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
|
Tan FHP, Azzam G, Najimudin N, Shamsuddin S, Zainuddin A, Kasihmuddin MSM. Salvianolic acid B ameliorates Aβ42 toxicity in Aβ42-expressing Drosophila model: behavioral and transcriptomic profiling. Metab Brain Dis 2025; 40:204. [PMID: 40377822 DOI: 10.1007/s11011-025-01625-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases worldwide. It is characterized by the accumulation of amyloid-beta (Aβ) plaques in which Aβ42 is the most toxic and aggressive species. This work investigates the possibility of salvianolic acid B (SalB), a natural compound with established neuroprotective activity, to counteract the Aβ42-induced toxicity in a Drosophila melanogaster model of AD. SalB's effect was assessed in the Aβ42-expressing Drosophila model by measuring three major AD-related behavioural symptoms: eye morphology (cytotoxicity), lifespan, and locomotor activity. The eye assay, longevity, and locomotion assays were employed, followed by RNA sequencing (RNA-seq) to identify molecular alterations following SalB treatment. Aβ42 expression in the Aβ42-expressing Drosophila model resulted in deformed eye morphology, reduced lifespan, and motor impairment. Treatment with SalB restored part of eye morphology, extended lifespan, and improved locomotion. RNA-seq revealed differential gene expression in oxidative phosphorylation, glutathione metabolism, and detoxification processes, suggesting the involvement of antioxidant defence in SalB-mediated neuroprotection. These findings indicate that SalB could be therapeutic for AD and other neurodegenerative disorders, possibly through the modulation of oxidative stress against Aβ42 toxicity. Further research is warranted to address its mechanisms and other uses in neurodegenerative therapy.
Collapse
Affiliation(s)
- Florence Hui Ping Tan
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia.
- School of Biological Sciences, Universiti Sains Malaysia, Georgetown, Penang, 11800, Malaysia.
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, Georgetown, Penang, 11800, Malaysia.
| | - Ghows Azzam
- School of Biological Sciences, Universiti Sains Malaysia, Georgetown, Penang, 11800, Malaysia.
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, Georgetown, Penang, 11800, Malaysia.
- Malaysia Genome and Vaccine Institute (MGVI), National Institutes of Biotechnology Malaysia (NIBM), Jalan Bangi, Kajang, 43000, Selangor, Malaysia.
| | - Nazalan Najimudin
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, Georgetown, Penang, 11800, Malaysia
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Georgetown, Penang, 11800, Malaysia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, Georgetown, Penang, 11800, Malaysia
- Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Georgetown, Penang, 11800, Malaysia
| | - Azalina Zainuddin
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| | | |
Collapse
|
3
|
Timofeeva AM, Aulova KS, Nevinsky GA. Modeling Alzheimer's Disease: A Review of Gene-Modified and Induced Animal Models, Complex Cell Culture Models, and Computational Modeling. Brain Sci 2025; 15:486. [PMID: 40426657 PMCID: PMC12109626 DOI: 10.3390/brainsci15050486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/30/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
Alzheimer's disease, a complex neurodegenerative disease, is characterized by the pathological aggregation of insoluble amyloid β and hyperphosphorylated tau. Multiple models of this disease have been employed to investigate the etiology, pathogenesis, and multifactorial aspects of Alzheimer's disease and facilitate therapeutic development. Mammals, especially mice, are the most common models for studying the pathogenesis of this disease in vivo. To date, the scientific literature has documented more than 280 mouse models exhibiting diverse aspects of Alzheimer's disease pathogenesis. Other mammalian species, including rats, pigs, and primates, have also been utilized as models. Selected aspects of Alzheimer's disease have also been modeled in simpler model organisms, such as Drosophila melanogaster, Caenorhabditis elegans, and Danio rerio. It is possible to model Alzheimer's disease not only by creating genetically modified animal lines but also by inducing symptoms of this neurodegenerative disease. This review discusses the main methods of creating induced models, with a particular focus on modeling Alzheimer's disease on cell cultures. Induced pluripotent stem cell (iPSC) technology has facilitated novel investigations into the mechanistic underpinnings of diverse diseases, including Alzheimer's. Progress in culturing brain tissue allows for more personalized studies on how drugs affect the brain. Recent years have witnessed substantial advancements in intricate cellular system development, including spheroids, three-dimensional scaffolds, and microfluidic cultures. Microfluidic technologies have emerged as cutting-edge tools for studying intercellular interactions, the tissue microenvironment, and the role of the blood-brain barrier (BBB). Modern biology is experiencing a significant paradigm shift towards utilizing big data and omics technologies. Computational modeling represents a powerful methodology for researching a wide array of human diseases, including Alzheimer's. Bioinformatic methodologies facilitate the analysis of extensive datasets generated via high-throughput experimentation. It is imperative to underscore the significance of integrating diverse modeling techniques in elucidating pathogenic mechanisms in their entirety.
Collapse
Affiliation(s)
- Anna M. Timofeeva
- SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk 630090, Russia
| | | | | |
Collapse
|
4
|
Yadav S, Graham A, Al Hammood F, Garbark C, Vasudevan D, Pandey U, Asara JM, Rajasundaram D, Parkhitko AA. Unique tau- and synuclein-dependent metabolic reprogramming in neurons distinct from normal aging. Aging Cell 2024; 23:e14277. [PMID: 39137949 PMCID: PMC11561663 DOI: 10.1111/acel.14277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 08/15/2024] Open
Abstract
Neuronal cells are highly specialized cells and have a specific metabolic profile to support their function. It has been demonstrated that the metabolic profiles of different cells/tissues undergo significant reprogramming with advancing age, which has often been considered a contributing factor towards aging-related diseases including Alzheimer's (AD) and Parkinson's (PD) diseases. However, it is unclear if the metabolic changes associated with normal aging predispose neurons to disease conditions or a distinct set of metabolic alterations happen in neurons in AD or PD which might contribute to disease pathologies. To decipher the changes in neuronal metabolism with age, in AD, or in PD, we performed high-throughput steady-state metabolite profiling on heads in wildtype Drosophila and in Drosophila models relevant to AD and PD. Intriguingly, we found that the spectrum of affected metabolic pathways is dramatically different between normal aging, Tau, or Synuclein overexpressing neurons. Genetic targeting of the purine and glutamate metabolism pathways, which were dysregulated in both old age and disease conditions partially rescued the neurodegenerative phenotype associated with the overexpression of wildtype and mutant tau. Our findings support a "two-hit model" to explain the pathological manifestations associated with AD where both aging- and Tau/Synuclein- driven metabolic reprogramming events cooperate with each other, and targeting both could be a potent therapeutic strategy.
