.18.2.155] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 02/12/2014] [Accepted: 02/28/2014] [Indexed: 11/15/2022]
Abstract
Overexpression of amyloid precursor protein with the Swedish mutation causes abnormal hyperphosphorylation of the microtubule-associated protein tau. Hyperphosphorylated isoforms of tau are major components of neurofibrillary tangles, which are histopathological hallmarks of Alzheimer's disease. Protein phosphatase 2A (PP2A), a major tau protein phosphatase, consists of a structural A subunit, catalytic C subunit, and a variety of regulatory B subunits. The B subunits have been reported to modulate function of the PP2A holoenzyme by regulating substrate binding, enzyme activity, and subcellular localization. In the current study, we characterized regulatory B subunit-specific regulation of tau protein phosphorylation. We showed that the PP2A B subunit PPP2R2A mediated dephosphorylation of tau protein at Ser-199, Ser-202/Thr-205, Thr-231, Ser-262, and Ser-422. Down-regulation of PPP2R5D expression decreased tau phosphorylation at Ser-202/Thr-205, Thr-231, and Ser-422, which indicates activation of the tau kinase glycogen synthase kinase 3 beta (GSK3β) by PP2A with PPP2R5D subunit. The level of activating phosphorylation of the GSK3β kinase Akt at Thr-308 and Ser-473 were both increased by PPP2R5D knockdown. We also characterized B subunit-specific phosphorylation sites in tau using mass spectrometric analysis. Liquid chromatography-mass spectrometry revealed that the phosphorylation status of the tau protein may be affected by PP2A, depending on the specific B subunits. These studies further our understanding of the function of various B subunits in mediating site-specific regulation of tau protein phosphorylation.
Collapse
Affiliation(s)
- Un Young Yu
- Department of Biochemistry, Ewha Womans University College of Medicine, Seoul 158-710, Korea
| | - Byong Chul Yoo
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang 410-768, Korea
| | - Jung-Hyuck Ahn
- Department of Biochemistry, Ewha Womans University College of Medicine, Seoul 158-710, Korea
| |
Collapse
|
28
|
Ortiz-Matamoros A, Salcedo-Tello P, Avila-Muñoz E, Zepeda A, Arias C. Role of wnt signaling in the control of adult hippocampal functioning in health and disease: therapeutic implications. Curr Neuropharmacol 2014; 11:465-76. [PMID: 24403870 PMCID: PMC3763754 DOI: 10.2174/1570159x11311050001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 03/16/2013] [Accepted: 03/16/2013] [Indexed: 12/12/2022] Open
Abstract
It is well recognized the role of the Wnt pathway in many developmental processes such as neuronal maturation, migration, neuronal connectivity and synaptic formation. Growing evidence is also demonstrating its function in the mature brain where is associated with modulation of axonal remodeling, dendrite outgrowth, synaptic activity, neurogenesis and behavioral plasticity. Proteins involved in Wnt signaling have been found expressed in the adult hippocampus suggesting that Wnt pathway plays a role in the hippocampal function through life. Indeed, Wnt ligands act locally to regulate neurogenesis, neuronal cell shape and pre- and postsynaptic assembly, events that are thought to underlie changes in synaptic function associated with long-term potentiation and with cognitive tasks such as learning and memory. Recent data have demonstrated the increased expression of the Wnt antagonist Dickkopf-1 (DKK1) in brains of Alzheimer´s disease (AD) patients suggesting that dysfunction of Wnt signaling could also contribute to AD pathology. We review here evidence of Wnt-associated molecules expression linked to physiological and pathological hippocampal functioning in the adult brain. The basic aspects of Wnt related mechanisms underlying hippocampal plasticity as well as evidence of how hippocampal dysfunction may rely on Wnt dysregulation is analyzed. This information would provide some clues about the possible therapeutic targets for developing treatments for neurodegenerative diseases associated with aberrant brain plasticity.
