1
|
Victor Atoki A, Aja PM, Shinkafi TS, Ondari EN, Adeniyi AI, Fasogbon IV, Dangana RS, Shehu UU, Akin-Adewumi A. Exploring the versatility of Drosophila melanogaster as a model organism in biomedical research: a comprehensive review. Fly (Austin) 2025; 19:2420453. [PMID: 39722550 DOI: 10.1080/19336934.2024.2420453] [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: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/28/2024] Open
Abstract
Drosophila melanogaster is a highly versatile model organism that has profoundly advanced our understanding of human diseases. With more than 60% of its genes having human homologs, Drosophila provides an invaluable system for modelling a wide range of pathologies, including neurodegenerative disorders, cancer, metabolic diseases, as well as cardiac and muscular conditions. This review highlights key developments in utilizing Drosophila for disease modelling, emphasizing the genetic tools that have transformed research in this field. Technologies such as the GAL4/UAS system, RNA interference (RNAi) and CRISPR-Cas9 have enabled precise genetic manipulation, with CRISPR-Cas9 allowing for the introduction of human disease mutations into orthologous Drosophila genes. These approaches have yielded critical insights into disease mechanisms, identified novel therapeutic targets and facilitated both drug screening and toxicological studies. Articles were selected based on their relevance, impact and contribution to the field, with a particular focus on studies offering innovative perspectives on disease mechanisms or therapeutic strategies. Our findings emphasize the central role of Drosophila in studying complex human diseases, underscoring its genetic similarities to humans and its effectiveness in modelling conditions such as Alzheimer's disease, Parkinson's disease and cancer. This review reaffirms Drosophila's critical role as a model organism, highlighting its potential to drive future research and therapeutic advancements.
Collapse
Affiliation(s)
| | - Patrick Maduabuchi Aja
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- Department of Biochemistry, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Erick Nyakundi Ondari
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- School of Pure and Applied Sciences, Department of Biological Sciences, Kisii University, Kisii, Kenya
| | | | | | | | - Umar Uthman Shehu
- Department of Physiology, Kampala International University, Ishaka, Uganda
| | | |
Collapse
|
2
|
Amin MA, Zehravi M, Sweilam SH, Shatu MM, Durgawale TP, Qureshi MS, Durgapal S, Haque MA, Vodeti R, Panigrahy UP, Ahmad I, Khan SL, Emran TB. Neuroprotective potential of epigallocatechin gallate in Neurodegenerative Diseases: Insights into molecular mechanisms and clinical Relevance. Brain Res 2025; 1860:149693. [PMID: 40350140 DOI: 10.1016/j.brainres.2025.149693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis pose significant challenges due to their complex pathophysiology and lack of effective treatments. Green tea, rich in the epigallocatechin gallate (EGCG) polyphenolic component, has demonstrated potential as a neuroprotective agent with numerous medicinal applications. EGCG effectively reduces tau and Aβ aggregation in ND models, promotes autophagy, and targets key signaling pathways like Nrf2-ARE, NF-κB, and MAPK. This review explores the molecular processes that underlie EGCG's neuroprotective properties, including its ability to regulate mitochondrial dysfunction, oxidative stress, neuroinflammation, and protein misfolding. Clinical research indicates that EGCG may enhance cognitive and motor abilities, potentially inhibiting disease progression despite absorption and dose optimization limitations. The substance has been proven to slow the amyloidogenic process, prevent protein aggregation, decrease amyloid cytotoxicity, inhibit fibrillogenesis, and restructure fibrils for synergistic therapeutic effects. The review highlights the potential of EGCG as a natural, multi-targeted strategy for NDs but emphasizes the need for further clinical trials to enhance its therapeutic efficacy.
Collapse
Affiliation(s)
- Md Al Amin
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1216, Bangladesh.
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia.
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo 11829, Egypt
| | - Mst Maharunnasa Shatu
- Department of Botany, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Trupti Pratik Durgawale
- Department of Pharmaceutical Chemistry, Krishna Institute of Pharmacy, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, Maharashtra 415539, India
| | - Mohammad Shamim Qureshi
- Department of Pharmacognosy & Phytochemistry, Anwarul Uloom College of Pharmacy, New Mallepally, Hyderabad 500001, India
| | - Sumit Durgapal
- Department of Pharmaceutics, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Premnagar, Dehradun 248007, Uttarakhand, India
| | | | | | - Uttam Prasad Panigrahy
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Sharuk L Khan
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa 413520, Maharashtra, India
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1216, Bangladesh.
| |
Collapse
|
3
|
Li N, Zhang Z, Shen L, Song G, Tian J, Liu Q, Ni J. Selenium metabolism and selenoproteins function in brain and encephalopathy. SCIENCE CHINA. LIFE SCIENCES 2025; 68:628-656. [PMID: 39546178 DOI: 10.1007/s11427-023-2621-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/09/2024] [Indexed: 11/17/2024]
Abstract
Selenium (Se) is an essential trace element of the utmost importance to human health. Its deficiency induces various disorders. Se species can be absorbed by organisms and metabolized to hydrogen selenide for the biosynthesis of selenoproteins, selenonucleic acids, or selenosugars. Se in mammals mainly acts as selenoproteins to exert their biological functions. The brain ranks highest in the specific hierarchy of organs to maintain the level of Se and the expression of selenoproteins under the circumstances of Se deficiency. Dyshomeostasis of Se and dysregulation of selenoproteins result in encephalopathy such as Alzheimer's disease, Parkinson's disease, depression, amyotrophic lateral sclerosis, and multiple sclerosis. This review provides a summary and discussion of Se metabolism, selenoprotein function, and their roles in modulating brain diseases based on the most currently published literature. It focuses on how Se is utilized and transported to the brain, how selenoproteins are biosynthesized and function physiologically in the brain, and how selenoproteins are involved in neurodegenerative diseases. At the end of this review, the perspectives and problems are outlined regarding Se and selenoproteins in the regulation of encephalopathy.
Collapse
Affiliation(s)
- Nan Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Zhonghao Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| | - Liming Shen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Guoli Song
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| | - Jing Tian
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China.
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China.
| | - Jiazuan Ni
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| |
Collapse
|
4
|
Yang M, Zhang X, Zhang D, Zhang Y, Wang J, Zhang Y, Gu C, Zhang X, Wei L. Body Fluid Biomarkers of Neurological Injury in HIV-1-Associated Neurocognitive Disorder. AIDS Res Hum Retroviruses 2025. [PMID: 39938886 DOI: 10.1089/aid.2024.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Since combined antiretroviral therapy for human immunodeficiency virus-associated neurocognitive dysfunction (HAND) only slows the disease's progression, early identification and timely intervention are crucial for effective therapy. In this article, we review the latest evidence in body fluid biomarkers of HAND, providing an overview of research conducted on cerebrospinal fluid and blood samples to draw conclusions on promising biomarkers. Although the significance of biomarkers such as amyloid metabolites, tau proteins, neurofilament light chain, myelin oligodendrocyte glycoprotein, and brain-derived neurotrophic factor in the early detection of HAND may not be immediately clear, they could potentially play a crucial role in evaluating prognosis and tracking the effectiveness of treatment.
Collapse
Affiliation(s)
- Meijuan Yang
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, China
| | - Xiaomei Zhang
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, China
| | - Dong Zhang
- Department of Anesthesiology, Gansu Provincial Hospital, Lanzhou, China
| | - Yamin Zhang
- Department of Neurology, Gansu Provincial Hospital, Lanzhou, China
| | - Jiamei Wang
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, China
| | - Yi Zhang
- Department of Neurology, Gansu Provincial Hospital, Lanzhou, China
| | - Cheng Gu
- Department of Neurology, Gansu Provincial Hospital, Lanzhou, China
| | - Xingwang Zhang
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, China
| | - Lianhua Wei
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
5
|
Krishnamurthy HK, Jayaraman V, Krishna K, Wang T, Bei K, Changalath C, Rajasekaran JJ. An overview of the genes and biomarkers in Alzheimer's disease. Ageing Res Rev 2025; 104:102599. [PMID: 39612989 DOI: 10.1016/j.arr.2024.102599] [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: 09/03/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and neurodegenerative disease characterized by neurofibrillary tangles (NFTs) and amyloid plaque. Familial AD is caused by mutations in the APP, PSEN1, and PSEN2 genes and these mutations result in the early onset of the disease. Sporadic AD usually affects older adults over the age of 65 years and is, therefore classified as late-onset AD (LOAD). Several risk factors associated with LOAD including the APOE gene have been identified. Moreover, GWAS studies have identified a wide array of genes and polymorphisms that are associated with LOAD risk. Currently, the diagnosis of AD involves the evaluation of memory and personality changes, cognitive impairment, and medical and family history to rule out other diseases. Laboratory tests to assess the biomarkers in the body fluids as well as MRI, CT, and PET scans to analyze the presence of plaques and NFTs are also included in the diagnosis of AD. It is important to diagnose AD before the onset of clinical symptoms, i.e. during the preclinical stage, to delay the progression and for better management of the disease. Research has been conducted to identify biomarkers of AD in the CSF, serum, saliva, and urine during the preclinical stage. Current research has identified several biomarkers and potential biomarkers in the body fluids that enhance diagnostic accuracy. Aside from genetics, other factors such as diet, physical activity, and lifestyle factors may influence the risk of developing AD. Clinical trials are underway to find potential biomarkers, diagnostic measures, and treatments for AD mainly in the preclinical stage. This review provides an overview of the genes and biomarkers of AD.
Collapse
Affiliation(s)
| | | | - Karthik Krishna
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | - Tianhao Wang
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | - Kang Bei
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | | | | |
Collapse
|
6
|
Lin P, Xu J, Yang F, Li D, Zhang R, Jiang Y, Zheng T. Elevated concentrations of amyloid-β oligomers and their proapoptotic effects on age-related cataract. FASEB J 2024; 38:e23861. [PMID: 39247969 DOI: 10.1096/fj.202301281rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 09/10/2024]
Abstract
Recently, amyloid-β oligomers (AβOs) have been studied as the primary pathogenic substances in Alzheimer's disease (AD). Our previous study revealed that the Aβ expression level is closely related to ARC progression. Here, we demonstrated that the accumulation of AβOs in the lens epithelium of age-related cataract (ARC) patients increased during ARC progression and that this alteration was consistent with the changes in mitochondrial function, oxidative stress, and cellular apoptosis. In vitro, human lens epithelial cells (HLECs) treated with AβOs exhibited Ca2+ dyshomeostasis, impaired mitochondrial function, elevated oxidative stress levels, and increased apoptosis. Moreover, the proapoptotic effect of AβOs was alleviated after the uptake of mitochondrial Ca2+ was inhibited. These results establish that AβOs may promote HLEC apoptosis by inducing mitochondrial Ca2+ overload, thus preliminarily revealing the possible association between the accumulation of AβOs and other pathological processes in ARC.
Collapse
Affiliation(s)
- Peimin Lin
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jie Xu
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Fan Yang
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Dan Li
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Rong Zhang
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yongxiang Jiang
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Tianyu Zheng
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
7
|
Jeon SH, Seong HJ, Kim H, Kim D, Yang KY, Nam SH. Improvement of branched-chain amino acid production by isolated high-producing protease from Bacillus amyloliquefaciens NY130 on isolated soy/whey proteins and their muscle cell protection. Food Chem 2024; 450:139327. [PMID: 38636380 DOI: 10.1016/j.foodchem.2024.139327] [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: 12/30/2023] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Abstract
Branched-chain amino acids (BCAAs) are vital components of human and animal nutrition that contribute to the building blocks of proteins. In this study, 170 protease-producing strains were isolated and screened from soy-fermented foods. Bacillus amyloliquefaciens NY130 was obtained from Cheonggukjang with high production of BCAAs. Optimal production of protease from B. amyloliquefaciens NY130 (protease NY130) was achieved at 42 °C and pH 6.0 for 21 h. It was purified and determined as 27- and 40 kDa. Protease NY130 showed maximum activity at pH 9.0 and 45 °C with Km value of 10.95 mg for ISP and 1.69 mg for WPI. Protease-treated ISP and WPI showed increased sweetness and saltiness via electronic tongue analysis and enhanced the protective effect against oxidative stress in C2C12 myocytes by increasing p-mTOR/mTOR protein expression to 160%. This work possesses potential in producing BCAAs by using protease for utilization in food.
