1
|
Bian X, Zhao Z, Gao X. Quantifying the association between stroke and dementia: a bibliometric study. Front Neurol 2024; 15:1438699. [PMID: 39440254 PMCID: PMC11493744 DOI: 10.3389/fneur.2024.1438699] [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: 05/26/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Background Stroke and dementia are two serious neurological disorders in modern medicine. Studies have revealed a significant link between the two, but there is still a lack of bibliometric analysis in this area. The objective of this study is to use bibliometric analysis to investigate the connection between stroke and dementia, as well as to assess the current state of research in this field and identify future trends. Methods The publications from the Web of Science were Collection and retrieved for the last 22 years (2002-2023). CiteSpace, VOSviewer, and the R package Bibliometrix were used to conduct bibliometric analysis. GraphPad Prism was used to plot. Results A total of 1,309 publications were included in the analysis. The number of articles on dementia and stroke has continued to grow steadily over the past 22 years. While China is the country with the most articles, the most influential and widely researched countries are England and the United States. The keyword analysis illustrates that the prevention of dementia through stroke prevention is a major focus and trend in this research area. Conclusion This study provides a visual analysis method for measuring the association between stroke and dementia, and examines the current state of research in this area and future research trends. In the future, dementia caused by stroke needs to be emphasized, and prevention of dementia through stroke prevention is a research priority.
Collapse
Affiliation(s)
- Xinyi Bian
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zibin Zhao
- The First School of Clinical Medicine, Bengbu Medical University, Bengbu, China
| | - Xiaoping Gao
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Uwishema O, Kassahun Bekele B, Nazir A, Filbert Luta E, Abdulnaser Al-Saab E, Jacques Desire I, Franklin Ozioma C, Wojtara M. Breaking barriers: addressing inequities in Alzheimer's disease diagnosis and treatment in Africa. Ann Med Surg (Lond) 2024; 86:5299-5303. [PMID: 39239000 PMCID: PMC11374311 DOI: 10.1097/ms9.0000000000002344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/24/2024] [Indexed: 09/07/2024] Open
Abstract
Introduction Alzheimer's disease represents a substantial and escalating public health threat across Africa. Alzheimer's disease leads to substantial cognitive impairment and memory loss, placing a heavy burden on the affected individuals and their families, friends, and caregivers. It affects 2.67 million people in Africa, the majority of whom live in sub-Saharan Africa. The prevalence of this disease is expected to rise drastically to approximately 150 million individuals worldwide by 2050, as estimated by the WHO. Aim This paper offers an integrative profile of Alzheimer's disease in Africa, spanning known genetic and modifiable risks, discusses the existing challenges in diagnosis and treatment, projections on prevalence and disability-adjusted life year burden through 2050, and priority policy responses needed to rebalance the equation. Methods This paper examines available literature to summarize current knowledge on risk factors, diagnosis, treatments, and burden of Alzheimer's disease in Africa. Gather epidemiological assessments, clinical guidelines, and commentary related to Alzheimer's disease in Africa. Results The data reveals concerning realities regarding Alzheimer's disease diagnosis and care in Africa. Diagnostic infrastructure shortcomings, resource limitations, and knowledge gaps emerge as recurring barriers. Positron emission tomography scans, cerebrospinal fluid assays, and other mainstay detection modalities common in developed countries show restricted availability. Conclusion Addressing Africa's Alzheimer's disease crisis demands a multipronged strategy to uplift diagnostic capacities, treatment availability, specialist training, public awareness, and coordinated policymaking. Prioritizing biomarkers and imaging to confirm early neurodegeneration is foundational, alongside drug access expansion.
Collapse
Affiliation(s)
| | - Bezawit Kassahun Bekele
- Oli Health Magazine Organization, Research and Education
- Department of medicine, University of Nigeria, Nsukka, Nigeria
- Addis Ababa University, School of Medicine, Addis Ababa, Ethiopia
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Erick Filbert Luta
- Oli Health Magazine Organization, Research and Education
- Department of medicine, Muhimbili University of Health and Allied Sciences (MUHAS), Dar es Salaam, Tanzania
- Department of medicine, Tanzania Medical Students Association (TAMSA)
| | - Elaf Abdulnaser Al-Saab
- Oli Health Magazine Organization, Research and Education
- Department of medicine, Al Iraqia University School of Medicine, Baghdad, Iraq
| | - Irakiza Jacques Desire
- Oli Health Magazine Organization, Research and Education
- Department of medicine, University of Rwanda, Kigali, Rwanda
| | - Chukwuma Franklin Ozioma
- Oli Health Magazine Organization, Research and Education
- Department of medicine, University of Nigeria, Nsukka, Nigeria
| | - Magda Wojtara
- Oli Health Magazine Organization, Research and Education
| |
Collapse
|
3
|
Momen YS, Mishra J, Kumar N. Brain-Gut and Microbiota-Gut-Brain Communication in Type-2 Diabetes Linked Alzheimer's Disease. Nutrients 2024; 16:2558. [PMID: 39125436 PMCID: PMC11313915 DOI: 10.3390/nu16152558] [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: 05/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 08/12/2024] Open
Abstract
The gastrointestinal (GI) tract, home to the largest microbial population in the human body, plays a crucial role in overall health through various mechanisms. Recent advancements in research have revealed the potential implications of gut-brain and vice-versa communication mediated by gut-microbiota and their microbial products in various diseases including type-2 diabetes and Alzheimer's disease (AD). AD is the most common type of dementia where most of cases are sporadic with no clearly identified cause. However, multiple factors are implicated in the progression of sporadic AD which can be classified as non-modifiable (e.g., genetic) and modifiable (e.g. Type-2 diabetes, diet etc.). Present review focusses on key players particularly the modifiable factors such as Type-2 diabetes (T2D) and diet and their implications in microbiota-gut-brain (MGB) and brain-gut (BG) communication and cognitive functions of healthy brain and their dysfunction in Alzheimer's Disease. Special emphasis has been given on elucidation of the mechanistic aspects of the impact of diet on gut-microbiota and the implications of some of the gut-microbial products in T2D and AD pathology. For example, mechanistically, HFD induces gut dysbiosis with driven metabolites that in turn cause loss of integrity of intestinal barrier with concomitant colonic and systemic chronic low-grade inflammation, associated with obesity and T2D. HFD-induced obesity and T2D parallel neuroinflammation, deposition of Amyloid β (Aβ), and ultimately cognitive impairment. The review also provides a new perspective of the impact of diet on brain-gut and microbiota-gut-brain communication in terms of transcription factors as a commonly spoken language that may facilitates the interaction between gut and brain of obese diabetic patients who are at a higher risk of developing cognitive impairment and AD. Other commonality such as tyrosine kinase expression and functions maintaining intestinal integrity on one hand and the phagocytic clarence by migratory microglial functions in brain are also discussed. Lastly, the characterization of the key players future research that might shed lights on novel potential pharmacological target to impede AD progression are also discussed.
Collapse
Affiliation(s)
| | | | - Narendra Kumar
- Department of Pharmaceutical Sciences, ILR College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, USA
| |
Collapse
|
4
|
Micocci S, Stefania R, Garello F, Fasoglio U, Hawala I, Tei L, Geninatti Crich S, Digilio G. Synthesis of fluorinated curcumin derivatives for detecting amyloid plaques by 19F-MRI. Org Biomol Chem 2024; 22:5948-5959. [PMID: 38979663 DOI: 10.1039/d4ob00730a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The most prominent pathophysiological hallmark of Alzheimer's disease is the aggregation of amyloid-β (Aβ) peptides into senile plaques. Curcumin and its derivatives exhibit a high affinity for binding to Aβ fibrils, effectively inhibiting their growth. This property holds promise for both therapeutic applications and diagnostic molecular imaging. In this study, curcumin was functionalized with perfluoro-tert-butyl groups to create candidate molecular probes specifically targeted to Aβ fibrils for use in 19F-magnetic resonance imaging. Two types of fluorinated derivatives were considered: mono-substituted (containing nine fluorine atoms per molecule) and disubstituted (containing eighteen fluorine atoms). The linker connecting the perfluoro moiety with the curcumin scaffold was evaluated for its impact on binding affinity and water solubility. All mono-substituted compounds and one disubstituted compound exhibited a binding affinity toward Aβ fibrils on the same order of magnitude as reference curcumin. The insertion of a charged carboxylate group into the linker enhanced the water solubility of the probes. Compound Curc-Glu-F9 (with one L-glutamyl moiety and a perfluoro-tert-butyl group), showed the best properties in terms of binding affinity towards Aβ fibrils, water solubility, and intensity of the 19F-NMR signal in the Aβ oligomer bound form.
Collapse
Affiliation(s)
- Sebastiano Micocci
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Torino, Italy
| | - Rachele Stefania
- Department of Science and Technological Innovation, University of Eastern Piedmont "Amedeo Avogadro", Viale Teresa Michel 11, 15120, Alessandria, Italy.
| | - Francesca Garello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Torino, Italy
| | - Umberto Fasoglio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Torino, Italy
| | - Ivan Hawala
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Torino, Italy
| | - Lorenzo Tei
- Department of Science and Technological Innovation, University of Eastern Piedmont "Amedeo Avogadro", Viale Teresa Michel 11, 15120, Alessandria, Italy.
| | - Simonetta Geninatti Crich
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Torino, Italy
| | - Giuseppe Digilio
- Department of Science and Technological Innovation, University of Eastern Piedmont "Amedeo Avogadro", Viale Teresa Michel 11, 15120, Alessandria, Italy.
| |
Collapse
|
5
|
Yao YG, Lu L, Ni RJ, Bi R, Chen C, Chen JQ, Fuchs E, Gorbatyuk M, Lei H, Li H, Liu C, Lv LB, Tsukiyama-Kohara K, Kohara M, Perez-Cruz C, Rainer G, Shan BC, Shen F, Tang AZ, Wang J, Xia W, Xia X, Xu L, Yu D, Zhang F, Zheng P, Zheng YT, Zhou J, Zhou JN. Study of tree shrew biology and models: A booming and prosperous field for biomedical research. Zool Res 2024; 45:877-909. [PMID: 39004865 PMCID: PMC11298672 DOI: 10.24272/j.issn.2095-8137.2024.199] [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: 06/17/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The tree shrew ( Tupaia belangeri) has long been proposed as a suitable alternative to non-human primates (NHPs) in biomedical and laboratory research due to its close evolutionary relationship with primates. In recent years, significant advances have facilitated tree shrew studies, including the determination of the tree shrew genome, genetic manipulation using spermatogonial stem cells, viral vector-mediated gene delivery, and mapping of the tree shrew brain atlas. However, the limited availability of tree shrews globally remains a substantial challenge in the field. Additionally, determining the key questions best answered using tree shrews constitutes another difficulty. Tree shrew models have historically been used to study hepatitis B virus (HBV) and hepatitis C virus (HCV) infection, myopia, and psychosocial stress-induced depression, with more recent studies focusing on developing animal models for infectious and neurodegenerative diseases. Despite these efforts, the impact of tree shrew models has not yet matched that of rodent or NHP models in biomedical research. This review summarizes the prominent advancements in tree shrew research and reflects on the key biological questions addressed using this model. We emphasize that intensive dedication and robust international collaboration are essential for achieving breakthroughs in tree shrew studies. The use of tree shrews as a unique resource is expected to gain considerable attention with the application of advanced techniques and the development of viable animal models, meeting the increasing demands of life science and biomedical research.
Collapse
Affiliation(s)
- Yong-Gang Yao
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China. E-mail:
| | - Li Lu
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Rong-Jun Ni
- Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, Sichuan 610044, China
| | - Rui Bi
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ceshi Chen
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Jia-Qi Chen
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Eberhard Fuchs
- German Primate Center, Leibniz Institute of Primate Research, Göttingen 37077, Germany
| | - Marina Gorbatyuk
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hao Lei
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Hongli Li
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Chunyu Liu
- Soong Ching Ling Institute of Maternity and Child Health, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Long-Bao Lv
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima-city, Kagoshima 890-8580, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | | | - Gregor Rainer
- Department of Medicine, University of Fribourg, Fribourg CH-1700, Switzerland
| | - Bao-Ci Shan
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fang Shen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China
| | - An-Zhou Tang
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi 530000, China
| | - Jing Wang
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wei Xia
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi 530000, China
| | - Xueshan Xia
- School of Public Health, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Ling Xu
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Dandan Yu
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Feng Zhang
- Soong Ching Ling Institute of Maternity and Child Health, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ping Zheng
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Yong-Tang Zheng
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Jumin Zhou
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Jiang-Ning Zhou
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- Institute of Brain Science, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| |
Collapse
|
6
|
Guan XJ, Deng ZQ, Liu J, Su CF, Tong BCK, Zhu Z, Sreenivasmurthy SG, Kan YX, Lu KJ, Chu CPK, Pi RB, Cheung KH, Iyaswamy A, Song JX, Li M. Corynoxine promotes TFEB/TFE3-mediated autophagy and alleviates Aβ pathology in Alzheimer's disease models. Acta Pharmacol Sin 2024; 45:900-913. [PMID: 38225393 PMCID: PMC11053156 DOI: 10.1038/s41401-023-01197-1] [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: 05/01/2023] [Accepted: 11/09/2023] [Indexed: 01/17/2024] Open
Abstract
Autophagy impairment is a key factor in Alzheimer's disease (AD) pathogenesis. TFEB (transcription factor EB) and TFE3 (transcription factor binding to IGHM enhancer 3) are nuclear transcription factors that regulate autophagy and lysosomal biogenesis. We previously showed that corynoxine (Cory), a Chinese medicine compound, protects neurons from Parkinson's disease (PD) by activating autophagy. In this study, we investigated the effect of Cory on AD models in vivo and in vitro. We found that Cory improved learning and memory function, increased neuronal autophagy and lysosomal biogenesis, and reduced pathogenic APP-CTFs levels in 5xFAD mice model. Cory activated TFEB/TFE3 by inhibiting AKT/mTOR signaling and stimulating lysosomal calcium release via transient receptor potential mucolipin 1 (TRPML1). Moreover, we demonstrated that TFEB/TFE3 knockdown abolished Cory-induced APP-CTFs degradation in N2aSwedAPP cells. Our findings suggest that Cory promotes TFEB/TFE3-mediated autophagy and alleviates Aβ pathology in AD models.
