1
|
Yoo J, Lee J, Ahn B, Han J, Lim MH. Multi-target-directed therapeutic strategies for Alzheimer's disease: controlling amyloid-β aggregation, metal ion homeostasis, and enzyme inhibition. Chem Sci 2025:d4sc06762b. [PMID: 39810997 PMCID: PMC11726323 DOI: 10.1039/d4sc06762b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia, marked by progressive cognitive decline and memory impairment. Despite advances in therapeutic research, single-target-directed treatments often fall short in addressing the complex, multifactorial nature of AD. This arises from various pathological features, including amyloid-β (Aβ) aggregate deposition, metal ion dysregulation, oxidative stress, impaired neurotransmission, neuroinflammation, mitochondrial dysfunction, and neuronal cell death. This review illustrates their interrelationships, with a particular emphasis on the interplay among Aβ, metal ions, and AD-related enzymes, such as β-site amyloid precursor protein cleaving enzyme 1 (BACE1), matrix metalloproteinase 9 (MMP9), lysyl oxidase-like 2 (LOXL2), acetylcholinesterase (AChE), and monoamine oxidase B (MAOB). We further underscore the potential of therapeutic strategies that simultaneously inhibit Aβ aggregation and address other pathogenic mechanisms. These approaches offer a more comprehensive and effective method for combating AD, overcoming the limitations of conventional therapies.
Collapse
Affiliation(s)
- Jeasang Yoo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jimin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Byeongha Ahn
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul Seoul 02504 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
2
|
Jiang X, Wang Y, Lin Z, Li C, Wang Q, Zhang J, Liu X, Li Z, Cui C. Polygonatum sibiricum polysaccharides: A promising strategy in the treatment of neurodegenerative disease. Neurochem Int 2024; 181:105902. [PMID: 39542041 DOI: 10.1016/j.neuint.2024.105902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Neurodegenerative diseases (NDDs), as a neurological disorder characterised by neuronal degeneration and death, are a serious threat to human health and have long attracted attention due to their complex pathogenesis and the ineffectiveness of therapeutic drugs. Existing studies have shown that Polygonatum Sibiricum polysaccharides (PSP) have immunoregulatory, antioxidant, anti-inflammatory and other pharmacological effects, and their neuroprotective effects have been demonstrated in several scientific studies. This paper reviews the main pharmacological effects and mechanisms of PSP in the protection and treatment of NDDs, to provide a reference for the clinical application and basic research of PSP in NDDs.
Collapse
Affiliation(s)
- Xue Jiang
- Shandong Medicine Technician College, 271000, Taian, China; Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, 271000, Taian, China
| | - Yumei Wang
- Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, 271000, Taian, China
| | - Zhaochen Lin
- Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, 271000, Taian, China
| | - Chao Li
- Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, 271000, Taian, China
| | - Qian Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Junyan Zhang
- College of Life Sciences, Northwest A & F University, 710000, Xi'an, China
| | - Xiuhua Liu
- Shandong Taishan Sealwort Biotechnology Limited Liability Company, 271000, Taian, China
| | - Ziye Li
- Xiangya School of Public Health, Central South University, 410078, Changsha, China
| | - Chao Cui
- Qilu Hospital of Shandong University Dezhou Hospital, 253000, Dezhou, China; Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, 271000, Taian, China.
| |
Collapse
|
3
|
Singh L. Daidzein's potential in halting neurodegeneration: unveiling mechanistic insights. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03356-5. [PMID: 39158734 DOI: 10.1007/s00210-024-03356-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/01/2024] [Indexed: 08/20/2024]
Abstract
Neurological conditions encompassing a wide range of disorders pose significant challenges globally. The complex interactions among signaling pathways and molecular elements play pivotal roles in the initiation and progression of neurodegenerative diseases. Isoflavones have emerged as a promising candidate to fight against neurodegenerative diseases. Daidzein, a 7-hydroxy-3-(4-hydroxyphenyl)-chromen-4-one, belongs to the isoflavone class and exhibits a diverse pharmacological profile. It is found primarily in soybeans and soy products, as well as in some other legumes and herbs. Investigations into daidzein have revealed that it confers neuroprotection by inhibiting oxidative stress, inflammation, and apoptosis, which are key contributors to neuronal damage and degeneration. Activating pathways like PI3K/Akt/mTOR and promoting neurotrophic factors like BDNF by daidzein underscore its potential in supporting neuronal function and combating neurodegeneration. Daidzein's effects on dopamine provide further avenues for intervention in conditions like Parkinson's disease. Additionally, the modulation of inflammatory and NRF-2-antioxidant signaling by daidzein reinforces its neuroprotective role. Moreover, daidzein's interaction with receptors and cellular processes like ER-β, GPR30, MAO, VEGF, and GnRH highlights its multifaceted effects across multiple pathways involved in neuroprotection and neuronal function. This review article delves into the mechanistic interplay of various mediators in mediating the neuroprotective effects of daidzein. The review article consolidates and analyzes research published over nearly two decades (2005-2024) from various databases, including PubMed, Scopus, ScienceDirect, and Web of Science, to provide a comprehensive understanding of daidzein's effects and mechanisms in neuroprotection.
Collapse
Affiliation(s)
- Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| |
Collapse
|
4
|
Xie W, Liu W, Wang L, Zhu B, Zhao C, Liao Z, Li Y, Jiang X, Liu J, Ren C. Roles of THEM4 in the Akt pathway: a double-edged sword. J Zhejiang Univ Sci B 2024; 25:541-556. [PMID: 39011675 PMCID: PMC11254685 DOI: 10.1631/jzus.b2300457] [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: 06/26/2023] [Accepted: 10/10/2023] [Indexed: 07/17/2024]
Abstract
The protein kinase B (Akt) pathway can regulate the growth, proliferation, and metabolism of tumor cells and stem cells through the activation of multiple downstream target genes, thus affecting the development and treatment of a range of diseases. Thioesterase superfamily member 4 (THEM4), a member of the thioesterase superfamily, is one of the Akt kinase-binding proteins. Some studies on the mechanism of cancers and other diseases have shown that THEM4 binds to Akt to regulate its phosphorylation. Initially, THEM4 was considered an endogenous inhibitor of Akt, which can inhibit the phosphorylation of Akt in diseases such as lung cancer, pancreatic cancer, and liver cancer, but subsequently, THEM4 was shown to promote the proliferation of tumor cells by positively regulating Akt activity in breast cancer and nasopharyngeal carcinoma, which contradicts previous findings. Considering these two distinct views, this review summarizes the important roles of THEM4 in the Akt pathway, focusing on THEM4 as an Akt-binding protein and its regulatory relationship with Akt phosphorylation in various diseases, especially cancer. This work provides a better understanding of the roles of THEM4 combined with Akt in the treatment of diseases.
Collapse
Affiliation(s)
- Wen Xie
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Weidong Liu
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Lei Wang
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Bin Zhu
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Cong Zhao
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Ziling Liao
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Yihan Li
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Xingjun Jiang
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Liu
- Department of Critical Care Medicine, Hainan Hospital of Chinese PLA General Hospital, Hainan Province Clinical Medical Center, Sanya 572013, China. ,
| | - Caiping Ren
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China.
| |
Collapse
|
5
|
Lu XY, Lv QY, Li QL, Zhang H, Chen CT, Tian HM. Impact of acupuncture on ischemia/reperfusion injury: Unraveling the role of miR-34c-5p and autophagy activation. Brain Res Bull 2024; 215:111031. [PMID: 39002935 DOI: 10.1016/j.brainresbull.2024.111031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
We have previously reported that the expression of miR-34c-5p was up-regulated during acupuncture treatment in the setting of a cerebral ischemia/reperfusion injury (CIRI), indicating that miR-34c-5p plays an important role in healing from a CIRI-induced brain injury. This study sought to evaluate the effects of acupuncture on miR-34c-5p expression and autophagy in the forward and reverse directions using a rat focal cerebral ischemia/reperfusion model. After 120 minutes of middle cerebral artery occlusion and reperfusion, rats were treated with acupuncture at the "Dazhui" (DU20), "Baihui" (DU26) and "Renzhong" (DU14) points. Neurologic function deficit score, cerebral infarct area ratio, neuronal apoptosis and miR-34c-5p expression were evaluated 72 hr after treatment. The autophagy agonist RAPA and the antagonist 3MA were used to evaluate the neuro protective effects of autophagy-mediated acupuncture. We found that acupuncture treatment improved autophagy in the brain tissue of CIRI rats. Acupuncture reversed the negative effects of 3MA on CIRI, and acupuncture combined with RAPA further enhanced autophagy. We also found that acupuncture could increase miR-34c-5p expression in hippocampal neurons after ischemia/reperfusion. Acupuncture and a miR-34c agomir were able to enhance autophagy, improve neurologic deficits, and reduce the cerebral infarct area ratio and apoptosis rate by promoting the expression of miR-34c-5p. Silencing miR-34c resulted in a significantly reduced activating effect of acupuncture on autophagy and increased apoptosis, neurologic deficit symptoms, and cerebral infarct area ratio. This confirms that acupuncture can upregulate miR-34c-5p expression, which is beneficial in the treatment of CIRI.
Collapse
Affiliation(s)
- Xiao-Ye Lu
- College of Acupuncture and Tuina and Rehabilitation, Hunan University of Traditional Chinese Medicine, Changsha, Hunan Province 410007, China; Department of Rehabilitation, Changsha Central Hospital, Changsha, Hunan Province 410004, China
| | - Qian-Yi Lv
- College of Acupuncture and Tuina and Rehabilitation, Hunan University of Traditional Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Qi-Long Li
- College of Acupuncture and Tuina and Rehabilitation, Hunan University of Traditional Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Hong Zhang
- College of Acupuncture and Tuina and Rehabilitation, Hunan University of Traditional Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Chu-Tao Chen
- College of Acupuncture and Tuina and Rehabilitation, Hunan University of Traditional Chinese Medicine, Changsha, Hunan Province 410007, China.
| | - Hao-Mei Tian
- College of Acupuncture and Tuina and Rehabilitation, Hunan University of Traditional Chinese Medicine, Changsha, Hunan Province 410007, China.
| |
Collapse
|
6
|
Hou Z, Sun L, Jiang Z, Zeng T, Wu P, Huang J, Liu H, Xiao P. Neuropharmacological insights into Gardenia jasminoides Ellis: Harnessing therapeutic potential for central nervous system disorders. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155374. [PMID: 38301302 DOI: 10.1016/j.phymed.2024.155374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND In China, Gardenia jasminoides Ellis (GJE) has a longstanding history of application. The Ministry of Health has listed it as one of the first pharmaceutical or food resources. In ethnic, traditional, and folk medicine, GJE has been used to treat fever and cold and relieve nervous anxiety. Recent studies have confirmed the significant efficacy of GJE for treating central nervous system (CNS) disorders, including Alzheimer's disease, Parkinson's disease, and major depressive disorder; however, GJE has not been systematically evaluated. PURPOSE This research systematically summarizes global studies on the use of GJE for treating CNS disorders and explores the potential applications and underlying mechanisms via intestinal flora analysis and network pharmacology, aiming to establish a scientific basis for innovative CNS disorder treatment with GJE. METHODS The PRISMA guidelines were used, and electronic databases such as the Web of Science, PubMed, and China National Knowledge Infrastructure were searched using the following search terms: "Gardenia jasminoides Ellis" with "central nervous system disease," "neuroprotection," "Alzheimer's disease," "Parkinson's disease," "ischemic stroke," "Epilepsy," and "major depressive disorder." The published literature up to September 2023 was searched to obtain relevant information on the application of GJE for treating CNS disorders. RESULTS There has been an increase in research on the material formulation and mechanisms of action of GJE for treating CNS disorders, with marked effects on CNS disorder treatment in different countries and regions. We summarized the research results related to the role of GJE in vitro and in vivo via multitargeted interventions in response to the complex mechanisms of action of CNS disorders. CONCLUSION We systematically reviewed the research progress on traditional treatment for GJE and preclinical mechanisms of CNS disorders and explored the potential of optimizing network pharmacology strategies and intestinal flora analysis to elucidate the mechanisms of action of GJE. The remarkable therapeutic efficacy of GJE, an important resource in traditional medicine, has been well documented in the literature, highlighting its significant medicinal potential.