Collapse
Affiliation(s)
- Sae Hyun Jeon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyeon-Jun Seong
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea; Graduate School of International Agricultural Technology, Seoul National University, Gangwon-do, 25354, Republic of Korea
| | - Hayeong Kim
- Institute of Food Industrialization, Institutes of Green Bioscience & Technology, Center for Food and Bioconvergece, Seoul National University, Gangwon-do, 25354, Republic of Korea
| | - Doman Kim
- Graduate School of International Agricultural Technology, Seoul National University, Gangwon-do, 25354, Republic of Korea; Institute of Food Industrialization, Institutes of Green Bioscience & Technology, Center for Food and Bioconvergece, Seoul National University, Gangwon-do, 25354, Republic of Korea
| | - Kwang-Yeol Yang
- Department of Applied Biology, College of Agriculture and Life Science, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Seung-Hee Nam
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea; Institute of Agricultural and Life Science Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
8
|
Nagayama T, Yagishita S, Shibata M, Furuno A, Saito T, Saido TC, Wakatsuki S, Araki T. Transient sleep apnea results in long-lasting increase in β-amyloid generation and tau hyperphosphorylation. Neurosci Res 2024; 205:40-46. [PMID: 38508957 DOI: 10.1016/j.neures.2024.03.003] [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: 12/10/2023] [Revised: 03/07/2024] [Accepted: 03/18/2024] [Indexed: 03/22/2024]
Abstract
Sleep apnea is regarded as an important risk factor in the pathogenesis of Alzheimer disease (AD). Chronic intermittent hypoxia treatment (IHT) given during the sleep period of the circadian cycle in experimental animals is a well-established sleep apnea model. Here we report that transient IHT for 4 days on AD model mice causes Aβ overproduction 2 months after IHT presumably via upregulation of synaptic BACE1, side-by-side with tau hyperphosphorylation. These results suggest that even transient IHT may be sufficient to cause long-lasting changes in the molecules measured as AD biomarkers in the brain.
Collapse
Affiliation(s)
- Takeru Nagayama
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | - Sosuke Yagishita
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | - Megumi Shibata
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | - Akiko Furuno
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, Nagoya, Aichi 467-8601, Japan; Laboratory of Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory of Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Shuji Wakatsuki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan.
| |
Collapse
|
9
|
Bongiorni S, Catalani E, Arisi I, Lazzarini F, Del Quondam S, Brunetti K, Cervia D, Prantera G. Pathological Defects in a Drosophila Model of Alzheimer's Disease and Beneficial Effects of the Natural Product Lisosan G. Biomolecules 2024; 14:855. [PMID: 39062569 PMCID: PMC11274821 DOI: 10.3390/biom14070855] [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: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) brains are histologically marked by the presence of intracellular and extracellular amyloid deposits, which characterize the onset of the disease pathogenesis. Increasing evidence suggests that certain nutrients exert a direct or indirect effect on amyloid β (Aβ)-peptide production and accumulation and, consequently, on AD pathogenesis. We exploited the fruit fly Drosophila melanogaster model of AD to evaluate in vivo the beneficial properties of Lisosan G, a fermented powder obtained from organic whole grains, on the intracellular Aβ-42 peptide accumulation and related pathological phenotypes of AD. Our data showed that the Lisosan G-enriched diet attenuates the production of neurotoxic Aβ peptides in fly brains and reduces neuronal apoptosis. Notably, Lisosan G exerted anti-oxidant effects, lowering brain levels of reactive oxygen species and enhancing mitochondrial activity. These aspects paralleled the increase in autophagy turnover and the inhibition of nucleolar stress. Our results give support to the use of the Drosophila model not only to investigate the molecular genetic bases of neurodegenerative disease but also to rapidly and reliably test the efficiency of potential therapeutic agents and diet regimens.
Collapse
Affiliation(s)
- Silvia Bongiorni
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, 01100 Viterbo, Italy; (S.B.); (F.L.); (G.P.)
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy; (E.C.); (S.D.Q.); (K.B.)
| | - Ivan Arisi
- Bioinformatics Facility, European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, 00161 Rome, Italy;
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 00133 Rome, Italy
| | - Francesca Lazzarini
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, 01100 Viterbo, Italy; (S.B.); (F.L.); (G.P.)
| | - Simona Del Quondam
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy; (E.C.); (S.D.Q.); (K.B.)
| | - Kashi Brunetti
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy; (E.C.); (S.D.Q.); (K.B.)
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy; (E.C.); (S.D.Q.); (K.B.)
| | - Giorgio Prantera
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, 01100 Viterbo, Italy; (S.B.); (F.L.); (G.P.)
| |
Collapse
|
10
|
Stahl A, Tomchik SM. Modeling neurodegenerative and neurodevelopmental disorders in the Drosophila mushroom body. Learn Mem 2024; 31:a053816. [PMID: 38876485 PMCID: PMC11199955 DOI: 10.1101/lm.053816.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/01/2024] [Indexed: 06/16/2024]
Abstract
The common fruit fly Drosophila melanogaster provides a powerful platform to investigate the genetic, molecular, cellular, and neural circuit mechanisms of behavior. Research in this model system has shed light on multiple aspects of brain physiology and behavior, from fundamental neuronal function to complex behaviors. A major anatomical region that modulates complex behaviors is the mushroom body (MB). The MB integrates multimodal sensory information and is involved in behaviors ranging from sensory processing/responses to learning and memory. Many genes that underlie brain disorders are conserved, from flies to humans, and studies in Drosophila have contributed significantly to our understanding of the mechanisms of brain disorders. Genetic mutations that mimic human diseases-such as Fragile X syndrome, neurofibromatosis type 1, Parkinson's disease, and Alzheimer's disease-affect MB structure and function, altering behavior. Studies dissecting the effects of disease-causing mutations in the MB have identified key pathological mechanisms, and the development of a complete connectome promises to add a comprehensive anatomical framework for disease modeling. Here, we review Drosophila models of human neurodevelopmental and neurodegenerative disorders via the effects of their underlying mutations on MB structure, function, and the resulting behavioral alterations.
Collapse
Affiliation(s)
- Aaron Stahl
- Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Seth M Tomchik
- Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa 52242, USA
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, Iowa 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
- Hawk-IDDRC, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
11
|
Sarkar D, Bhunia A. Delineating the Role of GxxxG Motif in Amyloidogenesis: A New Perspective in Targeting Amyloid-Beta Mediated AD Pathogenesis. ACS BIO & MED CHEM AU 2024; 4:4-19. [PMID: 38404748 PMCID: PMC10885112 DOI: 10.1021/acsbiomedchemau.3c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 02/27/2024]
Abstract
The pursuit of a novel structural motif that can shed light on the key functional attributes is a primary focus in the study of protein folding disorders. Decades of research on Alzheimer's disease (AD) have centered on the Amyloid β (Aβ) pathway, highlighting its significance in understanding the disorder. The diversity in the Aβ pathway and the possible silent tracks which are yet to discover, makes it exceedingly intimidating to the interdisciplinary scientific community. Over the course of AD research, Aβ has consistently been at the forefront of scientific inquiry and discussion. In this review, we epitomize the role of a potential structural motif (GxxxG motif) that may provide a new horizon to the Aβ conflict. We emphasize on how comprehensive understanding of this motif from a structure-function perspective may pave the way for designing novel therapeutics intervention in AD and related diseases.
Collapse
Affiliation(s)
- Dibakar Sarkar
- Department of Chemical Sciences, Bose Institute, Unified Academic Campus, Sector V, Salt Lake EN
80, Kolkata 700 091, India
| | - Anirban Bhunia
- Department of Chemical Sciences, Bose Institute, Unified Academic Campus, Sector V, Salt Lake EN
80, Kolkata 700 091, India
| |
Collapse
|
12
|
Dos Santos NCL, Malta SM, Franco RR, Silva HCG, Silva MH, Rodrigues TS, de Oliveira RM, Araújo TN, Augusto SC, Espindola FS, Ueira-Vieira C. Antioxidant and anti-Alzheimer's potential of Tetragonisca angustula (Jataí) stingless bee pollen. Sci Rep 2024; 14:308. [PMID: 38172290 PMCID: PMC10764861 DOI: 10.1038/s41598-023-51091-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/30/2023] [Indexed: 01/05/2024] Open
Abstract
Alzheimer's disease (AD) is considered the leading cause of dementia in the elderly worldwide. It results in progressive memory loss and impairment of cognitive and motor skills, leading to a high degree of disability and dependence. The development of AD is associated with the accumulation of senile plaques in the brain, caused by the amyloidogenic pathway of the disease. Several genetic and biochemical events are linked to AD development, with oxidative stress being one of them. Due to the scarcity of drugs aimed at treating AD, antioxidant compounds are increasingly studied as therapeutic targets for the disease. In this study, we investigate the antioxidant and anti-Alzheimer potential of the Tetragonisca angustula (Jataí) pollen extract in a Drosophila melanogaster Alzheimer's model. For this purpose, we utilized a D. melanogaster AD-like model, which expresses genes related to the amyloidogenic pathway of Alzheimer's disease. We explored the floral origin of the collected pollen, conducted phytochemical prospecting, and evaluated its antioxidant capacity in vitro. In vivo experiments involved assessing the survival and climbing ability of the D. melanogaster AD-like model with various concentrations of the pollen extract. Our findings revealed that the pollen extract of Tetragonisca angustula exhibits a significant antioxidant response and high concentrations of important phytochemicals, such as flavonoids and polyphenols. Furthermore, it enhanced the survival rate of D. melanogaster, and across all concentrations tested, it improved the climbing ability of the flies after 15 days of treatment with methanolic pollen extract. Additionally, the pollen extract reduced the neurodegeneration index in histopathological analysis. Thus, our study demonstrates the potential of Tetragonisca angustula pollen as an important subject for further investigation, aiming to isolate molecules that could potentially serve as therapeutic targets for Alzheimer's disease.
Collapse
Affiliation(s)
- Natalia Carine Lima Dos Santos
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
- Laboratório de Genética, Instituto de Biotecnologia, Universidade Federal de Uberlândia, Rua Acre, Bloco 2E, Sala 226, Uberlândia, MG, 38408-144, Brazil.
| | - Serena Mares Malta
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | | | | | | | - Thayane Nogueira Araújo
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | | | - Carlos Ueira-Vieira
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
- Laboratório de Genética, Instituto de Biotecnologia, Universidade Federal de Uberlândia, Rua Acre, Bloco 2E, Sala 226, Uberlândia, MG, 38408-144, Brazil.
| |
Collapse
|
13
|
Ho A, Ngala B, Yamada C, Garcia C, Duarte C, Akkaoui J, Ciolac D, Nusbaum A, Kochen W, Efremova D, Groppa S, Nathanson L, Bissel S, Oblak A, Kacena MA, Movila A. IL-34 exacerbates pathogenic features of Alzheimer's disease and calvaria osteolysis in triple transgenic (3x-Tg) female mice. Biomed Pharmacother 2023; 166:115435. [PMID: 37666180 DOI: 10.1016/j.biopha.2023.115435] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 09/06/2023] Open
Abstract
Hallmark features of Alzheimer's disease (AD) include elevated accumulation of aggregated Aβ40 and Aβ42 peptides, hyperphosphorylated Tau (p-Tau), and neuroinflammation. Emerging evidence indicated that interleukin-34 (IL-34) contributes to AD and inflammatory osteolysis via the colony-stimulating factor-1 receptor (CSF-1r). In addition, CSF-1r is also activated by macrophage colony-stimulating factor-1 (M-CSF). While the role of M-CSF in bone physiology and pathology is well addressed, it remains controversial whether IL-34-mediated signaling promotes osteolysis, neurodegeneration, and neuroinflammation in relation to AD. In this study, we injected 3x-Tg mice with mouse recombinant IL-34 protein over the calvaria bone every other day for 42 days. Then, behavioral changes, brain pathology, and calvaria osteolysis were evaluated using various behavioral maze and histological assays. We demonstrated that IL-34 administration dramatically elevated AD-like anxiety and memory loss, pathogenic amyloidogenesis, p-Tau, and RAGE expression in female 3x-Tg mice. Furthermore, IL-34 delivery promoted calvaria inflammatory osteolysis compared to the control group. In addition, we also compared the effects of IL-34 and M-CSF on macrophages, microglia, and RANKL-mediated osteoclastogenesis in relation to AD pathology in vitro. We observed that IL-34-exposed SIM-A9 microglia and 3x-Tg bone marrow-derived macrophages released significantly elevated amounts of pro-inflammatory cytokines, TNF-α, IL-1β, and IL-6, compared to M-CSF treatment in vitro. Furthermore, IL-34, but not M-CSF, elevated RANKL-primed osteoclastogenesis in the presence of Aβ40 and Aβ42 peptides in bone marrow derived macrophages isolated from female 3x-Tg mice. Collectively, our data indicated that IL-34 elevates AD-like features, including behavioral changes and neuroinflammation, as well as osteoclastogenesis in female 3x-Tg mice.