Collapse
Affiliation(s)
- Xin-Jie Guan
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Zhi-Qiang Deng
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Jia Liu
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Cheng-Fu Su
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Benjamin Chun-Kit Tong
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zhou Zhu
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Sravan Gopalkrishnashetty Sreenivasmurthy
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yu-Xuan Kan
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Ke-Jia Lu
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Carol Pui-Kei Chu
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Rong-Biao Pi
- School of Medicine, Sun Yat-sen University (Shenzhen), Shenzhen, 518107, China
| | - King-Ho Cheung
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Ashok Iyaswamy
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China.
| | - Ju-Xian Song
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Min Li
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China.
| |
Collapse
|
7
|
Zhao Y, Wang D. Bibliometric Insights Into the Evolution of Atrial Fibrillation and Dementia Research 2002-2022. Med Sci Monit 2024; 30:e943239. [PMID: 38504433 PMCID: PMC10936108 DOI: 10.12659/msm.943239] [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: 11/19/2023] [Accepted: 01/01/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND In response to the escalating prevalence of atrial fibrillation (AF) and its potential correlation with cognitive impairment and dementia, we conducted a comprehensive bibliometric analysis to assess current research trends, contributors, and collaborative networks in this evolving interdisciplinary field. MATERIAL AND METHODS Articles published between 2002 and 2022 were extracted from the Web of Science database and carefully screened, yielding 328 publications. Two reviewers independently conducted the screening and quality appraisal. We employed sophisticated tools such as CiteSpace, VOSviewer, and Bibliometrix (R-Studio's R tool) to succinctly summarize and thoroughly analyze the publications. RESULTS A total of 328 publications, comprising 262 papers and 66 reviews, were included in the final analysis. The number of publications exhibited a consistent year-on-year increase, demonstrating an average annual growth rate of 20.57%. These publications originated from 41 countries and regions, with the highest contributions observed from the United States, United Kingdom, Italy, and China. Notably, the University of Liverpool emerged as the most prolific institution, while the most prolific author was Lip GYH from the United Kingdom. The journal with the most publications is the journal of the American Heart Association (19 articles). The most popular keywords in order were: risk and stroke (n=101), dementia (n=100), decline (n=70), prevalence (n=67), and Alzheimer's disease. CONCLUSIONS This study shows the current research status and emerging trends in atrial fibrillation's link to dementia and cognitive impairment. It highlights global growth and collaboration patterns while offering a comprehensive view of their interrelationship, pointing toward future research directions.
Collapse
|
8
|
Wrzesień A, Andrzejewski K, Jampolska M, Kaczyńska K. Respiratory Dysfunction in Alzheimer's Disease-Consequence or Underlying Cause? Applying Animal Models to the Study of Respiratory Malfunctions. Int J Mol Sci 2024; 25:2327. [PMID: 38397004 PMCID: PMC10888758 DOI: 10.3390/ijms25042327] [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: 01/17/2024] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative brain disease that is the most common cause of dementia among the elderly. In addition to dementia, which is the loss of cognitive function, including thinking, remembering, and reasoning, and behavioral abilities, AD patients also experience respiratory disturbances. The most common respiratory problems observed in AD patients are pneumonia, shortness of breath, respiratory muscle weakness, and obstructive sleep apnea (OSA). The latter is considered an outcome of Alzheimer's disease and is suggested to be a causative factor. While this narrative review addresses the bidirectional relationship between obstructive sleep apnea and Alzheimer's disease and reports on existing studies describing the most common respiratory disorders found in patients with Alzheimer's disease, its main purpose is to review all currently available studies using animal models of Alzheimer's disease to study respiratory impairments. These studies on animal models of AD are few in number but are crucial for establishing mechanisms, causation, implementing potential therapies for respiratory disorders, and ultimately applying these findings to clinical practice. This review summarizes what is already known in the context of research on respiratory disorders in animal models, while pointing out directions for future research.
Collapse
Affiliation(s)
| | | | | | - Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.W.); (K.A.); (M.J.)
| |
Collapse
|
9
|
Sharma A, Ray A, Sathaye S, Singhal RS. A supercritical fluid co-extract of turmeric powder and dried coconut shreds shows neuroprotection against AlCl 3-induced Alzheimer's disease in rats through nose to brain delivery. Bioorg Chem 2024; 143:107046. [PMID: 38141332 DOI: 10.1016/j.bioorg.2023.107046] [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: 06/27/2023] [Revised: 12/06/2023] [Accepted: 12/16/2023] [Indexed: 12/25/2023]
Abstract
This study was aimed at investigating the neuroprotective potential of a co-extract obtained by supercritical fluid extraction (SFE) of turmeric powder and dried coconut shreds against aluminium chloride (AlCl3)-induced Alzheimer's disease (AD) in male Wistar rats. Fifty animals were allocated to five groups, which received saline (vehicle control, group 1), a combination of saline and aluminium chloride (AlCl3) (disease control, group 2), coconut oil (COO) (SFE extracted, treatment group 3), turmeric oleoresin (Cur) (SFE extracted, treatment group 4) and SFE co-extract of turmeric powder and coconut shreds (CurCOO) (treatment group 5). Animals were subjected to behavioural evaluation. In addition, the hippocampal section of the brain from all groups was subjected to biochemical, molecular and histopathological evaluations. The results showed CurCOO administered intranasally improved cognitive abilities, reversed histological alterations in the brain, reduced hippocampus inflammation studied through proinflammatory cytokine markers like TNF-α and IL-6 as compared to the disease control group. The impact of CurCOO on preventive neurodegeneration was also observed through a reduction in protein transcription factor NF-kB in the treated group 5 as compared to a disease control group. The effect of intranasal delivery of CurCOO on the neurons responsible for memory consolidation was evident from low acetylcholinesterase (AChE) enzyme activity in the treated groups with respect to AlCl3 induced group. Summarily, the results demonstrated intranasal delivery of CurCOO to show better efficacy than Cur and COO in preventing neurodegeneration associated with AlCl3 induced Alzheimer's disease.
Collapse
Affiliation(s)
- Abhinav Sharma
- Department of Food Engineering & Technology, Institute of Chemical Technology, Mumbai 400019, India
| | - Aratrika Ray
- Department of Food Engineering & Technology, Institute of Chemical Technology, Mumbai 400019, India
| | - Sadhana Sathaye
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India
| | - Rekha S Singhal
- Department of Food Engineering & Technology, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
10
|
Li M, Sun H, Hou Z, Hao S, Jin L, Wang B. Engineering the Physical Microenvironment into Neural Organoids for Neurogenesis and Neurodevelopment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306451. [PMID: 37771182 DOI: 10.1002/smll.202306451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/04/2023] [Indexed: 09/30/2023]
Abstract
Understanding the signals from the physical microenvironment is critical for deciphering the processes of neurogenesis and neurodevelopment. The discovery of how surrounding physical signals shape human developing neurons is hindered by the bottleneck of conventional cell culture and animal models. Notwithstanding neural organoids provide a promising platform for recapitulating human neurogenesis and neurodevelopment, building neuronal physical microenvironment that accurately mimics the native neurophysical features is largely ignored in current organoid technologies. Here, it is discussed how the physical microenvironment modulates critical events during the periods of neurogenesis and neurodevelopment, such as neural stem cell fates, neural tube closure, neuronal migration, axonal guidance, optic cup formation, and cortical folding. Although animal models are widely used to investigate the impacts of physical factors on neurodevelopment and neuropathy, the important roles of human stem cell-derived neural organoids in this field are particularly highlighted. Considering the great promise of human organoids, building neural organoid microenvironments with mechanical forces, electrophysiological microsystems, and light manipulation will help to fully understand the physical cues in neurodevelopmental processes. Neural organoids combined with cutting-edge techniques, such as advanced atomic force microscopes, microrobots, and structural color biomaterials might promote the development of neural organoid-based research and neuroscience.
Collapse
Affiliation(s)
- Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Heng Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| | - Zongkun Hou
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| |
Collapse
|
11
|
Jiang Z, Wang J, Qin Y, Liu S, Luo B, Bai F, Wei H, Zhang S, Wei J, Ding G, Ma L, He S, Chen R, Sun Y, Chen Y, Wang L, Xu H, Wang X, Chen G, Lei W. A nonhuman primate model with Alzheimer's disease-like pathology induced by hippocampal overexpression of human tau. Alzheimers Res Ther 2024; 16:22. [PMID: 38281031 PMCID: PMC10821564 DOI: 10.1186/s13195-024-01392-0] [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: 10/17/2023] [Accepted: 01/15/2024] [Indexed: 01/29/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is one of the most burdening diseases of the century with no disease-modifying treatment at this time. Nonhuman primates (NHPs) share genetic, anatomical, and physiological similarities with humans, making them ideal model animals for investigating the pathogenesis of AD and potential therapies. However, the use of NHPs in AD research has been hindered by the paucity of AD monkey models due to their long generation time, ethical considerations, and technical challenges in genetically modifying monkeys. METHODS Here, we developed an AD-like NHP model by overexpressing human tau in the bilateral hippocampi of adult rhesus macaque monkeys. We evaluated the pathological features of these monkeys with immunostaining, Nissl staining, cerebrospinal fluid (CSF) analysis, magnetic resonance imaging (MRI), positron emission tomography (PET), and behavioural tests. RESULTS We demonstrated that after hippocampal overexpression of tau protein, these monkeys displayed multiple pathological features of AD, including 3-repeat (3R)/4-repeat (4R) tau accumulation, tau hyperphosphorylation, tau propagation, neuronal loss, hippocampal atrophy, neuroinflammation, Aβ clearance deficits, blood vessel damage, and cognitive decline. More interestingly, the accumulation of both 3R and 4R tau is specific to NHPs but not found in adult rodents. CONCLUSIONS This work establishes a tau-induced AD-like NHP model with many key pathological and behavioural features of AD. In addition, our model may potentially become one of the AD NHP models adopted by researchers worldwide since it can be generated within 2 ~ 3 months through a single injection of AAVs into the monkey brains. Hence, our model NHPs may facilitate mechanistic studies and therapeutic treatments for AD.
Collapse
Affiliation(s)
- Zhouquan Jiang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Jing Wang
- Department of Neurosurgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Yongpeng Qin
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Shanggong Liu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Bin Luo
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Fan Bai
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Huiyi Wei
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Shaojuan Zhang
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Junjie Wei
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Guoyu Ding
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Long Ma
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Shu He
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Rongjie Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Ying Sun
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Yi Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Lu Wang
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Hao Xu
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Xiangyu Wang
- Department of Neurosurgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Gong Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China.
| | - Wenliang Lei
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
12
|
Plantone D, Pardini M, Caneva S, De Stefano N. Is There a Role of Vitamin D in Alzheimer's Disease? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:545-553. [PMID: 37246320 DOI: 10.2174/1871527322666230526164421] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/05/2023] [Accepted: 04/19/2023] [Indexed: 05/30/2023]
Abstract
Alzheimer's disease (AD) represents the most prevalent type of neurodegenerative dementia and the sixth leading cause of death worldwide. The so-called "non-calcemic actions" of vitamin D have been increasingly described, and its insufficiency has already been linked to the onset and progression of the main neurological diseases, including AD. Immune-mediated Aβ plaque's phagocytosis and clearance, immune response, oxidative stress, and mitochondrial function are all influenced by vitamin D, and these functions are considered relevant in AD pathogenesis. However, it has been shown that the genomic vitamin D signaling pathway is already impaired in the AD brain, making things more complicated. In this paper, we aim to summarise the role of vitamin D in AD and review the results of the supplementation trials in AD patients.