Collapse
Affiliation(s)
- Abril Ortiz-Matamoros
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México D.F
| | - Pamela Salcedo-Tello
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México D.F
| | - Evangelina Avila-Muñoz
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México D.F
| | - Angélica Zepeda
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México D.F
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México D.F
| |
Collapse
|
29
|
Liu AR, Liu L, Chen S, Yang Y, Zhao HJ, Liu L, Guo FM, Lu XM, Qiu HB. Activation of canonical wnt pathway promotes differentiation of mouse bone marrow-derived MSCs into type II alveolar epithelial cells, confers resistance to oxidative stress, and promotes their migration to injured lung tissue in vitro. J Cell Physiol 2013; 228:1270-83. [PMID: 23154940 DOI: 10.1002/jcp.24282] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 11/02/2012] [Indexed: 01/31/2023]
Abstract
The differentiation of mesenchymal stem cells (MSCs) into type II alveolar epithelial (AT II) cells in vivo and in vitro, is critical for reepithelization and recovery in acute lung injury (ALI), but the mechanisms responsible for differentiation are unclear. In the present study, we investigated the role of the canonical wnt pathway in the differentiation of mouse bone marrow-derived MSCs (mMSCs) into AT II cells. Using a modified co-culture system with murine lung epithelial-12 (MLE-12) cells and small airway growth media (SAGM) to efficiently drive mMSCs differentiation, we found that GSK 3β and β-catenin in the canonical wnt pathway were up-regulated during differentiation. The levels of surfactant protein (SP) C, SPB, and SPD, the specific markers of AT II cells, correspondingly increased in mMSCs when Wnt3a or LiCl was added to the co-culture system to activate wnt/β-catenin signaling. The expression of these factors was depressed to some extent by inhibiting the pathway with the addition of DKK 1. The differentiation rate of mMSCs also depends on their abilities to accumulate and survive in inflammatory tissue. Our results suggested that the activation of wnt/β-catenin signaling promoted mMSCs migration towards ALI mouse-derived lung tissue in a Transwell assay, and ameliorated the cell death and the reduction of Bcl-2/Bax induced by H(2) O(2), which simultaneously caused reduced GSK 3β and β-catenin in mMSCs. These data supports a potential mechanism for the differentiation of mMSCs into AT II cells involving canonical wnt pathway activation, which may be significant to their application in ALI.
Collapse
Affiliation(s)
- Ai-Ran Liu
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kang J, Rivest S. Lipid metabolism and neuroinflammation in Alzheimer's disease: a role for liver X receptors. Endocr Rev 2012; 33:715-46. [PMID: 22766509 DOI: 10.1210/er.2011-1049] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Liver X receptors (LXR) are nuclear receptors that have emerged as key regulators of lipid metabolism. In addition to their functions as cholesterol sensors, LXR have also been found to regulate inflammatory responses in macrophages. Alzheimer's disease (AD) is a neurodegenerative disease characterized by a progressive cognitive decline associated with inflammation. Evidence indicates that the initiation and progression of AD is linked to aberrant cholesterol metabolism and inflammation. Activation of LXR can regulate neuroinflammation and decrease amyloid-β peptide accumulation. Here, we highlight the role of LXR in orchestrating lipid homeostasis and neuroinflammation in the brain. In addition, diabetes mellitus is also briefly discussed as a significant risk factor for AD because of the appearing beneficial effects of LXR on glucose homeostasis. The ability of LXR to attenuate AD pathology makes them potential therapeutic targets for this neurodegenerative disease.
Collapse
Affiliation(s)
- Jihong Kang
- Department of Physiology and Pathophysiology and Key Laboratory of Molecular Cardiovascular Sciences, State Education Ministry, Peking University Health Science Center, Beijing 100191, China
| | | |
Collapse
|
31
|
Dutta B, Wallqvist A, Reifman J. PathNet: a tool for pathway analysis using topological information. SOURCE CODE FOR BIOLOGY AND MEDICINE 2012; 7:10. [PMID: 23006764 PMCID: PMC3563509 DOI: 10.1186/1751-0473-7-10] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 08/03/2012] [Indexed: 01/01/2023]
Abstract
Background Identification of canonical pathways through enrichment of differentially expressed genes in a given pathway is a widely used method for interpreting gene lists generated from high-throughput experimental studies. However, most algorithms treat pathways as sets of genes, disregarding any inter- and intra-pathway connectivity information, and do not provide insights beyond identifying lists of pathways. Results We developed an algorithm (PathNet) that utilizes the connectivity information in canonical pathway descriptions to help identify study-relevant pathways and characterize non-obvious dependencies and connections among pathways using gene expression data. PathNet considers both the differential expression of genes and their pathway neighbors to strengthen the evidence that a pathway is implicated in the biological conditions characterizing the experiment. As an adjunct to this analysis, PathNet uses the connectivity of the differentially expressed genes among all pathways to score pathway contextual associations and statistically identify biological relations among pathways. In this study, we used PathNet to identify biologically relevant results in two Alzheimer’s disease microarray datasets, and compared its performance with existing methods. Importantly, PathNet identified de-regulation of the ubiquitin-mediated proteolysis pathway as an important component in Alzheimer’s disease progression, despite the absence of this pathway in the standard enrichment analyses. Conclusions PathNet is a novel method for identifying enrichment and association between canonical pathways in the context of gene expression data. It takes into account topological information present in pathways to reveal biological information. PathNet is available as an R workspace image from
http://www.bhsai.org/downloads/pathnet/.