Collapse
Affiliation(s)
- Shweta Yadav
- Aging Institute of UPMC and the University of PittsburghPittsburghPennsylvaniaUSA
| | - Aidan Graham
- Aging Institute of UPMC and the University of PittsburghPittsburghPennsylvaniaUSA
| | - Farazdaq Al Hammood
- Aging Institute of UPMC and the University of PittsburghPittsburghPennsylvaniaUSA
| | - Chris Garbark
- Department of Cell BiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Deepika Vasudevan
- Department of Cell BiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Udai Pandey
- Department of Pediatrics, Children's Hospital of PittsburghUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - John M. Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, and Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Children's Hospital of PittsburghUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Andrey A. Parkhitko
- Aging Institute of UPMC and the University of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
5
|
Sethi P, Bhaskar R, Singh KK, Gupta S, Han SS, Avinash D, Abomughaid MM, Koul A, Rani B, Ghosh S, Jha NK, Sinha JK. Exploring advancements in early detection of Alzheimer's disease with molecular assays and animal models. Ageing Res Rev 2024; 100:102411. [PMID: 38986845 DOI: 10.1016/j.arr.2024.102411] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Alzheimer's Disease (AD) is a challenging neurodegenerative condition, with overwhelming implications for affected individuals and healthcare systems worldwide. Animal models have played a crucial role in studying AD pathogenesis and testing therapeutic interventions. Remarkably, studies on the genetic factors affecting AD risk, such as APOE and TREM2, have provided valuable insights into disease mechanisms. Early diagnosis has emerged as a crucial factor in effective AD management, as demonstrated by clinical studies emphasizing the benefits of initiating treatment at early stages. Novel diagnostic technologies, including RNA sequencing of microglia, offer promising avenues for early detection and monitoring of AD progression. Therapeutic strategies remain to evolve, with a focus on targeting amyloid beta (Aβ) and tau pathology. Advances in animal models, such as APP-KI mice, and the advancement of anti-Aβ drugs signify progress towards more effective treatments. Therapeutically, the focus has shifted towards intricate approaches targeting multiple pathological pathways simultaneously. Strategies aimed at reducing Aβ plaque accumulation, inhibiting tau hyperphosphorylation, and modulating neuroinflammation are actively being explored, both in preclinical models and clinical trials. While challenges continue in developing validated animal models and translating preclinical findings to clinical success, the continuing efforts in understanding AD at molecular, cellular, and clinical levels offer hope for improved management and eventual prevention of this devastating disease.
Collapse
Affiliation(s)
- Paalki Sethi
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune, Maharashtra 411057, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Apurva Koul
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Bindu Rani
- Department of Medicine, National Institute of Medical Sciences, NIMS University, Jaipur, Rajsthan, India
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India.
| | - Niraj Kumar Jha
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab 140401, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | | |
Collapse
|
6
|
Hendricks EL, Linskey N, Smith IR, Liebl FLW. Kismet/CHD7/CHD8 and Amyloid Precursor Protein-like Regulate Synaptic Levels of Rab11 at the Drosophila Neuromuscular Junction. Int J Mol Sci 2024; 25:8429. [PMID: 39125997 PMCID: PMC11313043 DOI: 10.3390/ijms25158429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
The transmembrane protein β-amyloid precursor protein (APP) is central to the pathophysiology of Alzheimer's disease (AD). The β-amyloid hypothesis posits that aberrant processing of APP forms neurotoxic β-amyloid aggregates, which lead to the cognitive impairments observed in AD. Although numerous additional factors contribute to AD, there is a need to better understand the synaptic function of APP. We have found that Drosophila APP-like (APPL) has both shared and non-shared roles at the synapse with Kismet (Kis), a chromatin helicase binding domain (CHD) protein. Kis is the homolog of CHD7 and CHD8, both of which are implicated in neurodevelopmental disorders including CHARGE Syndrome and autism spectrum disorders, respectively. Loss of function mutations in kis and animals expressing human APP and BACE in their central nervous system show reductions in the glutamate receptor subunit, GluRIIC, the GTPase Rab11, and the bone morphogenetic protein (BMP), pMad, at the Drosophila larval neuromuscular junction (NMJ). Similarly, processes like endocytosis, larval locomotion, and neurotransmission are deficient in these animals. Our pharmacological and epistasis experiments indicate that there is a functional relationship between Kis and APPL, but Kis does not regulate appl expression at the larval NMJ. Instead, Kis likely influences the synaptic localization of APPL, possibly by promoting rab11 transcription. These data identify a potential mechanistic connection between chromatin remodeling proteins and aberrant synaptic function in AD.
Collapse
Affiliation(s)
| | | | | | - Faith L. W. Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL 62026, USA
| |
Collapse
|
7
|
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
|
8
|
Taub DG, Woolf CJ. Age-dependent small fiber neuropathy: Mechanistic insights from animal models. Exp Neurol 2024; 377:114811. [PMID: 38723859 PMCID: PMC11131160 DOI: 10.1016/j.expneurol.2024.114811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/07/2024] [Accepted: 05/05/2024] [Indexed: 05/28/2024]
Abstract
Small fiber neuropathy (SFN) is a common and debilitating disease in which the terminals of small diameter sensory axons degenerate, producing sensory loss, and in many patients neuropathic pain. While a substantial number of cases are attributable to diabetes, almost 50% are idiopathic. An underappreciated aspect of the disease is its late onset in most patients. Animal models of human genetic mutations that produce SFN also display age-dependent phenotypes suggesting that aging is an important contributor to the risk of development of the disease. In this review we define how particular sensory neurons are affected in SFN and discuss how aging may drive the disease. We also evaluate how animal models of SFN can define disease mechanisms that will provide insight into early risk detection and suggest novel therapeutic interventions.
Collapse
Affiliation(s)
- Daniel G Taub
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| | - Clifford J Woolf
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Mou W, Tang Y, Huang Y, Wu Z, Cui Y. Upregulation of neuronal ER-phagy improves organismal fitness and alleviates APP toxicity. Cell Rep 2024; 43:114255. [PMID: 38761376 DOI: 10.1016/j.celrep.2024.114255] [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: 12/05/2023] [Revised: 03/31/2024] [Accepted: 05/04/2024] [Indexed: 05/20/2024] Open
Abstract
ER-phagy, a selective autophagy targeting the endoplasmic reticulum (ER) for lysosomal degradation through cargo receptors, plays a critical role in ER quality control and is linked to various diseases. However, its physiological and pathological roles remain largely unclear due to a lack of animal model studies. This study establishes Drosophila as an in vivo ER-phagy model. Starvation triggers ER-phagy across multiple fly tissues. Disturbing ER-phagy by either globally upregulating or downregulating ER-phagy receptors, Atl or Rtnl1, harms the fly. Notably, moderate upregulation of ER-phagy in fly brains by overexpressing Atl or Rtnl1 significantly attenuates age-associated neurodegenerations. Furthermore, in a Drosophila model of Alzheimer's disease expressing human amyloid precursor protein (APP), impaired ER-phagy is observed. Enhancing ER-phagy in the APP-expressing fly brain facilitates APP degradation, significantly alleviating disease symptoms. Therefore, our findings suggest that modulating ER-phagy may offer a therapeutic strategy to treat aging and diseases associated with ER protein aggregation.