Collapse
Affiliation(s)
- Domenico Plantone
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Stefano Caneva
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
13
|
Adiga D, Eswaran S, Srinath S, Khan NG, Kumar D, Kabekkodu SP. Noncoding RNAs in Alzheimer's Disease: Overview of Functional and Therapeutic Significance. Curr Top Med Chem 2024; 24:1615-1634. [PMID: 38616763 DOI: 10.2174/0115680266293212240405042540] [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: 11/16/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 04/16/2024]
Abstract
Alzheimer's disease (AD) is a multifactorial disorder resulting from the complex interaction between genetic, epigenetic, and environmental factors. It represents an impending epidemic and lacks effective pharmacological interventions. The emergence of high throughput sequencing techniques and comprehensive genome evaluation has uncovered a diverse spectrum of noncoding RNA (ncRNA) families. ncRNAs are the critical modulators of an eclectic array of biological processes and are now transpiring as imperative players in diagnosing and treating various diseases, including neurodegenerative disorders. Several ncRNAs are explicitly augmented in the brain, wherein they potentially regulate cognitive abilities and other functions of the central nervous system. Growing evidence suggests the substantial role of ncRNAs as modulators of tau phosphorylation, Aβ production, neuroinflammation, and neuronal survival. It indicates their therapeutic relevance as a biomarker and druggable targets against AD. The current review summarizes the existing literature on the functional significance of ncRNAs in AD pathogenesis and its imminent implications in clinics.
Collapse
Affiliation(s)
- Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
| | - Sangavi Eswaran
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
| | - Sriharikrishnaa Srinath
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
| | - Nadeem G Khan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Erandwane, Pune, 411038, Maharashtra, India
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, One Shields Avenue, Davis, CA95616, USA
| | - Shama P Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
| |
Collapse
|
14
|
Dighe S, Jog S, Momin M, Sawarkar S, Omri A. Intranasal Drug Delivery by Nanotechnology: Advances in and Challenges for Alzheimer's Disease Management. Pharmaceutics 2023; 16:58. [PMID: 38258068 PMCID: PMC10820353 DOI: 10.3390/pharmaceutics16010058] [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: 09/18/2023] [Revised: 10/11/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
Alzheimer's disease, a progressive neurodegenerative condition, is characterized by a gradual decline in cognitive functions. Current treatment approaches primarily involve the administration of medications through oral, parenteral, and transdermal routes, aiming to improve cognitive function and alleviate symptoms. However, these treatments face limitations, such as low bioavailability and inadequate permeation. Alternative invasive methods, while explored, often entail discomfort and require specialized assistance. Therefore, the development of a non-invasive and efficient delivery system is crucial. Intranasal delivery has emerged as a potential solution, although it is constrained by the unique conditions of the nasal cavity. An innovative approach involves the use of nano-carriers based on nanotechnology for intranasal delivery. This strategy has the potential to overcome current limitations by providing enhanced bioavailability, improved permeation, effective traversal of the blood-brain barrier, extended retention within the body, and precise targeting of the brain. The comprehensive review focuses on the advancements in designing various types of nano-carriers, including polymeric nanoparticles, metal nanoparticles, lipid nanoparticles, liposomes, nanoemulsions, Quantum dots, and dendrimers. These nano-carriers are specifically tailored for the intranasal delivery of therapeutic agents aimed at combatting Alzheimer's disease. In summary, the development and utilization of intranasal delivery systems based on nanotechnology show significant potential in surmounting the constraints of current Alzheimer's disease treatment strategies. Nevertheless, it is essential to acknowledge regulatory as well as toxicity concerns associated with this route; meticulous consideration is required when engineering a carrier. This comprehensive review underscores the potential to revolutionize Alzheimer's disease management and highlights the importance of addressing regulatory considerations for safe and effective implementations. Embracing this strategy could lead to substantial advancements in the field of Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Sayali Dighe
- Department of Pharmaceutics, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai 400056, India
| | - Sunil Jog
- Department of Pharmaceutics, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai 400056, India
- Indoco Remedies Private Limited, Mumbai 400098, India
| | - Munira Momin
- Department of Pharmaceutics, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai 400056, India
| | - Sujata Sawarkar
- Department of Pharmaceutics, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai 400056, India
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
15
|
Adiga D, Eswaran S, Sriharikrishnaa S, Khan NG, Prasada Kabekkodu S, Kumar D. Epigenetics of Alzheimer’s Disease: Past, Present and Future. ENZYMATIC TARGETS FOR DRUG DISCOVERY AGAINST ALZHEIMER'S DISEASE 2023:27-72. [DOI: 10.2174/9789815136142123010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Alzheimer’s disease (AD) exemplifies a looming epidemic lacking effective
treatment and manifests with the accumulation of neurofibrillary tangles, amyloid-β
plaques, neuroinflammation, behavioral changes, and acute cognitive impairments. It is
a complex, multifactorial disorder that arises from the intricate interaction between
environment and genetic factors, restrained via epigenetic machinery. Though the
research progress has improved the understanding of clinical manifestations and
disease advancement, the causal mechanism of detrimental consequences remains
undefined. Despite the substantial improvement in recent diagnostic modalities, it is
challenging to distinguish AD from other forms of dementia. Accurate diagnosis is a
major glitch in AD as it banks on the symptoms and clinical criteria. Several studies are
underway in exploring novel and reliable biomarkers for AD. In this direction,
epigenetic alterations have transpired as key modulators in AD pathogenesis with the
impeding inferences for the management of this neurological disorder. The present
chapter aims to discuss the significance of epigenetic modifications reported in the
pathophysiology of AD such as DNA methylation, hydroxy-methylation, methylation
of mtDNA, histone modifications, and noncoding RNAs. Additionally, the chapter also
describes the possible therapeutic avenues that target epigenetic modifications in AD.
Collapse
Affiliation(s)
- Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy
of Higher Education (MAHE), Manipal – 576104, Karnataka, India
| | - Sangavi Eswaran
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy
of Higher Education (MAHE), Manipal – 576104, Karnataka, India
| | - S. Sriharikrishnaa
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy
of Higher Education (MAHE), Manipal – 576104, Karnataka, India
| | - Nadeem G. Khan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy
of Higher Education (MAHE), Manipal – 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy
of Higher Education (MAHE), Manipal – 576104, Karnataka, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth
(Deemed to be University), Erandwane, Pune – 411038, Maharashtra, India
| |
Collapse
|
16
|
Bodily TA, Ramanathan A, Wei S, Karkisaval A, Bhatt N, Jerez C, Haque MA, Ramil A, Heda P, Wang Y, Kumar S, Leite M, Li T, Zhao J, Lal R. In pursuit of degenerative brain disease diagnosis: Dementia biomarkers detected by DNA aptamer-attached portable graphene biosensor. Proc Natl Acad Sci U S A 2023; 120:e2311565120. [PMID: 37956285 PMCID: PMC10666025 DOI: 10.1073/pnas.2311565120] [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: 07/10/2023] [Accepted: 09/25/2023] [Indexed: 11/15/2023] Open
Abstract
Dementia is a brain disease which results in irreversible and progressive loss of cognition and motor activity. Despite global efforts, there is no simple and reliable diagnosis or treatment option. Current diagnosis involves indirect testing of commonly inaccessible biofluids and low-resolution brain imaging. We have developed a portable, wireless readout-based Graphene field-effect transistor (GFET) biosensor platform that can detect viruses, proteins, and small molecules with single-molecule sensitivity and specificity. We report the detection of three important amyloids, namely, Amyloid beta (Aβ), Tau (τ), and α-Synuclein (αS) using DNA aptamer nanoprobes. These amyloids were isolated, purified, and characterized from the autopsied brain tissues of Alzheimer's Disease (AD) and Parkinson's Disease (PD) patients. The limit of detection (LoD) of the sensor is 10 fM, 1-10 pM, 10-100 fM for Aβ, τ, and αS, respectively. Synthetic as well as autopsied brain-derived amyloids showed a statistically significant sensor response with respect to derived thresholds, confirming the ability to define diseased vs. nondiseased states. The detection of each amyloid was specific to their aptamers; Aβ, τ, and αS peptides when tested, respectively, with aptamers nonspecific to them showed statistically insignificant cross-reactivity. Thus, the aptamer-based GFET biosensor has high sensitivity and precision across a range of epidemiologically significant AD and PD variants. This portable diagnostic system would allow at-home and POC testing for neurodegenerative diseases globally.
Collapse
Affiliation(s)
| | - Anirudh Ramanathan
- Department of Bioengineering, University of California, San Diego, CA92093
| | - Shanhong Wei
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Abhijith Karkisaval
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, CA92093
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Disorders, Department of Neurology, University of Texas Medical Branch, Galveston, TX77555
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Disorders, Department of Neurology, University of Texas Medical Branch, Galveston, TX77555
| | - Md Anzarul Haque
- Mitchell Center for Neurodegenerative Disorders, Department of Neurology, University of Texas Medical Branch, Galveston, TX77555
| | - Armando Ramil
- Department of Bioengineering, University of California, San Diego, CA92093
| | - Prachi Heda
- Department of Bioengineering, University of California, San Diego, CA92093
| | - Yi Wang
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
| | - Sanjeev Kumar
- Department of Computer Science, University of Illinois Urbana-Champaign, Champaign, IL61820
| | - Mikayla Leite
- Department of Bioengineering, University of California, San Diego, CA92093
| | - Tie Li
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
| | - Ratnesh Lal
- Department of Bioengineering, University of California, San Diego, CA92093
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, CA92093
- Materials Science and Engineering Program, University of California, San Diego, CA92093
| |
Collapse
|
17
|
Rodriguez-Lopez A, Torres-Paniagua AM, Acero G, Díaz G, Gevorkian G. Increased TSPO expression, pyroglutamate-modified amyloid beta (AβN3(pE)) accumulation and transient clustering of microglia in the thalamus of Tg-SwDI mice. J Neuroimmunol 2023; 382:578150. [PMID: 37467699 DOI: 10.1016/j.jneuroim.2023.578150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023]
Abstract
Epidemiological studies showed that Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA) frequently co-occur; however, the precise mechanism is not well understood. A unique animal model (Tg-SwDI mice) was developed to investigate the early-onset and robust accumulation of both parenchymal and vascular Aβ in the brain. Tg-SwDI mice have been extensively used to study the mechanisms of cerebrovascular dysfunction, neuroinflammation, neurodegeneration, and cognitive decline observed in AD/CAA patients and to design biomarkers and therapeutic strategies. In the present study, we documented interesting new features in the thalamus of Tg-SwDI mice: 1) a sharp increase in the expression of ionized calcium-binding adapter molecule 1 (Iba-1) in microglia in 6-month-old animals; 2) microglia clustering at six months that disappeared in old animals; 3) N-truncated/modified AβN3(pE) peptide in 9-month-old female and 12-month-old male mice; 4) an age-dependent increase in translocator protein (TSPO) expression. These findings reinforce the versatility of this model for studying multiple pathological issues involved in AD and CAA.
Collapse
Affiliation(s)
- Adrian Rodriguez-Lopez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Apartado Postal 70228, Cuidad Universitaria, CDMX, CP 04510, Mexico
| | - Alicia M Torres-Paniagua
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Apartado Postal 70228, Cuidad Universitaria, CDMX, CP 04510, Mexico
| | - Gonzalo Acero
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Apartado Postal 70228, Cuidad Universitaria, CDMX, CP 04510, Mexico
| | - Georgina Díaz
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Apartado Postal 70228, Cuidad Universitaria, CDMX, CP 04510, Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Apartado Postal 70228, Cuidad Universitaria, CDMX, CP 04510, Mexico.
| |
Collapse
|
18
|
Piotrowski SL, Tucker A, Jacobson S. The elusive role of herpesviruses in Alzheimer's disease: current evidence and future directions. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:253-266. [PMID: 38013835 PMCID: PMC10474380 DOI: 10.1515/nipt-2023-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/26/2023] [Indexed: 11/29/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. While pathologic hallmarks, such as extracellular beta-amyloid plaques, are well-characterized in affected individuals, the pathogenesis that causes plaque formation and eventual cognitive decline is not well understood. A recent resurgence of the decades-old "infectious hypothesis" has garnered increased attention on the potential role that microbes may play in AD. In this theory, it is thought that pathogens such as viruses may act as seeds for beta-amyloid aggregation, ultimately leading to plaques. Interest in the infectious hypothesis has also spurred further investigation into additional characteristics of viral infection that may play a role in AD progression, such as neuroinflammation, latency, and viral DNA integration. While a flurry of research in this area has been recently published, with herpesviruses being of particular interest, the role of pathogens in AD remains controversial. In this review, the insights gained thus far into the possible role of herpesviruses in AD are summarized. The challenges and potential future directions of herpesvirus research in AD and dementia are also discussed.
