1
|
He Y, Lu W, Zhou X, Mu J, Shen W. Unraveling Alzheimer's disease: insights from single-cell sequencing and spatial transcriptomic. Front Neurol 2024; 15:1515981. [PMID: 39741706 PMCID: PMC11685091 DOI: 10.3389/fneur.2024.1515981] [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: 10/23/2024] [Accepted: 12/05/2024] [Indexed: 01/03/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder marked by cognitive decline, primarily affecting memory and executive function. This review highlights recent advancements in single-cell sequencing and spatial transcriptomics, which provide detailed insights into the cellular heterogeneity and neuroimmune mechanisms of AD. The review addresses the need for understanding complex cellular interactions to identify novel therapeutic targets and biomarkers. Single-cell sequencing has revolutionized our understanding by mapping gene expression at the individual cell level, revealing distinct microglial and astrocytic states that contribute to neuroinflammation and neurodegeneration. These technologies have uncovered disease-associated microglial subpopulations and gene expression changes linked to AD risk genes, essential for developing targeted therapies. In conclusion, the integration of single-cell and spatial transcriptomics with other omics data is crucial for a comprehensive understanding of AD, paving the way for personalized medicine. Continued interdisciplinary collaboration will be vital in translating these findings into effective treatments, improving patient outcomes.
Collapse
Affiliation(s)
- Yibo He
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Wenqiang Lu
- Department of Thoracic Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu, China
| | - Xiao Zhou
- Department of Pharmacy, Shuangqiao Economic and Technological Development Zone People's Hospital, Chongqing, China
| | - Jie Mu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Wei Shen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Singh M, Ali H, Renuka Jyothi S, Kaur I, Kumar S, Sharma N, Siva Prasad GV, Pramanik A, Hassan Almalki W, Imran M. Tau proteins and senescent Cells: Targeting aging pathways in Alzheimer's disease. Brain Res 2024; 1844:149165. [PMID: 39155034 DOI: 10.1016/j.brainres.2024.149165] [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: 06/16/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by abnormal accumulation of tau proteins and amyloid-β, leading to neuronal death and cognitive impairment. Recent studies have implicated aging pathways, including dysregulation of tau and cellular senescence in AD pathogenesis. In AD brains, tau protein, which normally stabilizes microtubules, becomes hyperphosphorylated and forms insoluble neurofibrillary tangles. These tau aggregates impair neuronal function and are propagated across the brain's neurocircuitry. Meanwhile, the number of senescent cells accumulating in the aging brain is rising, releasing a pro-inflammatory SASP responsible for neuroinflammation and neurodegeneration. This review explores potential therapeutic interventions for AD targeting tau protein and senescent cells, and tau -directed compounds, senolytics, eliminating senescent cells, and agents that modulate the SASP-senomodulators. Ultimately, a combined approach that incorporates tau-directed medications and targeted senescent cell-based therapies holds promise for reducing the harmful impact of AD's shared aging pathways.
Collapse
Affiliation(s)
- Mahaveer Singh
- School of Pharmacy and Technology Management, SVKMs NMIMS University, Shirpur campus, Maharastra India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali 140307, Punjab, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
3
|
Zhu C, Liu J, Lin J, Xu J, Yu E. Investigating the effects of Ginkgo biloba leaf extract on cognitive function in Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14914. [PMID: 39238068 PMCID: PMC11377177 DOI: 10.1111/cns.14914] [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: 12/15/2023] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 09/07/2024] Open
Abstract
AIMS Alzheimer's disease (AD) is a neurodegenerative disorder with limited treatment options. This study aimed to investigate the therapeutic effects of Ginkgo biloba leaf extract (GBE) on AD and explore its potential mechanisms of action. METHODS Key chemical components of GBE, including quercetin, luteolin, and kaempferol, were identified using network pharmacology methods. Bioinformatics analysis revealed their potential roles in AD through modulation of the PI3K/AKT/NF-κB signaling pathway. RESULTS Mouse experiments demonstrated that GBE improved cognitive function, enhanced neuronal morphology, and reduced serum inflammatory factors. Additionally, GBE modulated the expression of relevant proteins and mRNA. CONCLUSION GBE shows promise as a potential treatment for AD. Its beneficial effects on cognitive function, neuronal morphology, and inflammation may be attributed to its modulation of the PI3K/AKT/NF-κB signaling pathway. These findings provide experimental evidence for the application of Ginkgo biloba leaf in AD treatment and highlight its potential mechanisms of action.