Collapse
Affiliation(s)
- Ziyu Hou
- Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Medicinal Plant Development (IMPLAD), No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Le Sun
- Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Medicinal Plant Development (IMPLAD), No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China.
| | - Zheyu Jiang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Medicinal Plant Development (IMPLAD), No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Tiexin Zeng
- Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Medicinal Plant Development (IMPLAD), No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Peiling Wu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Medicinal Plant Development (IMPLAD), No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Jiali Huang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Medicinal Plant Development (IMPLAD), No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Haibo Liu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Medicinal Plant Development (IMPLAD), No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China.
| | - Peigen Xiao
- Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Medicinal Plant Development (IMPLAD), No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| |
Collapse
|
7
|
Maiese K. Artificial Intelligence and Disease Signature Pathways: Driving Innovation to Elucidate Underlying Pathogenic Mechanisms. Curr Neurovasc Res 2024; 21:229-233. [PMID: 38910427 DOI: 10.2174/1567202621999240621122700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
|
8
|
Maiese K. The impact of aging and oxidative stress in metabolic and nervous system disorders: programmed cell death and molecular signal transduction crosstalk. Front Immunol 2023; 14:1273570. [PMID: 38022638 PMCID: PMC10663950 DOI: 10.3389/fimmu.2023.1273570] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Life expectancy is increasing throughout the world and coincides with a rise in non-communicable diseases (NCDs), especially for metabolic disease that includes diabetes mellitus (DM) and neurodegenerative disorders. The debilitating effects of metabolic disorders influence the entire body and significantly affect the nervous system impacting greater than one billion people with disability in the peripheral nervous system as well as with cognitive loss, now the seventh leading cause of death worldwide. Metabolic disorders, such as DM, and neurologic disease remain a significant challenge for the treatment and care of individuals since present therapies may limit symptoms but do not halt overall disease progression. These clinical challenges to address the interplay between metabolic and neurodegenerative disorders warrant innovative strategies that can focus upon the underlying mechanisms of aging-related disorders, oxidative stress, cell senescence, and cell death. Programmed cell death pathways that involve autophagy, apoptosis, ferroptosis, and pyroptosis can play a critical role in metabolic and neurodegenerative disorders and oversee processes that include insulin resistance, β-cell function, mitochondrial integrity, reactive oxygen species release, and inflammatory cell activation. The silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), AMP activated protein kinase (AMPK), and Wnt1 inducible signaling pathway protein 1 (WISP1) are novel targets that can oversee programmed cell death pathways tied to β-nicotinamide adenine dinucleotide (NAD+), nicotinamide, apolipoprotein E (APOE), severe acute respiratory syndrome (SARS-CoV-2) exposure with coronavirus disease 2019 (COVID-19), and trophic factors, such as erythropoietin (EPO). The pathways of programmed cell death, SIRT1, AMPK, and WISP1 offer exciting prospects for maintaining metabolic homeostasis and nervous system function that can be compromised during aging-related disorders and lead to cognitive impairment, but these pathways have dual roles in determining the ultimate fate of cells and organ systems that warrant thoughtful insight into complex autofeedback mechanisms.
Collapse
Affiliation(s)
- Kenneth Maiese
- Innovation and Commercialization, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
9
|
Maiese K. Cognitive Impairment in Multiple Sclerosis. Bioengineering (Basel) 2023; 10:871. [PMID: 37508898 PMCID: PMC10376413 DOI: 10.3390/bioengineering10070871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Almost three million individuals suffer from multiple sclerosis (MS) throughout the world, a demyelinating disease in the nervous system with increased prevalence over the last five decades, and is now being recognized as one significant etiology of cognitive loss and dementia. Presently, disease modifying therapies can limit the rate of relapse and potentially reduce brain volume loss in patients with MS, but unfortunately cannot prevent disease progression or the onset of cognitive disability. Innovative strategies are therefore required to address areas of inflammation, immune cell activation, and cell survival that involve novel pathways of programmed cell death, mammalian forkhead transcription factors (FoxOs), the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), and associated pathways with the apolipoprotein E (APOE-ε4) gene and severe acute respiratory syndrome coronavirus (SARS-CoV-2). These pathways are intertwined at multiple levels and can involve metabolic oversight with cellular metabolism dependent upon nicotinamide adenine dinucleotide (NAD+). Insight into the mechanisms of these pathways can provide new avenues of discovery for the therapeutic treatment of dementia and loss in cognition that occurs during MS.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
10
|
Maiese K. Cellular Metabolism: A Fundamental Component of Degeneration in the Nervous System. Biomolecules 2023; 13:816. [PMID: 37238686 PMCID: PMC10216724 DOI: 10.3390/biom13050816] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
It is estimated that, at minimum, 500 million individuals suffer from cellular metabolic dysfunction, such as diabetes mellitus (DM), throughout the world. Even more concerning is the knowledge that metabolic disease is intimately tied to neurodegenerative disorders, affecting both the central and peripheral nervous systems as well as leading to dementia, the seventh leading cause of death. New and innovative therapeutic strategies that address cellular metabolism, apoptosis, autophagy, and pyroptosis, the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), growth factor signaling with erythropoietin (EPO), and risk factors such as the apolipoprotein E (APOE-ε4) gene and coronavirus disease 2019 (COVID-19) can offer valuable insights for the clinical care and treatment of neurodegenerative disorders impacted by cellular metabolic disease. Critical insight into and modulation of these complex pathways are required since mTOR signaling pathways, such as AMPK activation, can improve memory retention in Alzheimer's disease (AD) and DM, promote healthy aging, facilitate clearance of β-amyloid (Aß) and tau in the brain, and control inflammation, but also may lead to cognitive loss and long-COVID syndrome through mechanisms that can include oxidative stress, mitochondrial dysfunction, cytokine release, and APOE-ε4 if pathways such as autophagy and other mechanisms of programmed cell death are left unchecked.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
11
|
Kumari S, Dhapola R, Reddy DH. Apoptosis in Alzheimer's disease: insight into the signaling pathways and therapeutic avenues. Apoptosis 2023:10.1007/s10495-023-01848-y. [PMID: 37186274 DOI: 10.1007/s10495-023-01848-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/17/2023]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of hyperphosphorylated tau and amyloid-β (Aβ) protein resulting in synaptic loss and apoptosis. Aβ and tau deposition trigger apoptotic pathways that result in neuronal death. Apoptosis is considered to be responsible for manifestations associated with AD under pathological conditions. It regulates via extrinsic and intrinsic pathways. It activates various proteins including Bcl-2 family proteins like Bax, Bad, Bid, Bcl-XS, Bcl-XL and caspases comprising of initiator, effector and inflammatory caspases carried out through a cascade of events that finally lead to cell disintegration. The apoptotic elements interact with trophic factors, signaling molecules including Ras-ERK, JNK, GSK-3β, BDNF/TrkB/CREB and PI3K/AKT/mTOR. Ras-ERK signaling is involved in the progression of cell cycle and apoptosis. JNK pathway is also upregulated in AD which results in decreased expression of anti-apoptotic proteins. JAK-STAT triggers caspase-3 mediated apoptosis leading to neurodegeneration. The imbalance between autophagy and apoptosis is regulated by PI3K/Akt/mTOR pathway. GSK-3β is involved in the stimulation of pro-apoptotic factors resulting in dysregulation of apoptosis. Drugs like filgrastim, epigallocatechin gallate, curcumin, nicergoline and minocycline are under development which target these pathways and modulate the disease condition. This study sheds light on apoptotic pathways that are cardinal for neuronal survival and perform crucial role in the occurrence of AD along with the trends in therapeutics targeting apoptosis induced AD. To develop prospective treatments for AD, it is desirable to elucidate potential targets including restoration apoptotic balance, regulation of caspases, Bcl-2 and other crucial proteins involved in apoptosis mediated AD.