Collapse
Affiliation(s)
- Bhaskar Dutta
- DoD Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U,S, Army Medical Research and Materiel Command, Ft, Detrick, MD, 21702, USA.
| | | | | |
Collapse
|
32
|
Maiese K, Chong ZZ, Shang YC, Wang S. Erythropoietin: new directions for the nervous system. Int J Mol Sci 2012; 13:11102-11129. [PMID: 23109841 PMCID: PMC3472733 DOI: 10.3390/ijms130911102] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/16/2012] [Accepted: 08/30/2012] [Indexed: 12/14/2022] Open
Abstract
New treatment strategies with erythropoietin (EPO) offer exciting opportunities to prevent the onset and progression of neurodegenerative disorders that currently lack effective therapy and can progress to devastating disability in patients. EPO and its receptor are present in multiple systems of the body and can impact disease progression in the nervous, vascular, and immune systems that ultimately affect disorders such as Alzheimer's disease, Parkinson's disease, retinal injury, stroke, and demyelinating disease. EPO relies upon wingless signaling with Wnt1 and an intimate relationship with the pathways of phosphoinositide 3-kinase (PI 3-K), protein kinase B (Akt), and mammalian target of rapamycin (mTOR). Modulation of these pathways by EPO can govern the apoptotic cascade to control β-catenin, glycogen synthase kinase-3β, mitochondrial permeability, cytochrome c release, and caspase activation. Yet, EPO and each of these downstream pathways require precise biological modulation to avert complications associated with the vascular system, tumorigenesis, and progression of nervous system disorders. Further understanding of the intimate and complex relationship of EPO and the signaling pathways of Wnt, PI 3-K, Akt, and mTOR are critical for the effective clinical translation of these cell pathways into robust treatments for neurodegenerative disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- Cancer Institute of New Jersey, New Brunswick, New Jersey 08901, USA
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| | - Zhao Zhong Chong
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| | - Yan Chen Shang
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| | - Shaohui Wang
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| |
Collapse
|
33
|
Pinto-Almazán R, Calzada-Mendoza CC, Campos-Lara MG, Guerra-Araiza C. Effect of chronic administration of estradiol, progesterone, and tibolone on the expression and phosphorylation of glycogen synthase kinase-3β and the microtubule-associated protein tau in the hippocampus and cerebellum of female rat. J Neurosci Res 2011; 90:878-86. [DOI: 10.1002/jnr.22808] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 08/25/2011] [Accepted: 09/01/2011] [Indexed: 01/26/2023]
|
34
|
Shang YC, Chong ZZ, Wang S, Maiese K. Erythropoietin and Wnt1 govern pathways of mTOR, Apaf-1, and XIAP in inflammatory microglia. Curr Neurovasc Res 2011; 8:270-85. [PMID: 22023617 PMCID: PMC3254854 DOI: 10.2174/156720211798120990] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 09/20/2011] [Accepted: 10/04/2011] [Indexed: 01/01/2023]
Abstract
Inflammatory microglia modulate a host of cellular processes in the central nervous system that include neuronal survival, metabolic fluxes, foreign body exclusion, and cellular regeneration. Elucidation of the pathways that oversee microglial survival and integrity may offer new avenues for the treatment of neurodegenerative disorders. Here we demonstrate that erythropoietin (EPO), an emerging strategy for immune system modulation, prevents microglial early and late apoptotic injury during oxidant stress through Wnt1, a cysteine-rich glycosylated protein that modulates cellular development and survival. Loss of Wnt1 through blockade of Wnt1 signaling or through the gene silencing of Wnt1 eliminates the protective capacity of EPO. Furthermore, endogenous Wnt1 in microglia is vital to preserve microglial survival since loss of Wnt1 alone increases microglial injury during oxidative stress. Cellular protection by EPO and Wnt1 intersects at the level of protein kinase B (Akt1), the mammalian target of rapamycin (mTOR), and p70S6K, which are necessary to foster cytoprotection for microglia. Downstream from these pathways, EPO and Wnt1 control "anti-apoptotic" pathways of microglia through the modulation of mitochondrial membrane permeability, the release of cytochrome c, and the expression of apoptotic protease activating factor-1 (Apaf-1) and X-linked inhibitor of apoptosis protein (XIAP). These studies offer new insights for the development of innovative therapeutic strategies for neurodegenerative disorders that focus upon inflammatory microglia and novel signal transduction pathways.