Collapse
Affiliation(s)
- Stacey L. Piotrowski
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Comparative Biomedical Scientist Training Program, National Institutes of Health, Bethesda, MD, USA
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Allison Tucker
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Steven Jacobson
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
Rochín-Hernández LJ, Jiménez-Acosta MA, Ramírez-Reyes L, Figueroa-Corona MDP, Sánchez-González VJ, Orozco-Barajas M, Meraz-Ríos MA. The Proteome Profile of Olfactory Ecto-Mesenchymal Stem Cells-Derived from Patients with Familial Alzheimer's Disease Reveals New Insights for AD Study. Int J Mol Sci 2023; 24:12606. [PMID: 37628788 PMCID: PMC10454072 DOI: 10.3390/ijms241612606] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disease and the first cause of dementia worldwide, has no effective treatment, and its pathological mechanisms are not yet fully understood. We conducted this study to explore the proteomic differences associated with Familial Alzheimer's Disease (FAD) in olfactory ecto-mesenchymal stem cells (MSCs) derived from PSEN1 (A431E) mutation carriers compared with healthy donors paired by age and gender through two label-free liquid chromatography-mass spectrometry approaches. The first analysis compared carrier 1 (patient with symptoms, P1) and its control (healthy donor, C1), and the second compared carrier 2 (patient with pre-symptoms, P2) with its respective control cells (C2) to evaluate whether the protein alterations presented in the symptomatic carrier were also present in the pre-symptom stages. Finally, we analyzed the differentially expressed proteins (DEPs) for biological and functional enrichment. These proteins showed impaired expression in a stage-dependent manner and are involved in energy metabolism, vesicle transport, actin cytoskeleton, cell proliferation, and proteostasis pathways, in line with previous AD reports. Our study is the first to conduct a proteomic analysis of MSCs from the Jalisco FAD patients in two stages of the disease (symptomatic and presymptomatic), showing these cells as a new and excellent in vitro model for future AD studies.
Collapse
Affiliation(s)
- Lory J. Rochín-Hernández
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico; (L.J.R.-H.); (M.A.J.-A.); (M.d.P.F.-C.)
| | - Miguel A. Jiménez-Acosta
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico; (L.J.R.-H.); (M.A.J.-A.); (M.d.P.F.-C.)
| | - Lorena Ramírez-Reyes
- Unidad de Genómica, Proteómica y Metabolómica, Laboratorio Nacional de Servicios Experimentales (LaNSE), Centro de Investigación y de Estudios Avanzados, Ciudad de México 07360, Mexico;
| | - María del Pilar Figueroa-Corona
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico; (L.J.R.-H.); (M.A.J.-A.); (M.d.P.F.-C.)
| | - Víctor J. Sánchez-González
- Centro Universitario de Los Altos, Universidad de Guadalajara, Tepatitlán de Morelos 47620, Mexico; (V.J.S.-G.); (M.O.-B.)
| | - Maribel Orozco-Barajas
- Centro Universitario de Los Altos, Universidad de Guadalajara, Tepatitlán de Morelos 47620, Mexico; (V.J.S.-G.); (M.O.-B.)
| | - Marco A. Meraz-Ríos
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico; (L.J.R.-H.); (M.A.J.-A.); (M.d.P.F.-C.)
| |
Collapse
|
20
|
Alluri R, Kilari EK, Pasala PK, Kopalli SR, Koppula S. Repurposing Diltiazem for Its Neuroprotective Anti-Dementia Role against Intra-Cerebroventricular Streptozotocin-Induced Sporadic Alzheimer's Disease-Type Rat Model. Life (Basel) 2023; 13:1688. [PMID: 37629545 PMCID: PMC10455909 DOI: 10.3390/life13081688] [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: 07/08/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neuropsychiatric disorder and a common cause of progressive dementia. Diltiazem (DTZ), the non-dihydropyridine benzothiazepine class of calcium channel blocker (CCB), used clinically in angina and other cardiovascular disorders, has proven neurological benefits. In the present study, the neuroprotective anti-dementia effects of DTZ against intra-cerebroventricular-streptozotocin (ICV-STZ)-induced sporadic AD (SAD)-type rat model was investigated. ICV-STZ-induced cognitive impairments were measured via passive avoidance and Morris water maze tasks. Anti-oxidative enzyme status, pro-inflammatory markers, and amyloid-beta (Aβ) protein expression in rat brain tissues were measured using ELISA kits, Western blotting, and immunostaining techniques. The data revealed that ICV-STZ injection in rats significantly induced cognitive deficits and altered the levels of oxidative and pro-inflammatory markers (p < 0.05~p < 0.001). Treatment with DTZ (10 mg/kg, 20 mg/kg, and 40 mg/kg, p.o.) daily for twenty-one days, 1 h before a single ICV-STZ (3 mg/kg) injection, significantly improved cognitive impairments and ameliorated the ICV-STZ-induced altered nitrite, pro-inflammatory cytokines (TNF-α, and IL-1β) and anti-oxidative enzyme levels (superoxide dismutase, lipid peroxidation, and glutathione). Further, DTZ restored the increased Aβ protein expression in ICV-STZ-induced brain tissue. Considering the results obtained, DTZ might have a potential therapeutic role in treating and managing AD and related dementia pathologies due to its anti-dementia activity in SAD-type conditions in rats induced by ICV-STZ.
Collapse
Affiliation(s)
- Ramesh Alluri
- Cognitive Science Research Initiative Lab., Department of Pharmacology, Vishnu Institute of Pharmaceutical Education and Research, Medak Dist., Narsapur 502313, India
| | - Eswar Kumar Kilari
- Department of Pharmacology, University College of Pharmaceutical Sciences, Andhra University, Visakhapatnam 530003, India
| | - Praveen Kumar Pasala
- Department of Pharmacology, Raghavendra Institute of Pharmaceutical Education and Research, Jawaharlal Nehru Technological University Anantapur—JNTUA, Anantapur 515721, India
| | - Spandana Rajendra Kopalli
- Department of Integrated Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Science, Konkuk University, Chungju-si 380-701, Republic of Korea
| |
Collapse
|
21
|
Hua L, Gao F, Xia X, Guo Q, Zhao Y, Huang S, Yuan Z. Individual-specific functional connectivity improves prediction of Alzheimer's disease's symptoms in elderly people regardless of APOE ε4 genotype. Commun Biol 2023; 6:581. [PMID: 37258640 PMCID: PMC10232409 DOI: 10.1038/s42003-023-04952-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
To date, reliable biomarkers remain unclear that could link functional connectivity to patients' symptoms for detecting and predicting the process from normal aging to Alzheimer's disease (AD) in elderly people with specific genotypes. To address this, individual-specific functional connectivity is constructed for elderly participants with/without APOE ε4 allele. Then, we utilize recursive feature selection-based machine learning to reveal individual brain-behavior relationships and to predict the symptom transition in different genotypes. Our findings reveal that compared with conventional atlas-based functional connectivity, individual-specific functional connectivity exhibits higher classification and prediction performance from normal aging to AD in both APOE ε4 groups, while no significant performance is detected when the data of two genotyping groups are combined. Furthermore, individual-specific between-network connectivity constitutes a major contributor to assessing cognitive symptoms. This study highlights the essential role of individual variation in cortical functional anatomy and the integration of brain and behavior in predicting individualized symptoms.
Collapse
Affiliation(s)
- Lin Hua
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
| | - Fei Gao
- Institute of Modern Languages and Linguistics, Fudan University, Shanghai, 200433, China
| | - Xiaoluan Xia
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
| | - Qiwei Guo
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
| | - Shaohui Huang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhen Yuan
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China.
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China.
| |
Collapse
|
22
|
Padmanabhan P, Götz J. Clinical relevance of animal models in aging-related dementia research. NATURE AGING 2023; 3:481-493. [PMID: 37202516 DOI: 10.1038/s43587-023-00402-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Alzheimer's disease (AD) and other, less prevalent dementias are complex age-related disorders that exhibit multiple etiologies. Over the past decades, animal models have provided pathomechanistic insight and evaluated countless therapeutics; however, their value is increasingly being questioned due to the long history of drug failures. In this Perspective, we dispute this criticism. First, the utility of the models is limited by their design, as neither the etiology of AD nor whether interventions should occur at a cellular or network level is fully understood. Second, we highlight unmet challenges shared between animals and humans, including impeded drug transport across the blood-brain barrier, limiting effective treatment development. Third, alternative human-derived models also suffer from the limitations mentioned above and can only act as complementary resources. Finally, age being the strongest AD risk factor should be better incorporated into the experimental design, with computational modeling expected to enhance the value of animal models.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
23
|
Khan AW, Farooq M, Hwang MJ, Haseeb M, Choi S. Autoimmune Neuroinflammatory Diseases: Role of Interleukins. Int J Mol Sci 2023; 24:7960. [PMID: 37175665 PMCID: PMC10178921 DOI: 10.3390/ijms24097960] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Autoimmune neuroinflammatory diseases are a group of disorders resulting from abnormal immune responses in the nervous system, causing inflammation and tissue damage. The interleukin (IL) family of cytokines, especially IL-1, IL-6, and IL-17, plays a critical role in the pathogenesis of these diseases. IL-1 is involved in the activation of immune cells, production of pro-inflammatory cytokines, and promotion of blood-brain barrier breakdown. IL-6 is essential for the differentiation of T cells into Th17 cells and has been implicated in the initiation and progression of neuroinflammation. IL-17 is a potent pro-inflammatory cytokine produced by Th17 cells that plays a crucial role in recruiting immune cells to sites of inflammation. This review summarizes the current understanding of the roles of different interleukins in autoimmune neuroinflammatory diseases, including multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's disease, neuromyelitis optica, and autoimmune encephalitis, and discusses the potential of targeting ILs as a therapeutic strategy against these diseases. We also highlight the need for further research to better understand the roles of ILs in autoimmune neuroinflammatory diseases and to identify new targets for treating these debilitating diseases.
Collapse
Affiliation(s)
- Abdul Waheed Khan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Mariya Farooq
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Moon-Jung Hwang
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Muhammad Haseeb
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Republic of Korea
| |
Collapse
|
24
|
Koul B, Farooq U, Yadav D, Song M. Phytochemicals: A Promising Alternative for the Prevention of Alzheimer's Disease. Life (Basel) 2023; 13:life13040999. [PMID: 37109528 PMCID: PMC10144079 DOI: 10.3390/life13040999] [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: 03/20/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a neurological condition that worsens with ageing and affects memory and cognitive function. Presently more than 55 million individuals are affected by AD all over the world, and it is a leading cause of death in old age. The main purpose of this paper is to review the phytochemical constituents of different plants that are used for the treatment of AD. A thorough and organized review of the existing literature was conducted, and the data under the different sections were found using a computerized bibliographic search through the use of databases such as PubMed, Web of Science, Google Scholar, Scopus, CAB Abstracts, MEDLINE, EMBASE, INMEDPLAN, NATTS, and numerous other websites. Around 360 papers were screened, and, out of that, 258 papers were selected on the basis of keywords and relevant information that needed to be included in this review. A total of 55 plants belonging to different families have been reported to possess different bioactive compounds (galantamine, curcumin, silymarin, and many more) that play a significant role in the treatment of AD. These plants possess anti-inflammatory, antioxidant, anticholinesterase, and anti-amyloid properties and are safe for consumption. This paper focuses on the taxonomic details of the plants, the mode of action of their phytochemicals, their safety, future prospects, limitations, and sustainability criteria for the effective treatment of AD.
Collapse
Affiliation(s)
- Bhupendra Koul
- Department of Biotechnology, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Usma Farooq
- Department of Botany, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
25
|
Costa T, Manuello J, Cauda F, Liloia D. Retrospective Bayesian Evidence of Null Effect in Two Decades of Alzheimer's Disease Clinical Trials. J Alzheimers Dis 2023; 91:531-535. [PMID: 36442201 DOI: 10.3233/jad-220942] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Despite intense research on Alzheimer's disease, no validated treatment able to reverse symptomatology or stop disease progression exists. A recent systematic review by Kim and colleagues evaluated possible reasons behind the failure of the majority of the clinical trials. As the focus was on methodological factors, no statistical trends were examined in detail. Here, we aim to complete this picture leveraging on Bayesian analysis. In particular, we tested whether the failure of those clinical trials was essentially due to insufficient statistical power or to lack of a true effect. The strong Bayes' Factor obtained supported the latter hypothesis.
Collapse
Affiliation(s)
- Tommaso Costa
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy.,FOCUS Lab, Department of Psychology, University of Turin, Turin, Italy
| | - Jordi Manuello
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy.,FOCUS Lab, Department of Psychology, University of Turin, Turin, Italy
| | - Franco Cauda
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy.,FOCUS Lab, Department of Psychology, University of Turin, Turin, Italy
| | - Donato Liloia
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy.,FOCUS Lab, Department of Psychology, University of Turin, Turin, Italy
| |
Collapse
|
26
|
Engels-Domínguez N, Koops EA, Prokopiou PC, Van Egroo M, Schneider C, Riphagen JM, Singhal T, Jacobs HIL. State-of-the-art imaging of neuromodulatory subcortical systems in aging and Alzheimer's disease: Challenges and opportunities. Neurosci Biobehav Rev 2023; 144:104998. [PMID: 36526031 PMCID: PMC9805533 DOI: 10.1016/j.neubiorev.2022.104998] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/30/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022]
Abstract
Primary prevention trials have shifted their focus to the earliest stages of Alzheimer's disease (AD). Autopsy data indicates that the neuromodulatory subcortical systems' (NSS) nuclei are specifically vulnerable to initial tau pathology, indicating that these nuclei hold great promise for early detection of AD in the context of the aging brain. The increasing availability of new imaging methods, ultra-high field scanners, new radioligands, and routine deep brain stimulation implants has led to a growing number of NSS neuroimaging studies on aging and neurodegeneration. Here, we review findings of current state-of-the-art imaging studies assessing the structure, function, and molecular changes of these nuclei during aging and AD. Furthermore, we identify the challenges associated with these imaging methods, important pathophysiologic gaps to fill for the AD NSS neuroimaging field, and provide future directions to improve our assessment, understanding, and clinical use of in vivo imaging of the NSS.