Collapse
Affiliation(s)
- Sneha Kumari
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Rishika Dhapola
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Dibbanti HariKrishna Reddy
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
12
|
HU K, GAO Y, CHU S, CHEN N. Review of the effects and Mechanisms of microglial autophagy in ischemic stroke. Int Immunopharmacol 2022; 108:108761. [DOI: 10.1016/j.intimp.2022.108761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/28/2022] [Accepted: 04/03/2022] [Indexed: 12/30/2022]
|
13
|
Maiese K. Biomarkers for Parkinson's Disease and Neurodegenerative Disorders: A Role for Non-coding RNAs. Curr Neurovasc Res 2022; 19:127-130. [PMID: 35657043 DOI: 10.2174/1567202619666220602125806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
14
|
Maiese K. Neurodegeneration, memory loss, and dementia: the impact of biological clocks and circadian rhythm. FRONT BIOSCI-LANDMRK 2021; 26:614-627. [PMID: 34590471 PMCID: PMC8756734 DOI: 10.52586/4971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 11/23/2022]
Abstract
Introduction: Dementia and cognitive loss impact a significant proportion of the global population and present almost insurmountable challenges for treatment since they stem from multifactorial etiologies. Innovative avenues for treatment are highly warranted. Methods and results: Novel work with biological clock genes that oversee circadian rhythm may meet this critical need by focusing upon the pathways of the mechanistic target of rapamycin (mTOR), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), mammalian forkhead transcription factors (FoxOs), the growth factor erythropoietin (EPO), and the wingless Wnt pathway. These pathways are complex in nature, intimately associated with autophagy that can maintain circadian rhythm, and have an intricate relationship that can lead to beneficial outcomes that may offer neuroprotection, metabolic homeostasis, and prevention of cognitive loss. However, biological clocks and alterations in circadian rhythm also have the potential to lead to devastating effects involving tumorigenesis in conjunction with pathways involving Wnt that oversee angiogenesis and stem cell proliferation. Conclusions: Current work with biological clocks and circadian rhythm pathways provide exciting possibilities for the treating dementia and cognitive loss, but also provide powerful arguments to further comprehend the intimate and complex relationship among these pathways to fully potentiate desired clinical outcomes.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
15
|
|
16
|
Sun XL, Zhang JB, Guo YX, Xia TS, Xu LC, Rahmand K, Wang GP, Li XJ, Han T, Wang NN, Xin HL. Xanthohumol ameliorates memory impairment and reduces the deposition of β-amyloid in APP/PS1 mice via regulating the mTOR/LC3II and Bax/Bcl-2 signalling pathways. J Pharm Pharmacol 2021; 73:1230-1239. [PMID: 33909081 DOI: 10.1093/jpp/rgab052] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/25/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Xanthohumol (XAN) is a unique component of Humulus lupulus L. and is known for its diverse biological activities. In this study, we investigated whether Xanthohumol could ameliorate memory impairment of APP/PS1 mice, and explored its potential mechanism of action. METHODS APP/PS1 mice were used for in vivo test and were treated with N-acetylcysteine and Xanthohumol for 2 months. Learning and memory levels were evaluated by the Morris water maze. Inflammatory and oxidative markers in serum and hippocampus and the deposition of Aβ in the hippocampus were determined. Moreover, the expression of autophagy and apoptosis proteins was also evaluated by western blot. KEY FINDINGS Xanthohumol significantly reduced the latency and increased the residence time of mice in the target quadrant. Additionally, Xanthohumol increased superoxide dismutase level and reduced Interleukin-6 and Interleukin-1β levels both in serum and hippocampus. Xanthohumol also significantly reduced Aβ deposition in the hippocampus and activated autophagy and anti-apoptotic signals. CONCLUSIONS Xanthohumol effectively ameliorates memory impairment of APP/PS1 mice by activating mTOR/LC3 and Bax/Bcl-2 signalling pathways, which provides new insight into the neuroprotective effects of Xanthohumol.
Collapse
Affiliation(s)
- Xiao-Lei Sun
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, ShanghaiChina.,School of Pharmacy, Shandong University of Traditional Chinese Medicine, JinanChina
| | - Jia-Bao Zhang
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, ShanghaiChina
| | - Yun-Xiang Guo
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, ShanghaiChina
| | - Tian-Shuang Xia
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, ShanghaiChina
| | - Ling-Chuan Xu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, JinanChina
| | - Khalid Rahmand
- Faculty of Science, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Guo-Ping Wang
- Xinjiang Institute of Chinese Materia Medica, Urumqi, China
| | - Xiao-Jin Li
- Xinjiang Institute of Chinese Materia Medica, Urumqi, China
| | - Ting Han
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, ShanghaiChina
| | - Na-Ni Wang
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, ShanghaiChina
| | - Hai-Liang Xin
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, ShanghaiChina
| |
Collapse
|
17
|
Sharma VK, Singh TG, Singh S, Garg N, Dhiman S. Apoptotic Pathways and Alzheimer's Disease: Probing Therapeutic Potential. Neurochem Res 2021; 46:3103-3122. [PMID: 34386919 DOI: 10.1007/s11064-021-03418-7] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/17/2022]
Abstract
Apoptosis is an intrinsic biochemical, cellular process that regulates cell death and is crucial for cell survival, cellular homeostasis, and maintaining the optimum functional status. Apoptosis in a predetermined and programmed manner regulates several molecular events, including cell turnover, embryonic development, and immune system functions but may be the exclusive contributor to several disorders, including neurodegenerative manifestations, when it functions in an aberrant and disorganized manner. Alzheimer's disease (AD) is a fatal, chronic neurodegenerative disorder where apoptosis has a compelling and divergent role. The well-characterized pathological features of AD, including extracellular plaques of amyloid-beta, intracellular hyperphosphorylated tangles of tau protein (NFTs), inflammation, mitochondrial dysfunction, oxidative stress, and excitotoxic cell death, also instigate an abnormal apoptotic cascade in susceptible brain regions (cerebral cortex, hippocampus). The apoptotic players in these regions affect cellular organelles (mitochondria and endoplasmic reticulum), interact with trophic factors, and several pathways, including PI3K/AKT, JNK, MAPK, mTOR signalling. This dysregulated apoptotic cascade end with an abnormal neuronal loss which is a primary event that may precede the other events of AD progression and correlates well with the degree of dementia. The present review provides insight into the diverse and versatile apoptotic mechanisms that are indispensable for neuronal survival and constitute an integral part of the pathological progression of AD. Identification of potential targets (restoring apoptotic and antiapoptotic balance, caspases, TRADD, RIPK1, FADD, TNFα, etc.) may be valuable and advantageous to decide the fate of neurons and to develop potential therapeutics for treatment of AD.
Collapse
Affiliation(s)
- Vivek Kumar Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India.,Government College of Pharmacy, Rohru, District Shimla, Himachal Pradesh, 171207, India
| | | | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| |
Collapse
|
18
|
Palomino O, García-Aguilar A, González A, Guillén C, Benito M, Goya L. Biological Actions and Molecular Mechanisms of Sambucus nigra L. in Neurodegeneration: A Cell Culture Approach. Molecules 2021; 26:molecules26164829. [PMID: 34443417 PMCID: PMC8399386 DOI: 10.3390/molecules26164829] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/03/2021] [Accepted: 08/07/2021] [Indexed: 12/03/2022] Open
Abstract
Sambucus nigra flowers (elderflower) have been widely used in traditional medicine for the relief of early symptoms of common cold. Its chemical composition mainly consists of polyphenolic compounds such as flavonoids, hydroxycinnamic acids, and triterpenes. Although the antioxidant properties of polyphenols are well known, the aim of this study is to assess the antioxidant and protective potentials of Sambucus nigra flowers in the human neuroblastoma (SH-SY5Y) cell line using different in vitro approaches. The antioxidant capacity is first evaluated by the oxygen radical absorbance capacity (ORAC) and the free radical scavenging activity (DPPH) methods. Cell viability is assessed by the crystal violet method; furthermore, the intracellular ROS formation (DCFH-DA method) is determined, together with the effect on the cell antioxidant defenses: reduced glutathione (GSH) and antioxidant enzyme activities (GPx, GR). On the other hand, mTORC1 hyperactivation and autophagy blockage have been associated with an increase in the formation of protein aggregates, this promoting the transference and expansion of neurodegenerative diseases. Then, the ability of Sambucus nigra flowers in the regulation of mTORC1 signaling activity and the reduction in oxidative stress through the activation of autophagy/mitophagy flux is also examined. In this regard, search for different molecules with a potential inhibitory effect on mTORC1 activation could have multiple positive effects either in the molecular pathogenic events and/or in the progression of several diseases including neurodegenerative ones.
Collapse
Affiliation(s)
- Olga Palomino
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, University Complutense of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (O.P.); (A.G.-A.); (A.G.)
| | - Ana García-Aguilar
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, University Complutense of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (O.P.); (A.G.-A.); (A.G.)
| | - Adrián González
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, University Complutense of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (O.P.); (A.G.-A.); (A.G.)
| | - Carlos Guillén
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University Complutense of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (C.G.); (M.B.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manuel Benito
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University Complutense of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (C.G.); (M.B.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Luis Goya
- Department of Metabolism and Nutrition, Institute of Science and Food Technology and Nutrition (ICTAN—CSIC), 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-91-549-2300
| |
Collapse
|
19
|
Maiese K. Cognitive Impairment and Dementia: Gaining Insight through Circadian Clock Gene Pathways. Biomolecules 2021; 11:1002. [PMID: 34356626 PMCID: PMC8301848 DOI: 10.3390/biom11071002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 01/18/2023] Open
Abstract
Neurodegenerative disorders affect fifteen percent of the world's population and pose a significant financial burden to all nations. Cognitive impairment is the seventh leading cause of death throughout the globe. Given the enormous challenges to treat cognitive disorders, such as Alzheimer's disease, and the inability to markedly limit disease progression, circadian clock gene pathways offer an exciting strategy to address cognitive loss. Alterations in circadian clock genes can result in age-related motor deficits, affect treatment regimens with neurodegenerative disorders, and lead to the onset and progression of dementia. Interestingly, circadian pathways hold an intricate relationship with autophagy, the mechanistic target of rapamycin (mTOR), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), mammalian forkhead transcription factors (FoxOs), and the trophic factor erythropoietin. Autophagy induction is necessary to maintain circadian rhythm homeostasis and limit cortical neurodegenerative disease, but requires a fine balance in biological activity to foster proper circadian clock gene regulation that is intimately dependent upon mTOR, SIRT1, FoxOs, and growth factor expression. Circadian rhythm mechanisms offer innovative prospects for the development of new avenues to comprehend the underlying mechanisms of cognitive loss and forge ahead with new therapeutics for dementia that can offer effective clinical treatments.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
20
|
Radulovic J, Gabbay V. PFC mTOR signaling as a biological signature for cognitive deficits in bipolar disorder without psychosis. Cell Rep Med 2021; 2:100282. [PMID: 34095884 PMCID: PMC8149463 DOI: 10.1016/j.xcrm.2021.100282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Vanderplow et al.1 report decreased PFC Akt-mTOR signaling in males with bipolar disorder (BD) without psychosis compared with those with psychosis, possibly related to cognitive deficits. Understanding how cognition differs between these BD subtypes clinically and biologically remains a challenge.