Collapse
Affiliation(s)
- Yan Chen Shang
- Laboratory of Cellular and Molecular Signaling, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
- Department of Neurology and Neurosciences, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
- Cancer Center - New Jersey Medical School, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
| | - Zhao Zhong Chong
- Laboratory of Cellular and Molecular Signaling, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
- Department of Neurology and Neurosciences, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
- Cancer Center - New Jersey Medical School, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
| | - Shaohui Wang
- Laboratory of Cellular and Molecular Signaling, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
- Department of Neurology and Neurosciences, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
- Cancer Center - New Jersey Medical School, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
| | - Kenneth Maiese
- Laboratory of Cellular and Molecular Signaling, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
- Department of Neurology and Neurosciences, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
- Cancer Center - New Jersey Medical School, University of Medicine and Dentistry, New Jersey Medical School, Newark, 07101 New Jersey
| |
Collapse
|
35
|
Salcedo-Tello P, Ortiz-Matamoros A, Arias C. GSK3 Function in the Brain during Development, Neuronal Plasticity, and Neurodegeneration. Int J Alzheimers Dis 2011; 2011:189728. [PMID: 21660241 PMCID: PMC3109514 DOI: 10.4061/2011/189728] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 03/07/2011] [Indexed: 02/06/2023] Open
Abstract
GSK3 has diverse functions, including an important role in brain pathology. In this paper, we address the primary functions of GSK3 in development and neuroplasticity, which appear to be interrelated and to mediate age-associated neurological diseases. Specifically, GSK3 plays a pivotal role in controlling neuronal progenitor proliferation and establishment of neuronal polarity during development, and the upstream and downstream signals modulating neuronal GSK3 function affect cytoskeletal reorganization and neuroplasticity throughout the lifespan. Modulation of GSK3 in brain areas subserving cognitive function has become a major focus for treating neuropsychiatric and neurodegenerative diseases. As a crucial node that mediates a variety of neuronal processes, GSK3 is proposed to be a therapeutic target for restoration of synaptic functioning and cognition, particularly in Alzheimer's disease.
Collapse
Affiliation(s)
- Pamela Salcedo-Tello
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510 Ciudad de México, Mexico
| | | | | |
Collapse
|
36
|
Jesky R, Hailong C. Are Herbal Compounds the Next Frontier for Alleviating Learning and Memory Impairments? An Integrative Look at Memory, Dementia and the Promising Therapeutics of Traditional Chinese Medicines. Phytother Res 2011; 25:1105-18. [DOI: 10.1002/ptr.3388] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 11/18/2010] [Accepted: 11/22/2010] [Indexed: 12/28/2022]
Affiliation(s)
- Robert Jesky
- Department of General Surgery-Integrated Traditional Chinese and Western Medicine; 1st Affiliated Hospital of Dalian Medical University; No. 222, Zhongshan Road, Xigang District Dalian China
| | - Chen Hailong
- Department of General Surgery-Integrated Traditional Chinese and Western Medicine; 1st Affiliated Hospital of Dalian Medical University; No. 222, Zhongshan Road, Xigang District Dalian China
| |
Collapse
|
37
|
Maiese K, Chong ZZ, Shang YC, Hou J. Novel avenues of drug discovery and biomarkers for diabetes mellitus. J Clin Pharmacol 2011; 51:128-52. [PMID: 20220043 PMCID: PMC3033756 DOI: 10.1177/0091270010362904] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Globally, developed nations spend a significant amount of their resources on health care initiatives that poorly translate into increased population life expectancy. As an example, the United States devotes 16% of its gross domestic product to health care, the highest level in the world, but falls behind other nations that enjoy greater individual life expectancy. These observations point to the need for pioneering avenues of drug discovery to increase life span with controlled costs. In particular, innovative drug development for metabolic disorders such as diabetes mellitus becomes increasingly critical given that the number of diabetic people will increase exponentially over the next 20 years. This article discusses the elucidation and targeting of novel cellular pathways that are intimately tied to oxidative stress in diabetes mellitus for new treatment strategies. Pathways that involve wingless, β-nicotinamide adenine dinucleotide (NAD(+)) precursors, and cytokines govern complex biological pathways that determine both cell survival and longevity during diabetes mellitus and its complications. Furthermore, the role of these entities as biomarkers for disease can further enhance their utility irrespective of their treatment potential. Greater understanding of the intricacies of these unique cellular mechanisms will shape future drug discovery for diabetes mellitus to provide focused clinical care with limited or absent long-term complications.