Collapse
Affiliation(s)
- Nina Engels-Domínguez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Elouise A Koops
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Prokopis C Prokopiou
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maxime Van Egroo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Christoph Schneider
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joost M Riphagen
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tarun Singhal
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi I L Jacobs
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
27
|
Jin J, Xu Z, Zhang L, Zhang C, Zhao X, Mao Y, Zhang H, Liang X, Wu J, Yang Y, Zhang J. Gut-derived β-amyloid: Likely a centerpiece of the gut-brain axis contributing to Alzheimer's pathogenesis. Gut Microbes 2023; 15:2167172. [PMID: 36683147 PMCID: PMC9872956 DOI: 10.1080/19490976.2023.2167172] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/03/2023] [Indexed: 01/24/2023] Open
Abstract
Peripheral β-amyloid (Aβ), including those contained in the gut, may contribute to the formation of Aβ plaques in the brain, and gut microbiota appears to exert an impact on Alzheimer's disease (AD) via the gut-brain axis, although detailed mechanisms are not clearly defined. The current study focused on uncovering the potential interactions among gut-derived Aβ in aging, gut microbiota, and AD pathogenesis. To achieve this goal, the expression levels of Aβ and several key proteins involved in Aβ metabolism were initially assessed in mouse gut, with key results confirmed in human tissue. The results demonstrated that a high level of Aβ was detected throughout the gut in both mice and human, and gut Aβ42 increased with age in wild type and mutant amyloid precursor protein/presenilin 1 (APP/PS1) mice. Next, the gut microbiome of mice was characterized by 16S rRNA sequencing, and we found the gut microbiome altered significantly in aged APP/PS1 mice and fecal microbiota transplantation (FMT) of aged APP/PS1 mice increased gut BACE1 and Aβ42 levels. Intra-intestinal injection of isotope or fluorescence labeled Aβ combined with vagotomy was also performed to investigate the transmission of Aβ from gut to brain. The data showed that, in aged mice, the gut Aβ42 was transported to the brain mainly via blood rather than the vagal nerve. Furthermore, FMT of APP/PS1 mice induced neuroinflammation, a phenotype that mimics early AD pathology. Taken together, this study suggests that the gut is likely a critical source of Aβ in the brain, and gut microbiota can further upregulate gut Aβ production, thereby potentially contributing to AD pathogenesis.
Collapse
Affiliation(s)
- Jinghua Jin
- Department of Pathology, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi Xu
- Department of Pathology, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, China
| | - Lina Zhang
- Department of Pathology, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Can Zhang
- Department of Pathology, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoduo Zhao
- Department of Pathology, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yuxuan Mao
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, China
| | - Haojian Zhang
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingguang Liang
- Central Laboratory, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Juanli Wu
- National Human Brain Bank for Health and Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Yang
- Department of Pathology, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Zhang
- Department of Pathology, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, China
- National Human Brain Bank for Health and Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
28
|
Mattke S, Corrêa Dos Santos Filho O, Hanson M, Mateus EF, Neto JPR, de Souza LC, Rizek Schultz R, Pereira Pinto R. Preparedness of the Brazilian health-care system to provide access to a disease-modifying Alzheimer's disease treatment. Alzheimers Dement 2023; 19:375-381. [PMID: 36063494 PMCID: PMC10087834 DOI: 10.1002/alz.12778] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND While the majority of patients with Alzheimer's disease resides in low and middle-income countries, little is known of their preparedness for emerging disease-modifying treatments. We analyze the preparedness of Brazil, one of the most populous middle-income countries, from a capacity and institutional preparedness perspective. METHODS Desk research and 12 interviews for background and capacity data. Markov model to estimate wait times for access to treatment. FINDINGS Brazil has no national dementia strategy or established pathway for evaluation of cognitive concerns, and dementia is typically diagnosed late if at all. While members of private health plans have ready access to elective specialty care, wait times in the public sector are long. Assuming potentially treatment-eligible patients are referred from primary to specialty care based on a brief cognitive exam and a blood test for the Alzheimer's pathology, available capacity will not be sufficient to match the projected demand. The biggest obstacle is availability of dementia specialist visits, and the effect of population growth and ageing means that the wait list for specialist appointment will continue to grow from around 400,000 in 2022 to over 2.2 million in 2040. We do not project substantial wait times for confirmatory biomarker testing and treatment delivery but note that this is a consequence of patients waiting for their specialist appointments. These queues will result in estimated persistent wait times for treatment of around two years on average with substantial differences between the public and private sectors, as capacity growth is insufficient to keep up with increasing demand. DISCUSSION Our findings suggest that Brazil is ill-prepared to provide timely access to an Alzheimer's treatment with predicted wait times of about two years, largely because of a limited number of dementia specialists.
Collapse
Affiliation(s)
- Soeren Mattke
- University of Southern California, Los Angeles, California, USA
| | | | - Mark Hanson
- University of Southern California, Los Angeles, California, USA
| | - Elaine Fernandes Mateus
- Febraz - Brazilian Federation of Alzheimer's Associations, Brazil and Londrina State University, Londrina, Brazil
| | - João Paulo Reis Neto
- CAPESESP - Caixa de Previdência e Assistência dos Servidores da Fundação Nacional de Saúde, Rio de Janeiro, Brazil
| | | | | | - Roney Pereira Pinto
- Programa Ciências da Saúde da Faculdade de Medicina, Federal University of Goias, Goiânia, Brazil
| |
Collapse
|
29
|
Naki T, Matshe WMR, Balogun MO, Sinha Ray S, Egieyeh SA, Aderibigbe BA. Polymer drug conjugates containing memantine, tacrine and cinnamic acid: promising nanotherapeutics for the treatment of Alzheimer's disease. J Microencapsul 2023; 40:15-28. [PMID: 36622880 DOI: 10.1080/02652048.2023.2167011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
AIM To prepare polymer-drug conjugates containing a combination of memantine, tacrine, and E)-N-(3-aminopropyl)cinnamide, promising therapeutics for the treatment of neurodegenerative disorders. METHODS The conjugates were characterised by 1HNMR, particle size analysis, SEM, LC-MS, TEM/EDX, and XRD, followed by in vitro anti-acetylcholinesterase and drug release studies. RESULTS 1H NMR analysis revealed successful drug conjugation with drug mass percentages in the range of 1.3-6.0% w/w. The drug release from the conjugates was sustained for 10 h in the range of 20-36%. The conjugates' capability to inhibit acetylcholinesterase (AChE) activity was significant with IC50 values in the range of 13-44.4 µm which was more effective than tacrine (IC50 =1698.8 µm). The docking studies further confirmed that the conjugation of the drugs into the polymer improved their anti-acetylcholinesterase activity. CONCLUSION The drug release profile, particle sizes, and in vitro studies revealed that the conjugates are promising therapeutics for treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Tobeka Naki
- Department of Chemistry, University of Fort Hare, Alice, South Africa
| | | | | | - Suprakas Sinha Ray
- DST/CSIR National Centre for Nanostructured Materials, Council for Scientific and Industrial Research, Pretoria, South Africa
| | | | | |
Collapse
|
30
|
Identification of Potential Repurposable Drugs in Alzheimer’s Disease Exploiting a Bioinformatics Analysis. J Pers Med 2022; 12:jpm12101731. [PMID: 36294870 PMCID: PMC9605472 DOI: 10.3390/jpm12101731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurologic disorder causing brain atrophy and the death of brain cells. It is a progressive condition marked by cognitive and behavioral impairment that significantly interferes with daily activities. AD symptoms develop gradually over many years and eventually become more severe, and no cure has been found yet to arrest this process. The present study is directed towards suggesting putative novel solutions and paradigms for fighting AD pathogenesis by exploiting new insights from network medicine and drug repurposing strategies. To identify new drug–AD associations, we exploited SAveRUNNER, a recently developed network-based algorithm for drug repurposing, which quantifies the vicinity of disease-associated genes to drug targets in the human interactome. We complemented the analysis with an in silico validation of the candidate compounds through a gene set enrichment analysis, aiming to determine if the modulation of the gene expression induced by the predicted drugs could be counteracted by the modulation elicited by the disease. We identified some interesting compounds belonging to the beta-blocker family, originally approved for treating hypertension, such as betaxolol, bisoprolol, and metoprolol, whose connection with a lower risk to develop Alzheimer’s disease has already been observed. Moreover, our algorithm predicted multi-kinase inhibitors such as regorafenib, whose beneficial effects were recently investigated for neuroinflammation and AD pathology, and mTOR inhibitors such as sirolimus, whose modulation has been associated with AD.
Collapse
|
31
|
Mumtaz I, Ayaz MO, Khan MS, Manzoor U, Ganayee MA, Bhat AQ, Dar GH, Alghamdi BS, Hashem AM, Dar MJ, Ashraf GM, Maqbool T. Clinical relevance of biomarkers, new therapeutic approaches, and role of post-translational modifications in the pathogenesis of Alzheimer's disease. Front Aging Neurosci 2022; 14:977411. [PMID: 36158539 PMCID: PMC9490081 DOI: 10.3389/fnagi.2022.977411] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/18/2022] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that causes progressive loss of cognitive functions like thinking, memory, reasoning, behavioral abilities, and social skills thus affecting the ability of a person to perform normal daily functions independently. There is no definitive cure for this disease, and treatment options available for the management of the disease are not very effective as well. Based on histopathology, AD is characterized by the accumulation of insoluble deposits of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs). Although several molecular events contribute to the formation of these insoluble deposits, the aberrant post-translational modifications (PTMs) of AD-related proteins (like APP, Aβ, tau, and BACE1) are also known to be involved in the onset and progression of this disease. However, early diagnosis of the disease as well as the development of effective therapeutic approaches is impeded by lack of proper clinical biomarkers. In this review, we summarized the current status and clinical relevance of biomarkers from cerebrospinal fluid (CSF), blood and extracellular vesicles involved in onset and progression of AD. Moreover, we highlight the effects of several PTMs on the AD-related proteins, and provide an insight how these modifications impact the structure and function of proteins leading to AD pathology. Finally, for disease-modifying therapeutics, novel approaches, and targets are discussed for the successful treatment and management of AD.
Collapse
Affiliation(s)
- Ibtisam Mumtaz
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| | - Mir Owais Ayaz
- Laboratory of Cell and Molecular Biology, Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Centre for Scientific and Innovative Research, Ghaziabad, Utter Pradesh, India
| | - Mohamad Sultan Khan
- Neurobiology and Molecular Chronobiology Laboratory, Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Umar Manzoor
- Laboratory of Immune and Inflammatory Disease, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, South Korea
| | - Mohd Azhardin Ganayee
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, India
| | - Aadil Qadir Bhat
- Laboratory of Cell and Molecular Biology, Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Centre for Scientific and Innovative Research, Ghaziabad, Utter Pradesh, India
| | - Ghulam Hassan Dar
- Sri Pratap College, Cluster University Srinagar, Jammu and Kashmir, India
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar M. Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohd Jamal Dar
- Laboratory of Cell and Molecular Biology, Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Centre for Scientific and Innovative Research, Ghaziabad, Utter Pradesh, India
| | - Gulam Md. Ashraf
- Pre-clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tariq Maqbool
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|
32
|
Banaras S, Paracha RZ, Nisar M, Arif A, Ahmad J, Tariq Saeed M, Mustansar Z, Shuja MN, Paracha RN. System level modeling and analysis of TNF- α mediated sphingolipid signaling pathway in neurological disorders for the prediction of therapeutic targets. Front Physiol 2022; 13:872421. [PMID: 36060699 PMCID: PMC9437628 DOI: 10.3389/fphys.2022.872421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 07/15/2022] [Indexed: 01/09/2023] Open
Abstract
Sphingomyelin (SM) belongs to a class of lipids termed sphingolipids. The disruption in the sphingomyelin signaling pathway is associated with various neurodegenerative disorders. TNF-α, a potent pro-inflammatory cytokine generated in response to various neurological disorders like Alzheimer's disease (AD), Parkinson's disease (PD), and Multiple Sclerosis (MS), is an eminent regulator of the sphingomyelin metabolic pathway. The immune-triggered regulation of the sphingomyelin metabolic pathway via TNF-α constitutes the sphingomyelin signaling pathway. In this pathway, sphingomyelin and its downstream sphingolipids activate various signaling cascades like PI3K/AKT and MAPK/ERK pathways, thus, controlling diverse processes coupled with neuronal viability, survival, and death. The holistic analysis of the immune-triggered sphingomyelin signaling pathway is imperative to make necessary predictions about its pivotal components and for the formulation of disease-related therapeutics. The current work offers a comprehensive in silico systems analysis of TNF-α mediated sphingomyelin and downstream signaling cascades via a model-based quantitative approach. We incorporated the intensity values of genes from the microarray data of control individuals from the AD study in the input entities of the pathway model. Computational modeling and simulation of the inflammatory pathway enabled the comprehensive study of the system dynamics. Network and sensitivity analysis of the model unveiled essential interaction parameters and entities during neuroinflammation. Scanning of the key entities and parameters allowed us to determine their ultimate impact on neuronal apoptosis and survival. Moreover, the efficacy and potency of the FDA-approved drugs, namely Etanercept, Nivocasan, and Scyphostatin allowed us to study the model's response towards inhibition of the respective proteins/enzymes. The network analysis revealed the pivotal model entities with high betweenness and closeness centrality values including recruit FADD, TNFR_TRADD, act CASP2, actCASP8, actCASP3 and 9, cytochrome C, and RIP_RAIDD which profoundly impacted the neuronal apoptosis. Whereas some of the entities with high betweenness and closeness centrality values like Gi-coupled receptor, actS1PR, Sphingosine, S1P, actAKT, and actERK produced a high influence on neuronal survival. However, the current study inferred the dual role of ceramide, both on neuronal survival and apoptosis. Moreover, the drug Nivocasan effectively reduces neuronal apoptosis via its inhibitory mechanism on the caspases.