Collapse
Affiliation(s)
- Jelena Radulovic
- Albert Einstein College of Medicine, Psychiatry Research Institute at Montefiore Einstein (PRIME), New York, NY, USA
| | - Vilma Gabbay
- Albert Einstein College of Medicine, Psychiatry Research Institute at Montefiore Einstein (PRIME), New York, NY, USA
- The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| |
Collapse
|
21
|
Xu Q, Guohui M, Li D, Bai F, Fang J, Zhang G, Xing Y, Zhou J, Guo Y, Kan Y. lncRNA C2dat2 facilitates autophagy and apoptosis via the miR-30d-5p/DDIT4/mTOR axis in cerebral ischemia-reperfusion injury. Aging (Albany NY) 2021; 13:11315-11335. [PMID: 33833132 PMCID: PMC8109078 DOI: 10.18632/aging.202824] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/20/2020] [Indexed: 12/19/2022]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) is an important pathophysiological process of ischemic stroke associated with various physiological and pathological processes, including autophagy and apoptosis. In this study, we examined the role and mechanism of long noncoding RNA CAMK2D-associated transcript 2 (C2dat2) in regulating CIRI in vivo and in vitro. C2dat2 up-regulation facilitated neuronal autophagy and apoptosis induced by CIRI. Mechanistically, C2dat2 acts as a competing endogenous RNA (ceRNA) to negatively regulate miR-30d-5p expression. More specifically, miR-30d-5p targeted the 3′-untranslated region of DNA damage-inducible transcript 4 (DDIT4) and silenced its target mRNA DDIT4. Additionally, C2dat2 binding with heat shock cognate 70/heat shock protein 90 blocked RNA-induced silencing complex assembly to abolish the miR-30d-5p targeting of DDIT4 and inhibited miR-30d-5p to silence its target mRNA DDIT4. Further analysis showed that C2dat2 knockdown conspicuously inhibited the up-regulation of DDIT4 and Beclin-1 levels and LC3B II/I ratio and the down-regulation of P62 and phosphorylated mammalian target of rapamycin (mTOR)/mTOR and phosphorylated-P70S6K/P70S6K ratio in Neuro-2a cells after oxygen-glucose deprivation/reoxygenation. This study first revealed that C2dat2/miR-30d-5p/DDIT4/mTOR forms a novel signaling pathway to facilitate autophagy and apoptosis induced by CIRI, contributing to the better understanding of the mechanisms of CIRI and enriching the ceRNA hypothesis in CIRI.
Collapse
Affiliation(s)
- Qian Xu
- Henan Provincial Engineering Laboratory of Insects Bio-Reactor, Nanyang Normal University, Nanyang 473000, China
| | - Ma Guohui
- Henan Provincial Engineering Laboratory of Insects Bio-Reactor, Nanyang Normal University, Nanyang 473000, China
| | - Dandan Li
- Henan Provincial Engineering Laboratory of Insects Bio-Reactor, Nanyang Normal University, Nanyang 473000, China
| | - Fanghui Bai
- Henan Provincial Nanyang Central Hospital, Nanyang 473000, China
| | - Jintao Fang
- Henan Provincial Engineering Laboratory of Insects Bio-Reactor, Nanyang Normal University, Nanyang 473000, China
| | - Gui Zhang
- Henan Provincial Engineering Laboratory of Insects Bio-Reactor, Nanyang Normal University, Nanyang 473000, China.,School of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473000, China
| | - Yuxin Xing
- Henan Provincial Engineering Laboratory of Insects Bio-Reactor, Nanyang Normal University, Nanyang 473000, China.,School of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473000, China
| | - Jiawei Zhou
- Henan Provincial Engineering Laboratory of Insects Bio-Reactor, Nanyang Normal University, Nanyang 473000, China.,School of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473000, China
| | - Yugang Guo
- Henan Provincial Engineering Laboratory of Insects Bio-Reactor, Nanyang Normal University, Nanyang 473000, China
| | - Yunchao Kan
- Henan Provincial Engineering Laboratory of Insects Bio-Reactor, Nanyang Normal University, Nanyang 473000, China
| |
Collapse
|
22
|
Yarahmadi A, Azarpira N, Mostafavi-Pour Z. Role of mTOR Complex 1 Signaling Pathway in the Pathogenesis of Diabetes Complications; A Mini Review. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2021; 10:181-189. [PMID: 35178356 PMCID: PMC8800458 DOI: 10.22088/ijmcm.bums.10.3.181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/15/2021] [Indexed: 11/25/2022]
Abstract
The mammalian target of rapamycin (mTOR) is an evolutionarily conserved serine/threonine-protein kinase that senses and combines various environmental signals to regulate the growth and homeostasis of human cells. This signaling pathway synchronizes many critical cellular processes and is involved in an increasing number of pathological conditions such as diabetes, cancer, obesity, and metabolic syndrome. Here, we review different complications of diabetes that are associated with mTOR complex 1 imbalance. We further discuss pharmacological approaches to treat diabetes complications linked to mTOR deregulation.
Collapse
Affiliation(s)
- Amir Yarahmadi
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Zohreh Mostafavi-Pour
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Corresponding author: Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
23
|
Maiese K. Dysregulation of metabolic flexibility: The impact of mTOR on autophagy in neurodegenerative disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:1-35. [PMID: 32854851 DOI: 10.1016/bs.irn.2020.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Non-communicable diseases (NCDs) that involve neurodegenerative disorders and metabolic disease impact over 400 million individuals globally. Interestingly, metabolic disorders, such as diabetes mellitus, are significant risk factors for the development of neurodegenerative diseases. Given that current therapies for these NCDs address symptomatic care, new avenues of discovery are required to offer treatments that affect disease progression. Innovative strategies that fill this void involve the mechanistic target of rapamycin (mTOR) and its associated pathways of mTOR complex 1 (mTORC1), mTOR complex 2 (mTORC2), AMP activated protein kinase (AMPK), trophic factors that include erythropoietin (EPO), and the programmed cell death pathways of autophagy and apoptosis. These pathways are intriguing in their potential to provide effective care for metabolic and neurodegenerative disorders. Yet, future work is necessary to fully comprehend the entire breadth of the mTOR pathways that can effectively and safely translate treatments to clinical medicine without the development of unexpected clinical disabilities.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY, United States.
| |
Collapse
|
24
|
Yan M, Li M, Gu S, Sun Z, Ma T, Ma X. Ginkgo biloba extract protects diabetic rats against cerebral ischemia‑reperfusion injury by suppressing oxidative stress and upregulating the expression of glutamate transporter 1. Mol Med Rep 2020; 21:1809-1818. [PMID: 32319622 PMCID: PMC7057817 DOI: 10.3892/mmr.2020.10990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 01/14/2020] [Indexed: 12/20/2022] Open
Abstract
The current study aimed to evaluate the neuroprotective effect of Ginkgo biloba extract (GbE) on the progression of acute cerebral ischemia-reperfusion injury in diabetic rats, and to determine the molecular mechanism associated with this effect. Streptozotocin (STZ) induced diabetic rats were pretreated with GbE (50, 100 and 200 mg/kg/day; intragastric) for 3 weeks. During this period, body weight changes and fasting blood glucose levels were assessed each week. Following pretreatment, rats were subjected to suture occlusion of the middle cerebral artery for 30 min, which was followed by 24 h of reperfusion. Neurological deficits were subsequently evaluated at 2 and 24 h following reperfusion. Rats were sacrificed after 24 h reperfusion, and infarct volume and S100B content were measured to evaluate the neuroprotective effect of GbE. The results of the present study demonstrated that GbE pretreatment improved neurological scores, and reduced cerebral infarct volume and S100B content. Oxidative stress markers, including glutathione (GSH) and superoxide dismutase (SOD) were increased, and malondialdehyde (MDA) contents were reduced following GbE treatment. The levels of p-Akt, p-mTOR and glutamate transporter 1 (GLT1) were observed to be increased in GbE-pretreated rats. These results indicated that GbE pretreatment may serve a protective role against cerebral ischemia-reperfusion injury in diabetic rats by inhibiting oxidative stress reaction, upregulating the expression of Akt/mTOR and promoting GLT1 expression. In conclusion, the current study revealed the protective role and molecular mechanisms of GbE in diabetic rats with cerebral ischemia-reperfusion injury, and may provide novel insight into the future clinical treatment of this condition.
Collapse
Affiliation(s)
- Miao Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Mei Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Shuling Gu
- Department of Pharmacology, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Zheng Sun
- Department of Pharmacology, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Tengfei Ma
- Department of Pharmacology, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Xing Ma
- Department of Pharmacology, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| |
Collapse
|
25
|
Li J, Liu G, Li L, Yao Z, Huang J. Research progress on the effect of autophagy-lysosomal pathway on tumor drug resistance. Exp Cell Res 2020; 389:111925. [DOI: 10.1016/j.yexcr.2020.111925] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 12/14/2022]
|
26
|
Neuroprotective effect of crocin against rotenone-induced Parkinson's disease in rats: Interplay between PI3K/Akt/mTOR signaling pathway and enhanced expression of miRNA-7 and miRNA-221. Neuropharmacology 2020; 164:107900. [DOI: 10.1016/j.neuropharm.2019.107900] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 10/14/2019] [Accepted: 11/30/2019] [Indexed: 11/23/2022]
|
27
|
Maiese K. Nicotinamide: Oversight of Metabolic Dysfunction Through SIRT1, mTOR, and Clock Genes. Curr Neurovasc Res 2020; 17:765-783. [PMID: 33183203 PMCID: PMC7914159 DOI: 10.2174/1567202617999201111195232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022]
Abstract
Metabolic disorders that include diabetes mellitus present significant challenges for maintaining the welfare of the global population. Metabolic diseases impact all systems of the body and despite current therapies that offer some protection through tight serum glucose control, ultimately such treatments cannot block the progression of disability and death realized with metabolic disorders. As a result, novel therapeutic avenues are critical for further development to address these concerns. An innovative strategy involves the vitamin nicotinamide and the pathways associated with the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), the mechanistic target of rapamycin (mTOR), mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), AMP activated protein kinase (AMPK), and clock genes. Nicotinamide maintains an intimate relationship with these pathways to oversee metabolic disease and improve glucose utilization, limit mitochondrial dysfunction, block oxidative stress, potentially function as antiviral therapy, and foster cellular survival through mechanisms involving autophagy. However, the pathways of nicotinamide, SIRT1, mTOR, AMPK, and clock genes are complex and involve feedback pathways as well as trophic factors such as erythropoietin that require a careful balance to ensure metabolic homeostasis. Future work is warranted to gain additional insight into these vital pathways that can oversee both normal metabolic physiology and metabolic disease.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022
| |
Collapse
|
28
|
Jęśko H, Stępień A, Lukiw WJ, Strosznajder RP. The Cross-Talk Between Sphingolipids and Insulin-Like Growth Factor Signaling: Significance for Aging and Neurodegeneration. Mol Neurobiol 2019; 56:3501-3521. [PMID: 30140974 PMCID: PMC6476865 DOI: 10.1007/s12035-018-1286-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/25/2018] [Indexed: 12/20/2022]
Abstract
Bioactive sphingolipids: sphingosine, sphingosine-1-phosphate (S1P), ceramide, and ceramide-1-phosphate (C1P) are increasingly implicated in cell survival, proliferation, differentiation, and in multiple aspects of stress response in the nervous system. The opposite roles of closely related sphingolipid species in cell survival/death signaling is reflected in the concept of tightly controlled sphingolipid rheostat. Aging has a complex influence on sphingolipid metabolism, disturbing signaling pathways and the properties of lipid membranes. A metabolic signature of stress resistance-associated sphingolipids correlates with longevity in humans. Moreover, accumulating evidence suggests extensive links between sphingolipid signaling and the insulin-like growth factor I (IGF-I)-Akt-mTOR pathway (IIS), which is involved in the modulation of aging process and longevity. IIS integrates a wide array of metabolic signals, cross-talks with p53, nuclear factor κB (NF-κB), or reactive oxygen species (ROS) and influences gene expression to shape the cellular metabolic profile and stress resistance. The multiple connections between sphingolipids and IIS signaling suggest possible engagement of these compounds in the aging process itself, which creates a vulnerable background for the majority of neurodegenerative disorders.