Collapse
Affiliation(s)
- Anny Ho
- Department of Oral Sciences and Translational Research, College of Dental Medicine, Nova Southeastern University, Davie, FL, USA
| | - Bidii Ngala
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chiaki Yamada
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christopher Garcia
- Department of Oral Sciences and Translational Research, College of Dental Medicine, Nova Southeastern University, Davie, FL, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Carolina Duarte
- Department of Oral Sciences and Translational Research, College of Dental Medicine, Nova Southeastern University, Davie, FL, USA
| | - Juliet Akkaoui
- Department of Oral Sciences and Translational Research, College of Dental Medicine, Nova Southeastern University, Davie, FL, USA; Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Dumitru Ciolac
- Laboratory of Neurobiology and Medical Genetics, "Nicolae Testemițanu" State University of Medicine and Pharmacology, Chisinau, Republic of Moldova; Department of Neurology, Institute of Emergency Medicine, Chisinau, Republic of Moldova
| | - Amilia Nusbaum
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - William Kochen
- College of Psychology, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Daniela Efremova
- Laboratory of Neurobiology and Medical Genetics, "Nicolae Testemițanu" State University of Medicine and Pharmacology, Chisinau, Republic of Moldova; Department of Neurology, Institute of Emergency Medicine, Chisinau, Republic of Moldova
| | - Stanislav Groppa
- Laboratory of Neurobiology and Medical Genetics, "Nicolae Testemițanu" State University of Medicine and Pharmacology, Chisinau, Republic of Moldova; Department of Neurology, Institute of Emergency Medicine, Chisinau, Republic of Moldova
| | - Lubov Nathanson
- Institute for Neuro Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Stephanie Bissel
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adrian Oblak
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Melissa A Kacena
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexandru Movila
- Department of Oral Sciences and Translational Research, College of Dental Medicine, Nova Southeastern University, Davie, FL, USA; Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA; Institute for Neuro Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA.
| |
Collapse
|
14
|
Lacham-Hartman S, Moshe R, Ben-Zichri S, Shmidov Y, Bitton R, Jelinek R, Papo N. APPI-Derived Cyclic Peptide Enhances Aβ42 Aggregation and Reduces Aβ42-Mediated Membrane Destabilization and Cytotoxicity. ACS Chem Neurosci 2023; 14:3385-3397. [PMID: 37579500 DOI: 10.1021/acschemneuro.3c00208] [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] [Indexed: 08/16/2023] Open
Abstract
An amyloid precursor protein inhibitor (APPI) and amyloid beta 42 (Aβ42) are both subdomains of the human transmembrane amyloid precursor protein (APP). In the brains of patients with Alzheimer's disease (AD), Aβ42 oligomerizes into aggregates of various sizes, with intermediate, low-molecular-weight Aβ42 oligomers currently being held to be the species responsible for the most neurotoxic effects associated with the disease. Strategies to ameliorate the toxicity of these intermediate Aβ42 oligomeric species include the use of short, Aβ42-interacting peptides that either inhibit the formation of the Aβ42 oligomeric species or promote their conversion to high-molecular-weight aggregates. We therefore designed such an Aβ42-interacting peptide that is based on the β-hairpin amino acid sequence of the APPI, which exhibits high similarity to the β-sheet-like aggregation site of Aβ42. Upon tight binding of this 20-mer cyclic peptide to Aβ42 (in a 1:1 molar ratio), the formation of Aβ42 aggregates was enhanced, and consequently, Aβ42-mediated cell toxicity was ameliorated. We showed that in the presence of the cyclic peptide, interactions of Aβ42 with both plasma and mitochondrial membranes and with phospholipid vesicles that mimic these membranes were inhibited. Specifically, the cyclic peptide inhibited Aβ42-mediated mitochondrial membrane depolarization and reduced Aβ42-mediated apoptosis and cell death. We suggest that the cyclic peptide modulates Aβ42 aggregation by enhancing the formation of large aggregates─as opposed to low-molecular-weight intermediates─and as such has the potential for further development as an AD therapeutic.
Collapse
Affiliation(s)
- Shiran Lacham-Hartman
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Reut Moshe
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Shani Ben-Zichri
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Yulia Shmidov
- Department of Chemical Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Ronit Bitton
- Department of Chemical Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
- Ilse Katz Institute for Nanoscale Science &Technology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Raz Jelinek
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
- Ilse Katz Institute for Nanoscale Science &Technology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Niv Papo
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| |
Collapse
|
15
|
Lenz M, Eichler A, Kruse P, Galanis C, Kleidonas D, Andrieux G, Boerries M, Jedlicka P, Müller U, Deller T, Vlachos A. The Amyloid Precursor Protein Regulates Synaptic Transmission at Medial Perforant Path Synapses. J Neurosci 2023; 43:5290-5304. [PMID: 37369586 PMCID: PMC10359033 DOI: 10.1523/jneurosci.1824-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 05/20/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The perforant path provides the primary cortical excitatory input to the hippocampus. Because of its important role in information processing and coding, entorhinal projections to the dentate gyrus have been studied in considerable detail. Nevertheless, synaptic transmission between individual connected pairs of entorhinal stellate cells and dentate granule cells remains to be characterized. Here, we have used mouse organotypic entorhino-hippocampal tissue cultures of either sex, in which the entorhinal cortex (EC) to dentate granule cell (GC; EC-GC) projection is present, and EC-GC pairs can be studied using whole-cell patch-clamp recordings. By using cultures of wild-type mice, the properties of EC-GC synapses formed by afferents from the lateral and medial entorhinal cortex were compared, and differences in short-term plasticity were identified. As the perforant path is severely affected in Alzheimer's disease, we used tissue cultures of amyloid precursor protein (APP)-deficient mice to examine the role of APP at this synapse. APP deficiency altered excitatory neurotransmission at medial perforant path synapses, which was accompanied by transcriptomic and ultrastructural changes. Moreover, presynaptic but not postsynaptic APP deletion through the local injection of Cre-expressing adeno-associated viruses in conditional APPflox/flox tissue cultures increased the neurotransmission efficacy at perforant path synapses. In summary, these data suggest a physiological role for presynaptic APP at medial perforant path synapses that may be adversely affected under altered APP processing conditions.SIGNIFICANCE STATEMENT The hippocampus receives input from the entorhinal cortex via the perforant path. These projections to hippocampal dentate granule cells are of utmost importance for learning and memory formation. Although there is detailed knowledge about perforant path projections, the functional synaptic properties at the level of individual connected pairs of neurons are not well understood. In this study, we investigated the role of APP in mediating functional properties and transmission rules in individually connected neurons using paired whole-cell patch-clamp recordings and genetic tools in organotypic tissue cultures. Our results show that presynaptic APP expression limits excitatory neurotransmission via the perforant path, which could be compromised in pathologic conditions such as Alzheimer's disease.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, 30625 Hannover, Germany
| | - Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Dimitrios Kleidonas
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- German Cancer Consortium, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Peter Jedlicka
- Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt am Main, Germany
| | - Ulrike Müller
- Institute of Pharmacy and Molecular Biotechnology, Functional Genomics, Ruprecht-Karls University Heidelberg, 69120 Heidelberg, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
16
|
Epremyan KK, Mamaev DV, Zvyagilskaya RA. Alzheimer's Disease: Significant Benefit from the Yeast-Based Models. Int J Mol Sci 2023; 24:9791. [PMID: 37372938 DOI: 10.3390/ijms24129791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related, multifaceted neurological disorder associated with accumulation of aggregated proteins (amyloid Aβ and hyperphosphorylated tau), loss of synapses and neurons, and alterations in microglia. AD was recognized by the World Health Organization as a global public health priority. The pursuit of a better understanding of AD forced researchers to pay attention to well-defined single-celled yeasts. Yeasts, despite obvious limitations in application to neuroscience, show high preservation of basic biological processes with all eukaryotic organisms and offer great advantages over other disease models due to the simplicity, high growth rates on low-cost substrates, relatively simple genetic manipulations, the large knowledge base and data collections, and availability of an unprecedented amount of genomic and proteomic toolboxes and high-throughput screening techniques, inaccessible to higher organisms. Research reviewed above clearly indicates that yeast models, together with other, more simple eukaryotic models including animal models, C. elegans and Drosophila, significantly contributed to understanding Aβ and tau biology. These models allowed high throughput screening of factors and drugs that interfere with Aβ oligomerization, aggregation and toxicity, and tau hyperphosphorylation. In the future, yeast models will remain relevant, with a focus on creating novel high throughput systems to facilitate the identification of the earliest AD biomarkers among different cellular networks in order to achieve the main goal-to develop new promising therapeutic strategies to treat or prevent the disease.
Collapse
Affiliation(s)
- Khoren K Epremyan
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33/2, 119071 Moscow, Russia
| | - Dmitry V Mamaev
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33/2, 119071 Moscow, Russia
| | - Renata A Zvyagilskaya
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33/2, 119071 Moscow, Russia
| |
Collapse
|
17
|
Guo ZH, Khattak S, Rauf MA, Ansari MA, Alomary MN, Razak S, Yang CY, Wu DD, Ji XY. Role of Nanomedicine-Based Therapeutics in the Treatment of CNS Disorders. Molecules 2023; 28:1283. [PMID: 36770950 PMCID: PMC9921752 DOI: 10.3390/molecules28031283] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/31/2023] Open
Abstract
Central nervous system disorders, especially neurodegenerative diseases, are a public health priority and demand a strong scientific response. Various therapy procedures have been used in the past, but their therapeutic value has been insufficient. The blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier is two of the barriers that protect the central nervous system (CNS), but are the main barriers to medicine delivery into the CNS for treating CNS disorders, such as brain tumors, Parkinson's disease, Alzheimer's disease, and Huntington's disease. Nanotechnology-based medicinal approaches deliver valuable cargos targeting molecular and cellular processes with greater safety, efficacy, and specificity than traditional approaches. CNS diseases include a wide range of brain ailments connected to short- and long-term disability. They affect millions of people worldwide and are anticipated to become more common in the coming years. Nanotechnology-based brain therapy could solve the BBB problem. This review analyzes nanomedicine's role in medication delivery; immunotherapy, chemotherapy, and gene therapy are combined with nanomedicines to treat CNS disorders. We also evaluated nanotechnology-based approaches for CNS disease amelioration, with the intention of stimulating the immune system by delivering medications across the BBB.
Collapse
Affiliation(s)
- Zi-Hua Guo
- Department of Neurology, Kaifeng Hospital of Traditional Chinese Medicine, No. 54 East Caizhengting St., Kaifeng 475000, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Mohd Ahmar Rauf
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Mohammad N. Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia
| | - Sufyan Razak
- Dow Medical College, John Hopkins Medical Center, School of Medicine, Baltimore, MD 21205, USA
| | - Chang-Yong Yang
- School of Nursing and Health, Henan University, Kaifeng 475004, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- School of Stomatology, Henan University, Kaifeng 475004, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| |
Collapse
|
18
|
Kamikubo Y, Jin H, Zhou Y, Niisato K, Hashimoto Y, Takasugi N, Sakurai T. Ex vivo analysis platforms for monitoring amyloid precursor protein cleavage. Front Mol Neurosci 2023; 15:1068990. [PMID: 36683852 PMCID: PMC9852844 DOI: 10.3389/fnmol.2022.1068990] [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] [Received: 10/13/2022] [Accepted: 12/14/2022] [Indexed: 01/09/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative brain disorder and the most common cause of dementia in the elderly. The presence of large numbers of senile plaques, neurofibrillary tangles, and cerebral atrophy is the characteristic feature of AD. Amyloid β peptide (Aβ), derived from the amyloid precursor protein (APP), is the main component of senile plaques. AD has been extensively studied using methods involving cell lines, primary cultures of neural cells, and animal models; however, discrepancies have been observed between these methods. Dissociated cultures lose the brain's tissue architecture, including neural circuits, glial cells, and extracellular matrix. Experiments with animal models are lengthy and require laborious monitoring of multiple parameters. Therefore, it is necessary to combine these experimental models to understand the pathology of AD. An experimental platform amenable to continuous observation and experimental manipulation is required to analyze long-term neuronal development, plasticity, and progressive neurodegenerative diseases. In the current study, we provide a practical method to slice and cultivate rodent hippocampus to investigate the cleavage of APP and secretion of Aβ in an ex vivo model. Furthermore, we provide basic information on Aβ secretion using slice cultures. Using our optimized method, dozens to hundreds of long-term stable slice cultures can be coordinated simultaneously. Our findings are valuable for analyses of AD mouse models and senile plaque formation culture models.