Collapse
Affiliation(s)
- Kenneth Maiese
- Department of Neurology, 8C-1 UHC, Wayne State University School of Medicine, 4201 St. Antoine, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
38
|
Chong ZZ, Shang YC, Hou J, Maiese K. Wnt1 neuroprotection translates into improved neurological function during oxidant stress and cerebral ischemia through AKT1 and mitochondrial apoptotic pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2010; 3:153-65. [PMID: 20716939 DOI: 10.4161/oxim.3.2.11758] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although essential for the development of the nervous system, Wnt1 also has been associated with neurodegenerative disease and cognitive loss during periods of oxidative stress. Here we show that endogenous expression of Wnt1 is suppressed during oxidative stress in both in vitro and in vivo experimental models. Loss of endogenous Wnt1 signaling directly correlates with neuronal demise and increased functional deficit, illustrating that endogenous neuronal Wnt1 offers a vital level of intrinsic cellular protection against oxidative stress. Furthermore, transient overexpression of Wnt1 or application of exogenous Wnt1 recombinant protein is necessary to preserve neurological function and rescue neurons from apoptotic membrane phosphatidylserine externalization and genomic DNA degradation, since blockade of Wnt1 signaling with a Wnt1 antibody or dickkopf related protein 1 abrogates neuronal protection by Wnt1. Wnt1 ultimately relies upon the activation of Akt1, the modulation of mitochondrial membrane permeability, and the release of cytochrome c to control the apoptotic cascade, since inhibition of Wnt1 signaling, the phosphatidylinositol 3-kinase pathway, or Akt1 activity abrogates the ability of Wnt1 to block these apoptotic components. Our work identifies Wnt1 and its downstream signaling as cellular targets with high clinical potential for novel treatment strategies for multiple disorders precipitated by oxidative stress.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | | | |
Collapse
|
39
|
Okamoto H, Voleti B, Banasr M, Sarhan M, Duric V, Girgenti MJ, Dileone RJ, Newton SS, Duman RS. Wnt2 expression and signaling is increased by different classes of antidepressant treatments. Biol Psychiatry 2010; 68:521-7. [PMID: 20570247 PMCID: PMC2929274 DOI: 10.1016/j.biopsych.2010.04.023] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 04/09/2010] [Accepted: 04/15/2010] [Indexed: 12/19/2022]
Abstract
BACKGROUND Despite recent interest in glycogen synthase kinase-3beta (GSK-3beta) as a target for the treatment of mood disorders, there has been very little work related to these illnesses on the upstream signaling molecules that regulate this kinase as well as downstream targets. METHODS With a focused microarray approach we examined the influence of different classes of antidepressants on Wnt signaling that controls GSK-3beta activity as well as the transcription factors that contribute to the actions of GSK-3beta. RESULTS The results demonstrate that Wnt2 is a common target of different classes of antidepressants and also show differential regulation of Wnt-GSK-3beta signaling genes. Increased expression and function of Wnt2 was confirmed by secondary measures. Moreover, with a viral vector approach we demonstrate that increased expression of Wnt2 in the hippocampus is sufficient to produce antidepressant-like behavioral actions in well-established models of depression and treatment response. CONCLUSIONS These findings demonstrate that Wnt2 expression and signaling is a common target of antidepressants and that increased Wnt2 is sufficient to produce antidepressant effects.
Collapse
Affiliation(s)
- Hideki Okamoto
- Connecticut Mental Health Center, Yale University School of Medicine, New Haven, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Shang YC, Chong ZZ, Hou J, Maiese K. Wnt1, FoxO3a, and NF-kappaB oversee microglial integrity and activation during oxidant stress. Cell Signal 2010; 22:1317-29. [PMID: 20462515 PMCID: PMC2893230 DOI: 10.1016/j.cellsig.2010.04.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 04/29/2010] [Indexed: 01/08/2023]
Abstract
Elucidating the underlying mechanisms that govern microglial activation and survival is essential for the development of new treatment strategies for neurodegenerative disorders, since microglia serve not only as guardian sentries of the nervous system, but also play a significant role in determining neuronal and vascular cell fate. Here we show that endogenous and exogenous Wnt1 in inflammatory microglial cells is necessary for the prevention of apoptotic early membrane phosphatidylserine exposure and later DNA degradation, since blockade of Wnt1 signaling abrogates cell survival during oxidative stress. Wnt1 prevents apoptotic demise through the post-translational phosphorylation and maintenance of FoxO3a in the cytoplasm to inhibit an apoptotic cascade that relies upon the loss of mitochondrial membrane permeability, cytochrome c release, Bad phosphorylation, and activation of caspase 3 and caspase 1 as demonstrated by complimentary gene knockdown studies of FoxO3a. Furthermore, subcellular trafficking and gene knockdown studies of NF-kappaB p65 illustrate that microglial cell survival determined by Wnt1 during oxidative stress requires NF-kappaB p65. Our work highlights Wnt1 and the control of novel downstream transcriptional pathways as critical components for the oversight of nervous system microglial cells.