Collapse
Affiliation(s)
- Henryk Jęśko
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Pawińskiego, 5, 02-106, Poland
| | - Adam Stępień
- Central Clinical Hospital of the Ministry of National Defense, Department of Neurology, Military Institute of Medicine, Warsaw, Szaserów, 128, 04-141, Poland
| | - Walter J Lukiw
- LSU Neuroscience Center and Departments of Neurology and Ophthalmology, Louisiana State University School of Medicine, New Orleans, USA
| | - Robert P Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Pawińskiego, 5, 02-106, Poland.
| |
Collapse
|
29
|
Sun G, Qu S, Wang S, Shao Y, Sun J. Taurine attenuates acrylamide-induced axonal and myelinated damage through the Akt/GSK3β-dependent pathway. Int J Immunopathol Pharmacol 2019; 32:2058738418805322. [PMID: 30354842 PMCID: PMC6202743 DOI: 10.1177/2058738418805322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Acrylamide (ACR), formed during the Maillard reaction induced by high temperature
in food processing, is one of the main causes of neurodegenerative diseases.
Taurine, a free intracellular β-amino acid, is characterized by many functions,
including antioxidation, anti-inflammatory, and neuroprotective properties. This
promotes its application in the treatment of neurodegenerative diseases. In this
study, the neuroprotective effects of taurine against ACR-induced neurotoxicity
and the potential underlying mechanisms were explored. Rats were intoxicated
with ACR and injected with taurine in different groups for totally 2 weeks
between January and July 2017. Electron microscopic analysis was used to observe
the changes in tissues of the rats. Meanwhile, the levels of proteins including
p-Akt, p-GSK3β, SIM312, and MBP were detected by Western blot. Furthermore, the
GSK3β phosphorylation in taurine-treated dorsal root ganglion (DRG) with ACR was
examined in the presence of the Akt inhibitor, MK-2206. The analysis of
behavioral performances and electron micrographs indicated that taurine
treatment significantly attenuated the toxic manifestations induced by ACR and
stimulated the growth of axons and the medullary sheath, which was associated
with the activation of the Akt/GSK3β signaling pathway. Mechanistically, it was
found that taurine activated GSK3β, leading to significant recovery of the
damage in ACR-induced sciatic nerves. Furthermore, MK-2206, an inhibitor of Akt,
was applied in DRG cells, suggesting that taurine-induced GSK3β phosphorylation
was Akt dependent. Our findings demonstrated that taurine attenuated ACR-induced
neuropathy in vivo, in an Akt/GSK3β-dependent manner. This confirmed the
treatment with taurine to be a novel strategy against ACR-induced
neurotoxicity.
Collapse
Affiliation(s)
- Guohua Sun
- The First Affiliated Hospital of Dalian
Medical University, Liaoning, China
| | - Shuxian Qu
- Institute of Cancer Stem Cell, Dalian
Medical University, Dalian, Liaoning, China
| | - Siyi Wang
- The First Affiliated Hospital of Dalian
Medical University, Liaoning, China
| | - Ying Shao
- The First Affiliated Hospital of Dalian
Medical University, Liaoning, China
| | - Jingsong Sun
- The First Affiliated Hospital of Dalian
Medical University, Liaoning, China
- Jingsong Sun, The First Affiliated Hospital
of Dalian Medical University, Liaoning 116011, China.
| |
Collapse
|
30
|
Shukla M, Chinchalongporn V, Govitrapong P, Reiter RJ. The role of melatonin in targeting cell signaling pathways in neurodegeneration. Ann N Y Acad Sci 2019; 1443:75-96. [PMID: 30756405 DOI: 10.1111/nyas.14005] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/17/2018] [Accepted: 01/02/2019] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are typified by neuronal loss associated with progressive dysfunction and clinical presentation. Neurodegenerative diseases are characterized by the intra- and extracellular conglomeration of misfolded proteins that occur because of abnormal protein dynamics and genetic manipulations; these trigger processes of cell death in these disorders. The disrupted signaling mechanisms involved are oxidative stress-mediated mitochondrial and calcium signaling deregulation, alterations in immune and inflammatory signaling, disruption of autophagic integrity, proteostasis dysfunction, and anomalies in the insulin, Notch, and Wnt/β-catenin signaling pathways. Herein, we accentuate some of the contemporary translational approaches made in characterizing the underlying mechanisms of neurodegeneration. Melatonin-induced cognitive enhancement and inhibition of oxidative signaling substantiates the efficacy of melatonin in combating neurodegenerative processes. Our review considers in detail the possible roles of melatonin in understanding the synergistic pathogenic mechanisms between aggregated proteins and in regulating, modulating, and preventing the altered signaling mechanisms discovered in cellular and animal models along with clinical evaluations pertaining to neurodegeneration. Furthermore, this review showcases the therapeutic potential of melatonin in preventing and treating neurodegenerative diseases with optimum prognosis.
Collapse
Affiliation(s)
- Mayuri Shukla
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Vorapin Chinchalongporn
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, Thailand.,Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, Thailand.,Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center San Antonio, San Antonio, Texas
| |
Collapse
|
31
|
A11, a novel diaryl acylhydrazone derivative, exerts neuroprotection against ischemic injury in vitro and in vivo. Acta Pharmacol Sin 2019; 40:160-169. [PMID: 29925921 DOI: 10.1038/s41401-018-0028-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/08/2018] [Accepted: 04/09/2018] [Indexed: 12/21/2022] Open
Abstract
There is an urgent need to develop effective therapies for ischemic stroke, but the complicated pathological processes after ischemia make doing so difficult. In the current study, we identified a novel diaryl acylhydrazone derivative, A11, which has multiple neuroprotective properties in ischemic stroke models. First, A11 was demonstrated to induce neuroprotection against ischemic injury in a dose-dependent manner (from 0.3 to 3 μM) in three in vitro experimental ischemic stroke models: oxygen glucose deprivation (OGD), hydrogen peroxide, and glutamate-stimulated neuronal cell injury models. Moreover, A11 was able to potently alleviate three critical pathological changes, apoptosis, oxidative stress, and mitochondrial dysfunction, following ischemic insult in neuronal cells. Further analysis revealed that A11 upregulated the phosphorylation levels of protein kinase B (AKT) and extracellular signal-regulated kinase (ERK) in OGD-exposed neuronal cells, suggesting joint activation of the phosphoinositide 3-kinase (PI3K)/AKT and mitogen-activated protein kinase (MEK)/ERK pathways. In rats with middle cerebral artery occlusion, single-dose administration of A11 (3 mg/kg per day, i.v.) at the onset of reperfusion significantly reduced the infarct volumes and ameliorated neurological deficits. Our study, for the first time, reports the anti-ischemic effect of diaryl acylhydrazone chemical entities, especially A11, which acts on multiple ischemia-associated pathological processes. Our results may provide new clues for the development of an effective therapeutic agent for ischemic stroke.
Collapse
|
32
|
Cheng CY, Kao ST, Lee YC. Ferulic acid ameliorates cerebral infarction by activating Akt/mTOR/4E‑BP1/Bcl‑2 anti‑apoptotic signaling in the penumbral cortex following permanent cerebral ischemia in rats. Mol Med Rep 2018; 19:792-804. [PMID: 30569126 DOI: 10.3892/mmr.2018.9737] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 11/23/2018] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to determine the effects of ferulic acid (FerA) administered immediately following the onset of permanent middle cerebral artery occlusion (MCAo) and then 7 days of ischemia, and also to explore the involvement of protein kinase B (Akt)‑induced signaling in the penumbral cortex. Immediately following the onset of MCAo, FerA was intravenously administered to rats at a dose of 60 mg/kg (FerA‑60 mg), 80 mg/kg (FerA‑80 mg), or 100 mg/kg (FerA‑100 mg). FerA‑80 mg and FerA‑100 mg effectively ameliorated cerebral infarction and neurological deficits 7 days following permanent cerebral ischemia. FerA‑80 mg and FerA‑100 mg significantly upregulated the expression of phospho‑Akt (p‑Akt), phospho‑mammalian target of rapamycin (p‑mTOR), and eukaryotic initiation factor 4E (eIF4E)‑binding protein 1 (4E‑BP1), and the phospho‑4E‑BP1 (p‑4E‑BP1)/4E‑BP1 and mitochondrial Bcl‑2/Bax ratios, and markedly downregulated the levels of cytochrome c‑, cleaved caspase‑3‑, and terminal deoxynucleotidyl transferase‑mediated dUTP‑biotin nick‑end labeling‑immunoreactive cells in the penumbral cortex at 7 days post‑ischemia. LY294002, a selective inhibitor of phosphoinositide 3‑kinase/Akt signaling, was administered 30 min prior to ischemia, which abrogated the upregulating effects of FerA‑100 mg on the expression of p‑Akt, p‑mTOR, 4E‑BP1, p‑4E‑BP1 and eIF4E, the mitochondrial Bcl‑2/Bax ratio and the ameliorating effect of FerA‑100 mg on cerebral infarction. FerA administered at doses of 80 and 100 mg/kg exerted beneficial effects against cerebral ischemia by activating Akt‑induced signaling. The effects of FerA at doses of 80 and 100 mg/kg on mitochondrial B‑cell lymphoma-2 (Bcl‑2)‑associated X protein‑related apoptosis were attributed to the activation of Akt/mTOR/4E‑BP1/Bcl‑2 anti‑apoptotic signaling, and eventually contributed to suppression of the cytochrome c/caspase‑3 activation pathway in the penumbral cortex 7 days following permanent cerebral ischemia.