Collapse
|
19
|
Mathew A, Balaji E V, Pai SRK, Kishore A, Pai V, Pemmireddy R, K S C. Current Drug Targets in Alzheimer's Associated Memory Impairment: A Comprehensive Review. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:255-275. [PMID: 35366787 DOI: 10.2174/1871527321666220401124719] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia among geriatrics. It is a progressive, degenerative neurologic disorder that causes memory and cognition loss. The accumulation of amyloid fibrils and neurofibrillary tangles in the brain of AD patients is a distinguishing feature of the disease. Therefore, most of the current therapeutic goals are targeting inhibition of beta-amyloid synthesis and aggregation as well as tau phosphorylation and aggregation. There is also a loss of the cholinergic neurons in the basal forebrain, and first-generation therapeutic agents were primarily focused on compensating for this loss of neurons. However, cholinesterase inhibitors can only alleviate cognitive symptoms of AD and cannot reduce the progression of the disease. Understanding the molecular and cellular changes associated with AD pathology has advanced significantly in recent decades. The etiology of AD is complex, with a substantial portion of sporadic AD emerging from unknown reasons and a lesser proportion of early-onset familial AD (FAD) caused by a mutation in several genes, such as the amyloid precursor protein (APP), presenilin 1 (PS1), and presenilin 2 (PS2) genes. Hence, efforts are being made to discover novel strategies for these targets for AD therapy. A new generation of AChE and BChE inhibitors is currently being explored and evaluated in human clinical trials for AD symptomatic treatment. Other approaches for slowing the progression of AD include serotonergic modulation, H3 receptor antagonism, phosphodiesterase, COX-2, and MAO-B inhibition. The present review provides an insight into the possible therapeutic strategies and their molecular mechanisms, enlightening the perception of classical and future treatment approaches.
Collapse
Affiliation(s)
- Anna Mathew
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Vignesh Balaji E
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Sreedhara Ranganath K Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Vasudev Pai
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Ramadevi Pemmireddy
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Chandrashekar K S
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| |
Collapse
|
20
|
Sánchez-Alegría K, Arias C. Functional consequences of brain exposure to saturated fatty acids: From energy metabolism and insulin resistance to neuronal damage. Endocrinol Diabetes Metab 2023; 6:e386. [PMID: 36321333 PMCID: PMC9836261 DOI: 10.1002/edm2.386] [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] [Received: 08/08/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Saturated fatty acids (FAs) are the main component of high-fat diets (HFDs), and high consumption has been associated with the development of insulin resistance, endoplasmic reticulum stress and mitochondrial dysfunction in neuronal cells. In particular, the reduction in neuronal insulin signaling seems to underlie the development of cognitive impairments and has been considered a risk factor for Alzheimer's disease (AD). METHODS This review summarized and critically analyzed the research that has impacted the field of saturated FA metabolism in neurons. RESULTS We reviewed the mechanisms for free FA transport from the systemic circulation to the brain and how they impact neuronal metabolism. Finally, we focused on the molecular and the physiopathological consequences of brain exposure to the most abundant FA in the HFD, palmitic acid (PA). CONCLUSION Understanding the mechanisms that lead to metabolic alterations in neurons induced by saturated FAs could help to develop several strategies for the prevention and treatment of cognitive impairment associated with insulin resistance, metabolic syndrome, or type II diabetes.
Collapse
Affiliation(s)
- Karina Sánchez-Alegría
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
21
|
Lordén G, Wozniak JM, Doré K, Dozier LE, Cates-Gatto C, Patrick GN, Gonzalez DJ, Roberts AJ, Tanzi RE, Newton AC. Enhanced activity of Alzheimer disease-associated variant of protein kinase Cα drives cognitive decline in a mouse model. Nat Commun 2022; 13:7200. [PMID: 36418293 PMCID: PMC9684486 DOI: 10.1038/s41467-022-34679-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/01/2022] [Indexed: 11/27/2022] Open
Abstract
Exquisitely tuned activity of protein kinase C (PKC) isozymes is essential to maintaining cellular homeostasis. Whereas loss-of-function mutations are generally associated with cancer, gain-of-function variants in one isozyme, PKCα, are associated with Alzheimer's disease (AD). Here we show that the enhanced activity of one variant, PKCα M489V, is sufficient to rewire the brain phosphoproteome, drive synaptic degeneration, and impair cognition in a mouse model. This variant causes a modest 30% increase in catalytic activity without altering on/off activation dynamics or stability, underscoring that enhanced catalytic activity is sufficient to drive the biochemical, cellular, and ultimately cognitive effects observed. Analysis of hippocampal neurons from PKCα M489V mice reveals enhanced amyloid-β-induced synaptic depression and reduced spine density compared to wild-type mice. Behavioral studies reveal that this mutation alone is sufficient to impair cognition, and, when coupled to a mouse model of AD, further accelerates cognitive decline. The druggability of protein kinases positions PKCα as a promising therapeutic target in AD.
Collapse
Affiliation(s)
- Gema Lordén
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jacob M Wozniak
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kim Doré
- Center for Neural Circuits and Behavior, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Lara E Dozier
- Section of Neurobiology. Division of Biological sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Chelsea Cates-Gatto
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Gentry N Patrick
- Section of Neurobiology. Division of Biological sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - David J Gonzalez
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Amanda J Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Alexandra C Newton
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
22
|
Incebacak Eltemur RD, Nguyen HP, Weber JJ. Calpain-mediated proteolysis as driver and modulator of polyglutamine toxicity. Front Mol Neurosci 2022; 15:1020104. [PMID: 36385755 PMCID: PMC9648470 DOI: 10.3389/fnmol.2022.1020104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 09/22/2023] Open
Abstract
Among posttranslational modifications, directed proteolytic processes have the strongest impact on protein integrity. They are executed by a variety of cellular machineries and lead to a wide range of molecular consequences. Compared to other forms of proteolytic enzymes, the class of calcium-activated calpains is considered as modulator proteases due to their limited proteolytic activity, which changes the structure and function of their target substrates. In the context of neurodegeneration and - in particular - polyglutamine disorders, proteolytic events have been linked to modulatory effects on the molecular pathogenesis by generating harmful breakdown products of disease proteins. These findings led to the formulation of the toxic fragment hypothesis, and calpains appeared to be one of the key players and auspicious therapeutic targets in Huntington disease and Machado Joseph disease. This review provides a current survey of the role of calpains in proteolytic processes found in polyglutamine disorders. Together with insights into general concepts behind toxic fragments and findings in polyglutamine disorders, this work aims to inspire researchers to broaden and deepen the knowledge in this field, which will help to evaluate calpain-mediated proteolysis as a unifying and therapeutically targetable posttranslational mechanism in neurodegeneration.
Collapse
Affiliation(s)
- Rana Dilara Incebacak Eltemur
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Jonasz Jeremiasz Weber
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| |
Collapse
|
23
|
Ou W, Ohno Y, Yang J, Chandrashekar DV, Abdullah T, Sun J, Murphy R, Roules C, Jagadeesan N, Cribbs DH, Sumbria RK. Efficacy and Safety of a Brain-Penetrant Biologic TNF-α Inhibitor in Aged APP/PS1 Mice. Pharmaceutics 2022; 14:2200. [PMID: 36297637 PMCID: PMC9612380 DOI: 10.3390/pharmaceutics14102200] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/04/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
Tumor necrosis factor alpha (TNF-α) plays a vital role in Alzheimer's disease (AD) pathology, and TNF-α inhibitors (TNFIs) modulate AD pathology. We fused the TNF-α receptor (TNFR), a biologic TNFI that sequesters TNF-α, to a transferrin receptor antibody (TfRMAb) to deliver the TNFI into the brain across the blood-brain barrier (BBB). TfRMAb-TNFR was protective in 6-month-old transgenic APP/PS1 mice in our previous work. However, the effects and safety following delayed chronic TfRMAb-TNFR treatment are unknown. Herein, we initiated the treatment when the male APP/PS1 mice were 10.7 months old (delayed treatment). Mice were injected intraperitoneally with saline, TfRMAb-TNFR, etanercept (non-BBB-penetrating TNFI), or TfRMAb for ten weeks. Biologic TNFIs did not alter hematology indices or tissue iron homeostasis; however, TfRMAb altered hematology indices, increased splenic iron transporter expression, and increased spleen and liver iron. TfRMAb-TNFR and etanercept reduced brain insoluble-amyloid beta (Aβ) 1-42, soluble-oligomeric Aβ, and microgliosis; however, only TfRMAb-TNFR reduced Aβ peptides, Thioflavin-S-positive Aβ plaques, and insoluble-oligomeric Aβ and increased plaque-associated phagocytic microglia. Accordingly, TfRMAb-TNFR improved spatial reference memory and increased BBB-tight junction protein expression, whereas etanercept did not. Overall, despite delayed treatment, TfRMAb-TNFR resulted in a better therapeutic response than etanercept without any TfRMAb-related hematology- or iron-dysregulation in aged APP/PS1 mice.
Collapse
Affiliation(s)
- Weijun Ou
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | - Yuu Ohno
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Dr, Claremont, CA 91711, USA
| | - Joshua Yang
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Dr, Claremont, CA 91711, USA
| | - Devaraj V. Chandrashekar
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | - Tamara Abdullah
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | - Jiahong Sun
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | - Riley Murphy
- Crean College of Health and Behavioral Sciences, Chapman University, Irvine, CA 92618, USA
| | - Chuli Roules
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | - Nataraj Jagadeesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | - David H. Cribbs
- MIND Institute, University of California, Irvine, CA 92697, USA
| | - Rachita K. Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
- Department of Neurology, University of California, Irvine, CA 92868, USA
| |
Collapse
|
24
|
The Role of Vitamin D in Alzheimer’s Disease: A Transcriptional Regulator of Amyloidopathy and Gliopathy. Biomedicines 2022; 10:biomedicines10081824. [PMID: 36009371 PMCID: PMC9404847 DOI: 10.3390/biomedicines10081824] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by amyloid-beta (Aβ) accumulation and cognitive mental decline. Epidemiological studies have suggested an association between low serum vitamin D levels and an increased risk of AD. Vitamin D regulates gene expression via the vitamin D receptor, a nuclear ligand-dependent transcription factor. However, the molecular mechanism underlying the pathogenic and therapeutic effects of vitamin D on AD is not fully understood yet. To better understand how vitamin D regulates the expression of genes related to AD pathology, first, we induced vitamin D deficiency in 5xFAD mice by providing a vitamin-D-deficient diet and observed the changes in the mRNA level of genes related to Aβ processing, which resulted in an increase in the Aβ load in the brain. The vitamin D-deficient diet also suppressed the expression of genes for microglial Aβ phagocytosis. Interestingly, vitamin D deficiency in the early stage of AD resulted in earlier memory impairment. In addition, we administered vitamin D intraperitoneally to 5xFAD mice with a normal diet and found lower Aβ levels with the suppressed expression of genes for Aβ generation and observed improved memory function, which may be potentially associated with reduced MAO-B expression. These findings strongly suggest the role of vitamin D as a crucial disease-modifying factor that may modulate the amyloid pathology with regard to reducing AD symptoms.
Collapse
|
25
|
Xiao M, Xiang W, Chen Y, Peng N, Du X, Lu S, Zuo Y, Li B, Hu Y, Li X. DHA Ameliorates Cognitive Ability, Reduces Amyloid Deposition, and Nerve Fiber Production in Alzheimer's Disease. Front Nutr 2022; 9:852433. [PMID: 35782939 PMCID: PMC9240638 DOI: 10.3389/fnut.2022.852433] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background The etiology of Alzheimer's disease (AD) is very complex. Docosahexaenoic acid (DHA) is important in cognitive ability and nervous system development. A limited number of studies have evaluated the efficacy of DHA in the treatment of AD. Introduction We detected neurofibrillary tangles (NFT) in the hippocampus and cortex of transgenic mice brain through silver glycine staining. We determined the activity of neurons by staining Nissl bodies, used liquid NMR to detect metabolites in the brain, and functional magnetic resonance imaging results to observe the connection signal value between brain regions. Materials and Methods We fed 3-month-old APP/PS1 double transgenic mice with DHA mixed feeds for 4 months to assess the effects of DHA on cognitive ability in AD mice through the Morris water maze and open field tests. To evaluate its effects with AD pathology, continuous feeding was done until the mice reached 9 months of age. Results Compared to AD mice, escape latency significantly decreased on the fifth day while swimming speed, target quadrant stay time, and the crossing number of platforms increased by varying degrees after DHA treatment. Brain tissue section staining revealed that DHA significantly reduced Aβ and nerve fibers in the brain of AD mice. Conclusion DHA significantly reduced the deposition of Aβ in the brain and inhibited the production of nerve fibers, thereby increasing cognitive abilities in AD mice. In addition, DHA suppressed blood lipid levels, and restored uric acid and urea levels, implying that DHA is a potential therapeutic option for early AD.