Collapse
Affiliation(s)
- Yan Chen Shang
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Zhao Zhong Chong
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Jinling Hou
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Kenneth Maiese
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201
- Departments of Neurology and Anatomy & Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201
- Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201
- Institute of Environmental Health Sciences, Wayne State University School of Medicine, Detroit, Michigan 48201
| |
Collapse
|
41
|
Maiese K, Shang YC, Chong ZZ, Hou J. Diabetes mellitus: channeling care through cellular discovery. Curr Neurovasc Res 2010; 7:59-64. [PMID: 20158461 DOI: 10.2174/156720210790820217] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 12/29/2009] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus (DM) impacts a significant portion of the world's population and care for this disorder places an economic burden on the gross domestic product for any particular country. Furthermore, both Type 1 and Type 2 DM are becoming increasingly prevalent and there is increased incidence of impaired glucose tolerance in the young. The complications of DM are protean and can involve multiple systems throughout the body that are susceptible to the detrimental effects of oxidative stress and apoptotic cell injury. For these reasons, innovative strategies are necessary for the implementation of new treatments for DM that are generated through the further understanding of cellular pathways that govern the pathological consequences of DM. In particular, both the precursor for the coenzyme beta-nicotinamide adenine dinucleotide (NAD(+)), nicotinamide, and the growth factor erythropoietin offer novel platforms for drug discovery that involve cellular metabolic homeostasis and inflammatory cell control. Interestingly, these agents and their tightly associated pathways that consist of cell cycle regulation, protein kinase B, forkhead transcription factors, and Wnt signaling also function in a broader sense as biomarkers for disease onset and progression.
Collapse
Affiliation(s)
- Kenneth Maiese
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201, USA.
| | | | | | | |
Collapse
|
42
|
Miao Y, Chen J, Zhang Q, Sun A. Deletion of tau attenuates heat shock-induced injury in cultured cortical neurons. J Neurosci Res 2010; 88:102-10. [PMID: 19642195 DOI: 10.1002/jnr.22188] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The microtubule-associated protein tau has been implicated in beta-amyloid- and glutamate-induced neurotoxicity. However, the potential role of tau in response to other insults to neurons remains unclear. In this study, we examined whether deletion of tau would change cell injury induced by heat shock in primary cultures of cortical neurons. After 30 min of a 45 degrees C heat shock, lactate dehydrogenase (LDH) release increased, reaching a peak at 6 hr in wild-type (WT) neurons. A significantly lower LDH release, with a peak delayed by 24 hr, was detected in tau knockout (TKO) neurons. After heat shock treatment, MAP-2 and tubulin staining of the processes of WT neurons revealed more dramatic abnormalities than in TKO neurons. Both WT and TKO neurons exhibited a similar elevation of HSP70 level but different time courses of Akt phosphorylation. In contrast to an early, brief response in WT neurons, TKO neurons displayed a late, but long-lasting increase in phosphorylation of Akt and its downstream target, glycogen synthase kinase 3beta. Additionally, inhibition of Akt activity aggravated the cell morbidity caused by heat shock exposure in both WT and TKO neurons, indicating a protective role of Akt against cell injury. In conclusion, our results demonstrate that deletion of tau attenuated heat shock-induced neuronal injury. Enhanced Akt response in the absence of endogenous tau is suggested to represent a compensatory mechanism for regulating cell reactions to stress stimuli.
Collapse
Affiliation(s)
- Yanying Miao
- Institute of Neurobiology, Fudan University, Shanghai, People's Republic of China.