Collapse
Affiliation(s)
- Sanam Banaras
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rehan Zafar Paracha
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Maryum Nisar
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Ayesha Arif
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Jamil Ahmad
- Computer Science and Information Technology (CS&IT), University of Malakand, Chakdara, Pakistan
| | - Muhammad Tariq Saeed
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Zartasha Mustansar
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | | | - Rizwan Nasir Paracha
- Department of Chemistry, University of Sargodha, Sub Campus Bhakkar, Bhakkar, Pakistan
| |
Collapse
|
33
|
Roy R, Bhattacharya P, Borah A. Targeting the Pathological Hallmarks of Alzheimer's Disease Through Nanovesicleaided Drug Delivery Approach. Curr Drug Metab 2022; 23:693-707. [PMID: 35619248 DOI: 10.2174/1389200223666220526094802] [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: 01/23/2022] [Revised: 02/23/2022] [Accepted: 03/02/2022] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Nanovesicle technology is making a huge contribution to the progress of treatment studies for various diseases, including Alzheimer's disease (AD). AD is the leading neurodegenerative disorder characterized by severe cognitive impairment. Despite the prevalence of several forms of anti-AD drugs, the accelerating pace of AD incidence cannot becurbed, and for rescue, nanovesicle technology has grabbed much attention. METHODOLOGY Comprehensive literature search was carried out using relevant keywords and online database platforms. The main concepts that have been covered included a complex pathomechanism underlying increased acetylcholinesterase (AchE) activity, β-amyloid aggregation, and tau-hyperphosphorylation forming neurofibrillary tangles (NFTs) in the brain, which are amongst the major hallmarks of AD pathology. Therapeutic recommendations exist in the form of AchE inhibitors, along with anti-amyloid and anti-tau therapeutics, which are being explored at a high pace. The degree of the therapeutic outcome, however, gets restricted by the pharmacological limitations. Susceptibility to peripheral metabolism and rapid elimination, inefficiency to cross the blood-brain barrier (BBB) and reach the target brain site are the factors that lower the biostability and bioavailability of anti-AD drugs. The nanovesicle technology has emerged as a route to preserve the therapeutic efficiency of the anti-AD drugs and promote AD treatment. The review hereby aims to summarize the developments made by the nanovesicle technology in aiding the delivery of synthetic and plant-based therapeutics targeting the molecular mechanism of AD pathology. CONCLUSION Nanovesicles appear to efficiently aid in target-specific delivery of anti-AD therapeutics and nullify the drawbacks posed by free drugs, besides reducing the dosage requirement and the adversities associated. In addition, the nanovesicle technology also appears to uplift the therapeutic potential of several phyto-compounds with immense anti-AD properties. Furthermore, the review also sheds light on future perspectives to mend the gaps that prevail in the nanovesicle-mediated drug delivery in AD treatment strategies.
Collapse
Affiliation(s)
- Rubina Roy
- Department of Life Science and Bioinformatics, Cellular and Molecular Neurobiology Laboratory, Assam University, Silchar- 788011, Assam, India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad - 382355, Gandhinagar, Gujarat, India
| | - Anupom Borah
- Department of Life Science and Bioinformatics, Cellular and Molecular Neurobiology Laboratory, Assam University, Silchar- 788011, Assam, India
| |
Collapse
|
34
|
Qian XH, Xie RY, Liu XL, Chen SD, Tang HD. Mechanisms of Short-Chain Fatty Acids Derived from Gut Microbiota in Alzheimer's Disease. Aging Dis 2022; 13:1252-1266. [PMID: 35855330 PMCID: PMC9286902 DOI: 10.14336/ad.2021.1215] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022] Open
Abstract
Short-chain fatty acids (SCFAs) are important metabolites derived from the gut microbiota through fermentation of dietary fiber. SCFAs participate a number of physiological and pathological processes in the human body, such as host metabolism, immune regulation, appetite regulation. Recent studies on gut-brain interaction have shown that SCFAs are important mediators of gut-brain interactions and are involved in the occurrence and development of many neurodegenerative diseases, including Alzheimer's disease. This review summarizes the current research on the potential roles and mechanisms of SCFAs in AD. First, we introduce the metabolic distribution, specific receptors and signaling pathways of SCFAs in human body. The concentration levels of SCFAs in AD patient/animal models are then summarized. In addition, we illustrate the effects and mechanisms of SCFAs on the cognitive level, pathological features (Aβ and tau) and neuroinflammation in AD. Finally, we analyze the translational value of SCFAs as potential therapeutic targets for the treatment of AD.
Collapse
Affiliation(s)
- Xiao-hang Qian
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Ru-yan Xie
- Shanghai Guangci Memorial hospital, Shanghai 200025, China.
| | - Xiao-li Liu
- Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital South Campus, Shanghai 201406, China.
| | - Sheng-di Chen
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
- Correspondence should be addressed to: Dr. Sheng-di Chen () and Dr. Hui-dong Tang (), Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hui-dong Tang
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
- Correspondence should be addressed to: Dr. Sheng-di Chen () and Dr. Hui-dong Tang (), Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
35
|
Long Y, Cheng Y, Yang J, Yang T, Lai Y. Abeta-induced Presynaptic Release of UBC9 through Extracellular Vesicles involves SNAP23. Neurosci Lett 2022; 785:136771. [DOI: 10.1016/j.neulet.2022.136771] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/20/2022] [Accepted: 06/30/2022] [Indexed: 11/26/2022]
|
36
|
Chen B, Marquez-Nostra B, Belitzky E, Toyonaga T, Tong J, Huang Y, Cai Z. PET Imaging in Animal Models of Alzheimer’s Disease. Front Neurosci 2022; 16:872509. [PMID: 35685772 PMCID: PMC9171374 DOI: 10.3389/fnins.2022.872509] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
The successful development and translation of PET imaging agents targeting β-amyloid plaques and hyperphosphorylated tau tangles have allowed for in vivo detection of these hallmarks of Alzheimer’s disease (AD) antemortem. Amyloid and tau PET have been incorporated into the A/T/N scheme for AD characterization and have become an integral part of ongoing clinical trials to screen patients for enrollment, prove drug action mechanisms, and monitor therapeutic effects. Meanwhile, preclinical PET imaging in animal models of AD can provide supportive information for mechanistic studies. With the recent advancement of gene editing technologies and AD animal model development, preclinical PET imaging in AD models will further facilitate our understanding of AD pathogenesis/progression and the development of novel treatments. In this study, we review the current state-of-the-art in preclinical PET imaging using animal models of AD and suggest future research directions.
Collapse
|
37
|
Shabbir A, Rehman K, Akash MSH, Akbar M, Chaudhary Z, Panichayupakaranant P, Shah MA. Differential neuroprotective effect of curcuminoid formulations in aluminum chloride-induced Alzheimer's disease. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:67981-67996. [PMID: 35525893 DOI: 10.1007/s11356-022-20593-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/29/2022] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a slowly progressive brain degenerative disorder which gradually impairs memory, thinking, and ability to perform easy routine tasks. This degenerative disorder mainly targets the elderly people and has imposed an endemic burden on society. Hence, there is a crucial need to investigate the efficacious herbal pharmacotherapies that can effectively mitigate and prevent the pathological hallmarks of AD. The current study aims to explore the potential efficacy of curcuminoid-rich extract (CRE) and its ternary complex (TC). Experimental rodents were administered with AlCl3 (300 mg/kg) to induce AD and treated with rivastigmine, curcuminoid crude extract, CRE, and TC orally for three consecutive weeks. Neurobehavioral, biochemical, and histopathological studies were performed from the last week of the study period. The mRNA expression of different pathological biomarkers was estimated by RT-qPCR analysis. The results of the study suggested that CRE and TC significantly improved the behavioral, biochemical parameters and acetylcholinesterase inhibitory activity in treatment groups. Histological analysis was also carried out indicating that the neurodegenerative changes and neuronal loss were stabilized by CRE and TC supplementation. CRE and TC supplementation remarkably downregulated the interleukin-1α, tumor necrosis factor-α, interleukin-1β, acetylcholinesterase, and β-secretase pathological gene expression. Hence, it was concluded that CRE and TC may act as promising candidates in the prevention of AD via numerous underlying signaling pathways.
Collapse
Affiliation(s)
- Anam Shabbir
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan.,LIAS College of Pharmacy, Faisalabad, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, The Women University, Multan, Pakistan
| | | | - Moazzama Akbar
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Zunera Chaudhary
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | | | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Government College University, Faisalabad, Pakistan
| |
Collapse
|
38
|
Song JG, Yu MS, Lee B, Lee J, Hwang SH, Na D, Kim HW. Analysis methods for the gut microbiome in neuropsychiatric and neurodegenerative disorders. Comput Struct Biotechnol J 2022; 20:1097-1110. [PMID: 35317228 PMCID: PMC8902474 DOI: 10.1016/j.csbj.2022.02.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022] Open
Abstract
For a long time, the central nervous system was believed to be the only regulator of cognitive functions. However, accumulating evidence suggests that the composition of the microbiome is strongly associated with brain functions and diseases. Indeed, the gut microbiome is involved in neuropsychiatric diseases (e.g., depression, autism spectrum disorder, and anxiety) and neurodegenerative diseases (e.g., Parkinson’s disease and Alzheimer’s disease). In this review, we provide an overview of the link between the gut microbiome and neuropsychiatric or neurodegenerative disorders. We also introduce analytical methods used to assess the connection between the gut microbiome and the brain. The limitations of the methods used at present are also discussed. The accurate translation of the microbiome information to brain disorder could promote better understanding of neuronal diseases and aid in finding alternative and novel therapies.
Collapse
Affiliation(s)
- Jae Gwang Song
- Department of Bio-integrated Science and Technology, College of Life Sciences, Sejong University, Seoul 05006, Republic of Korea
| | - Myeong-Sang Yu
- Department of Biomedical Engineering, Department of Biomedical Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Bomi Lee
- Department of Bio-integrated Science and Technology, College of Life Sciences, Sejong University, Seoul 05006, Republic of Korea
| | - Jingyu Lee
- Department of Biomedical Engineering, Department of Biomedical Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Su-Hee Hwang
- Department of Biomedical Engineering, Department of Biomedical Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Dokyun Na
- Department of Biomedical Engineering, Department of Biomedical Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
- Corresponding authors.
| | - Hyung Wook Kim
- Department of Bio-integrated Science and Technology, College of Life Sciences, Sejong University, Seoul 05006, Republic of Korea
- Corresponding authors.
| |
Collapse
|
39
|
Chan AWS, Cho IK, Li CX, Zhang X, Patel S, Rusnak R, Raper J, Bachevalier J, Moran SP, Chi T, Cannon KH, Hunter CE, Martin RC, Xiao H, Yang SH, Gumber S, Herndon JG, Rosen RF, Hu WT, Lah JJ, Levey AI, Smith Y, Walker LC. Cerebral Aβ deposition in an Aβ-precursor protein-transgenic rhesus monkey. AGING BRAIN 2022; 2:100044. [PMID: 36589695 PMCID: PMC9802652 DOI: 10.1016/j.nbas.2022.100044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
With the ultimate goal of developing a more representative animal model of Alzheimer's disease (AD), two female amyloid-β-(Aβ) precursor protein-transgenic (APPtg) rhesus monkeys were generated by lentiviral transduction of the APP gene into rhesus oocytes, followed by in vitro fertilization and embryo transfer. The APP-transgene included the AD-associated Swedish K670N/M671L and Indiana V717F mutations (APPSWE/IND) regulated by the human polyubiquitin-C promoter. Overexpression of APP was confirmed in lymphocytes and brain tissue. Upon sacrifice at 10 years of age, one of the monkeys had developed Aβ plaques and cerebral Aβ-amyloid angiopathy in the occipital, parietal, and caudal temporal neocortices. The induction of Aβ deposition more than a decade prior to its usual emergence in the rhesus monkey supports the feasibility of creating a transgenic nonhuman primate model for mechanistic analyses and preclinical testing of treatments for Alzheimer's disease and cerebrovascular amyloidosis.