Collapse
Affiliation(s)
- Min Xiao
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| | - Wei Xiang
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| | - Yashu Chen
- Key Laboratory of Oil Crop Biology and Genetic Breeding, Oil Crops Research Institute, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Wuhan, China
| | - Nan Peng
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiubo Du
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Shuhuan Lu
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| | - Yao Zuo
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| | - Boling Li
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| | - Yonggang Hu
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiangyu Li
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| |
Collapse
|
26
|
Advancements in the development of multi-target directed ligands for the treatment of Alzheimer's disease. Bioorg Med Chem 2022; 61:116742. [PMID: 35398739 DOI: 10.1016/j.bmc.2022.116742] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/01/2022] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial irreversible neurological disorder which results in cognitive impairment, loss of cholinergic neurons in synapses of the basal forebrain and neuronal death. Exact pathology of the disease is not yet known however, many hypotheses have been proposed for its treatment. The available treatments including monotherapies and combination therapies are not able to combat the disease effectively because of its complex pathological mechanism. A multipotent drug for AD has the potential to bind or inhibit multiple targets responsible for the progression of the disease like aggregated Aβ, hyperphosphorylated tau proteins, cholinergic and adrenergic receptors, MAO enzymes, overactivated N-methyl-d-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor etc. The traditional approach of one disease-one target-one drug has been rationalized to one drug-multi targets for the chronic diseases like AD and cancer. Thus, over the last decade research focus has been shifted towards the development of multi target directed ligands (MTDLs) which can simultaneously inhibit multiple targets and stop or slow the progression of the disease. The MTDLs can be more effective against AD and eliminate any possibility of drug-drug interactions. Many important active pharmacophore units have been fused, merged or incorporated into different scaffolds to synthesize new potent drugs. In the current article, we have described various hypothesis for AD and effectiveness of the MTDLs treatment strategy is discussed in detail. Different chemical scaffolds and their synthetic strategies have been described and important functionalities are identified in the chemical scaffold that have the potential to bind to the multiple targets. The important leads identified in this study with MTDL characteristics have the potential to be developed as drug candidates for the effective treatment of AD.
Collapse
|
27
|
Multifaceted neuroprotective effects of (-)-epigallocatechin-3-gallate (EGCG) in Alzheimer’s disease: an overview of pre-clinical studies focused on β-amyloid peptide. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2021.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
28
|
David Troncoso F, Alberto Sánchez D, Luján Ferreira M. Production of Plant Proteases and New Biotechnological Applications: An Updated Review. ChemistryOpen 2022; 11:e202200017. [PMID: 35286022 PMCID: PMC8919702 DOI: 10.1002/open.202200017] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
An updated review of emerging plant proteases with potential biotechnological application is presented. Plant proteases show comparable or even greater performance than animal or microbial proteases for by-product valorization through hydrolysis for, for example, cheese whey, bird feathers, collagen, keratinous materials, gelatin, fish protein, and soy protein. Active biopeptides can be obtained as high added value products, which have shown numerous beneficial effects on human health. Plant proteases can also be used for wastewater treatment. The production of new plant proteases is encouraged for the following advantages: low cost of isolation using simple procedures, remarkable stability over a wide range of operating conditions (temperature, pH, salinity, and organic solvents), substantial affinity to a broad variety of substrates, and possibility of immobilization. Vegetable proteases have enormous application potential for the valorization of industrial waste and its conversion into products with high added value through low-cost processes.
Collapse
Affiliation(s)
- Franco David Troncoso
- Departamento de Ingeniería QuímicaUniversidad Nacional del Sur (UNS)Bahía Blanca8000Argentina
- Planta Piloto de Ingeniería QuímicaPLAPIQUI (UNS-CONICET)Bahía Blanca8000Argentina
| | - Daniel Alberto Sánchez
- Departamento de Ingeniería QuímicaUniversidad Nacional del Sur (UNS)Bahía Blanca8000Argentina
- Planta Piloto de Ingeniería QuímicaPLAPIQUI (UNS-CONICET)Bahía Blanca8000Argentina
| | - María Luján Ferreira
- Departamento de QuímicaUniversidad Nacional del Sur (UNS)Bahía Blanca8000Argentina
- Planta Piloto de Ingeniería QuímicaPLAPIQUI (UNS-CONICET)Bahía Blanca8000Argentina
| |
Collapse
|
29
|
Xiao M, Yao C, Liu F, Xiang W, Zuo Y, Feng K, Lu S, Xiang L, Li M, Li X, Du X. Sialic Acid Ameliorates Cognitive Deficits by Reducing Amyloid Deposition, Nerve Fiber Production, and Neuronal Apoptosis in a Mice Model of Alzheimer's Disease. NEUROSCI 2022; 3:28-40. [PMID: 39484667 PMCID: PMC11523747 DOI: 10.3390/neurosci3010002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/22/2021] [Indexed: 11/03/2024] Open
Abstract
(1) Background: As a natural carbohydrate, sialic acid (SA) is helpful for brain development, cognitive ability, and the nervous system, but there are few reports about the effect of SA on Alzheimer's disease (AD). (2) Method: The present study evaluated the effect of SA on cognitive ability, neuronal activity, Aβ formation, and tau hyperphosphorylation in a double transgenic AD (2×Tg-AD) mice model. The 2×Tg-AD mice were randomly divided into four groups: the AD control group, 17 mg/kg SA-treated AD group, 84 mg/kg SA-treated AD group, and 420 mg/kg SA-treated AD group. Mice from all four groups were fed to 7 months of age for the behavioral test and to 9 months of age for the pathological factors investigation. (3) Results: In the Morris water maze, the escape latency significantly decreased on the fifth day in the SA-treated groups. The number of rearing and crossing times in the open field test also increased significantly, compared with the control group. SA treatment significantly reduced amyloid β-peptide (Aβ) and nerve fibers and increased the number of Nissl bodies in the brain of AD mice. (4) Conclusions: SA reduced the neuron damage by reducing Aβ and inhibited tau protein hyperphosphorylation, which improved the cognitive ability and mobility of AD mice.
Collapse
Affiliation(s)
- Min Xiao
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Chuangyu Yao
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China;
| | - Fang Liu
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Wei Xiang
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Yao Zuo
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Kejue Feng
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Shuhuan Lu
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Li Xiang
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Muzi Li
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Xiangyu Li
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Xiubo Du
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
30
|
Vitória JJM, Trigo D, da Cruz E Silva OAB. Revisiting APP secretases: an overview on the holistic effects of retinoic acid receptor stimulation in APP processing. Cell Mol Life Sci 2022; 79:101. [PMID: 35089425 PMCID: PMC11073327 DOI: 10.1007/s00018-021-04090-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide and is characterized by the accumulation of the β-amyloid peptide (Aβ) in the brain, along with profound alterations in phosphorylation-related events and regulatory pathways. The production of the neurotoxic Aβ peptide via amyloid precursor protein (APP) proteolysis is a crucial step in AD development. APP is highly expressed in the brain and is complexly metabolized by a series of sequential secretases, commonly denoted the α-, β-, and γ-cleavages. The toxicity of resulting fragments is a direct consequence of the first cleaving event. β-secretase (BACE1) induces amyloidogenic cleavages, while α-secretases (ADAM10 and ADAM17) result in less pathological peptides. Hence this first cleavage event is a prime therapeutic target for preventing or reverting initial biochemical events involved in AD. The subsequent cleavage by γ-secretase has a reduced impact on Aβ formation but affects the peptides' aggregating capacity. An array of therapeutic strategies are being explored, among them targeting Retinoic Acid (RA) signalling, which has long been associated with neuronal health. Additionally, several studies have described altered RA levels in AD patients, reinforcing RA Receptor (RAR) signalling as a promising therapeutic strategy. In this review we provide a holistic approach focussing on the effects of isoform-specific RAR modulation with respect to APP secretases and discuss its advantages and drawbacks in subcellular AD related events.
Collapse
Affiliation(s)
- José J M Vitória
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Diogo Trigo
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Odete A B da Cruz E Silva
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
31
|
Linking Diabetes Mellitus with Alzheimer's Disease: Bioinformatics Analysis for the Potential Pathways and Characteristic Genes. Biochem Genet 2021; 60:1049-1075. [PMID: 34779951 DOI: 10.1007/s10528-021-10154-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/03/2021] [Indexed: 01/22/2023]
Abstract
As the surging epidemics with significant disability, Alzheimer's disease (AD) and type II diabetes mellitus (T2DM) with microvascular complications are widely prevalent, sharing considerable similarities in putative pathomechanism. Despite a spurt of researches on the biology, knowledge about their interactive mechanisms is still rudimentary. Applying bioinformatics ways to explore the differentially co-expressed genes contributes to achieve our objectives to find new therapeutic targets. In this study, we firstly integrated gene expression omnibus datasets (GSE28146 and GSE43950) to identify differentially expressed genes. The enrichment analysis of pivotal genes, like gene ontology and pathway signaling proceeded subsequently. Besides, the related protein-protein interaction (PPI) network was then constructed. To further explain the inner connections, we ended up unearthing the biological significance of valuable targets. As a result, a set of 712, 630, 487, and 997 genes were differentially identified in T2DM with microvascular complications and AD at incipient, moderate, and severe, respectively. The enrichment analysis involving both diseases implicated the dominance of immune system, especially the noteworthy chemokine signaling. Multiple comparisons confirmed that CACNA2D3, NUMB, and IER3 were simultaneously participate in these two conditions, whose respective associations with neurological and endocrine diseases, and regulators including interacting chemicals, transcription factors, and miRNAs were analyzed. Bioinformatics analysis eventually concluded that immune-related biological functions and pathways closely link AD and T2DM with microvascular complications. Further exploration of the regulatory factors about CACNA2D3, NUMB, and IER3 in neuroendocrine field may provide us a promising direction to discover potential strategies for the comorbidity status.
Collapse
|
32
|
Armbrust F, Bickenbach K, Marengo L, Pietrzik C, Becker-Pauly C. The Swedish dilemma - the almost exclusive use of APPswe-based mouse models impedes adequate evaluation of alternative β-secretases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119164. [PMID: 34699873 DOI: 10.1016/j.bbamcr.2021.119164] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, however incurable so far. It is widely accepted that aggregated amyloid β (Aβ) peptides play a crucial role for the pathogenesis of AD, as they cause neurotoxicity and deposit as so-called Aβ plaques in AD patient brains. Aβ peptides derive from the amyloid precursor protein (APP) upon consecutive cleavage at the β- and γ-secretase site. Hence, mutations in the APP gene are often associated with autosomal dominant inherited AD. Almost thirty years ago, two mutations at the β-secretase site were observed in two Swedish families (termed Swedish APP (APPswe) mutations), which led to early-onset AD. Consequently, APPswe was established in almost every common AD mouse model, as it contributes to early Aβ plaque formation and cognitive impairments. Analyzing these APPswe-based mouse models, the aspartyl protease BACE1 has been evolving as the prominent β-secretase responsible for Aβ release in AD and as the most important therapeutic target for AD treatment. However, with respect to β-secretase processing, the very rare occurring APPswe variant substantially differs from wild-type APP. BACE1 dominates APPswe processing resulting in the release of Aβ1-x, whereas N-terminally truncated Aβ forms are scarcely generated. However, these N-terminally truncated Aβ species such as Aβ2-x, Aβ3-x and Aβ4-x are elevated in AD patient brains and exhibit an increased potential to aggregate compared to Aβ1-x peptides. Proteases such as meprin β, cathepsin B and ADAMTS4 were identified as alternative β-secretases being capable of generating these N-terminally truncated Aβ species from wild-type APP. However, neither meprin β nor cathepsin B are capable of generating N-terminally truncated Aβ peptides from APPswe. Hence, the role of BACE1 for the Aβ formation during AD might be overrepresented through the excessive use of APPswe mouse models. In this review we critically discuss the consideration of BACE1 as the most promising therapeutic target. Shifting the focus of AD research towards alternative β secretases might unveil promising alternatives to BACE1 inhibitors constantly failing in clinical trials due to ineffectiveness and harmful side effects.