Collapse
Affiliation(s)
- Wenqing Mou
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yinglu Tang
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX 75275, USA
| | - Yunpeng Huang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Zhihao Wu
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX 75275, USA.
| | - Yixian Cui
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
10
|
Li L, Wei Z, Tang Y, Jin M, Yao H, Li X, Li Q, Tan J, Xiao B. Icaritin greatly attenuates β-amyloid-induced toxicity in vivo. CNS Neurosci Ther 2024; 30:e14527. [PMID: 37990437 PMCID: PMC11017459 DOI: 10.1111/cns.14527] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/23/2023] Open
Abstract
AIMS The accumulation and deposition of β-amyloid (Aβ) has always been considered a major pathological feature of Alzheimer's disease (AD). The latest and mainstream amyloid cascade hypothesis indicates that all the main pathological changes in AD are attributed to the accumulation of soluble Aβ. However, the exploration of therapeutic drugs for Aβ toxicity has progressed slowly. This study aims to investigate the protective effects of Icaritin on the Aβ-induced Drosophila AD model and its possible mechanism. METHODS To identify the effects of Icaritin on AD, we constructed an excellent Drosophila AD model named Aβarc (arctic mutant Aβ42) Drosophila. Climbing ability, flight ability, and longevity were used to evaluate the effects of Icaritin on AD phenotypes. Aβarc was determined by immunostaining and ELISA. To identify the effects of Icaritin on oxidative stress, we performed the detection of ROS, hydrogen peroxide, MDA, SOD, catalase, GST, and Caspase-3. To identify the effects of Icaritin on energy metabolism, we performed the detection of ATP and lactate. Transcriptome analysis and qRT-PCR verifications were used to detect the genes directly involved in oxidative stress and energy metabolism. Mitochondrial structure and function were detected by an electron microscopy assay, a mitochondrial membrane potential assay, and a mitochondrial respiration assay. RESULTS We discovered that Icaritin almost completely rescues the climbing ability, flight ability, and longevity of Aβarc Drosophila. Aβarc was dramatically reduced by Icaritin treatment. We also found that Icaritin significantly reduces oxidative stress and greatly improves impaired energy metabolism. Importantly, transcriptome analysis and qRT-PCR verifications showed that many key genes, directly involved in oxidative stress and energy metabolism, are restored by Icaritin. Next, we found that Icaritin perfectly restores the integrity of mitochondrial structure and function damaged by Aβarc toxicity. CONCLUSION This study suggested that Icaritin is a potential drug to deal with the toxicity of Aβarc, at least partially realized by restoring the mitochondria/oxidative stress/energy metabolism axis, and holds potential for translation to human AD.
Collapse
Affiliation(s)
- Liangxian Li
- Laboratory of Respiratory DiseaseAffiliated Hospital of Guilin Medical UniversityGuilinChina
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinChina
| | - Zaiwa Wei
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinChina
| | - Yafang Tang
- Clinical Research Center for Neurological Diseases of Guangxi ProvinceAffiliated Hospital of Guilin Medical UniversityGuilinChina
| | - Mingyue Jin
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinChina
| | - Hua Yao
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinChina
| | - Xia Li
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinChina
| | - Qinghua Li
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinChina
- Clinical Research Center for Neurological Diseases of Guangxi ProvinceAffiliated Hospital of Guilin Medical UniversityGuilinChina
- Guangxi Engineering Research Center for Digital Medicine and Clinical TranslationAffiliated Hospital of Guilin Medical UniversityGuilinChina
| | - Jie Tan
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinChina
| | - Bo Xiao
- Laboratory of Respiratory DiseaseAffiliated Hospital of Guilin Medical UniversityGuilinChina
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinChina
- The Key Laboratory of Respiratory DiseasesEducation Department of Guangxi Zhuang Autonomous RegionGuilinChina
| |
Collapse
|
11
|
Nystuen KL, McNamee SM, Akula M, Holton KM, DeAngelis MM, Haider NB. Alzheimer's Disease: Models and Molecular Mechanisms Informing Disease and Treatments. Bioengineering (Basel) 2024; 11:45. [PMID: 38247923 PMCID: PMC10813760 DOI: 10.3390/bioengineering11010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's Disease (AD) is a complex neurodegenerative disease resulting in progressive loss of memory, language and motor abilities caused by cortical and hippocampal degeneration. This review captures the landscape of understanding of AD pathology, diagnostics, and current therapies. Two major mechanisms direct AD pathology: (1) accumulation of amyloid β (Aβ) plaque and (2) tau-derived neurofibrillary tangles (NFT). The most common variants in the Aβ pathway in APP, PSEN1, and PSEN2 are largely responsible for early-onset AD (EOAD), while MAPT, APOE, TREM2 and ABCA7 have a modifying effect on late-onset AD (LOAD). More recent studies implicate chaperone proteins and Aβ degrading proteins in AD. Several tests, such as cognitive function, brain imaging, and cerebral spinal fluid (CSF) and blood tests, are used for AD diagnosis. Additionally, several biomarkers seem to have a unique AD specific combination of expression and could potentially be used in improved, less invasive diagnostics. In addition to genetic perturbations, environmental influences, such as altered gut microbiome signatures, affect AD. Effective AD treatments have been challenging to develop. Currently, there are several FDA approved drugs (cholinesterase inhibitors, Aß-targeting antibodies and an NMDA antagonist) that could mitigate AD rate of decline and symptoms of distress.
Collapse
Affiliation(s)
- Kaden L. Nystuen
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Shannon M. McNamee
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Monica Akula
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Kristina M. Holton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Margaret M. DeAngelis
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Neena B. Haider
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Wu S, Liu X, Yang H, Ma W, Qin Z. The effect of lipid metabolism on age-associated cognitive decline: Lessons learned from model organisms and human. IBRO Neurosci Rep 2023; 15:165-169. [PMID: 38204577 PMCID: PMC10776322 DOI: 10.1016/j.ibneur.2023.08.2194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/26/2023] [Accepted: 08/29/2023] [Indexed: 01/12/2024] Open
Abstract
Lipids are required as integral building blocks of cells to support cellular structures and functions. The intricate mechanisms underpinning lipid homeostasis are essential for the health and maintenance of the central nervous system. Here we summarize the recent advances in dissecting the effect of lipid metabolism on cognitive function and its age-associated decline by reviewing relevant studies ranging from invertebrate model organisms to mammals including human.
