1
|
Cheng M, Dou Y. Nanomaterial-based approaches to neurotoxin neutralization in neurodegenerative diseases. Nanomedicine (Lond) 2025:1-13. [PMID: 40181662 DOI: 10.1080/17435889.2025.2487409] [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: 01/27/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025] Open
Abstract
Neurodegenerative diseases (NDs) are intricately linked to the accumulation of various neurotoxins, mainly including toxic proteins, inflammatory mediators, excess metal ions, and viral pathogens. Biological neutralization strategies that use agents to competitively bind harmful substances and thus inhibit their pathogenic activity hold promise for direct removal of neurotoxins but face many limitations and challenges in NDs. Nanomaterials provide a potential solution for neurotoxin neutralization in NDs due to their unique physicochemical and biological properties. This review summarizes recent advancements in nanomaterial-based approaches to neurotoxin neutralization in NDs, highlighting the diverse design principles and mechanisms of action. We also discuss the critical role of targeted delivery to optimize neutralization efficiency and the advantages of combining different neutralization mechanisms or introducing other therapeutic components to exert the synergistic effects. Furthermore, we reveal current limitations and future research directions aimed at paving the way for nanomedicine development based on neurotoxin neutralization for the treatment of NDs.
Collapse
Affiliation(s)
- Meng Cheng
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Yan Dou
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, P. R. China
| |
Collapse
|
2
|
Liu TT, Yang X, Lei HP, Hu YT, Wu LN, Wei AH, Ji XH, Liu J, Jin H, Shi JS, Zhou SY, Jin F. Gastrodin alleviates Aβ 25-35-induced glycolytic dysfunction via activating PI3K/AKT/BACH1 signaling in Alzheimer's disease models. Exp Neurol 2025; 389:115225. [PMID: 40127855 DOI: 10.1016/j.expneurol.2025.115225] [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: 11/30/2024] [Revised: 02/22/2025] [Accepted: 03/16/2025] [Indexed: 03/26/2025]
Abstract
Cerebral glycolytic alteration has been identified as an important contributor to the pathological progress of Alzheimer's disease (AD). Research has shown that gastrodin (GAS) possesses neuroprotection in various experimental models of AD, but its specific mechanism remains unclear. In this study, we determined whether GAS exerted neuroprotective effects on AD models through regulating PI3K/AKT/BACH1 signaling axis. Eight-week-old C57BL/6 J male mice were intracerebroventricularly injected with Aβ25-35, to establish an AD model, followed by the administration of GAS (30, 60 mg·kg-1·d-1, i.g.) for 21 days. Treatment of GAS markedly alleviated the downregulation of p-PI3K Tyr199/458, p-AKT Ser473, BACH1 and HK1 in the hippocampus of the Aβ25-35-induced AD mice. To further explore the mechanism of GAS-mediated neuroprotection, an in vitro AD cellular model was established by challenging HT22 cells with Aβ25-35. In the Aβ25-35 induced cells, the expression of BACH1, p-PI3K Tyr199/458 and p-AKT Ser473 was reduced, the mRNA and protein levels of HK1 were decreased, and the levels of pyruvate and ATP were reduced. After treatment of GAS, the decline of these indicators was reversed. In addition, overexpression of BACH1 by lentivirus transfection significantly upregulated the mRNA and protein levels of HK1, thereby enhancing glycolytic function and protecting HT22 cells from Aβ25-35-induced injury. The results of chromatin immunoprecipitation assay-real-time quantitative PCR revealed that BACH1 directly bound to the HK1 promoter region. Collectively, these findings suggest that GAS can play a protective role in Aβ25-35-induced experimental AD models by increasing HK1 expression and ameliorating glycolytic dysfunction through activation of the PI3K/AKT/BACH1 signaling axis.
Collapse
Affiliation(s)
- Ting-Ting Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China; Department of Hospital Pharmacy, People's Hospital of Changshan County, Quzhou, Zhejiang Province, China
| | - Xue Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Hui-Ping Lei
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Yue-Ting Hu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Ling-Nan Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Ai-Hong Wei
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Xin-Hao Ji
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Ju Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Jing-Shan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Shao-Yu Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China.
| | - Feng Jin
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China.