Collapse
Affiliation(s)
- Fred Armbrust
- Biochemical Institute, Unit for Degradomics of the Protease Web, University of Kiel, Kiel, Germany
| | - Kira Bickenbach
- Biochemical Institute, Unit for Degradomics of the Protease Web, University of Kiel, Kiel, Germany
| | - Liana Marengo
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Claus Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Christoph Becker-Pauly
- Biochemical Institute, Unit for Degradomics of the Protease Web, University of Kiel, Kiel, Germany.
| |
Collapse
|
33
|
Chen J, Luo B, Zhong BR, Li KY, Wen QX, Song L, Xiang XJ, Zhou GF, Hu LT, Deng XJ, Ma YL, Chen GJ. Sulfuretin exerts diversified functions in the processing of amyloid precursor protein. Genes Dis 2021; 8:867-881. [PMID: 34522714 PMCID: PMC8427253 DOI: 10.1016/j.gendis.2020.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 01/03/2023] Open
Abstract
Sulfuretin is a flavonoid that protects cell from damage induced by reactive oxygen species and inflammation. In this study, we investigated the role of sulfuretin in the processing of amyloid precursor protein (APP), in association with the two catalytic enzymes the α-secretase a disintegrin and metalloproteinase (ADAM10), and the beta-site APP cleaving enzyme 1 (BACE1) that play important roles in the generation of β amyloid protein (Aβ) in Alzheimer's disease (AD). We found that sulfuretin increased the levels of the immature but not the mature form of ADAM10 protein. The enhanced ADAM10 transcription by sulfuretin was mediated by the nucleotides −444 to −300 in the promoter region, and was attenuated by silencing or mutation of transcription factor retinoid X receptor (RXR) and by GW6471, a specific inhibitor of peroxisome proliferator-activated receptor α (PPAR-α). We further found that sulfuretin preferentially increased protein levels of the immature form of APP (im-APP) but significantly reduced those of BACE1, sAPPβ and β-CTF, whereas Aβ1-42 levels were slightly increased. Finally, the effect of sulfuretin on BACE1 and im-APP was selectively attenuated by the translation inhibitor cycloheximide and by lysosomal inhibitor chloroquine, respectively. Taken together, (1) RXR/PPAR-α signaling was involved in sulfuretin-mediated ADAM10 transcription. (2) Alteration of Aβ protein level by sulfuretin was not consistent with that of ADAM10 and BACE1 protein levels, but was consistent with the elevated level of im-APP protein, suggesting that im-APP, an isoform mainly localized to trans-Golgi network, plays an important role in Aβ generation.
Collapse
Affiliation(s)
- Jian Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Biao Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Bi-Rou Zhong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Kun-Yi Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Qi-Xin Wen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Li Song
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Xiao-Jiao Xiang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Gui-Feng Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Li-Tian Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China.,Department of Neurology, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan Province, 637000, PR China
| | - Xiao-Juan Deng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Yuan-Lin Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Guo-Jun Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| |
Collapse
|
34
|
Zhang X, Zhang CM, Prokopenko D, Liang Y, Zhen SY, Weigle IQ, Han W, Aryal M, Tanzi RE, Sisodia SS. An APP ectodomain mutation outside of the Aβ domain promotes Aβ production in vitro and deposition in vivo. J Exp Med 2021; 218:211936. [PMID: 33822840 PMCID: PMC8034382 DOI: 10.1084/jem.20210313] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 12/28/2022] Open
Abstract
Familial Alzheimer’s disease (FAD)–linked mutations in the APP gene occur either within the Aβ-coding region or immediately proximal and are located in exons 16 and 17, which encode Aβ peptides. We have identified an extremely rare, partially penetrant, single nucleotide variant (SNV), rs145081708, in APP that corresponds to a Ser198Pro substitution in exon 5. We now report that in stably transfected cells, expression of APP harboring the S198P mutation (APPS198P) leads to elevated production of Aβ peptides by an unconventional mechanism in which the folding and exit of APPS198P from the endoplasmic reticulum is accelerated. More importantly, coexpression of APP S198P and the FAD-linked PS1ΔE9 variant in the brains of male and female transgenic mice leads to elevated steady-state Aβ peptide levels and acceleration of Aβ deposition compared with age- and gender-matched mice expressing APP and PS1ΔE9. This is the first AD-linked mutation in APP present outside of exons 16 and 17 that enhances Aβ production and deposition.
Collapse
Affiliation(s)
- Xulun Zhang
- Department of Neurobiology, University of Chicago, Chicago, IL
| | - Can Martin Zhang
- Department of Neurology, Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Charlestown, MA
| | - Dmitry Prokopenko
- Department of Neurology, Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Charlestown, MA
| | - Yingxia Liang
- Department of Neurology, Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Charlestown, MA
| | - Sherri Y Zhen
- Department of Neurology, Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Charlestown, MA
| | - Ian Q Weigle
- Department of Neurobiology, University of Chicago, Chicago, IL
| | - Weinong Han
- Department of Neurobiology, University of Chicago, Chicago, IL
| | - Manish Aryal
- Department of Neurobiology, University of Chicago, Chicago, IL
| | - Rudolph E Tanzi
- Department of Neurology, Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Charlestown, MA
| | | |
Collapse
|
35
|
Crivelli SM, Luo Q, Stevens JA, Giovagnoni C, van Kruining D, Bode G, den Hoedt S, Hobo B, Scheithauer AL, Walter J, Mulder MT, Exley C, Mold M, Mielke MM, De Vries HE, Wouters K, van den Hove DLA, Berkes D, Ledesma MD, Verhaagen J, Losen M, Bieberich E, Martinez-Martinez P. CERT L reduces C16 ceramide, amyloid-β levels, and inflammation in a model of Alzheimer's disease. Alzheimers Res Ther 2021; 13:45. [PMID: 33597019 PMCID: PMC7890977 DOI: 10.1186/s13195-021-00780-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 01/31/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Dysregulation of ceramide and sphingomyelin levels have been suggested to contribute to the pathogenesis of Alzheimer's disease (AD). Ceramide transfer proteins (CERTs) are ceramide carriers which are crucial for ceramide and sphingomyelin balance in cells. Extracellular forms of CERTs co-localize with amyloid-β (Aβ) plaques in AD brains. To date, the significance of these observations for the pathophysiology of AD remains uncertain. METHODS A plasmid expressing CERTL, the long isoform of CERTs, was used to study the interaction of CERTL with amyloid precursor protein (APP) by co-immunoprecipitation and immunofluorescence in HEK cells. The recombinant CERTL protein was employed to study interaction of CERTL with amyloid-β (Aβ), Aβ aggregation process in presence of CERTL, and the resulting changes in Aβ toxicity in neuroblastoma cells. CERTL was overexpressed in neurons by adeno-associated virus (AAV) in a mouse model of familial AD (5xFAD). Ten weeks after transduction, animals were challenged with behavior tests for memory, anxiety, and locomotion. At week 12, brains were investigated for sphingolipid levels by mass spectrometry, plaques, and neuroinflammation by immunohistochemistry, gene expression, and/or immunoassay. RESULTS Here, we report that CERTL binds to APP, modifies Aβ aggregation, and reduces Aβ neurotoxicity in vitro. Furthermore, we show that intracortical injection of AAV, mediating the expression of CERTL, decreases levels of ceramide d18:1/16:0 and increases sphingomyelin levels in the brain of male 5xFAD mice. CERTL in vivo over-expression has a mild effect on animal locomotion, decreases Aβ formation, and modulates microglia by decreasing their pro-inflammatory phenotype. CONCLUSION Our results demonstrate a crucial role of CERTL in regulating ceramide levels in the brain, in amyloid plaque formation and neuroinflammation, thereby opening research avenues for therapeutic targets of AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Simone M. Crivelli
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY USA
- Veterans Affairs Medical Center, Lexington, KY 40502 USA
| | - Qian Luo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Jo A.A. Stevens
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Caterina Giovagnoni
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Daan van Kruining
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Gerard Bode
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Sandra den Hoedt
- Department of Internal Medicine, Laboratory Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Barbara Hobo
- Laboratory for Neuroregeneration, Netherlands institute for Neuroscience, Amsterdam, the Netherlands
| | - Anna-Lena Scheithauer
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jochen Walter
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Monique T. Mulder
- Department of Internal Medicine, Laboratory Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, UK
| | - Matthew Mold
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, UK
| | - Michelle M. Mielke
- Division of Epidemiology, Department of Health Science Research, and Department of Neurology, Mayo Clinic Rochester, Rochester, MN USA
| | - Helga E. De Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands
| | - Kristiaan Wouters
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, the Netherlands
| | - Daniel L. A. van den Hove
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Dusan Berkes
- Department of Organic Chemistry, Slovak University of Technology, Radlinského 9, 81237 Bratislava, Slovak Republic
| | - María Dolores Ledesma
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Madrid, Spain
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, Netherlands institute for Neuroscience, Amsterdam, the Netherlands
| | - Mario Losen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY USA
- Veterans Affairs Medical Center, Lexington, KY 40502 USA
| | - Pilar Martinez-Martinez
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
36
|
Latif-Hernandez A, Sabanov V, Ahmed T, Craessaerts K, Saito T, Saido T, Balschun D. The two faces of synaptic failure in App NL-G-F knock-in mice. Alzheimers Res Ther 2020; 12:100. [PMID: 32838792 PMCID: PMC7445922 DOI: 10.1186/s13195-020-00667-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/10/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Intensive basic and preclinical research into Alzheimer's disease (AD) has yielded important new findings, but they could not yet been translated into effective therapies. One of the reasons is the lack of animal models that sufficiently reproduce the complexity of human AD and the response of human brain circuits to novel treatment approaches. As a step in overcoming these limitations, new App knock-in models have been developed that avoid transgenic APP overexpression and its associated side effects. These mice are proposed to serve as valuable models to examine Aß-related pathology in "preclinical AD." METHODS Since AD as the most common form of dementia progresses into synaptic failure as a major cause of cognitive deficits, the detailed characterization of synaptic dysfunction in these new models is essential. Here, we addressed this by extracellular and whole-cell patch-clamp recordings in AppNL-G-F mice compared to AppNL animals which served as controls. RESULTS We found a beginning synaptic impairment (LTP deficit) at 3-4 months in the prefrontal cortex of AppNL-G-F mice that is further aggravated and extended to the hippocampus at 6-8 months. Measurements of miniature EPSCs and IPSCs point to a marked increase in excitatory and inhibitory presynaptic activity, the latter accompanied by a moderate increase in postsynaptic inhibitory function. CONCLUSIONS Our data reveal a marked impairment of primarily postsynaptic processes at the level of synaptic plasticity but the dominance of a presumably compensatory presynaptic upregulation at the level of elementary miniature synaptic function.
Collapse
Affiliation(s)
- Amira Latif-Hernandez
- Brain and Cognition, KU Leuven, Tiensestraat 102, Box 3714, 3000, Leuven, Belgium
- Present Address: Neurology and Neurological Sciences, Stanford Medicine, Stanford, USA
| | - Victor Sabanov
- Brain and Cognition, KU Leuven, Tiensestraat 102, Box 3714, 3000, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Tariq Ahmed
- Brain and Cognition, KU Leuven, Tiensestraat 102, Box 3714, 3000, Leuven, Belgium
- Present Address: Qatar Biomedical Research Institute, Ar-Rayyan, Qatar
| | - Katleen Craessaerts
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for the Biology of Disease, Leuven, Belgium
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan
- Present Address: Department of Neurocognitive Science, Nagoya City University Graduate School of Medical Science, Nagoya, Aichi, Japan
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan
| | - Detlef Balschun
- Brain and Cognition, KU Leuven, Tiensestraat 102, Box 3714, 3000, Leuven, Belgium.
- Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
37
|
Hanif S, Muhammad P, Chesworth R, Rehman FU, Qian RJ, Zheng M, Shi BY. Nanomedicine-based immunotherapy for central nervous system disorders. Acta Pharmacol Sin 2020; 41:936-953. [PMID: 32467570 PMCID: PMC7468531 DOI: 10.1038/s41401-020-0429-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022]
Abstract
Central nervous system (CNS) disorders represent a broad spectrum of brain ailments with short- and long-term disabilities, and nanomedicine-based approaches provide a new therapeutic approach to treating CNS disorders. A variety of potential drugs have been discovered to treat several neuronal disorders; however, their therapeutic success can be limited by the presence of the blood-brain barrier (BBB). Furthermore, unique immune functions within the CNS provide novel target mechanisms for the amelioration of CNS diseases. Recently, various therapeutic approaches have been applied to fight brain-related disorders, with moderate outcomes. Among the various therapeutic strategies, nanomedicine-based immunotherapeutic systems represent a new era that can deliver useful cargo with promising pharmacokinetics. These approaches exploit the molecular and cellular targeting of CNS disorders for enhanced safety, efficacy, and specificity. In this review, we focus on the efficacy of nanomedicines that utilize immunotherapy to combat CNS disorders. Furthermore, we detailed summarize nanomedicine-based pathways for CNS ailments that aim to deliver drugs across the BBB by mimicking innate immune actions. Overview of how nanomedicines can utilize multiple immunotherapy pathways to combat CNS disorders. ![]()
Collapse
|
38
|
Mousa YM, Abdallah IM, Hwang M, Martin DR, Kaddoumi A. Amylin and pramlintide modulate γ-secretase level and APP processing in lipid rafts. Sci Rep 2020; 10:3751. [PMID: 32111883 PMCID: PMC7048857 DOI: 10.1038/s41598-020-60664-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
A major characteristic of Alzheimer's disease (AD) is the accumulation of misfolded amyloid-β (Aβ) peptide. Several studies linked AD with type 2 diabetes due to similarities between Aβ and human amylin. This study investigates the effect of amylin and pramlintide on Aβ pathogenesis and the predisposing molecular mechanism(s) behind the observed effects in TgSwDI mouse, a cerebral amyloid angiopathy (CAA) and AD model. Our findings showed that thirty days of intraperitoneal injection with amylin or pramlintide increased Aβ burden in mice brains. Mechanistic studies revealed both peptides altered the amyloidogenic pathway and increased Aβ production by modulating amyloid precursor protein (APP) and γ-secretase levels in lipid rafts. In addition, both peptides increased levels of B4GALNT1 enzyme and GM1 ganglioside, and only pramlintide increased the level of GM2 ganglioside. Increased levels of GM1 and GM2 gangliosides play an important role in regulating amyloidogenic pathway proteins in lipid rafts. Increased brain Aβ burden by amylin and pramlintide was associated with synaptic loss, apoptosis, and microglia activation. In conclusion, our findings showed amylin or pramlintide increase Aβ levels and related pathology in TgSwDI mice brains, and suggest that increased amylin levels or the therapeutic use of pramlintide could increase the risk of AD.
Collapse
Affiliation(s)
- Youssef M Mousa
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, USA
| | - Ihab M Abdallah
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, USA
| | - Misako Hwang
- Scott-Ritchey Research Center, Auburn University, Auburn, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, Auburn University, Auburn, USA.,Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, USA.,Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, USA. .,Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA.
| |
Collapse
|
39
|
Ahmad SS, Khan S, Kamal MA, Wasi U. The Structure and Function of α, β and γ-Secretase as Therapeutic Target Enzymes in the Development of Alzheimer’s Disease: A Review. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 18:657-667. [DOI: 10.2174/1871527318666191011145941] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/20/2019] [Accepted: 09/04/2019] [Indexed: 12/22/2022]
Abstract
:Alzheimer's disease is a progressive neurodegenerative disorder that affects the central nervous system. There are several factors that cause AD, like, intracellular hyperphosphorylated Tau tangles, collection of extracellular Amyloid-β42 and generation of reactive oxygen species due to mitochondrial dysfunction. This review analyses the most active target of AD and both types of AD-like early-onset AD and late-onset AD. BACE1 is a β-secretase involved in the cleavage of amyloid precursor protein and the pathogenesis of Alzheimer's disease. The presenilin proteins play a critical role in the pathogenesis of Alzheimer malady by intervening the intramembranous cleavage of amyloid precursor protein and the generation of amyloid β. The two homologous proteins PS1 and PS2 speak to the reactant subunits of particular γ-secretase edifices that intercede an assortment of cellular processes. Natural products are common molecular platforms in drug development in AD. Many natural products are being tested in various animal model systems for their role as a potential therapeutic target in AD. Presently, there are a few theories clarifying the early mechanisms of AD pathogenesis. Recently, research advancements in the field of nanotechnology, which utilize macromolecular strategies to make drugs in nanoscale measurements, offer nanotechnology-based diagnostic tools and drug carriers which are highly sensitive for effective drug targeting in the treatment of Alzheimer’s disease.
Collapse
Affiliation(s)
- Syed S. Ahmad
- Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, India
| | - Shahzad Khan
- Wuhan University, School of Medicine, Wuhan, Hubei, China
| | - Mohammad A. Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| | - Umam Wasi
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, India
| |
Collapse
|
40
|
Vyas S, Kothari S, Kachhwaha S. Nootropic medicinal plants: Therapeutic alternatives for Alzheimer’s disease. J Herb Med 2019. [DOI: 10.1016/j.hermed.2019.100291] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
Saadipour K, Tiberi A, Lombardo S, Grajales E, Montroull L, Mañucat-Tan NB, LaFrancois J, Cammer M, Mathews PM, Scharfman HE, Liao FF, Friedman WJ, Zhou XF, Tesco G, Chao MV. Regulation of BACE1 expression after injury is linked to the p75 neurotrophin receptor. Mol Cell Neurosci 2019; 99:103395. [PMID: 31422108 DOI: 10.1016/j.mcn.2019.103395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/24/2019] [Accepted: 08/08/2019] [Indexed: 12/25/2022] Open
Abstract
BACE1 is a transmembrane aspartic protease that cleaves various substrates and it is required for normal brain function. BACE1 expression is high during early development, but it is reduced in adulthood. Under conditions of stress and injury, BACE1 levels are increased; however, the underlying mechanisms that drive BACE1 elevation are not well understood. One mechanism associated with brain injury is the activation of injurious p75 neurotrophin receptor (p75), which can trigger pathological signals. Here we report that within 72 h after controlled cortical impact (CCI) or laser injury, BACE1 and p75 are increased and tightly co-expressed in cortical neurons of mouse brain. Additionally, BACE1 is not up-regulated in p75 null mice in response to focal cortical injury, while p75 over-expression results in BACE1 augmentation in HEK-293 and SY5Y cell lines. A luciferase assay conducted in SY5Y cell line revealed that BACE1 expression is regulated at the transcriptional level in response to p75 transfection. Interestingly, this effect does not appear to be dependent upon p75 ligands including mature and pro-neurotrophins. In addition, BACE1 activity on amyloid precursor protein (APP) is enhanced in SY5Y-APP cells transfected with a p75 construct. Lastly, we found that the activation of c-jun n-terminal kinase (JNK) by p75 contributes to BACE1 up-regulation. This study explores how two injury-induced molecules are intimately connected and suggests a potential link between p75 signaling and the expression of BACE1 after brain injury.
Collapse
Affiliation(s)
- Khalil Saadipour
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA.
| | - Alexia Tiberi
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA; Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7, Pisa, 56126, Italy
| | - Sylvia Lombardo
- Alzheimer's Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Elena Grajales
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA
| | - Laura Montroull
- Department of Biological Sciences, Rutgers Life Sciences Center, Rutgers University, Newark, NJ 07102, USA
| | - Noralyn B Mañucat-Tan
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, South Australia, 5000, Australia
| | - John LaFrancois
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Michael Cammer
- DART Microscopy Laboratory, NYU Langone Medical Center, New York, NY 10016, USA
| | - Paul M Mathews
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Helen E Scharfman
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Francesca-Fang Liao
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Wilma J Friedman
- Department of Biological Sciences, Rutgers Life Sciences Center, Rutgers University, Newark, NJ 07102, USA
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, South Australia, 5000, Australia
| | - Giueseppina Tesco
- Alzheimer's Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Moses V Chao
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA.
| |
Collapse
|
42
|
A fluorescent protein-readout for transcriptional activity reveals regulation of APP nuclear signaling by phosphorylation sites. Biol Chem 2019; 400:1191-1203. [DOI: 10.1515/hsz-2019-0125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/14/2019] [Indexed: 11/15/2022]
Abstract
Abstract
Signaling pathways that originate at the plasma membrane, including regulated intramembrane proteolysis (RIP), enable extracellular cues to control transcription. We modified the yeast Gal4 transcription system to study the nuclear translocation of transcriptionally active complexes using the fluorescent protein citrine (Cit) as a reporter. This enabled highly sensitive quantitative analysis of transcription in situ at the single cell level. The Gal4/UAS-Cit transcription assay displayed a sigmoidal response limited by the number of integrated reporter cassettes. We validated the assay by analyzing nuclear translocation of the amyloid precursor protein (APP) intracellular domain (AICD) and confirmed the requirement of Fe65 for nuclear translocation of AICD. In addition to the strong on-off effects on transcriptional activity, the results of this assay establish that phosphorylation modifies nuclear signaling. The Y682F mutation in APP showed the strongest increase in Cit expression, underscoring its role in regulating Fe65 binding. Together, we established a highly sensitive fluorescent protein-based assay that can monitor transcriptional activity at the single cell level and demonstrate that AICD phosphorylation affects Fe65 nuclear activity. This assay also introduces a platform for future single cell-based drug screening methods for nuclear translocation.
Collapse
|
43
|
Tao CC, Cheng KM, Ma YL, Hsu WL, Chen YC, Fuh JL, Lee WJ, Chao CC, Lee EHY. Galectin-3 promotes Aβ oligomerization and Aβ toxicity in a mouse model of Alzheimer's disease. Cell Death Differ 2019; 27:192-209. [PMID: 31127200 PMCID: PMC7206130 DOI: 10.1038/s41418-019-0348-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 04/13/2019] [Accepted: 05/02/2019] [Indexed: 12/23/2022] Open
Abstract
Amyloid-β (Aβ) oligomers largely initiate the cascade underlying the pathology of Alzheimer's disease (AD). Galectin-3 (Gal-3), which is a member of the galectin protein family, promotes inflammatory responses and enhances the homotypic aggregation of cancer cells. Here, we examined the role and action mechanism of Gal-3 in Aβ oligomerization and Aβ toxicities. Wild-type (WT) and Gal-3-knockout (KO) mice, APP/PS1;WT mice, APP/PS1;Gal-3+/- mice and brain tissues from normal subjects and AD patients were used. We found that Aβ oligomerization is reduced in Gal-3 KO mice injected with Aβ, whereas overexpression of Gal-3 enhances Aβ oligomerization in the hippocampi of Aβ-injected mice. Gal-3 expression shows an age-dependent increase that parallels endogenous Aβ oligomerization in APP/PS1 mice. Moreover, Aβ oligomerization, Iba1 expression, GFAP expression and amyloid plaque accumulation are reduced in APP/PS1;Gal-3+/- mice compared with APP/PS1;WT mice. APP/PS1;Gal-3+/- mice also show better acquisition and retention performance compared to APP/PS1;WT mice. In studying the mechanism underlying Gal-3-promoted Aβ oligomerization, we found that Gal-3 primarily co-localizes with Iba1, and that microglia-secreted Gal-3 directly interacts with Aβ. Gal-3 also interacts with triggering receptor expressed on myeloid cells-2, which then mediates the ability of Gal-3 to activate microglia for further Gal-3 expression. Immunohistochemical analyses show that the distribution of Gal-3 overlaps with that of endogenous Aβ in APP/PS1 mice and partially overlaps with that of amyloid plaque. Moreover, the expression of the Aβ-degrading enzyme, neprilysin, is increased in Gal-3 KO mice and this is associated with enhanced integrin-mediated signaling. Consistently, Gal-3 expression is also increased in the frontal lobe of AD patients, in parallel with Aβ oligomerization. Because Gal-3 expression is dramatically increased as early as 3 months of age in APP/PS1 mice and anti-Aβ oligomerization is believed to protect against Aβ toxicity, Gal-3 could be considered a novel therapeutic target in efforts to combat AD.
Collapse
Affiliation(s)
- Chih-Chieh Tao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Kuang-Min Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Institute of Neuroscience, National Cheng-chi University, Taipei, Taiwan
| | - Yun-Li Ma
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wei-Lun Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yan-Chu Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Jong-Ling Fuh
- Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wei-Ju Lee
- Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Chih-Chang Chao
- Institute of Neuroscience, National Cheng-chi University, Taipei, Taiwan
| | - Eminy H Y Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan. .,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan. .,Institute of Neuroscience, National Cheng-chi University, Taipei, Taiwan.
| |
Collapse
|
44
|
Flores-León M, Pérez-Domínguez M, González-Barrios R, Arias C. Palmitic Acid-Induced NAD + Depletion is Associated with the Reduced Function of SIRT1 and Increased Expression of BACE1 in Hippocampal Neurons. Neurochem Res 2019; 44:1745-1754. [PMID: 31073968 DOI: 10.1007/s11064-019-02810-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 12/15/2022]
Abstract
Increased levels of circulating fatty acids, such as palmitic acid (PA), are associated with the development of obesity, insulin resistance, type-2 diabetes and metabolic syndrome. Furthermore, these diseases are linked to an increased risk of cancer, cardiovascular diseases, mild cognitive impairment and even Alzheimer's disease (AD). However, the precise actions of elevated PA levels on neurons and their association with neuronal metabolic disruption that leads to the expression of pathological markers of AD, such as the overproduction and accumulation of the amyloid-β peptide, represent an area of intense investigation. A possible molecular mechanism involved in the effects of PA may be through dysfunction of the NAD+ sensor enzyme, SIRT1. Therefore, the aim of the present study was to analyze the relationship between the effects of PA metabolism on the function of SIRT1 and the upregulation of BACE1 in cultured hippocampal neurons. PA reduced the total amount of NAD+ in neurons that caused an increase in p65 K310 acetylation due to inhibition of SIRT1 activity and low protein content. Furthermore, BACE1 protein and its activity were increased, and BACE1 was relocated in neurites after PA exposure.