Collapse
Affiliation(s)
- Shihao Wu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Department of Geriatric Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xiaoli Liu
- Punan Branch of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
| | - Haiyan Yang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China
| | - Wenlin Ma
- Department of Geriatric Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Shanghai Clinical Research Center for Aging and Medicine, Shanghai 200040, China
| | - Zhao Qin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China
| |
Collapse
|
13
|
Jiang Y, MacNeil LT. Simple model systems reveal conserved mechanisms of Alzheimer's disease and related tauopathies. Mol Neurodegener 2023; 18:82. [PMID: 37950311 PMCID: PMC10638731 DOI: 10.1186/s13024-023-00664-x] [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: 04/02/2023] [Accepted: 10/04/2023] [Indexed: 11/12/2023] Open
Abstract
The lack of effective therapies that slow the progression of Alzheimer's disease (AD) and related tauopathies highlights the need for a more comprehensive understanding of the fundamental cellular mechanisms underlying these diseases. Model organisms, including yeast, worms, and flies, provide simple systems with which to investigate the mechanisms of disease. The evolutionary conservation of cellular pathways regulating proteostasis and stress response in these organisms facilitates the study of genetic factors that contribute to, or protect against, neurodegeneration. Here, we review genetic modifiers of neurodegeneration and related cellular pathways identified in the budding yeast Saccharomyces cerevisiae, the nematode Caenorhabditis elegans, and the fruit fly Drosophila melanogaster, focusing on models of AD and related tauopathies. We further address the potential of simple model systems to better understand the fundamental mechanisms that lead to AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Yuwei Jiang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada.
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada.
| |
Collapse
|
14
|
Tan FHP, Ting ACJ, Najimudin N, Watanabe N, Shamsuddin S, Zainuddin A, Osada H, Azzam G. 3-[[(3S)-1,2,3,4-Tetrahydroisoquinoline-3-Carbonyl]Amino]Propanoic Acid (THICAPA) Is Protective Against Aβ42-Induced Toxicity In Vitro and in an Alzheimer's Disease Drosophila. J Gerontol A Biol Sci Med Sci 2023; 78:1944-1952. [PMID: 37453137 DOI: 10.1093/gerona/glad169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Indexed: 07/18/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia globally. The accumulation of amyloid-beta (Aβ) extracellular senile plaques in the brain is one of the hallmark mechanisms found in AD. Aβ42 is the most damaging and aggressively aggregating Aβ isomer produced in the brain. Although Aβ42 has been extensively researched as a crucial peptide connected to the development of the characteristic amyloid fibrils in AD, the specifics of its pathophysiology are still unknown. Therefore, the main objective was to identify novel compounds that could potentially mitigate the negative effects of Aβ42. 3-[[(3S)-1,2,3,4-Tetrahydroisoquinoline-3-carbonyl]amino]propanoic acid (THICAPA) was identified as a ligand for Aβ42 and for reducing fibrillary Aβ42 aggregation. THICAPA also improved cell viability when administered to PC12 neuronal cells that were exposed to Aβ42. Additionally, this compound diminished Aβ42 toxicity in the current AD Drosophila model by rescuing the rough eye phenotype, prolonging the life span, and enhancing motor functions. Through next-generation RNA-sequencing, immune response pathways were downregulated in response to THICAPA treatment. Thus, this study suggests THICAPA as a possible disease-modifying treatment for AD.
Collapse
Affiliation(s)
- Florence Hui Ping Tan
- School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, Penang, Malaysia
| | | | - Nazalan Najimudin
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, Penang, Malaysia
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Nobumoto Watanabe
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, Penang, Malaysia
- Bioprobe Application Research Unit, RIKEN CSRS, Wako, Saitama, Japan
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
- Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| | - Azalina Zainuddin
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Hiroyuki Osada
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, Penang, Malaysia
- Chemical Biology Research Group, RIKEN CSRS, Wako, Saitama, Japan
| | - Ghows Azzam
- School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia
- Malaysia Genome and Vaccine Institute (MGVI), National Institutes of Biotechnology Malaysia (NIBM), Jalan Bangi, Selangor, Malaysia
| |
Collapse
|
15
|
Tan FHP, Najimudin N, Watanabe N, Shamsuddin S, Azzam G. p-Coumaric acid attenuates the effects of Aβ42 in vitro and in a Drosophila Alzheimer's disease model. Behav Brain Res 2023; 452:114568. [PMID: 37414223 DOI: 10.1016/j.bbr.2023.114568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative condition in civilizations worldwide. The distinctive occurrence of amyloid-beta (Aβ) accumulation into insoluble fibrils is part of the disease pathophysiology with Aβ42 being the most toxic and aggressive Aβ species. The polyphenol, p-Coumaric acid (pCA), has been known to boost a number of therapeutic benefits. Here, pCA's potential to counteract the negative effects of Aβ42 was investigated. First, pCA was confirmed to reduce Aβ42 fibrillation using an in vitro activity assay. The compound was next examined on Aβ42-exposed PC12 neuronal cells and was found to significantly decrease Aβ42-induced cell mortality. pCA was then examined using an AD Drosophila melanogaster model. Feeding of pCA partially reversed the rough eye phenotype, significantly lengthened AD Drosophila's lifespan, and significantly enhanced the majority of the AD Drosophila's mobility in a sex-dependent manner. The findings of this study suggest that pCA may have therapeutic benefits for AD.
Collapse
Affiliation(s)
- Florence Hui Ping Tan
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Penang, Malaysia.
| | - Nazalan Najimudin
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia; USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Nobumoto Watanabe
- USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Penang, Malaysia; Bioprobe Application Research Unit, RIKEN Centre for Sustainable Resource Science, RIKEN, Japan; Chemical Resource Development Research Unit, RIKEN CSRS, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Penang, Malaysia; Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Ghows Azzam
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia; USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Penang, Malaysia; Malaysia Genome and Vaccine Institute (MGVI), National Institutes of Biotechnology Malaysia (NIBM), Jalan Bangi, 43000 Kajang, Selangor, Malaysia.
| |
Collapse
|
16
|
Adedayo BC, Akinniyi ST, Ogunsuyi OB, Oboh G. In the quest for the ideal sweetener: Aspartame exacerbates selected biomarkers in the fruit fly ( Drosophila melanogaster) model of Alzheimer's disease more than sucrose. AGING BRAIN 2023; 4:100090. [PMID: 37559954 PMCID: PMC10407236 DOI: 10.1016/j.nbas.2023.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 08/11/2023] Open
Abstract
This study evaluated the effect of dietary inclusions of aspartame and sucrose on some selected behavioral and biochemical indices linked with Alzheimer's disease in a transgenic fruit fly (Drosophila melanogaster) model expressing human amyloid precursor protein and secretase. Flies were raised on a diet supplemented with sucrose and aspartame for 14 days. Thereafter, the flies were assessed for their survival rate, learning and memory, as well as locomotor performance, 14 days post-treatment. This was followed by homogenising the fly heads, and the homogenates were assayed for acetylcholinesterase and monoamine oxidase activities, as well as levels of lipid peroxidation, reactive oxygen species, and total thiol. The results showed aspartame at all levels of dietary intake and a high proportion of sucrose significantly aggravated the mortality rate, locomotor deficiency, and impaired biomarkers of oxidative stress and antioxidant status in the transgenic flies, while no significant effect was found on acetylcholinesterase activity or memory function. These findings therefore suggest that while low dietary inclusions of sucrose are tolerable under AD-like phenotypes in the flies, high inclusions of sucrose and all proportions of aspartame tested aggravated mortality rate, locomotion and oxidative stress in the flies.