Collapse
Affiliation(s)
- Cheng Zhu
- School of Mental Health, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Jie Liu
- The Second People's Hospital of Chuzhou Sleep Disorders Department, Chuzhou, China
| | - Jixin Lin
- Second Clinical Medicine Faculty, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaxi Xu
- General Psychiatric Department, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Enyan Yu
- Clinical Psychology Department, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
4
|
Shobeiri P, Alilou S, Jaberinezhad M, Zare F, Karimi N, Maleki S, Teixeira AL, Perry G, Rezaei N. Circulating long non-coding RNAs as novel diagnostic biomarkers for Alzheimer's disease (AD): A systematic review and meta-analysis. PLoS One 2023; 18:e0281784. [PMID: 36947499 PMCID: PMC10032479 DOI: 10.1371/journal.pone.0281784] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 02/01/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have been reported to be involved in the pathogenesis of neurodegenerative diseases. It has also been hypothesized that plasma exosomal lncRNAs may be used as Alzheimer's disease (AD) biomarkers. In this systematic review, we compiled all studies on the subject to evaluate the accuracy of lncRNAs in identifying AD cases through meta-analysis. METHODS A PRISMA-compliant systematic search was conducted in PubMed/MEDLINE, EMBASE, and Web of Science databases for English publications till September 2022. We included all observational studies published which investigated the sensitivity and specificity of various lncRNAs in plasma samples of AD diagnosis. Our search strategy included lncRNA and all the related spelling and abbreviation variations combined with the keyword Alzheimer's disease. Methodological quality was assessed using the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) guidelines and the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-II) tool. The meta-analysis was carried out using the area under the Receiver Operator Characteristic (ROC) curves (AUC) and sensitivity and specificity values to assess the accuracy of the identified lncRNAs in AD diagnosis. To account for the predicted heterogeneity of the study, a random-effects model was used. All the statistical analyses and visualizations were conducted using Stata 17.0 software. RESULTS A total of seven studies (AD patients = 553, healthy controls = 513) were included in the meta-analysis. Three lncRNAs were upregulated (RNA BACE-AS1, RNA NEAT1, RNA GAS5), and one lncRNA (MALAT1) was downregulated in plasma samples of AD patients. RNA 51A and RNA BC200 were reported to have variable expression patterns. A lncRNA (RNA 17A) was not significantly different between AD and control groups. The pooled sensitivity, specificity, and AUC values of lncRNAs in identifying AD were (0.74; 95% CI [0.63, 0.82], I2 = 79.2%), (0.88; 95% CI [0.75, 0.94], I2 = 88.9%), and 0.86; 95% CI [0.82, 0.88], respectively. In addition, the pooled diagnostic odds ratio (DOR) of the five individual lncRNAs in AD diagnosis was 20. CONCLUSION lncRNAs had high accuracy in identifying AD and must be seen as a promising diagnostic biomarker of the disease.