Collapse
Affiliation(s)
- Manuel Flores-León
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico
| | - Martha Pérez-Domínguez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico
| | - Rodrigo González-Barrios
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología (INCan)-Instituto de Investigaciones Biomédicas (IIB), Universidad Nacional Autónoma de México (UNAM), 14080, México, DF, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico.
| |
Collapse
|
45
|
Eremenko E, Mittal K, Berner O, Kamenetsky N, Nemirovsky A, Elyahu Y, Monsonego A. BDNF-producing, amyloid β-specific CD4 T cells as targeted drug-delivery vehicles in Alzheimer's disease. EBioMedicine 2019; 43:424-434. [PMID: 31085101 PMCID: PMC6557914 DOI: 10.1016/j.ebiom.2019.04.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The delivery of therapeutic proteins to selected sites within the central nervous system (CNS) parenchyma is a major challenge in the treatment of various neurodegenerative disorders. As brain-derived neurotrophic factor (BDNF) is reduced in the brain of people with Alzheimer's disease (AD) and its administration has shown promising therapeutic effects in mouse model of the disease, we generated a novel platform for T cell-based BDNF delivery into the brain parenchyma. METHODS We generated amyloid beta-protein (Aβ)-specific CD4 T cells (Aβ-T cells), genetically engineered to express BDNF, and injected them intracerebroventricularly into the 5XFAD mouse model of AD. FINDINGS The BDNF-secreting Aβ-T cells migrated efficiently to amyloid plaques, where they significantly increased the levels of BDNF, its receptor TrkB, and various synaptic proteins known to be reduced in AD. Furthermore, the injected mice demonstrated reduced levels of beta-secretase 1 (BACE1)-a protease essential in the cleavage process of the amyloid precursor protein-and ameliorated amyloid pathology and inflammation within the brain parenchyma. INTERPRETATION A T cell-based delivery of proteins into the brain can serve as a platform to modulate neurotoxic inflammation and to promote neuronal repair in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ekaterina Eremenko
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Kritika Mittal
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Omer Berner
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Nikita Kamenetsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Anna Nemirovsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Yehezqel Elyahu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Alon Monsonego
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
46
|
Liu SY, Ma YL, Hsu WL, Chiou HY, Lee EHY. Protein inhibitor of activated STAT1 Ser 503 phosphorylation-mediated Elk-1 SUMOylation promotes neuronal survival in APP/PS1 mice. Br J Pharmacol 2019; 176:1793-1810. [PMID: 30849179 DOI: 10.1111/bph.14656] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/29/2019] [Accepted: 02/10/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Protein inhibitor of activated STAT1 (PIAS1) is phosphorylated by IKKα at Ser90 in a PIAS1 E3 ligase activity-dependent manner. Whether PIAS1 is also phosphorylated at other residues and the functional significance of these additional phosphorylation events are not known. The transcription factor Elk-1 remains SUMOylated under basal conditions, but the role of Elk-1 SUMOylation in brain is unknown. Here, we examined the functional significance of PIAS1-mediated Elk-1 SUMOylation in Alzheimer's disease (AD) using the APP/PS1 mouse model of AD and amyloid β (Aβ) microinjections in vivo. EXPERIMENTAL APPROACH Novel phosphorylation site(s) on PIAS1 were identified by LC-MS/MS, and MAPK/ERK-mediated phosphorylation of Elk-1 demonstrated using in vitro kinase assays. Elk-1 SUMOylation by PIAS1 in brain was determined using in vitro SUMOylation assays. Apoptosis in hippocampus was assessed by measuring GADD45α expression by western blotting, and apoptosis of hippocampal neurons in APP/PS1 mice was assessed by TUNEL assay. KEY RESULTS Using LC-MS/MS, we identified a novel MAPK/ERK-mediated phosphorylation site on PIAS1 at Ser503 and showed this phosphorylation determines PIAS1 E3 ligase activity. In rat brain, Elk-1 was SUMOylated by PIAS1, which decreased Elk-1 phosphorylation and down-regulated GADD45α expression. Moreover, lentiviral-mediated transduction of Elk-1-SUMO1 reduced the number of hippocampal apoptotic neurons in APP/PS1 mice. CONCLUSIONS AND IMPLICATIONS MAPK/ERK-mediated phosphorylation of PIAS1 at Ser503 determines PIAS1 E3 ligase activity. Moreover, PIAS1 mediates SUMOylation of Elk-1, which functions as an endogenous defence mechanism against Aβ toxicity in vivo. Targeting Elk-1 SUMOylation could be considered a novel therapeutic strategy against AD.
Collapse
Affiliation(s)
- Shau-Yu Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yun-Li Ma
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wei-Lun Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsin-Ying Chiou
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Eminy H Y Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
47
|
Li M, Luo Z, Yu S, Tang Z. Proton pump inhibitor use and risk of dementia: Systematic review and meta-analysis. Medicine (Baltimore) 2019; 98:e14422. [PMID: 30762748 PMCID: PMC6408083 DOI: 10.1097/md.0000000000014422] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/14/2018] [Accepted: 01/11/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Proton pump inhibitors (PPIs) are an established kind of drugs used to the treatment of most acid-related diseases. Some prospective studies have noticed that PPI use was associated with increased dementia risk. However, the results of those studies were inconsistent and controversial. This meta-analysis aims to determine the association of PPI use and risk of dementia among older people. METHODS Relevant articles were systematically identified by searching the PubMed, EMBASE, and Cochrane Library databases from inception to February 2018. Cohort studies that reported the risk of dementia or Alzheimer's disease (AD) among PPI users compared with non-PPI users were included. The quality of studies was assessed using the Newcastle-Ottawa Scale (NOS). The publication bias was detected by a funnel plot and Egger test. The meta-analysis will abstract risk estimates including relative risks (RRs), hazard ratios (HRs), and odds ratios (ORs) with a 95% confidence interval (CI) for the associations between PPI use and dementia or Alzheimer's risk. Study-specific results were pooled using a random-effects model. RESULTS Six cohort studies were selected finally. The pooled RRs of dementia and AD were 1.23 (95% CI: 0.90-1.67) and 1.01 (95% CI: 0.78-1.32), respectively, compared with those of non-PPI use. The Egger test and funnel plot showed no existence of publication bias. Overall, there was no statistically significant association between PPI use and risk of dementia or AD (P >.05). CONCLUSIONS This meta-analysis suggests that there was no statistical association between PPIs use and increased risk of dementia or AD.
Collapse
Affiliation(s)
- Min Li
- Department of Neurology, The Second Affiliated Hospital of Nanchang University
| | - Zheng Luo
- Department of Neurology, The Second Affiliated Hospital of Nanchang University
| | - Sisi Yu
- Department of Nursing School of Nanchang University, Nanchang, PR China
| | - Zhenyu Tang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University
| |
Collapse
|
48
|
Diagnostic and prognostic biomarkers for HAND. J Neurovirol 2019; 25:686-701. [PMID: 30607890 DOI: 10.1007/s13365-018-0705-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 02/06/2023]
Abstract
In 2007, the nosology for HIV-1-associated neurocognitive disorders (HAND) was updated to a primarily neurocognitive disorder. However, currently available diagnostic tools lack the sensitivity and specificity needed for an accurate diagnosis for HAND. Scientists and clinicians, therefore, have been on a quest for an innovative biomarker to diagnose (i.e., diagnostic biomarker) and/or predict (i.e., prognostic biomarker) the progression of HAND in the post-combination antiretroviral therapy (cART) era. The present review examined the utility and challenges of four proposed biomarkers, including neurofilament light (NFL) chain concentration, amyloid (i.e., sAPPα, sAPPβ, amyloid β) and tau proteins (i.e., total tau, phosphorylated tau), resting-state functional magnetic resonance imaging (fMRI), and prepulse inhibition (PPI). Although significant genotypic differences have been observed in NFL chain concentration, sAPPα, sAPPβ, amyloid β, total tau, phosphorylated tau, and resting-state fMRI, inconsistencies and/or assessment limitations (e.g., invasive procedures, lack of disease specificity, cost) challenge their utility as a diagnostic and/or prognostic biomarker for milder forms of neurocognitive impairment (NCI) in the post-cART era. However, critical evaluation of the literature supports the utility of PPI as a powerful diagnostic biomarker with high accuracy (i.e., 86.7-97.1%), sensitivity (i.e., 89.3-100%), and specificity (i.e., 79.5-94.1%). Additionally, the inclusion of multiple CSF and/or plasma markers, rather than a single protein, may provide a more sensitive diagnostic biomarker for HAND; however, a pressing need for additional research remains. Most notably, PPI may serve as a prognostic biomarker for milder forms of NCI, evidenced by its ability to predict later NCI in higher-order cognitive domains with regression coefficients (i.e., r) greater than 0.8. Thus, PPI heralds an opportunity for the development of a brief, noninvasive diagnostic and promising prognostic biomarker for milder forms of NCI in the post-cART era.
Collapse
|
49
|
C-terminal fragments of amyloid precursor proteins increase cofilin phosphorylation by LIM kinase in cultured rat primary neurons. Neuroreport 2019; 30:38-45. [PMID: 30444792 DOI: 10.1097/wnr.0000000000001162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Amyloid precursor proteins (APPs) are processed by β-, γ-, and ε-secretases and caspase-3 to generate C-terminal fragments of APP (APP-CTFs), which may contribute to the pathology of Alzheimer's disease (AD). In addition to amyloid plaques and neurofibrillary tangles, AD brains contain Hirano bodies, which are rod-like structures mostly composed of actin and the actin-binding protein, cofilin. However, the mechanisms underlying the formation of cofilin-actin rods are still unknown. In this study, we aim to elucidate the effects of APP-CTFs on the actin-depolymerizing factor [(ADF)/cofilin]. Our data indicate that transfection with APP-CT99 and APP-CT57 may increase the phosphorylation level of Ser3 of ADF/cofilin and Thr508 of LIM-kinase 1 in rat primary cortical neuronal cultures. S3 peptide, a synthetic peptide competitor of LIM-kinase 1 for ADF/cofilin phosphorylation and an inhibitor of APP-CTFs, induced ADF/cofilin phosphorylation. In comparison with the wild-type mouse, the APP-CT transgenic mouse showed increased immunoreactivity of phosphorylated cofilin (p-cofilin) in the brain. Treatment with DAPT, an inhibitor of γ-secretase, resulted in a decrease in p-cofilin protein level in the group transfected with full-length APP-695. Transfection with the mutant APP-CTF with a deleted YENPTY domain resulted in no significant increase in p-cofilin level. Thus, APP-CTFs induced cofilin phosphorylation to facilitate nuclear translocation. These results suggest a relationship between APP-CTFs and ADF/cofilin that may be suggestive of a new toxic pathway in the pathology of AD.
Collapse
|
50
|
Galvão F, Grokoski KC, da Silva BB, Lamers ML, Siqueira IR. The amyloid precursor protein (APP) processing as a biological link between Alzheimer's disease and cancer. Ageing Res Rev 2019; 49:83-91. [PMID: 30500566 DOI: 10.1016/j.arr.2018.11.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/12/2018] [Accepted: 11/26/2018] [Indexed: 01/07/2023]
Abstract
Aging is a risk factor for several illnesses, such as Alzheimer's Disease and various cancers. However, an inverse correlation between malignancies and Alzheimer's Disease has been suggested. This review addressed the potential role of non-amyloidogenic and amyloidogenic pathways of amyloid precursor protein processing as a relevant biochemical mechanism to clarify this association. Amyloidogenic and non-amyloidogenic pathways have been related to Alzheimer's Disease and certain malignancies, respectively. Several known molecules involved in APP processing, including its regulation and final products, were summarized. Among them some candidate mechanisms emerged, such as extracellular-regulated kinase (Erk) and protein kinase C (PKC). Therefore, the imbalance of APP processing may be involved with the negative correlation between cancer and Alzheimer Disease.
Collapse
|