Collapse
Affiliation(s)
- Bukola Christiana Adedayo
- Biochemistry Department, The Federal University of Technology, Akure P.M.B. 704, Nigeria
- Drosophila Research Lab, Functional Foods and Nutraceuticals Unit, The Federal University of Technology, Akure P.M.B. 704, Nigeria
| | - Stephanie Tolulope Akinniyi
- Biochemistry Department, The Federal University of Technology, Akure P.M.B. 704, Nigeria
- Drosophila Research Lab, Functional Foods and Nutraceuticals Unit, The Federal University of Technology, Akure P.M.B. 704, Nigeria
| | - Opeyemi Babatunde Ogunsuyi
- Drosophila Research Lab, Functional Foods and Nutraceuticals Unit, The Federal University of Technology, Akure P.M.B. 704, Nigeria
- Department of Biomedical Technology, The Federal University of Technology, Akure P.M.B. 704, Nigeria
| | - Ganiyu Oboh
- Biochemistry Department, The Federal University of Technology, Akure P.M.B. 704, Nigeria
- Drosophila Research Lab, Functional Foods and Nutraceuticals Unit, The Federal University of Technology, Akure P.M.B. 704, Nigeria
| |
Collapse
|
17
|
Tan FHP, Azzam G, Najimudin N, Shamsuddin S, Zainuddin A. Behavioural Effects and RNA-seq Analysis of Aβ42-Mediated Toxicity in a Drosophila Alzheimer's Disease Model. Mol Neurobiol 2023:10.1007/s12035-023-03368-x. [PMID: 37145377 DOI: 10.1007/s12035-023-03368-x] [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: 02/01/2023] [Accepted: 04/22/2023] [Indexed: 05/06/2023]
Abstract
Alzheimer's disease (AD) is the most common neurological ailment worldwide. Its process comprises the unique aggregation of extracellular senile plaques composed of amyloid-beta (Aβ) in the brain. Aβ42 is the most neurotoxic and aggressive of the Aβ42 isomers released in the brain. Despite much research on AD, the complete pathophysiology of this disease remains unknown. Technical and ethical constraints place limits on experiments utilizing human subjects. Thus, animal models were used to replicate human diseases. The Drosophila melanogaster is an excellent model for studying both physiological and behavioural aspects of human neurodegenerative illnesses. Here, the negative effects of Aβ42-expression on a Drosophila AD model were investigated through three behavioural assays followed by RNA-seq. The RNA-seq data was verified using qPCR. AD Drosophila expressing human Aβ42 exhibited degenerated eye structures, shortened lifespan, and declined mobility function compared to the wild-type Control. RNA-seq revealed 1496 genes that were differentially expressed from the Aβ42-expressing samples against the control. Among the pathways that were identified from the differentially expressed genes include carbon metabolism, oxidative phosphorylation, antimicrobial peptides, and longevity-regulating pathways. While AD is a complicated neurological condition whose aetiology is influenced by a number of factors, it is hoped that the current data will be sufficient to give a general picture of how Aβ42 influences the disease pathology. The discovery of molecular connections from the current Drosophila AD model offers fresh perspectives on the usage of this Drosophila which could aid in the discovery of new anti-AD medications.
Collapse
Affiliation(s)
- Florence Hui Ping Tan
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, 11800, Penang, Malaysia.
| | - Ghows Azzam
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, 11800, Penang, Malaysia.
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia.
- Malaysia Genome and Vaccine Institute (MGVI), National Institutes of Biotechnology Malaysia (NIBM), Jalan Bangi, 43000, Kajang, Selangor, Malaysia.
| | - Nazalan Najimudin
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, 11800, Penang, Malaysia
- Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Azalina Zainuddin
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
18
|
Varte V, Munkelwitz JW, Rincon-Limas DE. Insights from Drosophila on Aβ- and tau-induced mitochondrial dysfunction: mechanisms and tools. Front Neurosci 2023; 17:1184080. [PMID: 37139514 PMCID: PMC10150963 DOI: 10.3389/fnins.2023.1184080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia in older adults worldwide. Sadly, there are no disease-modifying therapies available for treatment due to the multifactorial complexity of the disease. AD is pathologically characterized by extracellular deposition of amyloid beta (Aβ) and intracellular neurofibrillary tangles composed of hyperphosphorylated tau. Increasing evidence suggest that Aβ also accumulates intracellularly, which may contribute to the pathological mitochondrial dysfunction observed in AD. According with the mitochondrial cascade hypothesis, mitochondrial dysfunction precedes clinical decline and thus targeting mitochondria may result in new therapeutic strategies. Unfortunately, the precise mechanisms connecting mitochondrial dysfunction with AD are largely unknown. In this review, we will discuss how the fruit fly Drosophila melanogaster is contributing to answer mechanistic questions in the field, from mitochondrial oxidative stress and calcium dysregulation to mitophagy and mitochondrial fusion and fission. In particular, we will highlight specific mitochondrial insults caused by Aβ and tau in transgenic flies and will also discuss a variety of genetic tools and sensors available to study mitochondrial biology in this flexible organism. Areas of opportunity and future directions will be also considered.
Collapse
Affiliation(s)
- Vanlalrinchhani Varte
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
| | - Jeremy W. Munkelwitz
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
| | - Diego E. Rincon-Limas
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- Genetics Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
19
|
Bujdoso R, Smith A, Fleck O, Spiropoulos J, Andréoletti O, Thackray AM. Prion disease modelled in Drosophila. Cell Tissue Res 2023; 392:47-62. [PMID: 35092497 PMCID: PMC10113284 DOI: 10.1007/s00441-022-03586-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/17/2022] [Indexed: 11/02/2022]
Abstract
Prion diseases are fatal neurodegenerative conditions of humans and various vertebrate species that are transmissible between individuals of the same or different species. A novel infectious moiety referred to as a prion is considered responsible for transmission of these conditions. Prion replication is believed to be the cause of the neurotoxicity that arises during prion disease pathogenesis. The prion hypothesis predicts that the transmissible prion agent consists of PrPSc, which is comprised of aggregated misfolded conformers of the normal host protein PrPC. It is important to understand the biology of transmissible prions and to identify genetic modifiers of prion-induced neurotoxicity. This information will underpin the development of therapeutic and control strategies for human and animal prion diseases. The most reliable method to detect prion infectivity is by in vivo transmission in a suitable experimental host, which to date have been mammalian species. Current prion bioassays are slow, cumbersome and relatively insensitive to low titres of prion infectivity, and do not lend themselves to rapid genetic analysis of prion disease. Here, we provide an overview of our novel studies that have led to the establishment of Drosophila melanogaster, a genetically well-defined invertebrate host, as a sensitive, versatile and economically viable animal model for the detection of mammalian prion infectivity and genetic modifiers of prion-induced toxicity.