Collapse
Affiliation(s)
- Chin-Yi Cheng
- School of Post‑Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Yu-Chen Lee
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan, R.O.C
| |
Collapse
|
33
|
Rad SK, Arya A, Karimian H, Madhavan P, Rizwan F, Koshy S, Prabhu G. Mechanism involved in insulin resistance via accumulation of β-amyloid and neurofibrillary tangles: link between type 2 diabetes and Alzheimer's disease. Drug Des Devel Ther 2018; 12:3999-4021. [PMID: 30538427 PMCID: PMC6255119 DOI: 10.2147/dddt.s173970] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The pathophysiological link between type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) has been suggested in several reports. Few findings suggest that T2DM has strong link in the development process of AD, and the complete mechanism is yet to be revealed. Formation of amyloid plaques (APs) and neurofibrillary tangles (NFTs) are two central hallmarks in the AD. APs are the dense composites of β-amyloid protein (Aβ) which accumulates around the nerve cells. Moreover, NFTs are the twisted fibers containing hyperphosphorylated tau proteins present in certain residues of Aβ that build up inside the brain cells. Certain factors contribute to the aetiogenesis of AD by regulating insulin signaling pathway in the brain and accelerating the formation of neurotoxic Aβ and NFTs via various mechanisms, including GSK3β, JNK, CamKII, CDK5, CK1, MARK4, PLK2, Syk, DYRK1A, PPP, and P70S6K. Progression to AD could be influenced by insulin signaling pathway that is affected due to T2DM. Interestingly, NFTs and APs lead to the impairment of several crucial cascades, such as synaptogenesis, neurotrophy, and apoptosis, which are regulated by insulin, cholesterol, and glucose metabolism. The investigation of the molecular cascades through insulin functions in brain contributes to probe and perceive progressions of diabetes to AD. This review elaborates the molecular insights that would help to further understand the potential mechanisms linking T2DM and AD.
Collapse
Affiliation(s)
- Sima Kianpour Rad
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Aditya Arya
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia,
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia,
- Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), Bukit Gambir, Gelugor, Pulau Pinang, Malaysia,
| | - Hamed Karimian
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia,
| | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Farzana Rizwan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Shajan Koshy
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Girish Prabhu
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| |
Collapse
|
34
|
Guo T, Cao G, Li Y, Zhang Z, Nör J, Clarkson B, Liu J. Signals in Stem Cell Differentiation on Fluorapatite-Modified Scaffolds. J Dent Res 2018; 97:1331-1338. [PMID: 29995454 PMCID: PMC6728582 DOI: 10.1177/0022034518788037] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Previously, we reported that the fluorapatite (FA)-modified polycaprolactone (PCL) nanofiber could be an odontogenic/osteogenic inductive tissue-engineering scaffold by inducing stem cell differentiation and mineralization. The present study aimed to explore which of the signal pathways affected this differentiation and mineralization process. The Human Signal Transduction PathwayFinder RT2 Profiler PCR Array was used to analyze the involvement of potential signal transduction pathways during human dental pulp stem cell (DPSCs) osteogenic differentiation induced by FA-modified PCL nanofiber scaffolds. Based on the results, perturbation studies of the signaling pathways hedgehog, insulin, and Wnt were performed. Moreover, the autophagy process was studied, as indicated by the expression of the microtubule-associated protein 1 light chain 3A/B-II (LC3-II) and the cell osteogenic phenotypic changes. In a comparison of the cells grown on PCL + FA scaffolds and those on PCL-only scaffolds, the transcript expression of BMP2, BMP4, FOXA2, PTCH1, WNT1, and WNT2 (PCR array-labeled signal proteins of the hedgehog pathway); CEBPB, FASN, and HK2 (PCR array-labeled signal proteins of the insulin pathway); and CCND1, JUN, MYC, TCF7, and WISP1 (PCR array-labeled signal proteins of the Wnt pathway) doubled at day 14 when obvious cell osteogenic differentiation occurred. Phenotypically, in all the perturbation groups at day 14, ALP activity, OPN, and autophagy marker LC3-II expression were coincidently decreased. Consistently, no positive alizarin red staining or von Kossa staining was observed in the specimens from these perturbation groups at day 28. The results showed that when obvious cell differentiation occurred at day 14 on PCL + FA control groups, the inhibition of the hedgehog, insulin, and Wnt pathways significantly decreased DPSC osteogenic differentiation and mineralization. The osteogenic differentiation of DPSCs grown on FA-modified PCL scaffolds appeared to be positively modulated by the hedgehog, insulin, and Wnt signal pathways, which were coordinated with and/or mediated by the cell autophagy process.
Collapse
Affiliation(s)
- T. Guo
- Nanjing Stomatological Hospital, Medical
School of Nanjing University, Nanjing, China
- Department of Cariology, Restorative
Sciences and Endodontics, Dental School, University of Michigan, Ann Arbor, MI,
USA
- Department of Stomatology, Nanjing
Jinling Hospital, Nanjing, China
| | - G. Cao
- Department of Stomatology, Nanjing
Jinling Hospital, Nanjing, China
| | - Y. Li
- Department of Cariology, Restorative
Sciences and Endodontics, Dental School, University of Michigan, Ann Arbor, MI,
USA
- Department of Oral and Maxillofacial
Surgery, State Key Laboratory of Military Stomatology, School of Stomatology, The
Fourth Military Medical University, Xian, China
| | - Z. Zhang
- Department of Cariology, Restorative
Sciences and Endodontics, Dental School, University of Michigan, Ann Arbor, MI,
USA
| | - J.E. Nör
- Department of Cariology, Restorative
Sciences and Endodontics, Dental School, University of Michigan, Ann Arbor, MI,
USA
| | - B.H. Clarkson
- Department of Cariology, Restorative
Sciences and Endodontics, Dental School, University of Michigan, Ann Arbor, MI,
USA
| | - J. Liu
- Department of Cariology, Restorative
Sciences and Endodontics, Dental School, University of Michigan, Ann Arbor, MI,
USA
| |
Collapse
|
35
|
Abstract
Olfactory receptors are expressed by different cell types throughout the body and regulate physiological cell functions beyond olfaction. In particular, the olfactory receptor OR2AT4 has been shown to stimulate keratinocyte proliferation in the skin. Here, we show that the epithelium of human hair follicles, particularly the outer root sheath, expresses OR2AT4, and that specific stimulation of OR2AT4 by a synthetic sandalwood odorant (Sandalore®) prolongs human hair growth ex vivo by decreasing apoptosis and increasing production of the anagen-prolonging growth factor IGF-1. In contrast, co-administration of the specific OR2AT4 antagonist Phenirat® and silencing of OR2AT4 inhibit hair growth. Together, our study identifies that human hair follicles can engage in olfactory receptor-dependent chemosensation and require OR2AT4-mediated signaling to sustain their growth, suggesting that olfactory receptors may serve as a target in hair loss therapy. Increasing evidence suggest that olfactory receptors can carry additional functions besides olfaction. Here, Chéret et al. show that stimulation of the olfactory receptor ORT2A4 by the odorant Sandalore® stimulates growth of human scalp hair follicles ex vivo, suggesting the use of ORT2A4-targeting odorants as hair growth-promoting agents.
Collapse
|
36
|
Fang Y, Jiang D, Wang Y, Wang Q, Lv D, Liu J, Liu C. Neuroprotection of rhGLP‐1 in diabetic rats with cerebral ischemia/reperfusion injury via regulation of oxidative stress, EAAT2, and apoptosis. Drug Dev Res 2018; 79:249-259. [PMID: 30222190 DOI: 10.1002/ddr.21439] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/05/2018] [Accepted: 06/07/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Yi Fang
- Department of Pharmacy Peking University People's Hospital Beijing People's Republic of China
| | - Daoli Jiang
- Department of Pharmacy The Affiliated Hospital of Xuzhou Medical University Xuzhou People's Republic of China
| | - Yitong Wang
- Department of Pharmacy Peking University People's Hospital Beijing People's Republic of China
- Department of Pharmacy Administration and Clinical Pharmacy Peking University Health Science Center Beijing People's Republic of China
| | - Qian Wang
- Department of Pharmacy Peking University People's Hospital Beijing People's Republic of China
| | - Dongmei Lv
- Department of Pharmacy The Affiliated Hospital of Xuzhou Medical University Xuzhou People's Republic of China
| | - Jichao Liu
- Animal Experimental Center Peking University People's Hospital Beijing People's Republic of China
| | - Chang Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy Xuzhou Medical University Xuzhou People's Republic of China
| |
Collapse
|
37
|
Yang M, Teng S, Ma C, Yu Y, Wang P, Yi C. Ascorbic acid inhibits senescence in mesenchymal stem cells through ROS and AKT/mTOR signaling. Cytotechnology 2018; 70:1301-1313. [PMID: 29777434 DOI: 10.1007/s10616-018-0220-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cell (MSC) aging seriously affects its function in stem cell transplantation for treatment. Extensive studies have focused on how to inhibit senescence in MSCs. However, the mechanism of senescence in MSC was not clear. In this study, we used D-galactose to induce MSC aging. Then we found that the number of aging cells was increased compared with untreated MSCs. We discovered that ascorbic acid could inhibit the production of reactive oxygen species (ROS) and activation of AKT/mTOR signaling in MSCs caused by D-galactose. Especially, when treated together with a ROS scavenger or AKT inhibitor, the senescent cells were obviously decreased in D-galactose-induced MSCs. Taken together, we identify that ascorbic acid owns the potential to inhibit the senescence of MSCs through ROS and Akt/mTOR signaling. Together, our data supports that ascorbic acid can be used to prevent MSCs from senescence, which can enhance the efficiency of stem cell transplantation in the clinic.