Collapse
Affiliation(s)
- Parnian Shobeiri
- Children’s Medical Center Hospital, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanam Alilou
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Jaberinezhad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farshad Zare
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nastaran Karimi
- School of Medicine, Sari Branch, Islamic Azad University, Sari, Iran
| | - Saba Maleki
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Antonio L. Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX, United States of America
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Khoury R, Gallop A, Roberts K, Grysman N, Lu J, Grossberg GT. Pharmacotherapy for Alzheimer’s disease: what’s new on the horizon? Expert Opin Pharmacother 2022; 23:1305-1323. [DOI: 10.1080/14656566.2022.2097868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Rita Khoury
- Department of Psychiatry and Clinical Psychology, St. Georges Hospital University Medical Center, Beirut, Lebanon
- University of Balamand, Faculty of Medicine, Beirut, Lebanon
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| | - Amy Gallop
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| | - Kelsey Roberts
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| | - Noam Grysman
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| | - Jiaxi Lu
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| | - George T. Grossberg
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
6
|
Fernández-Bolaños JG, López Ó. Butyrylcholinesterase inhibitors as potential anti-Alzheimer's agents: an updated patent review (2018-present). Expert Opin Ther Pat 2022; 32:913-932. [PMID: 35623095 DOI: 10.1080/13543776.2022.2083956] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) constitutes one of the most complex and devastating diseases, with an extraordinarily high increase expected for the next few years. Despite the numerous efforts accomplished so far there is still no cure but just palliative treatments. AREAS COVERED The main topic covered herein has been the development of butyrylcholinesterase (BuChE) inhibitors with the aim of increasing the levels of the neurotransmitter acetylcholine (ACh). Two main groups of compounds have been considered: multitarget and non-multitarget ligands, depending if the structural design is focused or not on other key targets and pathogenic factors of the disease. Seventeen patents regarding multitarget-directed ligands (MTDLs), twelve for not multitarget derivatives, and three for miscellaneous uses have been covered in the period 2018‒2021. EXPERT OPINION BuChE is an attractive target in the treatment of AD for many reasons. It is the most prevalent cholinesterase within more advanced stages of the disease, so drugs inhibiting it would be suitable for the treatment of mid- to severe Alzheimer's patients. Moreover, BuChE has been proved to be connected with some other key hallmarks of the disease, like amyloidogenesis; hybridization of a BuChE-targeting pharmacophore with other scaffolds designed for other therapeutic targets is quite a promising design for potential anti-Alzheimer's drugs.
Collapse
Affiliation(s)
- José G Fernández-Bolaños
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Sevilla, Spain
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Sevilla, Spain
| |
Collapse
|
7
|
Jana A, Bhattacharjee A, Das SS, Srivastava A, Choudhury A, Bhattacharjee R, De S, Perveen A, Iqbal D, Gupta PK, Jha SK, Ojha S, Singh SK, Ruokolainen J, Jha NK, Kesari KK, Ashraf GM. Molecular Insights into Therapeutic Potentials of Hybrid Compounds Targeting Alzheimer's Disease. Mol Neurobiol 2022; 59:3512-3528. [PMID: 35347587 PMCID: PMC9148293 DOI: 10.1007/s12035-022-02779-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/21/2022] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is one of the most complex progressive neurological disorders involving degeneration of neuronal connections in brain cells leading to cell death. AD is predominantly detected among elder people (> 65 years), mostly diagnosed with the symptoms of memory loss and cognitive dysfunctions. The multifarious pathogenesis of AD comprises the accumulation of pathogenic proteins, decreased neurotransmission, oxidative stress, and neuroinflammation. The conventional therapeutic approaches are limited to symptomatic benefits and are ineffective against disease progression. In recent years, researchers have shown immense interest in the designing and fabrication of various novel therapeutics comprised of naturally isolated hybrid molecules. Hybrid therapeutic compounds are developed from the combination of pharmacophores isolated from bioactive moieties which specifically target and block various AD-associated pathogenic pathways. The method of designing hybrid molecules has numerous advantages over conventional multitarget drug development methods. In comparison to in silico high throughput screening, hybrid molecules generate quicker results and are also less expensive than fragment-based drug development. Designing hybrid-multitargeted therapeutic compounds is thus a prospective approach in developing an effective treatment for AD. Nevertheless, several issues must be addressed, and additional researches should be conducted to develop hybrid therapeutic compounds for clinical usage while keeping other off-target adverse effects in mind. In this review, we have summarized the recent progress on synthesis of hybrid compounds, their molecular mechanism, and therapeutic potential in AD. Using synoptic tables, figures, and schemes, the review presents therapeutic promise and potential for the development of many disease-modifying hybrids into next-generation medicines for AD.