Collapse
Affiliation(s)
- Raymond Bujdoso
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 OES, UK.
| | - Andrew Smith
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 OES, UK
| | - Oliver Fleck
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 OES, UK
| | - John Spiropoulos
- Pathology Department, Animal and Plant Health Agency (APHA), Weybridge, Woodham Lane, New Haw, Surrey, KT15 3NB, Addlestone, UK
| | - Olivier Andréoletti
- UMR INRA ENVT 1225-Hôtes-Agents Pathogènes, Ecole Nationale Vétérinaire de Toulouse, 23 Chemin des Capelles, 31076, Toulouse, France
| | - Alana M Thackray
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 OES, UK.
| |
Collapse
|
20
|
Ghalayini J, Boulianne GL. Deciphering mechanisms of action of ACE inhibitors in neurodegeneration using Drosophila models of Alzheimer's disease. Front Neurosci 2023; 17:1166973. [PMID: 37113150 PMCID: PMC10126366 DOI: 10.3389/fnins.2023.1166973] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder for which there is no cure. Recently, several studies have reported a significant reduction in the incidence and progression of dementia among some patients receiving antihypertensive medications such as angiotensin-converting enzyme inhibitors (ACE-Is) and angiotensin receptor blockers (ARBs). Why these drugs are beneficial in some AD patients and not others is unclear although it has been shown to be independent of their role in regulating blood pressure. Given the enormous and immediate potential of ACE-Is and ARBs for AD therapeutics it is imperative that we understand how they function. Recently, studies have shown that ACE-Is and ARBs, which target the renin angiotensin system in mammals, are also effective in suppressing neuronal cell death and memory defects in Drosophila models of AD despite the fact that this pathway is not conserved in flies. This suggests that the beneficial effects of these drugs may be mediated by distinct and as yet, identified mechanisms. Here, we discuss how the short lifespan and ease of genetic manipulations available in Drosophila provide us with a unique and unparalleled opportunity to rapidly identify the targets of ACE-Is and ARBs and evaluate their therapeutic effectiveness in robust models of AD.
Collapse
Affiliation(s)
- Judy Ghalayini
- Program in Developmental and Stem Cell Biology, Peter Gilgin Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Gabrielle L. Boulianne
- Program in Developmental and Stem Cell Biology, Peter Gilgin Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- *Correspondence: Gabrielle L. Boulianne,
| |
Collapse
|
21
|
Akhtar A, Gupta SM, Dwivedi S, Kumar D, Shaikh MF, Negi A. Preclinical Models for Alzheimer's Disease: Past, Present, and Future Approaches. ACS OMEGA 2022; 7:47504-47517. [PMID: 36591205 PMCID: PMC9798399 DOI: 10.1021/acsomega.2c05609] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 05/13/2023]
Abstract
A robust preclinical disease model is a primary requirement to understand the underlying mechanisms, signaling pathways, and drug screening for human diseases. Although various preclinical models are available for several diseases, clinical models for Alzheimer's disease (AD) remain underdeveloped and inaccurate. The pathophysiology of AD mainly includes the presence of amyloid plaques and neurofibrillary tangles (NFT). Furthermore, neuroinflammation and free radical generation also contribute to AD. Currently, there is a wide gap in scientific approaches to preventing AD progression. Most of the available drugs are limited to symptomatic relief and improve deteriorating cognitive functions. To mimic the pathogenesis of human AD, animal models like 3XTg-AD and 5XFAD are the primarily used mice models in AD therapeutics. Animal models for AD include intracerebroventricular-streptozotocin (ICV-STZ), amyloid beta-induced, colchicine-induced, etc., focusing on parameters such as cognitive decline and dementia. Unfortunately, the translational rate of the potential drug candidates in clinical trials is poor due to limitations in imitating human AD pathology in animal models. Therefore, the available preclinical models possess a gap in AD modeling. This paper presents an outline that critically assesses the applicability and limitations of the current approaches in disease modeling for AD. Also, we attempted to provide key suggestions for the best-fit model to evaluate potential therapies, which might improve therapy translation from preclinical studies to patients with AD.
Collapse
Affiliation(s)
- Ansab Akhtar
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shraddha M. Gupta
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shubham Dwivedi
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Devendra Kumar
- Faculty
of Pharmacy, DIT University, Uttarakhand, Dehradun 248009, India
| | - Mohd. Farooq Shaikh
- Neuropharmacology
Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia
| | - Arvind Negi
- Department
of Bioproducts and Biosystems, Aalto University, FI-00076 Espoo, Finland
- E-mail:
| |
Collapse
|
22
|
Asadzadeh J, Ruchti E, Jiao W, Limoni G, MacLachlan C, Small SA, Knott G, Santa-Maria I, McCabe BD. Retromer deficiency in Tauopathy models enhances the truncation and toxicity of Tau. Nat Commun 2022; 13:5049. [PMID: 36030267 PMCID: PMC9420134 DOI: 10.1038/s41467-022-32683-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/10/2022] [Indexed: 11/24/2022] Open
Abstract
Alteration of the levels, localization or post-translational processing of the microtubule associated protein Tau is associated with many neurodegenerative disorders. Here we develop adult-onset models for human Tau (hTau) toxicity in Drosophila that enable age-dependent quantitative measurement of central nervous system synapse loss and axonal degeneration, in addition to effects upon lifespan, to facilitate evaluation of factors that may contribute to Tau-dependent neurodegeneration. Using these models, we interrogate the interaction of hTau with the retromer complex, an evolutionarily conserved cargo-sorting protein assembly, whose reduced activity has been associated with both Parkinson’s and late onset Alzheimer’s disease. We reveal that reduction of retromer activity induces a potent enhancement of hTau toxicity upon synapse loss, axon retraction and lifespan through a specific increase in the production of a C-terminal truncated isoform of hTau. Our data establish a molecular and subcellular mechanism necessary and sufficient for the depletion of retromer activity to exacerbate Tau-dependent neurodegeneration. Tau and the Retromer complex are both linked to Parkinson’s and Alzheimer’s disease. Using Drosophila neurodegeneration models, this study finds that low retromer activity induces a specific increase of a highly toxic truncated form of human Tau.
Collapse
Affiliation(s)
- Jamshid Asadzadeh
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Evelyne Ruchti
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Wei Jiao
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Greta Limoni
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Catherine MacLachlan
- BioEM Facility, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Scott A Small
- Department of Neurology, Columbia University, New York, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA
| | - Graham Knott
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland.,BioEM Facility, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Ismael Santa-Maria
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA.,Department of Pathology & Cell Biology, Columbia University, New York, USA.,Facultad Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Brian D McCabe
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland.
| |
Collapse
|
23
|
Siddique YH, Rahul, Ara G, Afzal M, Varshney H, Gaur K, Subhan I, Mantasha I, Shahid M. Beneficial effects of apigenin on the transgenic Drosophila model of Alzheimer's disease. Chem Biol Interact 2022; 366:110120. [PMID: 36027948 DOI: 10.1016/j.cbi.2022.110120] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 11/03/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder. The available drugs improve the symptoms but do not play role in modifying disease effects. Currently, the treatment strategies focus on inhibiting the production of Aβ-42 aggregates and tau filaments. In this context the natural plant products could act as a potent candidate. Therefore, we decided to study the effect of apigenin on the transgenic Drosophila model of AD i.e., expressing Aβ-42 in the neurons. The AD flies were allowed to feed on the diet having 25, 50, 75 and 100μM of apigenin for 30 days. The exposure of AD flies to apigenin showed a dose dependent significant decrease in the oxidative stress and delay in the loss of climbing ability. Apigenin also inhibits the activity of acetylcholinesterase. The immunostaining and molecular docking studies suggest that apigenin inhibits the formation of Aβ-42 aggregates. Apigenin is potent in reducing the AD symptoms being mimicked in the transgenic Drosophila model of AD.