Collapse
Affiliation(s)
- Anthony W S Chan
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - In Ki Cho
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Chun-Xia Li
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Xiaodong Zhang
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Sudeep Patel
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Rebecca Rusnak
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jessica Raper
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jocelyne Bachevalier
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Psychology, Emory College, Atlanta, GA 30322, USA
| | - Sean P Moran
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Tim Chi
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Katherine H Cannon
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Carissa E Hunter
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Ryan C Martin
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Hailian Xiao
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shang-Hsun Yang
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sanjeev Gumber
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - James G Herndon
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Rebecca F Rosen
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James J Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yoland Smith
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lary C Walker
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
40
|
Andrographolide promotes hippocampal neurogenesis and spatial memory in the APPswe/PS1ΔE9 mouse model of Alzheimer's disease. Sci Rep 2021; 11:22904. [PMID: 34824314 PMCID: PMC8616902 DOI: 10.1038/s41598-021-01977-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/08/2021] [Indexed: 02/07/2023] Open
Abstract
In Alzheimer´s disease (AD) there is a reduction in hippocampal neurogenesis that has been associated to cognitive deficits. Previously we showed that Andrographolide (ANDRO), the main bioactive component of Andrographis paniculate, induces proliferation in the hippocampus of the APPswe/PSEN1ΔE9 (APP/PS1) mouse model of AD as assessed by staining with the mitotic marker Ki67. Here, we further characterized the effect of ANDRO on hippocampal neurogenesis in APP/PS1 mice and evaluated the contribution of this process to the cognitive effect of ANDRO. Treatment of 8-month-old APP/PS1 mice with ANDRO for 4 weeks increased proliferation in the dentate gyrus as evaluated by BrdU incorporation. Although ANDRO had no effect on neuronal differentiation of newborn cells, it strongly increased neural progenitors, neuroblasts and newborn immature neurons, cell populations that were decreased in APP/PS1 mice compared to age-matched wild-type mice. ANDRO had no effect on migration or in total dendritic length, arborization and orientation of immature neurons, suggesting no effects on early morphological development of newborn neurons. Finally, ANDRO treatment improved the performance of APP/PS1 mice in the object location memory task. This effect was not completely prevented by co-treatment with the anti-mitotic drug TMZ, suggesting that other effects of ANDRO in addition to the increase in neurogenesis might underlie the observed cognitive improvement. Altogether, our data indicate that in APP/PS1 mice ANDRO stimulates neurogenesis in the hippocampus by inducing proliferation of neural precursor cells and improves spatial memory performance.
Collapse
|
41
|
Hu D, Liu Z, Qi X. Mitochondrial Quality Control Strategies: Potential Therapeutic Targets for Neurodegenerative Diseases? Front Neurosci 2021; 15:746873. [PMID: 34867159 PMCID: PMC8633545 DOI: 10.3389/fnins.2021.746873] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 10/19/2021] [Indexed: 12/30/2022] Open
Abstract
Many lines of evidence have indicated the therapeutic potential of rescuing mitochondrial integrity by targeting specific mitochondrial quality control pathways in neurodegenerative diseases, such as Parkinson's disease, Huntington's disease, and Alzheimer's disease. In addition to ATP synthesis, mitochondria are critical regulators of ROS production, lipid metabolism, calcium buffering, and cell death. The mitochondrial unfolded protein response, mitochondrial dynamics, and mitophagy are the three main quality control mechanisms responsible for maintaining mitochondrial proteostasis and bioenergetics. The proper functioning of these complex processes is necessary to surveil and restore mitochondrial homeostasis and the healthy pool of mitochondria in cells. Mitochondrial dysfunction occurs early and causally in disease pathogenesis. A significant accumulation of mitochondrial damage resulting from compromised quality control pathways leads to the development of neuropathology. Moreover, genetic or pharmaceutical manipulation targeting the mitochondrial quality control mechanisms can sufficiently rescue mitochondrial integrity and ameliorate disease progression. Thus, therapies that can improve mitochondrial quality control have great promise for the treatment of neurodegenerative diseases. In this review, we summarize recent progress in the field that underscores the essential role of impaired mitochondrial quality control pathways in the pathogenesis of neurodegenerative diseases. We also discuss the translational approaches targeting mitochondrial function, with a focus on the restoration of mitochondrial integrity, including mitochondrial dynamics, mitophagy, and mitochondrial proteostasis.
Collapse
Affiliation(s)
- Di Hu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Zunren Liu
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Xin Qi
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Center for Mitochondrial Disease, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
42
|
Deciphering the Potential Neuroprotective Effects of Luteolin against Aβ 1- 42-Induced Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22179583. [PMID: 34502488 PMCID: PMC8430819 DOI: 10.3390/ijms22179583] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/17/2022] Open
Abstract
The current study was undertaken to unveil the protective effects of Luteolin, a natural flavonoid, against amyloid-beta (Aβ1–42)-induced neuroinflammation, amyloidogenesis, and synaptic dysfunction in mice. For the development of an AD mouse model, amyloid-beta (Aβ1–42, 5 μL/5 min/mouse) oligomers were injected intracerebroventricularly (i.c.v.) into mice’s brain by using a stereotaxic frame. After that, the mice were treated with Luteolin for two weeks at a dose of 80 mg/kg/day. To monitor the biochemical changes, we conducted western blotting and immunofluorescence analysis. According to our findings, the infusion of amyloid-beta activated c-Jun N-terminal kinases (p-JNK), p38 mitogen-activated protein kinases, glial fibrillary acidic protein (GFAP), and ionized calcium adaptor molecule 1 (Iba-1) in the cortex and hippocampus of the experimental mice; these changes were significantly inhibited in Aβ1–42 + Luteolin-treated mice. Likewise, we also checked the expression of inflammatory markers, such as p-nuclear factor-kB p65 (p-NF-kB p65 (Ser536), tissue necrosis factor (TNF-α), and Interleukin1-β (IL-1β), in Aβ1–42-injected mice brain, which was attenuated in Aβ1–42 + Luteolin-treated mice brains. Further, we investigated the expression of pro- and anti-apoptotic cell death markers such as Bax, Bcl-2, Caspase-3, and Cox-2, which was significantly reduced in Aβ1–42 + Lut-treated mice brains compared to the brains of the Aβ-injected group. The results also indicated that with the administration of Aβ1–42, the expression levels of β-site amyloid precursor protein cleaving enzyme (BACE-1) and amyloid-beta (Aβ1–42) were significantly enhanced, while they were reduced in Aβ1–42 + Luteolin-treated mice. We also checked the expression of synaptic markers such as PSD-95 and SNAP-25, which was significantly enhanced in Aβ1–42 + Lut-treated mice. To unveil the underlying factors responsible for the protective effects of Luteolin against AD, we used a specific JNK inhibitor, which suggested that Luteolin reduced Aβ-associated neuroinflammation and neurodegeneration via inhibition of JNK. Collectively, our results indicate that Luteolin could serve as a novel therapeutic agent against AD-like pathological changes in mice.
Collapse
|
43
|
Kim JH, Lim DK, Suh YH, Chang KA. Long-Term Treatment of Cuban Policosanol Attenuates Abnormal Oxidative Stress and Inflammatory Response via Amyloid Plaques Reduction in 5xFAD Mice. Antioxidants (Basel) 2021; 10:antiox10081321. [PMID: 34439569 PMCID: PMC8389325 DOI: 10.3390/antiox10081321] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/27/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder resulting in cognitive decline or dementia, the number of patients with AD is continuously increasing. Although a lot of great progress has been made in research and development of AD therapeutics, there is no fundamental cure for this disease yet. This study demonstrated the memory-improving effects of Cuban policosanol (PCO) in 5xFAD mice, which is an animal model of AD. Following 4-months of treatment with PCO in 5xFAD mice, we found that the number of amyloid plaques decreased in the brain compared to the vehicle-treated 5xFAD mice. Long-term PCO treatment in 5xFAD mice resulted in the reduction of gliosis and abnormal inflammatory cytokines level (interleukin [IL]-1β, IL-6, and tumor necrosis factor [TNF]-α) in the cortex and hippocampus. Levels of lipid peroxide (4-hydroxynonenal [4-HNE]) and superoxide dismutase (SOD1 and SOD2) levels were also recoverd in the brains of PCO-treated 5xFAD mice. Notably, PCO administration reduced memory deficits in the passive avoidance test, as well as synaptic loss (PSD-95, synaptophysin) in 5xFAD mice. Collectively, we identified the potential effects of PCO as a useful supplement to delay or prevent AD progression by inhibiting the formation of Aβ plaques in the brain.
Collapse
Affiliation(s)
- Jin-Ho Kim
- Department of Health Sciences and Technology, Gachon Advanced Insiue for Healh Sciences & Technology (GAIHST), Gachon University, Incheon 21999, Korea;
| | - Dong-Kyun Lim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (D.-K.L.); (Y.-H.S.)
| | - Yoo-Hun Suh
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (D.-K.L.); (Y.-H.S.)
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul 03080, Korea
| | - Keun-A Chang
- Department of Health Sciences and Technology, Gachon Advanced Insiue for Healh Sciences & Technology (GAIHST), Gachon University, Incheon 21999, Korea;
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (D.-K.L.); (Y.-H.S.)
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 21999, Korea
- Correspondence: ; Tel.: +82-32-899-6411
| |
Collapse
|
44
|
Jabir NR, Rehman MT, Tabrez S, Alserihi RF, AlAjmi MF, Khan MS, Husain FM, Ahmed BA. Identification of Butyrylcholinesterase and Monoamine Oxidase B Targeted Ligands and their Putative Application in Alzheimer's Treatment: A Computational Strategy. Curr Pharm Des 2021; 27:2425-2434. [PMID: 33634754 DOI: 10.2174/1381612827666210226123240] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 02/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND With the burgeoning worldwide aging population, the incidence of Alzheimer's disease (AD) and its associated disorders is continuously rising. To appraise other relevant drug targets that could lead to potent enzyme targeting, 13 previously predicted ligands (shown favorable binding with AChE (acetylcholinesterase) and GSK-3 (glycogen synthase kinase) were screened for targeting 3 different enzymes, namely butyrylcholinesterase (BChE), monoamine oxidase A (MAO-A), and monoamine oxidase B (MAO-B) to possibly meet the unmet medical need of better AD treatment. MATERIALS AND METHODS The study utilized in silico screening of 13 ligands against BChE, MAO-A and MAOB using PyRx-Python prescription 0.8. The visualization of the active interaction of studied compounds with targeted proteins was performed by Discovery Studio 2020 (BIOVIA). RESULTS The computational screening of studied ligands revealed the docking energies in the range of -2.4 to -11.3 kcal/mol for all the studied enzymes. Among the 13 ligands, 8 ligands (55E, 6Z2, 6Z5, BRW, F1B, GVP, IQ6, and X37) showed the binding energies of ≤ -8.0 kcal/mol towards BChE, MAO-A and MAO-B. The ligand 6Z5 was found to be the most potent inhibitor of BChE and MAO-B, with a binding energy of -9.7 and -10.4 kcal mol, respectively. Molecular dynamics simulation of BChE-6Z5 and MAO-B-6Z5 complex confirmed the formation of a stable complex. CONCLUSION Our computational screening, molecular docking, and molecular dynamics simulation studies revealed that the above-mentioned enzymes targeted ligands might expedite the future design of potent anti-AD drugs generated on this chemical scaffold.