| | | | | | | |
Collapse
|
43
|
Oxidative stress: Biomarkers and novel therapeutic pathways. Exp Gerontol 2010; 45:217-34. [PMID: 20064603 DOI: 10.1016/j.exger.2010.01.004] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 12/28/2009] [Accepted: 01/07/2010] [Indexed: 01/12/2023]
Abstract
Oxidative stress significantly impacts multiple cellular pathways that can lead to the initiation and progression of varied disorders throughout the body. It therefore becomes imperative to elucidate the components and function of novel therapeutic strategies against oxidative stress to further clinical diagnosis and care. In particular, both the growth factor and cytokine erythropoietin (EPO) and members of the mammalian forkhead transcription factors of the O class (FoxOs) may offer the greatest promise for new treatment regimens since these agents and the cellular pathways they oversee cover a range of critical functions that directly influence progenitor cell development, cell survival and degeneration, metabolism, immune function, and cancer cell invasion. Furthermore, both EPO and FoxOs function not only as therapeutic targets, but also as biomarkers of disease onset and progression, since their cellular pathways are closely linked and overlap with several unique signal transduction pathways. However, biological outcome with EPO and FoxOs may sometimes be both unexpected and undesirable that can raise caution for these agents and warrant further investigations. Here we present the exciting as well as complicated role EPO and FoxOs possess to uncover the benefits as well as the risks of these agents for cell biology and clinical care in processes that range from stem cell development to uncontrolled cellular proliferation.
Collapse
|
44
|
Amorim MA, Guerra-Araiza C, Garcia-Segura LM. Progesterone as a regulator of phosphorylation in the central nervous system. Horm Mol Biol Clin Investig 2010; 4:601-7. [DOI: 10.1515/hmbci.2010.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/05/2010] [Indexed: 11/15/2022]
Abstract
AbstractProgesterone exerts a variety of actions in the central nervous system under physiological and pathological conditions. As in other tissues, progesterone acts in the brain through classical progesterone receptors and through alternative mechanisms. Here, we review the role of progesterone as a regulator of kinases and phosphatases, such as extracellular-signal regulated kinases, phosphoinositide 3-kinase, Akt, glycogen synthase kinase 3, protein phosphatase 2A and phosphatase and tensin homolog deleted on chromosome 10. In addition, we analyzed the effects of progesterone on the phosphorylation of Tau, a protein that is involved in microtubule stabilization in neurons.
Collapse
|
45
|
Maiese K, Chong ZZ, Hou J, Shang YC. New strategies for Alzheimer's disease and cognitive impairment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2009; 2:279-89. [PMID: 20716915 PMCID: PMC2835916 DOI: 10.4161/oxim.2.5.9990] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 08/24/2009] [Accepted: 09/02/2009] [Indexed: 02/06/2023]
Abstract
Approximately five million people suffer with Alzheimer's disease (AD) and more than twenty-four million people are diagnosed with AD, pre-senile dementia, and other disorders of cognitive loss worldwide. Furthermore, the annual cost per patient with AD can approach $200,000 with an annual population aggregate cost of $100 billion. Yet, complete therapeutic prevention or reversal of neurovascular injury during AD and cognitive loss is not achievable despite the current understanding of the cellular pathways that modulate nervous system injury during these disorders. As a result, identification of novel therapeutic targets for the treatment of neurovascular injury would be extremely beneficial to reduce or eliminate disability from diseases that lead to cognitive loss or impairment. Here we describe the capacity of intrinsic cellular mechanisms for the novel pathways of erythropoietin and forkhead transcription factors that may offer not only new strategies for disorders such as AD and cognitive loss, but also function as biomarkers for disease onset and progression.
Collapse
Affiliation(s)
- Kenneth Maiese
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, USA.
| | | | | | | |
Collapse
|
46
|
Maiese K, Hou J, Chong ZZ, Shang YC. Erythropoietin, forkhead proteins, and oxidative injury: biomarkers and biology. ScientificWorldJournal 2009; 9:1072-104. [PMID: 19802503 PMCID: PMC2762199 DOI: 10.1100/tsw.2009.121] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Oxidative stress significantly impacts multiple cellular pathways that can lead to the initiation and progression of varied disorders throughout the body. It therefore becomes imperative to elucidate the components and function of novel therapeutic strategies against oxidative stress to further clinical diagnosis and care. In particular, both the growth factor and cytokine erythropoietin (EPO), and members of the mammalian forkhead transcription factors of the O class (FoxOs), may offer the greatest promise for new treatment regimens, since these agents and the cellular pathways they oversee cover a range of critical functions that directly influence progenitor cell development, cell survival and degeneration, metabolism, immune function, and cancer cell invasion. Furthermore, both EPO and FoxOs function not only as therapeutic targets, but also as biomarkers of disease onset and progression, since their cellular pathways are closely linked and overlap with several unique signal transduction pathways. Yet, EPO and FoxOs may sometimes have unexpected and undesirable effects that can raise caution for these agents and warrant further investigations. Here we present the exciting as well as the complex role that EPO and FoxOs possess to uncover the benefits as well as the risks of these agents for cell biology and clinical care in processes that range from stem cell development to uncontrolled cellular proliferation.