| |
Collapse
|
3
|
Anitha K, Singh MK, Kohat K, Sri Varshini T, Chenchula S, Padmavathi R, Amerneni LS, Vishnu Vardhan K, Mythili Bai K, Chavan MR, Bhatt S. Recent Insights into the Neurobiology of Alzheimer's Disease and Advanced Treatment Strategies. Mol Neurobiol 2025; 62:2314-2332. [PMID: 39102108 DOI: 10.1007/s12035-024-04384-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024]
Abstract
In recent years, significant advancements have been made in understanding Alzheimer's disease from both neurobiological and clinical perspectives. Exploring the complex systems underlying AD has unveiled insights that could potentially revolutionize therapeutic approaches. Recent investigations have highlighted intricate interactions among genetic, molecular, and environmental factors in AD. Optimism arises from neurobiological advancements and diverse treatment options, potentially slowing or halting disease progression. Amyloid-beta plaques and tau protein tangles crucially influence AD onset and progression. Emerging treatments involve diverse strategies, such as approaches targeting multiple pathways involved in AD pathogenesis, such as inflammation, oxidative stress, and synaptic dysfunction pathways. Clinical trials using humanized monoclonal antibodies, focusing on immunotherapies eliminating amyloid-beta, have shown promise. Nonpharmacological interventions such as light therapy, electrical stimulation, cognitive training, physical activity, and dietary changes have drawn attention for their potential to slow cognitive aging and enhance brain health. Precision medicine, which involves tailoring therapies to individual genetic and molecular profiles, has gained traction. Ongoing research and interdisciplinary collaboration are expected to yield more effective treatments.
Collapse
Affiliation(s)
- Anitha K
- School of Pharmacy and Technology Management (SPTM), SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed to University, Shirpur, 425405, India
| | | | - Komal Kohat
- All India Institute of Medical Sciences, Madhya Pradesh, Bhopal, 462020, India
| | - Sri Varshini T
- All India Institute of Medical Sciences, Raipur, 462020, India
| | - Santenna Chenchula
- Department of Pharmacology, All India Institute of Medical Sciences, Bhopal, 462020, India.
| | - Padmavathi R
- SVS Medical College, Hyderabad, Telangana, India
| | | | - Vishnu Vardhan K
- All India Institute of Medical Sciences, Madhya Pradesh, Bhopal, 462020, India
| | | | - Madhav Rao Chavan
- All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - Shvetank Bhatt
- School of Health Sciences and Technology, MIT World Peace University, Dr. Vishwanath Karad, Pune, 411038, Maharashtra, India
| |
Collapse
|
4
|
Ai X, Cao Z, Ma Z, Liu Q, Huang W, Sun T, Li J, Yang C. Proteomic Analysis Reveals Physiological Activities of Aβ Peptide for Alzheimer's Disease. Int J Mol Sci 2024; 25:8336. [PMID: 39125907 PMCID: PMC11313142 DOI: 10.3390/ijms25158336] [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: 06/30/2024] [Revised: 07/27/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
With the rapid progress in deciphering the pathogenesis of Alzheimer's disease (AD), it has been widely accepted that the accumulation of misfolded amyloid β (Aβ) in the brain could cause the neurodegeneration in AD. Although much evidence demonstrates the neurotoxicity of Aβ, the role of Aβ in the nervous system are complex. However, more comprehensive studies are needed to understand the physiological effect of Aβ40 monomers in depth. To explore the physiological mechanism of Aβ, we employed mass spectrometry to investigate the altered proteomic events induced by a lower submicromolar concentration of Aβ. Human neuroblastoma SH-SY5Y cells were exposed to five different concentrations of Aβ1-40 monomers and collected at four time points. The proteomic analysis revealed the time-course behavior of proteins involved in biological processes, such as RNA splicing, nuclear transport and protein localization. Further biological studies indicated that Aβ40 monomers may activate PI3K/AKT signaling to regulate p-Tau, Ezrin and MAP2. These three proteins are associated with dendritic morphogenesis, neuronal polarity, synaptogenesis, axon establishment and axon elongation. Moreover, Aβ40 monomers may regulate their physiological forms by inhibiting the expression of BACE1 and APP via activation of the ERK1/2 pathway. A comprehensive exploration of pathological and physiological mechanisms of Aβ is beneficial for exploring novel treatment.