Collapse
Affiliation(s)
- Yasir Hasan Siddique
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India.
| | - Rahul
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Gulshan Ara
- Women's College, Zoology Section, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Mohammad Afzal
- Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Himanshi Varshney
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Kajal Gaur
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Iqra Subhan
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - I Mantasha
- Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India
| | - M Shahid
- Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India
| |
Collapse
|
24
|
Chocron ES, Munkácsy E, Kim HS, Karpowicz P, Jiang N, Van Skike CE, DeRosa N, Banh AQ, Palavicini JP, Wityk P, Kalinowski L, Galvan V, Osmulski PA, Jankowska E, Gaczynska M, Pickering AM. Genetic and pharmacologic proteasome augmentation ameliorates Alzheimer's-like pathology in mouse and fly APP overexpression models. SCIENCE ADVANCES 2022; 8:eabk2252. [PMID: 35675410 PMCID: PMC9177073 DOI: 10.1126/sciadv.abk2252] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 04/21/2022] [Indexed: 05/27/2023]
Abstract
The proteasome has key roles in neuronal proteostasis, including the removal of misfolded and oxidized proteins, presynaptic protein turnover, and synaptic efficacy and plasticity. Proteasome dysfunction is a prominent feature of Alzheimer's disease (AD). We show that prevention of proteasome dysfunction by genetic manipulation delays mortality, cell death, and cognitive deficits in fly and cell culture AD models. We developed a transgenic mouse with neuronal-specific proteasome overexpression that, when crossed with an AD mouse model, showed reduced mortality and cognitive deficits. To establish translational relevance, we developed a set of TAT-based proteasome-activating peptidomimetics that stably penetrated the blood-brain barrier and enhanced 20S/26S proteasome activity. These agonists protected against cell death, cognitive decline, and mortality in cell culture, fly, and mouse AD models. The protective effects of proteasome overexpression appear to be driven, at least in part, by the proteasome's increased turnover of the amyloid precursor protein along with the prevention of overall proteostatic dysfunction.
Collapse
Affiliation(s)
- E. Sandra Chocron
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
| | - Erin Munkácsy
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
| | - Harper S. Kim
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
- Center for Neurodegeneration and Experimental Therapeutics (CNET), Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Przemyslaw Karpowicz
- Department of Organic Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Nisi Jiang
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Candice E. Van Skike
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, TX, USA
| | - Nicholas DeRosa
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, TX, USA
| | - Andy Q. Banh
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, TX, USA
| | - Juan P. Palavicini
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
| | - Paweł Wityk
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
- Department of Medical Laboratory Diagnostics–Fahrenheit Biobank BBMRI.pl, Medical University of Gdańsk, Gdańsk, Poland
- BioTechMed Centre/Department of Mechanics of Materials and Structures, Gdańsk University of Technology, Gdańsk, Poland
| | - Leszek Kalinowski
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
- Department of Medical Laboratory Diagnostics–Fahrenheit Biobank BBMRI.pl, Medical University of Gdańsk, Gdańsk, Poland
- BioTechMed Centre/Department of Mechanics of Materials and Structures, Gdańsk University of Technology, Gdańsk, Poland
| | - Veronica Galvan
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- College of Medicine, Oklahoma Health Science Center, Oklahoma City, OK, USA
- Department of Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- South Texas VA Health Care System, San Antonio, TX, USA
- Oklahoma City VA Health Care System, Oklahoma City, OK, USA
| | - Pawel A. Osmulski
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Elzbieta Jankowska
- Department of Organic Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Maria Gaczynska
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Andrew M. Pickering
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
- Center for Neurodegeneration and Experimental Therapeutics (CNET), Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
25
|
Ogunsuyi O, Olasehinde T, Oboh G. Neuroprotective properties of Solanum leaves in Transgenic Drosophila melanogaster model of Alzheimer's disease. Biomarkers 2022; 27:587-598. [PMID: 35546534 DOI: 10.1080/1354750x.2022.2077446] [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: 11/02/2022]
Abstract
INTRODUCTION We investigated the effect of African eggplant (AE) (Solanum macrocarpon L) and Black nightshade (BN) (Solanum nigrum L) leaves; two tropical vegetables consumed by humans on behavioral, biochemical and histological indices in Drosophila melanogaster model of Alzheimer's disease (AD). MATERIALS AND METHOD Transgenic flies expressing human Amyloid Precursor Protein (hAPP) and β-secretase (hBACE 1) were exposed to the pulverized leaf samples (0.1 and 1.0%) in their diets for fourteen days. Thereafter, the flies were assessed for their behavioral indices and routine histology of brain cells. Furthermore, fly head homogenates were assayed for β-amyloid level, activities of acetylcholinesterase (AChE) and β-secretase (BACE-1), as well as oxidative stress markers. RESULTS Result showed that the significantly lower (p < 0.05) behavioral parameters (survival, locomotor performance and memory index), higher AChE and BACE-1 activities, β-amyloid, ROS and lipid peroxidation levels, as well as reduced antioxidant indices observed in the AD flies, were significantly ameliorated (p < 0.05) in AD flies treated with the leaf samples. DISCUSSION This study has showed that leaves of AE and BN ameliorated behavioral and biochemical indices in AD flies via neural enzyme modulatory, and antioxidant mechanisms. CONCLUSION Hence, this study further justifies the neuroprotective properties of both AE and BN.
Collapse
Affiliation(s)
- Opeyemi Ogunsuyi
- Department of Biochemistry, Federal University of Technology, P.M.B. 704, Akure, Nigeria.,Department of Biomedical Technology, Federal University of Technology, P.M.B. 704, Akure, Nigeria
| | - Tosin Olasehinde
- Nutrition and Toxicology Division, Food Technology Department, Federal Institute of Industrial Research, Lagos, Nigeria.,Discipline of Microbiology, School of Life Sciences, University of Kwazulu-Natal, Westville, Durban, Kwazulu-Natal Province, South Africa
| | - Ganiyu Oboh
- Department of Biochemistry, Federal University of Technology, P.M.B. 704, Akure, Nigeria
| |
Collapse
|
26
|
Chakrabarty R, Yousuf S, Singh MP. Contributive Role of Hyperglycemia and Hypoglycemia Towards the Development of Alzheimer's Disease. Mol Neurobiol 2022; 59:4274-4291. [PMID: 35503159 DOI: 10.1007/s12035-022-02846-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/20/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is one of the causes of dementia that results from several infections/biological conditions leading to either cell disruption or loss of neuronal communication. Studies have documented the accumulation of two proteins, beta-amyloid (Aβ), which accumulates on the exteriors of neurons, and tau (Tau), which assembles at the interiors of brain cells and is chiefly liable for the progression of the disease. Several molecular and cellular pathways account for the accumulation of amyloid-β and the formation of neurofibrillary tangles, which are phosphorylated variants of Tau protein. Moreover, research has revealed a potential connection between AD and diabetes. It has also been demonstrated that both hypoglycemia and hyperglycemia have a significant role in the development of AD. In addition, SUMO (small ubiquitin-like modifier protein) plays a crucial role in the pathogenesis of AD. SUMOylation is the process by which modification of amyloid precursor protein (APP) and Tau takes place. Furthermore, Drosophila melanogaster has proven to be an efficient model organism in studies to establish the relationship between AD and variations in blood glucose levels. In addition, the review successfully identifies the common pathway that links the effects of fluctuations in glucose levels on AD pathogenesis and advancements.