Collapse
Affiliation(s)
- Nasimudeen R Jabir
- Department of Biochemistry, Centre for Research and Development, PRIST University, Vallam, Thanjavur, Tamil Nadu, India
| | - Md Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Raed F Alserihi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed F AlAjmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Protein Research Chair, Department of Biochemistry, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, Faculty of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Bakrudeen Ali Ahmed
- Department of Biochemistry, Centre for Research and Development, PRIST University, Vallam, Thanjavur, Tamil Nadu, India
| |
Collapse
|
45
|
Darusman HS, Saepuloh U, Mariya SS, Sajuthi D, Schapiro SJ, Hau J. Increased expression of GAPDH in cynomolgus monkeys with spontaneous cognitive decline and amyloidopathy reminiscent of an Alzheimer's-type disease is reflected in the circulation. Am J Primatol 2021; 83:e23296. [PMID: 34196425 DOI: 10.1002/ajp.23296] [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: 11/01/2020] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 01/09/2023]
Abstract
Previous studies of aging cynomolgus monkeys from our group identified spontaneous age-associated cognitive declines associated with biomarkers and brain lesions reminiscent of Alzheimer's Disease (AD), in a proportion of aged monkeys. However, the molecular mechanisms that underlie the spontaneous amyloid disorders and cognitive declines observed in these affected monkeys have yet to be investigated in detail. Using reverse transcriptase quantitative real time PCR techniques, normalized to the ACTB housekeeping gene, we analyzed the expression patterns of a number of genes which have been implicated in amyloid and tau abnormalities, in well-characterized aged cynomolgus monkeys with cognitive decline. A significantly increased expression of the genes coding for glyceraldehyde 3-phosphate dehydrogenase (GAPDH), was found in aged-cognitive decline monkeys compared to age-matched healthy controls. GAPDH has been implicated in several neurodegenerative diseases and interacts with beta amyloid precursor proteins. These findings provide support for the utilization of cynomolgus macaques in translational preclinical research as valid spontaneous models in experimental investigations of the relationships among aging, cognitive decline, and the neuropathy of AD.
Collapse
Affiliation(s)
- Huda S Darusman
- Primate Research Center, Institute of Research and Community Service, Bogor Agricultural University (IPB University), Bogor, Indonesia.,Faculty of Veterinary Medicine, IPB University, Bogor, Indonesia
| | - Uus Saepuloh
- Primate Research Center, Institute of Research and Community Service, Bogor Agricultural University (IPB University), Bogor, Indonesia
| | - Sela S Mariya
- Primate Research Center, Institute of Research and Community Service, Bogor Agricultural University (IPB University), Bogor, Indonesia
| | - Dondin Sajuthi
- Primate Research Center, Institute of Research and Community Service, Bogor Agricultural University (IPB University), Bogor, Indonesia.,Faculty of Veterinary Medicine, IPB University, Bogor, Indonesia
| | - Steven J Schapiro
- Department of Comparative Medicine, UTMD Anderson Cancer Center, Bastrop, Texas, USA.,Department of Experimental Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jann Hau
- Department of Experimental Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
46
|
Analyzing Olfactory Neuron Precursors Non-Invasively Isolated through NADH FLIM as a Potential Tool to Study Oxidative Stress in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22126311. [PMID: 34204595 PMCID: PMC8231156 DOI: 10.3390/ijms22126311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/29/2021] [Accepted: 04/29/2021] [Indexed: 12/22/2022] Open
Abstract
Among all the proposed pathogenic mechanisms to understand the etiology of Alzheimer’s disease (AD), increased oxidative stress seems to be a robust and early disease feature where many of those hypotheses converge. However, despite the significant lines of evidence accumulated, an effective diagnosis and treatment of AD are not yet available. This limitation might be partially explained by the use of cellular and animal models that recapitulate partial aspects of the disease and do not account for the particular biology of patients. As such, cultures of patient-derived cells of peripheral origin may provide a convenient solution for this problem. Peripheral cells of neuronal lineage such as olfactory neuronal precursors (ONPs) can be easily cultured through non-invasive isolation, reproducing AD-related oxidative stress. Interestingly, the autofluorescence of key metabolic cofactors such as reduced nicotinamide adenine dinucleotide (NADH) can be highly correlated with the oxidative state and antioxidant capacity of cells in a non-destructive and label-free manner. In particular, imaging NADH through fluorescence lifetime imaging microscopy (FLIM) has greatly improved the sensitivity in detecting oxidative shifts with minimal intervention to cell physiology. Here, we discuss the translational potential of analyzing patient-derived ONPs non-invasively isolated through NADH FLIM to reveal AD-related oxidative stress. We believe this approach may potentially accelerate the discovery of effective antioxidant therapies and contribute to early diagnosis and personalized monitoring of this devastating disease.
Collapse
|
47
|
Tajmim A, Cuevas-Ocampo AK, Siddique AB, Qusa MH, King JA, Abdelwahed KS, Sonju JJ, El Sayed KA. (-)-Oleocanthal Nutraceuticals for Alzheimer's Disease Amyloid Pathology: Novel Oral Formulations, Therapeutic, and Molecular Insights in 5xFAD Transgenic Mice Model. Nutrients 2021; 13:nu13051702. [PMID: 34069842 PMCID: PMC8157389 DOI: 10.3390/nu13051702] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/03/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is a complex progressive neurodegenerative disorder affecting humans mainly through the deposition of Aβ-amyloid (Aβ) fibrils and accumulation of neurofibrillary tangles in the brain. Currently available AD treatments only exhibit symptomatic relief but do not generally intervene with the amyloid and tau pathologies. The extra-virgin olive oil (EVOO) monophenolic secoiridoid S-(–)-oleocanthal (OC) showed anti-inflammatory activity through COX system inhibition with potency comparable to the standard non-steroidal anti-inflammatory drug (NSAID) like ibuprofen. OC also showed positive in vitro, in vivo, and clinical therapeutic effects against cardiovascular diseases, many malignancies, and AD. Due to its pungent, astringent, and irritant taste, OC should be formulated in acceptable dosage form before its oral use as a potential nutraceutical. The objective of this study is to develop new OC oral formulations, assess whether they maintained OC activity on the attenuation of β-amyloid pathology in a 5xFAD mouse model upon 4-month oral dosing use. Exploration of potential OC formulations underlying molecular mechanism is also within this study scope. OC powder formulation (OC-PF) and OC-solid dispersion formulation with erythritol (OC-SD) were prepared and characterized using FT-IR spectroscopy, powder X-ray diffraction, and scanning electron microscopy (ScEM) analyses. Both formulations showed an improved OC dissolution profile. OC-PF and OC-SD improved memory deficits of 5xFAD mice in behavioral studies. OC-PF and OC-SD exhibited significant attenuation of the accumulation of Aβ plaques and tau phosphorylation in the brain of 5xFAD female mice. Both formulations markedly suppressed C3AR1 (complement component 3a receptor 1) activity by targeting the downstream marker STAT3. Collectively, these results demonstrate the potential for the application of OC-PF as a prospective nutraceutical or dietary supplement to control the progression of amyloid pathogenesis associated with AD.
Collapse
Affiliation(s)
- Afsana Tajmim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
| | - Areli K. Cuevas-Ocampo
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA 71103, USA; (A.K.C.-O.); (J.A.K.)
| | - Abu Bakar Siddique
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
| | - Mohammed H. Qusa
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
| | - Judy Ann King
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA 71103, USA; (A.K.C.-O.); (J.A.K.)
| | - Khaldoun S. Abdelwahed
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
| | - Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
| | - Khalid A. El Sayed
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
- Correspondence: ; Tel.: +1-318-342-1725
| |
Collapse
|
48
|
Kim SH, Yang S, Lim KH, Ko E, Jang HJ, Kang M, Suh PG, Joo JY. Prediction of Alzheimer's disease-specific phospholipase c gamma-1 SNV by deep learning-based approach for high-throughput screening. Proc Natl Acad Sci U S A 2021; 118:e2011250118. [PMID: 33397809 PMCID: PMC7826347 DOI: 10.1073/pnas.2011250118] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/05/2020] [Indexed: 12/23/2022] Open
Abstract
Exon splicing triggered by unpredicted genetic mutation can cause translational variations in neurodegenerative disorders. In this study, we discover Alzheimer's disease (AD)-specific single-nucleotide variants (SNVs) and abnormal exon splicing of phospholipase c gamma-1 (PLCγ1) gene, using genome-wide association study (GWAS) and a deep learning-based exon splicing prediction tool. GWAS revealed that the identified single-nucleotide variations were mainly distributed in the H3K27ac-enriched region of PLCγ1 gene body during brain development in an AD mouse model. A deep learning analysis, trained with human genome sequences, predicted 14 splicing sites in human PLCγ1 gene, and one of these completely matched with an SNV in exon 27 of PLCγ1 gene in an AD mouse model. In particular, the SNV in exon 27 of PLCγ1 gene is associated with abnormal splicing during messenger RNA maturation. Taken together, our findings suggest that this approach, which combines in silico and deep learning-based analyses, has potential for identifying the clinical utility of critical SNVs in AD prediction.
Collapse
Affiliation(s)
- Sung-Hyun Kim
- Neurodegenerative Disease Research Group, 41062 Daegu, Republic of Korea
- Korea Brain Research Institute, 41062 Daegu, Republic of Korea
| | - Sumin Yang
- Neurodegenerative Disease Research Group, 41062 Daegu, Republic of Korea
- Korea Brain Research Institute, 41062 Daegu, Republic of Korea
| | - Key-Hwan Lim
- Neurodegenerative Disease Research Group, 41062 Daegu, Republic of Korea
- Korea Brain Research Institute, 41062 Daegu, Republic of Korea
| | - Euiseng Ko
- Department of Computer Science, University of Nevada, Las Vegas, NV 89154
| | - Hyun-Jun Jang
- School of Life Sciences, Ulsan National Institute of Science and Technology, 44919 Ulsan, Republic of Korea
| | - Mingon Kang
- Department of Computer Science, University of Nevada, Las Vegas, NV 89154
| | - Pann-Ghill Suh
- Korea Brain Research Institute, 41062 Daegu, Republic of Korea
| | - Jae-Yeol Joo
- Neurodegenerative Disease Research Group, 41062 Daegu, Republic of Korea;
- Korea Brain Research Institute, 41062 Daegu, Republic of Korea
| |
Collapse
|
49
|
Sorby-Adams AJ, Schneider WT, Goncalves RP, Knolle F, Morton AJ. Measuring executive function in sheep (Ovis aries) using visual stimuli in a semi-automated operant system. J Neurosci Methods 2020; 351:109009. [PMID: 33340554 DOI: 10.1016/j.jneumeth.2020.109009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/27/2020] [Accepted: 11/19/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cognitive impairment is a distinguishing feature of many neurodegenerative diseases. The intra-dimensional (ID) extra-dimensional (ED) attentional set shift task is part of a clinical battery of tests used to evaluate executive function in Huntington's and Alzheimer's disease patients. The IDED task, however, has not translated well to pre-clinical rodent models of neurological disease. NEW METHOD The ability to perform executive tasks coupled with a long lifespan makes sheep (Ovis aries) an ideal species for modelling cognitive decline in progressive neurodegenerative conditions. We describe the methodology for testing the performance of sheep in the IDED task using a semi-automated system in which visual stimuli are presented as coloured letters on computer screens. RESULTS During each stage of IDED testing, all sheep (n = 12) learned successfully to discriminate between different colours and letters. Sheep were quick to learn the rules of acquisition at each stage. They required significantly more trials to reach criterion (p < 0.05) and made more errors (p < 0.05) following stimulus reversal, with the exception of the ED shift (p > 0.05). COMPARISON WITH EXISTING METHOD(S) Previous research shows that sheep can perform IDED set shifting in a walk-through maze using solid objects with two changeable dimensions (colour and shape) as the stimuli. Presenting the stimuli on computer screens provides better validity, greater task flexibility and higher throughput than the walk-through maze. CONCLUSION All sheep completed each stage of the task, with a range of abilities expected in an outbred population. The IDED task described is ideally suited as a quantifiable and clinically translatable measure of executive function in sheep.
Collapse
Affiliation(s)
- A J Sorby-Adams
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - W T Schneider
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - R P Goncalves
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - F Knolle
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom; Department of Neurology, Klinikum recht der Isar, Technical University Munich, Munich, Germany
| | - A J Morton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom.
| |
Collapse
|
50
|
Tian M, Li B, Shen L. Hierarchical Self-Assembly Mechanism of Ladder-Like Orientated Aβ40 Single-Stranded Protofibrils into Multistranded Mature Fibrils. ACS Macro Lett 2020; 9:1759-1765. [PMID: 35653679 DOI: 10.1021/acsmacrolett.0c00622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The complex self-assembly processes in three dimensions of Alzheimer's β-peptide (Aβ) amyloid protofibrils into polymorphic mature fibrils, particularly the relative protofibril orientation and packing mechanism, are poorly understood. We report here the identification and quantification of the hierarchical self-assembly details among distinct Aβ40 fibrils, particularly the winding pictures of two, three, and four individual single-stranded protofibrils into two-, three-, and four-stranded mature fibrils, respectively, via cross-sectional analysis of atomic force microscopy (AFM) images. The statistical polymer physics analysis of fibril flexibilities from AFM characterizations as well as molecular dynamics (MD) simulations reveal a ladder-like packing mechanism rather than a closed-packing manner for the interprotofibril association into Aβ40 mature fibrils. Moreover, our MD results show atomic packing polymorphism at the well-packing interfaces even within the same multistranded fibril. This work provides mechanistic insights into the polymorphic transition of single-stranded Aβ40 protofibrils into multistranded mature fibrils at the mesoscopic level, which is useful for a more comprehensive understanding of Alzheimer's β-peptide amyloidosis.
Collapse
Affiliation(s)
- Mengting Tian
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Bei Li
- Research Center for Materials Genome Engineering, School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, China
| | - Lei Shen
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|