Collapse
Affiliation(s)
- Kenneth Maiese
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan, USA.
| | | | | | | |
Collapse
|
47
|
Regulation of Epithelial-Mesenchymal Transition in Palatal Fusion. Exp Biol Med (Maywood) 2009; 234:483-91. [DOI: 10.3181/0812-mr-365] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
During palatal fusion, the midline epithelial seam between the palatal shelves degrades to achieve mesenchymal confluence. Morphological and molecular evidence support the theory that the epithelial-mesenchymal transition is one mechanism that regulates palatal fusion. It appears that transforming growth factor (TGF)-β signaling plays a role in palatal EMT. TGFβ3 is the main inducer in palatal fusion and activates both Smad-dependent and -independent signaling pathways, including the key EMT transcription factors, Lef1, Twist, and Snail1, in the MEE prior to the palatal EMT program. The roles and interactions among these transcription factors will be discussed.
Collapse
|
48
|
Kihira T, Suzuki A, Kondo T, Wakayama I, Yoshida S, Hasegawa K, Garruto RM. Immunohistochemical expression of IGF-I and GSK in the spinal cord of Kii and Guamanian ALS patients. Neuropathology 2009; 29:548-58. [PMID: 19323791 DOI: 10.1111/j.1440-1789.2009.01010.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insulin-like growth factor-I (IGF-I) is a potent survival factor for motor neurons in animals, and glycogen synthase kinase-3beta (GSK-3beta) is suspected to play roles in apoptosis and tau phosphorylation. Here we report the immunological expression of IGF-I, GSK-3beta, phosphorylated-GSK-3alpha/beta (p-GSK-3alpha/beta) and phosphorylated-tau in the spinal cord and hippocampus of Kii and Guam amyotrophic lateral sclerosis (ALS) patients. Sixteen ALS patients (10 Japanese sporadic, 3 Kii and 3 Guam ALS) and 14 neurological controls (10 Japanese and 4 Guamanian) were examined. The immunoreactivity for each antibody was rated by the percentages of positive neurons to total anterior horn neurons in each patient and was analyzed statistically. Many normal-looking neurons from Japanese sporadic ALS, Kii ALS and Guam ALS patients, as well as from Japanese and Guam controls, were positive for anti-IGF-I antibody. A positive correlation between IR scores for anti-IGF-I antibody and clinical durations of Japanese sporadic ALS patients was found in this study (P < 0.0001). This suggested that IGF-I might have a protective effect against ALS degeneration. In Japanese sporadic ALS patients, abnormal as well as normal-looking neurons showed significant high IR scores for anti-GSK-3beta antibody than those of controls. Anterior horn neurons from Guam and Kii ALS patients characteristically showed weak staining for anti-GSK-3beta antibody but were markedly positive for anti-pGSK-3alpha/beta antibody compared to those from both Japanese controls and Japanese sporadic ALS patients, and showed the co-localization of IGF-I and p-GSK-3alpha/beta. This suggested that the IGF-I signaling pathway in Guam and Kii ALS patients might function to phosphorylate GSK-3beta to protect neurons from ALS degeneration. Neurofibrillary tangles (NFTs) in the hippocampus and spinal cord from Kii and Guam ALS patients showed the co-localization of PHF-tau and p-GSK-3alpha/beta by a confocal laser scanning technique. The predominant expression of p-GSK-3alpha/beta compared to GSK-3beta in spinal motor neurons and the co-localization of p-GSK-3alpha/beta and PHF-tau in NFT-laden neurons in the hippocampus and spinal cord were characteristic findings of Kii and Guam ALS patients.
Collapse
Affiliation(s)
- Tameko Kihira
- Department of Neurology, Wakayama Medical University, Wakayama City, Wakayama Prefecture, Japan.
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Alzheimer's disease is characterized by abnormal elevation of Abeta peptide and abnormal hyperphosphorylation of the tau protein. The "amyloid hypothesis," which is based on molecular defects observed in autosomal-dominant early-onset Alzheimer's disease (EOAD), suggests a serial model of causality, whereby elevation of Abeta drives other disease features including tau hyperphosphorylation. Here, we review recent evidence from drug trials, genetic studies, and experimental work in animal models that suggests that an alternative model might exist in late-onset AD (LOAD), the complex and more common form of the disease. Specifically, we hypothesize a "dual pathway" model of causality, whereby Abeta and tau can be linked by separate mechanisms driven by a common upstream driver. This model may account for the results of recent drug trials and, if confirmed, may guide future drug development.
Collapse
Affiliation(s)
- Scott A. Small
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Departments of Neurology and Pathology, Columbia University College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Karen Duff
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Departments of Neurology and Pathology, Columbia University College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|