Collapse
Affiliation(s)
- Mengkai Yang
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Songsong Teng
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Chunhui Ma
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yinxian Yu
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Peilin Wang
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Chengqing Yi
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
38
|
Maiese K. Moving to the Rhythm with Clock (Circadian) Genes, Autophagy, mTOR, and SIRT1 in Degenerative Disease and Cancer. Curr Neurovasc Res 2018; 14:299-304. [PMID: 28721811 DOI: 10.2174/1567202614666170718092010] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/22/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND The mammalian circadian clock and its associated clock genes are increasingly been recognized as critical components for a number of physiological and disease processes that extend beyond hormone release, thermal regulation, and sleep-wake cycles. New evidence suggests that clinical behavior disruptions that involve prolonged shift work and even space travel may negatively impact circadian rhythm and lead to multi-system disease. METHODS In light of the significant role circadian rhythm can hold over the body's normal physiology as well as disease processes, we examined and discussed the impact circadian rhythm and clock genes hold over lifespan, neurodegenerative disorders, and tumorigenesis. RESULTS In experimental models, lifespan is significantly reduced with the introduction of arrhythmic mutants and leads to an increase in oxidative stress exposure. Interestingly, patients with Alzheimer's disease and Parkinson's disease may suffer disease onset or progression as a result of alterations in the DNA methylation of clock genes as well as prolonged pharmacological treatment for these disorders that may lead to impairment of circadian rhythm function. Tumorigenesis also can occur with the loss of a maintained circadian rhythm and lead to an increased risk for nasopharyngeal carcinoma, breast cancer, and metastatic colorectal cancer. Interestingly, the circadian clock system relies upon the regulation of the critical pathways of autophagy, the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), and silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) as well as proliferative mechanisms that involve the wingless pathway of Wnt/β-catenin pathway to foster cell survival during injury and block tumor cell growth. CONCLUSION Future targeting of the pathways of autophagy, mTOR, SIRT1, and Wnt that control mammalian circadian rhythm may hold the key for the development of novel and effective therapies against aging- related disorders, neurodegenerative disease, and tumorigenesis.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, NY. United States
| |
Collapse
|
39
|
The mechanistic target of rapamycin (mTOR) and the silent mating-type information regulation 2 homolog 1 (SIRT1): oversight for neurodegenerative disorders. Biochem Soc Trans 2018. [PMID: 29523769 DOI: 10.1042/bst20170121] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As a result of the advancing age of the global population and the progressive increase in lifespan, neurodegenerative disorders continue to increase in incidence throughout the world. New strategies for neurodegenerative disorders involve the novel pathways of the mechanistic target of rapamycin (mTOR) and the silent mating-type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) that can modulate pathways of apoptosis and autophagy. The pathways of mTOR and SIRT1 are closely integrated. mTOR forms the complexes mTOR Complex 1 and mTOR Complex 2 and can impact multiple neurodegenerative disorders that include Alzheimer's disease, Huntington's disease, and Parkinson's disease. SIRT1 can control stem cell proliferation, block neuronal injury through limiting programmed cell death, drive vascular cell survival, and control clinical disorders that include dementia and retinopathy. It is important to recognize that oversight of programmed cell death by mTOR and SIRT1 requires a fine degree of precision to prevent the progression of neurodegenerative disorders. Additional investigations and insights into these pathways should offer effective and safe treatments for neurodegenerative disorders.
Collapse
|
40
|
Maiese K. Sirtuins: Developing Innovative Treatments for Aged-Related Memory Loss and Alzheimer's Disease. Curr Neurovasc Res 2018; 15:367-371. [PMID: 30484407 PMCID: PMC6538488 DOI: 10.2174/1567202616666181128120003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/14/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023]
Abstract
The world's population continues to age at a rapid pace. By the year 2050, individuals over the age of 65 will account for sixteen percent of the world's population and life expectancy will increase well over eighty years of age. Accompanied by the aging of the global population is a significant rise in Non-Communicable Diseases (NCDs). Neurodegenerative disorders will form a significant component for NCDs. Currently, dementia is the 7th leading cause of death and can be the result of multiple causes that include diabetes mellitus, vascular disease, and Alzheimer's Disease (AD). AD may represent at least sixty percent of these cases. Current treatment for these disorders is extremely limited to provide only some symptomatic relief at present. Sirtuins and in particular, the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), represent innovative strategies for the treatment of cognitive loss. New work has revealed that SIRT1 provides protection against memory loss through mechanisms that involve oxidative stress, Aβ toxicity, neurofibrillary degeneration, vascular injury, mitochondrial dysfunction, and neuronal loss. In addition, SIRT1 relies upon other avenues that can include trophic factors, such as erythropoietin, and signaling pathways, such as Wnt1 inducible signaling pathway protein 1 (WISP1/CCN4). Yet, SIRT1 can have detrimental effects as well that involve tumorigenesis and blockade of stem cell differentiation and maturation that can limit reparative processes for cognitive loss. Further investigations with sirtuins and SIRT1 should be able to capitalize upon these novel targets for dementia and cognitive loss.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101
| |
Collapse
|
41
|
Maiese K. Novel Treatment Strategies for the Nervous System: Circadian Clock Genes, Non-coding RNAs, and Forkhead Transcription Factors. Curr Neurovasc Res 2018; 15:81-91. [PMID: 29557749 PMCID: PMC6021214 DOI: 10.2174/1567202615666180319151244] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/23/2018] [Accepted: 02/07/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND With the global increase in lifespan expectancy, neurodegenerative disorders continue to affect an ever-increasing number of individuals throughout the world. New treatment strategies for neurodegenerative diseases are desperately required given the lack of current treatment modalities. METHODS Here, we examine novel strategies for neurodegenerative disorders that include circadian clock genes, non-coding Ribonucleic Acids (RNAs), and the mammalian forkhead transcription factors of the O class (FoxOs). RESULTS Circadian clock genes, non-coding RNAs, and FoxOs offer exciting prospects to potentially limit or remove the significant disability and death associated with neurodegenerative disorders. Each of these pathways has an intimate relationship with the programmed death pathways of autophagy and apoptosis and share a common link to the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) and the mechanistic target of rapamycin (mTOR). Circadian clock genes are necessary to modulate autophagy, limit cognitive loss, and prevent neuronal injury. Non-coding RNAs can control neuronal stem cell development and neuronal differentiation and offer protection against vascular disease such as atherosclerosis. FoxOs provide exciting prospects to block neuronal apoptotic death and to activate pathways of autophagy to remove toxic accumulations in neurons that can lead to neurodegenerative disorders. CONCLUSION Continued work with circadian clock genes, non-coding RNAs, and FoxOs can offer new prospects and hope for the development of vital strategies for the treatment of neurodegenerative diseases. These innovative investigative avenues have the potential to significantly limit disability and death from these devastating disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101
| |
Collapse
|
42
|
Ginsenoside Rg1 Decreases Oxidative Stress and Down-Regulates Akt/mTOR Signalling to Attenuate Cognitive Impairment in Mice and Senescence of Neural Stem Cells Induced by D-Galactose. Neurochem Res 2017; 43:430-440. [PMID: 29147958 DOI: 10.1007/s11064-017-2438-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 02/03/2023]
Abstract
Adult hippocampal neurogenesis plays a pivotal role in learning and memory. The suppression of hippocampal neurogenesis induced by an increase of oxidative stress is closely related to cognitive impairment. Neural stem cells which persist in the adult vertebrate brain keep up the production of neurons over the lifespan. The balance between pro-oxidants and anti-oxidants is important for function and surviving of neural stem cells. Ginsenoside Rg1 is one of the most active components of Panax ginseng, and many studies suggest that ginsenosides have antioxidant properties. This research explored the effects and underlying mechanisms of ginsenoside Rg1 on protecting neural stem cells (NSCs) from oxidative stress. The sub-acute ageing of C57BL/6 mice was induced by subcutaneous injection of D-gal (120 mg kg-1 day-1) for 42 day. On the 14th day of D-gal injection, the mice were treated with ginsenoside Rg1 (20 mg kg-1 day-1, intraperitoneally) or normal saline for 28 days. The study monitored the effects of Rg1 on proliferation, senescence-associated and oxidative stress biomarkers, and Akt/mTOR signalling pathway in NSCs. Compared with the D-gal group, Rg1 improved cognitive impairment induced by D-galactose in mice by attenuating senescence of neural stem cells. Rg1 also decreased the level of oxidative stress, with increased the activity of superoxide dismutase and glutathione peroxidase in vivo and in vitro. Rg1 furthermore reduced the phosphorylation levels of protein kinase B (Akt) and the mechanistic target of rapamycin (mTOR) and down-regulated the levels of downstream p53, p16, p21 and Rb in D-gal treated NSCs. The results suggested that the protective effect of ginsenoside Rg1 on attenuating cognitive impairment in mice and senescence of NSCs induced by D-gal might be related to the reduction of oxidative stress and the down-regulation of Akt/mTOR signaling pathway.
Collapse
|
43
|
Na EJ, Nam HY, Park J, Chung MA, Woo HA, Kim HJ. PI3K-mTOR-S6K Signaling Mediates Neuronal Viability via Collapsin Response Mediator Protein-2 Expression. Front Mol Neurosci 2017; 10:288. [PMID: 28966575 PMCID: PMC5605571 DOI: 10.3389/fnmol.2017.00288] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/25/2017] [Indexed: 01/22/2023] Open
Abstract
Collapsin response mediator protein (CRMP)-2 and the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway are associated with common physiological functions such as neuronal polarity, axonal outgrowth and synaptic strength, as well as various brain disorders including epilepsy. But, their regulatory and functional links are unclear. Alterations in CRMP-2 expression that lead to its functional changes are implicated in brain disorders such as epilepsy. Here, we investigate whether changes in CRMP-2 expression, possibly regulated by mTOR-related signaling, correlates with neuronal growth and viability. Inhibition of mTOR and/or phosphoinositol-3-kinase (PI3K) led to deceased p-S6K, and p-S6 signals also reduced CRMP-2 expression. These changes corresponded to inhibition of neuronal viability and proliferation in cultured hippocampal HT-22 cells under both basal serum-free and serum- or insulin-induced mTOR pathway-activated conditions. CRMP-2 expression tended to be increased by mTOR activation, indicated by an increase in p-S6/S6 level, in pentylentetrazole (PTZ)-induced epileptic rat hippocampal tissues was also significantly reduced by mTOR inhibition. Knockdown of CRMP-2 by si-RNA reduced the neuronal viability without changes in mTOR signaling, and overexpression of CRMP-2 recovered the glutamate-induced neurotoxicity and decrease of mTOR signaling in HT-22 cells. In conclusion, CRMP-2 protein expression controlled by the PI3K-mTOR-S6K signaling axis exerts its important functional roles in neuronal growth and survival.
Collapse
Affiliation(s)
- Eun J Na
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| | - Hye Yeon Nam
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| | - Jiyoung Park
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| | - Myung Ah Chung
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| | - Hyun Ae Woo
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| | - Hwa-Jung Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| |
Collapse
|
44
|
Cognitive Function of Artemisia argyi H. Fermented by Monascus purpureus under TMT-Induced Learning and Memory Deficits in ICR Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:5809370. [PMID: 29081819 PMCID: PMC5610803 DOI: 10.1155/2017/5809370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 07/05/2017] [Accepted: 07/17/2017] [Indexed: 11/23/2022]
Abstract
The cognitive effect of Artemisia argyi H. under liquid-state fermentation by Monascus purpureus (AAFM), which has cellular antioxidant activity and neuronal cell viability, on trimethyltin- (TMT-) induced learning and memory impairment in Institute of Cancer Research (ICR) mice was confirmed. Tests were conducted to determine the neuroprotective effects against H2O2-induced oxidative stress, and the results showed that AAFM has protective effects through the repression of mitochondrial injury and cellular membrane damage against H2O2-induced neurotoxicity. In animal experiments, such as the Y-maze, passive avoidance, and Morris water maze tests, AAFM also showed excellent ameliorating effects on TMT-induced cognitive dysfunction. After behavioral tests, brain tissues were extracted to assess damage to brain tissue. According to the experimental results, AAFM improved the cholinergic system by upregulating acetylcholine (ACh) contents and inhibiting acetylcholinesterase (AChE) activity. AAFM effectively improved the decline of the superoxide dismutase (SOD) level and the increase of the oxidized glutathione (GSH) ratio and lipid peroxidation (malondialdehyde (MDA) production) caused by TMT-induced oxidative stress. The occurrence of mitochondrial dysfunction and apoptosis was also decreased compared with the TMT group. Finally, quinic acid derivatives were identified as the major phenolic compounds in AAFM using ultra-performance liquid chromatography quadrupole-time-of-flight (UPLC-Q-TOF) MS analysis.