Collapse
Affiliation(s)
- Xiaorui Ai
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (X.A.); (Z.C.); (Z.M.); (Q.L.); (W.H.); (T.S.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zeyu Cao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (X.A.); (Z.C.); (Z.M.); (Q.L.); (W.H.); (T.S.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zhaoru Ma
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (X.A.); (Z.C.); (Z.M.); (Q.L.); (W.H.); (T.S.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Qinghuan Liu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (X.A.); (Z.C.); (Z.M.); (Q.L.); (W.H.); (T.S.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Wei Huang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (X.A.); (Z.C.); (Z.M.); (Q.L.); (W.H.); (T.S.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (X.A.); (Z.C.); (Z.M.); (Q.L.); (W.H.); (T.S.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Jing Li
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (X.A.); (Z.C.); (Z.M.); (Q.L.); (W.H.); (T.S.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Chenxi Yang
- School of Biological Science & Medical Engineering, Southeast University, No. 2 Sipailou, Nanjing 210096, China
| |
Collapse
|
5
|
Dhapola R, Sharma P, Kumari S, Bhatti JS, HariKrishnaReddy D. Environmental Toxins and Alzheimer's Disease: a Comprehensive Analysis of Pathogenic Mechanisms and Therapeutic Modulation. Mol Neurobiol 2024; 61:3657-3677. [PMID: 38006469 DOI: 10.1007/s12035-023-03805-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/13/2023] [Indexed: 11/27/2023]
Abstract
Alzheimer's disease is a leading cause of mortality worldwide. Inorganic and organic hazards, susceptibility to harmful metals, pesticides, agrochemicals, and air pollution are major environmental concerns. As merely 5% of AD cases are directly inherited indicating that these environmental factors play a major role in disease development. Long-term exposure to environmental toxins is believed to progress neuropathology, which leads to the development of AD. Numerous in-vitro and in-vivo studies have suggested the harmful impact of environmental toxins at cellular and molecular level. Common mechanisms involved in the toxicity of these environmental pollutants include oxidative stress, neuroinflammation, mitochondrial dysfunction, abnormal tau, and APP processing. Increased expression of GSK-3β, BACE-1, TNF-α, and pro-apoptotic molecules like caspases is observed upon exposure to these environmental toxins. In addition, the expression of neurotrophins like BDNF and GAP-43 have been found to be reduced as a result of toxicity. Further, modulation of signaling pathways involving PARP-1, PGC-1α, and MAPK/ERK induced by toxins have been reported to contribute in AD pathogenesis. These pathways are a promising target for developing novel AD therapeutics. Drugs like epigallocatechin-gallate, neflamapimod, salsalate, dexmedetomidine, and atabecestat are in different phases of clinical trials targeting the pathways for possible treatment of AD. This review aims to culminate the correlation between environmental toxicants and AD development. We emphasized upon the signaling pathways involved in the progression of the disease and the therapeutics under clinical trial targeting the altered pathways for possible treatment of AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151 401, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India.
| |
Collapse
|
6
|
Venkatesan D, Muthukumar S, Iyer M, Babu HWS, Gopalakrishnan AV, Yadav MK, Vellingiri B. Heavy metals toxicity on epigenetic modifications in the pathogenesis of Alzheimer's disease (AD). J Biochem Mol Toxicol 2024; 38:e23741. [PMID: 38816991 DOI: 10.1002/jbt.23741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/09/2024] [Accepted: 05/09/2024] [Indexed: 06/01/2024]
Abstract
Alzheimer's disease (AD) is a progressive decline in cognitive ability and behavior which eventually disrupts daily activities. AD has no cure and the progression rate varies unlikely. Among various causative factors, heavy metals are reported to be a significant hazard in AD pathogenesis. Metal-induced neurodegeneration has been focused globally with thorough research to unravel the mechanistic insights in AD. Recently, heavy metals suggested to play an important role in epigenetic alterations which might provide evidential results on AD pathology. Epigenetic modifications are known to play towards novel therapeutic approaches in treating AD. Though many studies focus on epigenetics and heavy metal implications in AD, there is a lack of research on heavy metal influence on epigenetic toxicity in neurological disorders. The current review aims to elucidate the plausible role of cadmium (Cd), iron (Fe), arsenic (As), copper (Cu), and lithium (Li) metals on epigenetic factors and the increase in amyloid beta and tau phosphorylation in AD. Also, the review discusses the common methods of heavy metal detection to implicate in AD pathogenesis. Hence, from this review, we can extend the need for future research on identifying the mechanistic behavior of heavy metals on epigenetic toxicity and to develop diagnostic and therapeutic markers in AD.