Collapse
Affiliation(s)
- Ankit Jana
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed To Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Arkadyuti Bhattacharjee
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed To Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Sabya Sachi Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Avani Srivastava
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed To Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Akshpita Choudhury
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed To Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Rahul Bhattacharjee
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed To Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Swagata De
- Department of English, DDE Unit, The University of Burdwan, GolapbagBurdwan, West Bengal, 713104, India
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Danish Iqbal
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, 11952, Saudi Arabia
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research (SBSR), Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, 15551, Al Ain, United Arab Emirates
| | - Sandeep Kumar Singh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Janne Ruokolainen
- Department of Applied Physics, School of Science, Aalto University, 00076, Espoo, Finland
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, 201310, India.
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, 00076, Espoo, Finland.
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia. .,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
8
|
Jeremic D, Jiménez-Díaz L, Navarro-López JD. Past, present and future of therapeutic strategies against amyloid-β peptides in Alzheimer's disease: a systematic review. Ageing Res Rev 2021; 72:101496. [PMID: 34687956 DOI: 10.1016/j.arr.2021.101496] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/30/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease in ageing, affecting around 46 million people worldwide but few treatments are currently available. The etiology of AD is still puzzling, and new drugs development and clinical trials have high failure rates. Urgent outline of an integral (multi-target) and effective treatment of AD is needed. Accumulation of amyloid-β (Aβ) peptides is considered one of the fundamental neuropathological pillars of the disease, and its dyshomeostasis has shown a crucial role in AD onset. Therefore, many amyloid-targeted therapies have been investigated. Here, we will systematically review recent (from 2014) investigational, follow-up and review studies focused on anti-amyloid strategies to summarize and analyze their current clinical potential. Combination of anti-Aβ therapies with new developing early detection biomarkers and other therapeutic agents acting on early functional AD changes will be highlighted in this review. Near-term approval seems likely for several drugs acting against Aβ, with recent FDA approval of a monoclonal anti-Aβ oligomers antibody -aducanumab- raising hopes and controversies. We conclude that, development of oligomer-epitope specific Aβ treatment and implementation of multiple improved biomarkers and risk prediction methods allowing early detection, together with therapies acting on other factors such as hyperexcitability in early AD, could be the key to slowing this global pandemic.
Collapse
|
9
|
Khan NH, Mir M, Ngowi EE, Zafar U, Khakwani MMAK, Khattak S, Zhai YK, Jiang ES, Zheng M, Duan SF, Wei JS, Wu DD, Ji XY. Nanomedicine: A Promising Way to Manage Alzheimer's Disease. Front Bioeng Biotechnol 2021; 9:630055. [PMID: 33996777 PMCID: PMC8120897 DOI: 10.3389/fbioe.2021.630055] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/08/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating disease of the aging population characterized by the progressive and slow brain decay due to the formation of extracellular plaques in the hippocampus. AD cells encompass tangles of twisted strands of aggregated microtubule binding proteins surrounded by plaques. Delivering corresponding drugs in the brain to deal with these clinical pathologies, we face a naturally built strong, protective barrier between circulating blood and brain cells called the blood-brain barrier (BBB). Nanomedicines provide state-of-the-art alternative approaches to overcome the challenges in drug transport across the BBB. The current review presents the advances in the roles of nanomedicines in both the diagnosis and treatment of AD. We intend to provide an overview of how nanotechnology has revolutionized the approaches used to manage AD and highlight the current key bottlenecks and future perspective in this field. Furthermore, the emerging nanomedicines for managing brain diseases like AD could promote the booming growth of research and their clinical availability.