Collapse
|
45
|
Lee KH, Lee SJ, Lee HJ, Choi GE, Jung YH, Kim DI, Gabr AA, Ryu JM, Han HJ. Amyloid β1-42 (Aβ1-42) Induces the CDK2-Mediated Phosphorylation of Tau through the Activation of the mTORC1 Signaling Pathway While Promoting Neuronal Cell Death. Front Mol Neurosci 2017; 10:229. [PMID: 28790888 PMCID: PMC5522873 DOI: 10.3389/fnmol.2017.00229] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/05/2017] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, characterized by cognitive impairment and memory loss. Amyloid β1-42 (Aβ) and hyper-phosphorylation of microtubule-associated protein tau have been considered as major histological features in AD. However, the mechanism of how Aβ induces the hyper-phosphorylation of tau remains to be clarified. In the present study, we investigated the underlying cellular mechanisms of Aβ with regard to the cell cycle regulatory protein-mediated phosphorylation of tau in promoting neuronal cell death. The oligomer Aβ (5 μM) significantly increased the level of caspase 3 cleavage and has the ability to induce cytotoxicity in human neuroblastoma SK-N-MC cells. Aβ induced the degree of extracellular calcium influx via the L-type channel to facilitate the production of reactive oxygen species (ROS). Aβ signaling through ROS production is uniquely mediated by the activation of PI3K/Akt, which is in turn required for mammalian target of rapamycin complex 1 (mTORC1) phosphorylation. mTORC1 activated by Aβ further increased the phosphorylation of eukaryotic translation initiation factor 4E (eIF4E), a binding protein (4E-BP1) and p70S6K1 to stimulate the HIF1α synthesis responsible for the induction of cyclinD1/cyclin-dependent kinase 4 (CDK4) and cyclinE/CDK2, whereas it significantly attenuated the activation of autophagy. Aβ distinctively induced the CDK2-mediated phosphorylation of tau, which is responsible for microtubule destabilization in promoting neuronal apoptosis. In mouse hippocampal primary neurons, the apoptotic cell death induced by Aβ is highly susceptible to the mTORC1 signaling pathway. These results demonstrate that Aβ efficiently stimulates the mTORC1 signaling pathway to facilitate HIF1α synthesis and autophagy inhibition to promote the expression of cell cycle regulatory proteins, during which CDK2 uniquely stimulates tau phosphorylation for microtubule destabilization-mediated neuronal apoptosis.
Collapse
Affiliation(s)
- Ki Hoon Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| | - Sei-Jung Lee
- Department of Pharmaceutical Engineering, Daegu Haany UniversityGyeongsan, South Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| | - Dah Ihm Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| | - Amr Ahmed Gabr
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea.,Department of Physiology, Faculty of Veterinary Medicine, Cairo UniversityGiza, Egypt
| | - Jung Min Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National UniversityGwangju, South Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| |
Collapse
|
46
|
Finelli MJ, Oliver PL. TLDc proteins: new players in the oxidative stress response and neurological disease. Mamm Genome 2017; 28:395-406. [PMID: 28707022 PMCID: PMC5614904 DOI: 10.1007/s00335-017-9706-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/03/2017] [Indexed: 12/14/2022]
Abstract
Oxidative stress (OS) arises from an imbalance in the cellular redox state, which can lead to intracellular damage and ultimately cell death. OS occurs as a result of normal ageing, but it is also implicated as a common etiological factor in neurological disease; thus identifying novel proteins that modulate the OS response may facilitate the design of new therapeutic approaches applicable to many disorders. In this review, we describe the recent progress that has been made using a range of genetic approaches to understand a family of proteins that share the highly conserved TLDc domain. We highlight their shared ability to prevent OS-related cell death and their unique functional characteristics, as well as discussing their potential application as new neuroprotective factors. Furthermore, with an increasing number of pathogenic mutations leading to epilepsy and hearing loss being discovered in the TLDc protein TBC1D24, understanding the function of this family has important implications for a range of inherited neurological diseases.
Collapse
Affiliation(s)
- Mattéa J Finelli
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Peter L Oliver
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK.
| |
Collapse
|
47
|
Zhu R, Chen YX, Ke QF, Gao YS, Guo YP. SC79-loaded ZSM-5/chitosan porous scaffolds with enhanced stem cell osteogenic differentiation and bone regeneration. J Mater Chem B 2017; 5:5009-5018. [PMID: 32264018 DOI: 10.1039/c7tb00897j] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
For effectively treating bone defects, the design of novel therapeutic scaffolds is an important strategy for enhancing stem cell osteogenic differentiation and new bone formation. Herein, we, for the first time, fabricated SC79-loaded ZSM-5/chitosan (ZSM-5/CS/SC79) porous scaffolds via the freeze-drying synthesis of ZSM-5/CS porous scaffolds followed by loading SC79 drug molecules. The ZSM-5/CS scaffolds possessed a three-dimensional (3D) interconnected porous structure, and the nanostructured ZSM-5 ellipsoids were uniformly dispersed on the CS films. The ZSM-5/CS/SC79 scaffolds had appropriate drug loading-release properties due to the hierarchically porous structures of ZSM-5 zeolites and the hydrogen bonding between the CS and SC97. In vitro cell tests demonstrated that both the ZSM-5/CS and ZSM-5/CS/SC79 scaffolds could promote the adhesion, spreading and proliferation of human bone mesenchymal stem cells (hBMSCs). Interestingly, the SC97 released from the scaffolds not only promoted the proliferation of hBMSCs, but also enhanced the osteogenic differentiation. As compared with the ZSM-5/CS control group, the ZSM-5/CS/SC79 scaffolds promoted the ALP activity of hBMSCs, improved the mRNA relative expression levels of osteocalcin (OCN), bone morphogenetic protein-2 (BMP-2) and alkaline phosphatise (ALP), and increased the protein level of β-catenin. The enhanced proliferation and osteogenic differentiation of hBMSCs contributed to the upregulation of Akt kinase by an activated Wnt/β-catenin signaling pathway. Moreover, in vivo animal tests indicated that SC79 released from the ZSM-5/CS/SC79 scaffolds promoted the new bone regeneration without systemic side effects in cranial defects. Therefore, ZSM-5/CS/SC79 scaffolds as novel and promising therapeutic scaffolds have promising applications in defined local bone regeneration.
Collapse
Affiliation(s)
- Rong Zhu
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China.
| | | | | | | | | |
Collapse
|
48
|
Lumkwana D, du Toit A, Kinnear C, Loos B. Autophagic flux control in neurodegeneration: Progress and precision targeting—Where do we stand? Prog Neurobiol 2017; 153:64-85. [DOI: 10.1016/j.pneurobio.2017.03.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 02/09/2023]
|
49
|
Can Co-Activation of Nrf2 and Neurotrophic Signaling Pathway Slow Alzheimer's Disease? Int J Mol Sci 2017; 18:ijms18061168. [PMID: 28561773 PMCID: PMC5485992 DOI: 10.3390/ijms18061168] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/22/2017] [Accepted: 05/27/2017] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is a multifaceted disease that is hard to treat by single-modal treatment. AD starts with amyloid peptides, mitochondrial dysfunction, and oxidative stress and later is accompanied with chronic endoplasmic reticulum (ER) stress and autophagy dysfunction, resulting in more complicated pathogenesis. Currently, few treatments can modify the complicated pathogenic progress of AD. Compared to the treatment with exogenous antioxidants, the activation of global antioxidant defense system via Nrf2 looks more promising in attenuating oxidative stress in AD brains. Accompanying the activation of the Nrf2-mediated antioxidant defense system that reduce the AD-causative factor, oxidative stress, it is also necessary to activate the neurotrophic signaling pathway that replaces damaged organelles and molecules with new ones. Thus, the dual actions to activate both the Nrf2 antioxidant system and neurotrophic signaling pathway are expected to provide a better strategy to modify AD pathogenesis. Here, we review the current understanding of AD pathogenesis and neuronal defense systems and discuss a possible way to co-activate the Nrf2 antioxidant system and neurotrophic signaling pathway with the hope of helping to find a better strategy to slow AD.
Collapse
|
50
|
Kang JY, Park SK, Guo TJ, Ha JS, Lee DS, Kim JM, Lee U, Kim DO, Heo HJ. Reversal of Trimethyltin-Induced Learning and Memory Deficits by 3,5-Dicaffeoylquinic Acid. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6981595. [PMID: 28105250 PMCID: PMC5221408 DOI: 10.1155/2016/6981595] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/21/2016] [Indexed: 11/30/2022]
Abstract
The antiamnesic effect of 3,5-dicaffeoylquinic acid (3,5-diCQA) as the main phenolic compound in Artemisia argyi H. extract on cognitive dysfunction induced by trimethyltin (TMT) (7.1 μg/kg of body weight; intraperitoneal injection) was investigated in order to assess its ameliorating function in mice. In several behavioral tests, namely, the Y-maze, passive avoidance, and Morris water maze (MWM) test, 3,5-diCQA significantly ameliorated learning and memory deficits. After the behavioral tests, brain tissues from the mice were analyzed to characterize the basis of the neuroprotective effect. Acetylcholine (ACh) levels increased, whereas the activity of acetylcholinesterase (AChE) decreased upon administration of 3,5-diCQA. In addition, 3,5-diCQA effectively protected against an increase in malondialdehyde (MDA) content, an increase in the oxidized glutathione (GSH) ratio, and a decline of total superoxide dismutase (SOD) level. 3,5-diCQA may prevent neuronal apoptosis through the protection of mitochondrial activities and the repression of apoptotic signaling molecules such as p-Akt, BAX, and p-tau (Ser 404).
Collapse
Affiliation(s)
- Jin Yong Kang
- Division of Applied Life Science (BK21 Plus), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Seon Kyeong Park
- Division of Applied Life Science (BK21 Plus), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Tian Jiao Guo
- Division of Applied Life Science (BK21 Plus), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jeong Su Ha
- Division of Applied Life Science (BK21 Plus), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Du Sang Lee
- Division of Applied Life Science (BK21 Plus), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jong Min Kim
- Division of Applied Life Science (BK21 Plus), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Uk Lee
- Division of Special Purpose Tree, National Institute of Forest Science, Suwon 16631, Republic of Korea
| | - Dae Ok Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Ho Jin Heo
- Division of Applied Life Science (BK21 Plus), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|