Collapse
Affiliation(s)
- Dhivya Venkatesan
- Centre for Neuroscience, Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore, India
| | - Sindduja Muthukumar
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Mahalaxmi Iyer
- Centre for Neuroscience, Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore, India
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Harysh Winster Suresh Babu
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Mukesh Kumar Yadav
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
7
|
Singh N, Nandy SK, Jyoti A, Saxena J, Sharma A, Siddiqui AJ, Sharma L. Protein Kinase C (PKC) in Neurological Health: Implications for Alzheimer's Disease and Chronic Alcohol Consumption. Brain Sci 2024; 14:554. [PMID: 38928554 PMCID: PMC11201589 DOI: 10.3390/brainsci14060554] [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: 05/07/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Protein kinase C (PKC) is a diverse enzyme family crucial for cell signalling in various organs. Its dysregulation is linked to numerous diseases, including cancer, cardiovascular disorders, and neurological problems. In the brain, PKC plays pivotal roles in synaptic plasticity, learning, memory, and neuronal survival. Specifically, PKC's involvement in Alzheimer's Disease (AD) pathogenesis is of significant interest. The dysregulation of PKC signalling has been linked to neurological disorders, including AD. This review elucidates PKC's pivotal role in neurological health, particularly its implications in AD pathogenesis and chronic alcohol addiction. AD, characterised by neurodegeneration, implicates PKC dysregulation in synaptic dysfunction and cognitive decline. Conversely, chronic alcohol consumption elicits neural adaptations intertwined with PKC signalling, exacerbating addictive behaviours. By unravelling PKC's involvement in these afflictions, potential therapeutic avenues emerge, offering promise for ameliorating their debilitating effects. This review navigates the complex interplay between PKC, AD pathology, and alcohol addiction, illuminating pathways for future neurotherapeutic interventions.
Collapse
Affiliation(s)
- Nishtha Singh
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| | - Shouvik Kumar Nandy
- School of Pharmacy, Techno India University, Sector-V, Kolkata 700091, West Bengal, India;
| | - Anupam Jyoti
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara 391760, Gujarat, India;
| | - Juhi Saxena
- Department of Biotechnology, Parul Institute of Technology, Parul University, Vadodara 391760, Gujarat, India;
| | - Aditi Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail 55476, Saudi Arabia
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| |
Collapse
|
8
|
Hoang NMH, Nguyen HD, Jo W, Kim MS. Role of prolactin in the protective effect of amisulpride against 1,2-Diacetylbenzene's neurotoxicity. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 107:104418. [PMID: 38493881 DOI: 10.1016/j.etap.2024.104418] [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: 07/19/2023] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Exposure to organic solvents is associated with various health problems, including neurodegenerative diseases. Among these solvents, 1,2-diethylbenzene is notable for its ability to produce a toxic metabolite, 1,2-Diacetylbenzene (DAB), which can cause memory impairment. Prolactin (PRL) is theorized to protect the central nervous system. Certain antipsychotic drugs, known for increasing PRL secretion, have shown to improve cognitive performance in psychotic Alzheimer's patients. Among these, amisulpride stands out for its high efficacy, limited side effects, and high selectivity for dopamine D2 receptors. In our study, we explored the potential of amisulpride to inhibit DAB-induced neurotoxicity via PRL activation. Our results show that amisulpride enhances the PRL/JAK/STAT, PI3K/AKT, and BDNF/ERK/CREB pathways, playing critical roles in PRL's neuroprotection pathways and memory formation. Additionally, amisulpride inhibited DAB-triggered NLRP3 inflammasome activation and apoptosis. Collectively, these findings suggest that amisulpride may be a promising therapeutic intervention for DAB-induced neurotoxicity, partly through activating the PRL pathway.
Collapse
Affiliation(s)
- Ngoc Minh-Hong Hoang
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Jeonnam 57922, Republic of Korea
| | - Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Jeonnam 57922, Republic of Korea
| | - Wonhee Jo
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Jeonnam 57922, Republic of Korea
| | - Min-Sun Kim
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Jeonnam 57922, Republic of Korea.
| |
Collapse
|
9
|
Xu Y, Nie J, Lu C, Hu C, Chen Y, Ma Y, Huang Y, Lu L. Effects and mechanisms of bisphenols exposure on neurodegenerative diseases risk: A systemic review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 919:170670. [PMID: 38325473 DOI: 10.1016/j.scitotenv.2024.170670] [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: 10/18/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Environmental bisphenols (BPs) pose a global threat to human health because of their extensive use as additives in plastic products. BP residues are increasing in various environmental media (i.e., water, soil, and indoor dust) and biological and human samples (i.e., serum and brain). Both epidemiological and animal studies have determined an association between exposure to BPs and an increased risk of neurodegenerative diseases (e.g., Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis), including cognitive abnormalities and behavioral disturbances. Hence, understanding the biological responses to different BPs is essential for prevention, and treatment. This study provides an overview of the underlying pathogenic molecular mechanisms as a valuable basis for understanding neurodegenerative disease responses to BPs, including accumulation of misfolded proteins, reduction of tyrosine hydroxylase and dopamine, abnormal hormone signaling, neuronal death, oxidative stress, calcium homeostasis, and inflammation. These findings provide new insights into the neurotoxic potential of BPs and ultimately contribute to a comprehensive health risk evaluation.