Collapse
Affiliation(s)
- Nazeer Hussain Khan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Maria Mir
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ebenezeri Erasto Ngowi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Department of Biological Sciences, Faculty of Sciences, Dar es Salaam University College of Education, Dar es Salaam, Tanzania
| | - Ujala Zafar
- School of Natural Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | | | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yuan-Kun Zhai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- School of Stomatology, Henan University, Kaifeng, China
| | - En-She Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institutes of Nursing and Health, School of Nursing and Health, Henan University, Kaifeng, China
| | - Meng Zheng
- International Joint Center for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China
| | - Shao-Feng Duan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Jian-She Wei
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Brain Research Laboratory, School of Life Sciences, Henan University, Kaifeng, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- School of Stomatology, Henan University, Kaifeng, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
10
|
Microglial Heterogeneity and Its Potential Role in Driving Phenotypic Diversity of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22052780. [PMID: 33803478 PMCID: PMC7967159 DOI: 10.3390/ijms22052780] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer’s disease (AD) is increasingly recognized as a highly heterogeneous disorder occurring under distinct clinical and neuropathological phenotypes. Despite the molecular determinants of such variability not being well defined yet, microglial cells may play a key role in this process by releasing distinct pro- and/or anti-inflammatory cytokines, potentially affecting the expression of the disease. We carried out a neuropathological and biochemical analysis on a series of AD brain samples, gathering evidence about the heterogeneous involvement of microglia in AD. The neuropathological studies showed differences concerning morphology, density and distribution of microglial cells among AD brains. Biochemical investigations showed increased brain levels of IL-4, IL-6, IL-13, CCL17, MMP-7 and CXCL13 in AD in comparison with control subjects. The molecular profiling achieved by measuring the brain levels of 25 inflammatory factors known to be involved in neuroinflammation allowed a stratification of the AD patients in three distinct “neuroinflammatory clusters”. These findings strengthen the relevance of neuroinflammation in AD pathogenesis suggesting, in particular, that the differential involvement of neuroinflammatory molecules released by microglial cells during the development of the disease may contribute to modulate the characteristics and the severity of the neuropathological changes, driving—at least in part—the AD phenotypic diversity.
Collapse
|
11
|
Kim SJ, Li J, Mahairaki V. Stem cell-derived three-dimensional (organoid) models of Alzheimer's disease: a precision medicine approach. Neural Regen Res 2021; 16:1546-1547. [PMID: 33433475 PMCID: PMC8323699 DOI: 10.4103/1673-5374.303019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Affiliation(s)
- Sujung Jun Kim
- Department of Neurology; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiaxin Li
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Vasiliki Mahairaki
- Department of Neurology, Johns Hopkins School of Medicine; The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| |
Collapse
|
12
|
Zhao Y, Jaber V, Alexandrov PN, Vergallo A, Lista S, Hampel H, Lukiw WJ. microRNA-Based Biomarkers in Alzheimer's Disease (AD). Front Neurosci 2020; 14:585432. [PMID: 33192270 PMCID: PMC7664832 DOI: 10.3389/fnins.2020.585432] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is a multifactorial, age-related neurological disease characterized by complex pathophysiological dynamics taking place at multiple biological levels, including molecular, genetic, epigenetic, cellular and large-scale brain networks. These alterations account for multiple pathophysiological mechanisms such as brain protein accumulation, neuroinflammatory/neuro-immune processes, synaptic dysfunction, and neurodegeneration that eventually lead to cognitive and behavioral decline. Alterations in microRNA (miRNA) signaling have been implicated in the epigenetics and molecular genetics of all neurobiological processes associated with AD pathophysiology. These changes encompass altered miRNA abundance, speciation and complexity in anatomical regions of the CNS targeted by the disease, including modified miRNA expression patterns in brain tissues, the systemic circulation, the extracellular fluid (ECF) and the cerebrospinal fluid (CSF). miRNAs have been investigated as candidate biomarkers for AD diagnosis, disease prediction, prognosis and therapeutic purposes because of their involvement in multiple brain signaling pathways in both health and disease. In this review we will: (i) highlight the significantly heterogeneous nature of miRNA expression and complexity in AD tissues and biofluids; (ii) address how information may be extracted from these data to be used as a diagnostic, prognostic and/or screening tools across the entire continuum of AD, from the preclinical stage, through the prodromal, i.e., mild cognitive impairment (MCI) phase all the way to clinically overt dementia; and (iii) consider how specific miRNA expression patterns could be categorized using miRNA reporters that span AD pathophysiological initiation and disease progression.