Collapse
Affiliation(s)
- Yeqing Xu
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jun Nie
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Chenghao Lu
- College of Mathematics and Computer Science, Zhejiang A & F University, Hangzhou 311300, China
| | - Chao Hu
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yunlu Chen
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ying Ma
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yuru Huang
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Liping Lu
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
10
|
Xiong J, Pang X, Song X, Yang L, Pang C. The coherence between PSMC6 and α-ring in the 26S proteasome is associated with Alzheimer's disease. Front Mol Neurosci 2024; 16:1330853. [PMID: 38357597 PMCID: PMC10864545 DOI: 10.3389/fnmol.2023.1330853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/22/2023] [Indexed: 02/16/2024] Open
Abstract
Alzheimer's disease (AD) is a heterogeneous age-dependent neurodegenerative disorder. Its hallmarks involve abnormal proteostasis, which triggers proteotoxicity and induces neuronal dysfunction. The 26S proteasome is an ATP-dependent proteolytic nanomachine of the ubiquitin-proteasome system (UPS) and contributes to eliminating these abnormal proteins. This study focused on the relationship between proteasome and AD, the hub genes of proteasome, PSMC6, and 7 genes of α-ring, are selected as targets to study. The following three characteristics were observed: 1. The total number of proteasomes decreased with AD progression because the proteotoxicity damaged the expression of proteasome proteins, as evidenced by the downregulation of hub genes. 2. The existing proteasomes exhibit increased activity and efficiency to counterbalance the decline in total proteasome numbers, as evidenced by enhanced global coordination and reduced systemic disorder of proteasomal subunits as AD advances. 3. The synergy of PSMC6 and α-ring subunits is associated with AD. Synergistic downregulation of PSMC6 and α-ring subunits reflects a high probability of AD risk. Regarding the above discovery, the following hypothesis is proposed: The aggregation of pathogenic proteins intensifies with AD progression, then proteasome becomes more active and facilitates the UPS selectively targets the degradation of abnormal proteins to maintain CNS proteostasis. In this paper, bioinformatics and support vector machine learning methods are applied and combined with multivariate statistical analysis of microarray data. Additionally, the concept of entropy was used to detect the disorder of proteasome system, it was discovered that entropy is down-regulated continually with AD progression against system chaos caused by AD. Another conception of the matrix determinant was used to detect the global coordination of proteasome, it was discovered that the coordination is enhanced to maintain the efficiency of degradation. The features of entropy and determinant suggest that active proteasomes resist the attack caused by AD like defenders, on the one hand, to protect themselves (entropy reduces), and on the other hand, to fight the enemy (determinant reduces). It is noted that these are results from biocomputing and need to be supported by further biological experiments.
Collapse
Affiliation(s)
- Jing Xiong
- College of Computer Science, Sichuan Normal University, Chengdu, China
| | - Xinping Pang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xianghu Song
- College of Computer Science, Sichuan Normal University, Chengdu, China
| | - Lin Yang
- College of Computer Science, Sichuan Normal University, Chengdu, China
| | - Chaoyang Pang
- College of Computer Science, Sichuan Normal University, Chengdu, China
| |
Collapse
|
11
|
Liu MH, Tang Y, Qu LQ, Song LL, Lo HH, Zhang RL, Yun XY, Wang HM, Chan JTW, Wu JH, Wang CR, Wong VKW, Wu AG, Law BYK. Raddeanin A isolated from Anemone raddeana Regel improves pathological and cognitive deficits of the mice model of Alzheimer's disease by targeting β-amyloidosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155121. [PMID: 37856988 DOI: 10.1016/j.phymed.2023.155121] [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: 02/18/2023] [Revised: 06/30/2023] [Accepted: 09/27/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Raddeanin A is a triterpenoid isolated from Anemone raddeana Regel. It exhibits a broad spectrum of biological activities such as anti-tumor and anti-inflammatory, however, its neuroprotective effect in targeting Alzheimer's disease (AD) remains uninvestigated. PURPOSE To provide scientific base for the development of novel AD drug by clarifying the neuroprotective effect and molecular mechanisms of raddeanin A in both in vitro and in vivo AD model. STUDY DESIGN To confirm the neuroprotective role of raddeanin A in the treatment of AD, its mechanisms and effects on β-amyloidosis and Aβ fibrillation was studied in U87 cells. Besides, the improvement on cognitive deficit, pathological defects, reactive astrocyte clusters, inhibition on neuronal inflammation and apoptosis were further studied in 3 x Tg-AD mice model of AD. METHODS Real-time PCR, western blot, dot blot, biolayer interferometry and bioinformatics analysis were used to confirm the in vitro effect and targets of raddeanin A on β-amyloidosis and its associated protein network. A series of experiments including Morris water maze, H&E staining, nissl staining and immunofluorescence analysis were conducted to confirm the protective behavioral effect of raddeanin A in the in vivo AD mice model. RESULTS Raddeanin A was identified to reduce β-amyloidosis in U87 cells and 3 x Tg-AD mice model of AD by decreasing level of BACE1, APP, APP-β and Aβ. Raddeanin A improved behavioral, spatial memory and learning ability in the AD mice. In the cortex and hippocampus, raddeanin A improved the morphology and arrangement of neurons, lower the level of reactive astrocyte marker GFAP and apoptotic marker proteins Bax/Bcl2 ratio. Moreover, raddeanin A upregulated the mRNA and protein level of Prkcα in the hippocampus of AD mice whose neuroprotective effect was exerted possibly via the activation of protein kinase C. CONCLUSION As a novel natural agent targeting β-amyloidosis, our results provide the first evidence of the multiple in vitro and in vivo neuroprotective effect of raddeanin A, suggesting its potential therapeutic application in preventing or alleviating the symptoms of AD.
Collapse
Affiliation(s)
- Meng Han Liu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yong Tang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Li Qun Qu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Lin Lin Song
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Hang Hong Lo
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Rui Long Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xiao Yun Yun
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Hui Miao Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Joyce Tsz Wai Chan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jian Hui Wu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Cai Ren Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Vincent Kam Wai Wong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - An Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
12
|
Lee SW, Lim JM, Lee GM, Park JH, Seralathan KK, Oh BT. Evaluation of Lentilactobacillus parafarraginis A6-2 strain for aluminum removal and anti-inflammatory effects: implications for alleviating Al toxicity. J Appl Microbiol 2023; 134:lxad271. [PMID: 37989872 DOI: 10.1093/jambio/lxad271] [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: 07/19/2023] [Revised: 09/30/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
AIM To assess the effectiveness of Lentilactobacillus parafarraginis A6-2 cell lysate for the removal of aluminum (Al), which induces neurotoxicity, and its protective effect at cellular level. METHODS AND RESULTS The cell lysate of the selected L. parafarraginis A6-2 strain demonstrated superior Al removal compared to live or dead cells. The Al removal efficiency of L. parafarraginis A6-2 cell lysate increased with decreasing pH and increasing temperature, primarily through adsorption onto peptidoglycan. Neurotoxicity mitigation potential of L. parafarraginis A6-2 was evaluated using C6 glioma cells. C6 cells exposed with increasing concentration of Al led to elevated toxicity and inflammation, which were gradually alleviated upon treatment with L. parafarraginis A6-2. Moreover, Al-induced oxidative stress in C6 cells showed a concentration-dependent reduction upon treatment with L. parafarraginis A6-2. CONCLUSIONS This study demonstrated that L. parafarraginis A6-2 strain, particularly in its lysate form, exhibited enhanced capability for Al removal. Furthermore, it effectively mitigated Al-induced toxicity, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Se-Won Lee
- Division of Biotechnology, Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Jeong-Muk Lim
- Division of Biotechnology, Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Gwang-Min Lee