Collapse
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Department of Cell Biology and Anatomy, Louisiana State University Health Science Center, New Orleans, LA, United States
| | - Vivian Jaber
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | | | - Andrea Vergallo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Simone Lista
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’Hôpital, Paris, France
- Institute of Memory and Alzheimer’s Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l’hôpital, Paris, France
| | - Harald Hampel
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Russian Academy of Medical Sciences, Moscow, Russia
- Department of Ophthalmology, LSU Neuroscience Center Louisiana State University Health Science Center, New Orleans, LA, United States
- Department of Neurology, LSU Neuroscience Center Louisiana State University Health Science Center, New Orleans, LA, United States
| |
Collapse
|
13
|
Barbagallo C, Di Martino MT, Grasso M, Salluzzo MG, Scionti F, Cosentino FII, Caruso G, Barbagallo D, Di Pietro C, Ferri R, Caraci F, Purrello M, Ragusa M. Uncharacterized RNAs in Plasma of Alzheimer's Patients Are Associated with Cognitive Impairment and Show a Potential Diagnostic Power. Int J Mol Sci 2020; 21:ijms21207644. [PMID: 33076555 PMCID: PMC7588983 DOI: 10.3390/ijms21207644] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) diagnosis is actually based on clinical evaluation and brain-imaging tests, and it can often be confirmed only post-mortem. Therefore, new non-invasive molecular biomarkers are necessary to improve AD diagnosis. As circulating microRNA biomarkers have been proposed for many diseases, including AD, we aimed to identify new diagnostic non-small RNAs in AD. Whole transcriptome analysis was performed on plasma samples of five AD and five unaffected individuals (CTRL) using the Clariom D Pico Assay, followed by validation in real-time PCR on 37 AD patients and 37 CTRL. Six differentially expressed (DE) transcripts were identified: GS1-304P7.3 (upregulated), NONHSAT090268, TC0100011037, TC0400008478, TC1400008125, and UBE2V1 (downregulated). Peripheral blood mononuclear cells (PBMCs) may influence the expression of circulating RNAs and their analysis has been proposed to improve AD clinical management. Accordingly, DE transcript expression was also evaluated in PBMCs, showing no difference between AD and CTRL. ROC (receiver operating characteristic) curve analysis was performed to evaluate the diagnostic accuracy of each DE transcript and a signature including all of them. A correlation between cognitive impairment and GS1-304P7.3, NONHSAT090268, TC0100011037, and TC0400008478 was detected, suggesting a potential association between their extracellular abundance and AD clinical phenotype. Finally, this study identified six transcripts showing altered expression in the plasma of AD patients. Given the need for new, accurate blood biomarkers for AD diagnosis, these transcripts may be considered for further analyses in larger cohorts, also in combination with other biomarkers, aiming to identify specific RNA-based biomarkers to be eventually applied to clinical practice.
Collapse
Affiliation(s)
- Cristina Barbagallo
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of Catania, 95123 Catania, Italy; (C.B.); (D.B.); (C.D.P.); (M.P.); (M.R.)
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.D.M.); (F.S.)
| | - Margherita Grasso
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
| | - Maria Grazia Salluzzo
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
| | - Francesca Scionti
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.D.M.); (F.S.)
| | | | - Giuseppe Caruso
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
| | - Davide Barbagallo
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of Catania, 95123 Catania, Italy; (C.B.); (D.B.); (C.D.P.); (M.P.); (M.R.)
| | - Cinzia Di Pietro
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of Catania, 95123 Catania, Italy; (C.B.); (D.B.); (C.D.P.); (M.P.); (M.R.)
| | - Raffaele Ferri
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
| | - Filippo Caraci
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
- Correspondence:
| | - Michele Purrello
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of Catania, 95123 Catania, Italy; (C.B.); (D.B.); (C.D.P.); (M.P.); (M.R.)
| | - Marco Ragusa
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of Catania, 95123 Catania, Italy; (C.B.); (D.B.); (C.D.P.); (M.P.); (M.R.)
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
| |
Collapse
|