- Division of Biotechnology, Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Jung-Hee Park
- Division of Biotechnology, Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Kamala-Kannan Seralathan
- Division of Biotechnology, Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Byung-Taek Oh
- Division of Biotechnology, Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea
| |
Collapse
|
13
|
Oummadi A, Menuet A, Méresse S, Laugeray A, Guillemin G, Mortaud S. The herbicides glyphosate and glufosinate and the cyanotoxin β-N-methylamino-l-alanine induce long-term motor disorders following postnatal exposure: the importance of prior asymptomatic maternal inflammatory sensitization. Front Neurosci 2023; 17:1172693. [PMID: 37360165 PMCID: PMC10288190 DOI: 10.3389/fnins.2023.1172693] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Background Prenatal maternal immune activation (MIA) and/or perinatal exposure to various xenobiotics have been identified as risk factors for neurological disorders, including neurodegenerative diseases. Epidemiological data suggest an association between early multi-exposures to various insults and neuropathologies. The "multiple-hit hypothesis" assumes that prenatal inflammation makes the brain more susceptible to subsequent exposure to several kinds of neurotoxins. To explore this hypothesis and its pathological consequences, a behavioral longitudinal procedure was performed after prenatal sensitization and postnatal exposure to low doses of pollutants. Methods Maternal exposure to an acute immune challenge (first hit) was induced by an asymptomatic lipopolysaccharide (LPS) dose (0.008 mg/kg) in mice. This sensitization was followed by exposing the offspring to environmental chemicals (second hit) postnatally, by the oral route. The chemicals used were low doses of the cyanotoxin β-N-methylamino-l-alanine (BMAA; 50 mg/kg), the herbicide glufosinate ammonium (GLA; 0.2 mg/kg) or the pesticide glyphosate (GLY; 5 mg/kg). After assessing maternal parameters, a longitudinal behavioral assessment was carried out on the offspring in order to evaluate motor and emotional abilities in adolescence and adulthood. Results We showed that the low LPS immune challenge was an asymptomatic MIA. Even though a significant increase in systemic pro-inflammatory cytokines was detected in the dams, no maternal behavioral defects were observed. In addition, as shown by rotarod assays and open field tests, this prenatal LPS administration alone did not show any behavioral disruption in offspring. Interestingly, our data showed that offspring subjected to both MIA and post-natal BMAA or GLA exposure displayed motor and anxiety behavioral impairments during adolescence and adulthood. However, this synergistic effect was not observed in the GLY-exposed offspring. Conclusion These data demonstrated that prenatal and asymptomatic immune sensitization represents a priming effect to subsequent exposure to low doses of pollutants. These double hits act in synergy to induce motor neuron disease-related phenotypes in offspring. Thus, our data strongly emphasize that multiple exposures for developmental neurotoxicity regulatory assessment must be considered. This work paves the way for future studies aiming at deciphering cellular pathways involved in these sensitization processes.
Collapse
Affiliation(s)
- Asma Oummadi
- Experimental and Molecular Immunology and Neurogenetics, UMR7355 CNRS, Orléans, France
- Faculty of Medicine and Human Health Sciences, Center for MND Research, Macquarie University, Sydney, NSW, Australia
| | - Arnaud Menuet
- Experimental and Molecular Immunology and Neurogenetics, UMR7355 CNRS, Orléans, France
- UFR Sciences et Techniques, University of Orléans, Orléans, France
| | - Sarah Méresse
- Experimental and Molecular Immunology and Neurogenetics, UMR7355 CNRS, Orléans, France
- UFR Sciences et Techniques, University of Orléans, Orléans, France
| | - Anthony Laugeray
- Faculty of Biology and Medicine, Department of Fundamental Neurosciences, Lausanne, Switzerland
| | - Gilles Guillemin
- Faculty of Medicine and Human Health Sciences, Center for MND Research, Macquarie University, Sydney, NSW, Australia
| | - Stéphane Mortaud
- Experimental and Molecular Immunology and Neurogenetics, UMR7355 CNRS, Orléans, France
- UFR Sciences et Techniques, University of Orléans, Orléans, France
| |
Collapse
|
14
|
Migliore L, Coppedè F. Gene-environment interactions in Alzheimer disease: the emerging role of epigenetics. Nat Rev Neurol 2022; 18:643-660. [PMID: 36180553 DOI: 10.1038/s41582-022-00714-w] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2022] [Indexed: 12/15/2022]
Abstract
With the exception of a few monogenic forms, Alzheimer disease (AD) has a complex aetiology that is likely to involve multiple susceptibility genes and environmental factors. The role of environmental factors is difficult to determine and, until a few years ago, the molecular mechanisms underlying gene-environment (G × E) interactions in AD were largely unknown. Here, we review evidence that has emerged over the past two decades to explain how environmental factors, such as diet, lifestyle, alcohol, smoking and pollutants, might interact with the human genome. In particular, we discuss how various environmental AD risk factors can induce epigenetic modifications of key AD-related genes and pathways and consider how epigenetic mechanisms could contribute to the effects of oxidative stress on AD onset. Studies on early-life exposures are helping to uncover critical time windows of sensitivity to epigenetic influences from environmental factors, thereby laying the foundations for future primary preventative approaches. We conclude that epigenetic modifications need to be considered when assessing G × E interactions in AD.
Collapse
Affiliation(s)
- Lucia Migliore
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy. .,Department of Laboratory Medicine, Pisa University Hospital, Pisa, Italy.
| | - Fabio Coppedè
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| |
Collapse
|
15
|
Stecker MM, Peltier MR, Reiss AB. The role of massive demographic databases in intractable illnesses: Denomics for dementia. AIMS Public Health 2022; 9:618-629. [PMID: 36330282 PMCID: PMC9581740 DOI: 10.3934/publichealth.2022043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/10/2022] [Accepted: 08/14/2022] [Indexed: 07/26/2023] Open
Abstract
Despite intensive research, effective treatments for many common and devastating diseases are lacking. For example, huge efforts and billions of dollars have been invested in Alzheimer's disease (AD), which affects over 50 million people worldwide. However, there is still no effective drug that can slow or cure AD. This relates, in part, to the absence of an animal model or cellular system that incorporates all the relevant features of the disease. Therefore, large scale studies on human populations and tissues will be key to better understanding dementia and developing methods to prevent or treat it. This is especially difficult because the dementia phenotype can result from many different processes and is likely to be affected by multiple personal and environmental variables. We hypothesize that analyzing massive volumes of demographic data that are currently available and combining this with genomic, proteomic, and metabolomic profiles of AD patients and their families, new insights into pathophysiology and treatment of AD may arise. While this requires much coordination and cooperation among large institutions, the potential for advancement would be life-changing for millions of people. In many ways this represents the next step in the information revolution started by the Human Genome Project.
Collapse
Affiliation(s)
| | - Morgan R. Peltier
- Department of Psychiatry, Hackensack Meridian Health, Neptune City, NJ 07753, USA
| | | |
Collapse
|
16
|
Nguyen HD, Kim MS. Exposure to a mixture of heavy metals induces cognitive impairment: Genes and microRNAs involved. Toxicology 2022; 471:153164. [PMID: 35346790 DOI: 10.1016/j.tox.2022.153164] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/10/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022]
Abstract
Converging evidence demonstrates that microRNAs (miRNAs) play an important role in the etiology of cognitive impairment. Thus, we aim to: (i) identify the molecular mechanisms of heavy metals, particularly miRNAs involved in the development of cognitive impairment; and (ii) generate miRNA sponges to prevent them from binding with their target messenger RNAs. The Comparative Toxicogenomics Database (CTD; http://ctd.mdibl.org), MicroRNA ENrichment TURned NETwork (MIENTURNET, http://userver.bio.uniroma1.it/apps/mienturnet/) and the microRNA sponge generator and tester (miRNAsong, http://www.med.muni.cz/histology/miRNAsong) were used as the core data-mining approaches in the current study. We observed that lead acetate, arsenic, gold, copper, iron, and aluminum, as well as their mixtures, had significant effects on the development of cognitive impairment. Although prevalent genes obtained from investigated heavy metals of cognitive impairment were different, the "PI3K-Akt signaling pathway", "pathways of neurodegeneration-multiple diseases", "apoptosis", "apoptosis-multiple species", "p53 signaling pathway", "NF-kappa B signaling pathway", and "Alzheimer's disease pathway" were highlighted. The mixed heavy metals altered the genes BAX, CASP3, BCL2, TNF, and IL-1B, indicating the significance of apoptosis and pro-inflammatory cytokines in the pathogenesis of cognitive impairment and the possibility of targeting these genes in future neuroprotective therapy. In addition, we used a network-based approach to identify key genes, miRNAs, pathways, and diseases related to the development of cognitive impairment. We also found 16 significant miRNAs related to cognitive impairment (hsa-miR-1-3p, hsa-let-7a-5p, hsa-miR-9-5p, hsa-miR-16-5p, hsa-miR-17-5p, hsa-miR-20a-5p, hsa-miR-26a-5p, hsa-miR-26b-5p, hsa-miR-34a-5p, hsa-miR-101-3p, hsa-miR-106a-5p, hsa-miR-128-3p, hsa-miR-144-3p, hsa-miR-199a-3p, hsa-miR-204-5p, and hsa-miR-335-5p). Finally, we created and evaluated miRNA sponge sequences for these miRNAs in silico. Further studies, including in vivo and in vitro, are needed to assess the link between these genes, miRNAs, pathways, and cognitive impairment.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Min-Sun Kim
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea.
| |
Collapse
|