Collapse
Affiliation(s)
- Riya Chakrabarty
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar-Ludhiana National Highway, Phagwara, Punjab, 144411, India
| | - Sumaira Yousuf
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar-Ludhiana National Highway, Phagwara, Punjab, 144411, India
| | - Mahendra P Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar-Ludhiana National Highway, Phagwara, Punjab, 144411, India.
| |
Collapse
|
27
|
Blanchette CR, Scalera AL, Harris KP, Zhao Z, Dresselhaus EC, Koles K, Yeh A, Apiki JK, Stewart BA, Rodal AA. Local regulation of extracellular vesicle traffic by the synaptic endocytic machinery. J Cell Biol 2022; 221:e202112094. [PMID: 35320349 PMCID: PMC8952828 DOI: 10.1083/jcb.202112094] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
Neuronal extracellular vesicles (EVs) are locally released from presynaptic terminals, carrying cargoes critical for intercellular signaling and disease. EVs are derived from endosomes, but it is unknown how these cargoes are directed to the EV pathway rather than for conventional endolysosomal degradation. Here, we find that endocytic machinery plays an unexpected role in maintaining a release-competent pool of EV cargoes at synapses. Endocytic mutants, including nervous wreck (nwk), shibire/dynamin, and AP-2, unexpectedly exhibit local presynaptic depletion specifically of EV cargoes. Accordingly, nwk mutants phenocopy synaptic plasticity defects associated with loss of the EV cargo synaptotagmin-4 (Syt4) and suppress lethality upon overexpression of the EV cargo amyloid precursor protein (APP). These EV defects are genetically separable from canonical endocytic functions in synaptic vesicle recycling and synaptic growth. Endocytic machinery opposes the endosomal retromer complex to regulate EV cargo levels and acts upstream of synaptic cargo removal by retrograde axonal transport. Our data suggest a novel molecular mechanism that locally promotes cargo loading into synaptic EVs.
Collapse
Affiliation(s)
| | | | - Kathryn P. Harris
- Department of Biology, University of Toronto Mississauga, Mississauga, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Zechuan Zhao
- Department of Biology, Brandeis University, Waltham, MA
| | | | - Kate Koles
- Department of Biology, Brandeis University, Waltham, MA
| | - Anna Yeh
- Department of Biology, Brandeis University, Waltham, MA
| | | | - Bryan A. Stewart
- Department of Biology, University of Toronto Mississauga, Mississauga, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | | |
Collapse
|
28
|
Neuroprotective Effects of PARP Inhibitors in Drosophila Models of Alzheimer’s Disease. Cells 2022; 11:cells11081284. [PMID: 35455964 PMCID: PMC9027574 DOI: 10.3390/cells11081284] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 12/10/2022] Open
Abstract
Alzheimer’s disease (AD) is an irreversible age-related neurodegenerative disorder clinically characterized by severe memory impairment, language deficits and cognitive decline. The major neuropathological hallmarks of AD include extracellular deposits of the β-amyloid (Aβ) peptides and cytoplasmic neurofibrillary tangles (NFTs) of hyperphosphorylated tau protein. The accumulation of plaques and tangles in the brain triggers a cascade of molecular events that culminate in neuronal damage and cell death. Despite extensive research, our understanding of the molecular basis of AD pathogenesis remains incomplete and a cure for this devastating disease is still not available. A growing body of evidence in different experimental models suggests that poly(ADP-ribose) polymerase-1 (PARP-1) overactivation might be a crucial component of the molecular network of interactions responsible for AD pathogenesis. In this work, we combined genetic, molecular and biochemical approaches to investigate the effects of two different PARP-1 inhibitors (olaparib and MC2050) in Drosophila models of Alzheimer’s disease by exploring their neuroprotective and therapeutic potential in vivo. We found that both pharmacological inhibition and genetic inactivation of PARP-1 significantly extend lifespan and improve the climbing ability of transgenic AD flies. Consistently, PARP-1 inhibitors lead to a significant decrease of Aβ42 aggregates and partially rescue the epigenetic alterations associated with AD in the brain. Interestingly, olaparib and MC2050 also suppress the AD-associated aberrant activation of transposable elements in neuronal tissues of AD flies.
Collapse
|
29
|
Zhang C, Inamdar SM, Swaminathan S, Marenda DR, Saunders AJ. Association of the Protein-Quality-Control Protein Ubiquilin-1 With Alzheimer’s Disease Both in vitro and in vivo. Front Neurosci 2022; 16:821059. [PMID: 35401099 PMCID: PMC8992708 DOI: 10.3389/fnins.2022.821059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) belongs to a class of diseases characterized by progressive accumulation and aggregation of pathogenic proteins, particularly Aβ proteins. Genetic analysis has identified UBQLN1 as an AD candidate gene. Ubiquilin-1 levels reduce with AD progression, suggesting a potential loss-of-function mechanism. The ubiquilin-1 protein is involved in protein quality control (PQC), which plays essential roles in cellular growth and normal cell function. Ubiquilin-1 regulates γ-secretase by increasing endoproteolysis of PS1, a key γ-secretase component. Presently, the effects of ubiquilin-1 on cellular physiology as well as Aβ-related events require further investigation. Here, we investigated the effects of ubiquilin-1 on cellular growth and viability in association with APP (amyloid-β protein precursor), APP processing-related β-secretase (BACE1, BACE) and γ-secretase using cell and animal-based models. We showed that loss-of-function in Drosophila ubqn suppresses human APP and human BACE phenotypes in wing veins and altered cell number and tissue compartment size in the wing. Additionally, we performed cell-based studies and showed that silencing UBQLN1 reduced cell viability and increased caspase-3 activity. Overexpression of UBQLN1 significantly reduced Aβ levels. Furthermore, pharmacological inhibition of γ-secretase increased ubiquilin-1 protein levels, suggesting a mechanism that regulates ubiquilin-1 levels which may associate with reduced Aβ reduction by inhibiting γ-secretase. Collectively, our results support not only a loss-of-function mechanism of ubiquilin-1 in association with AD, but also support the significance of targeting ubiquilin-1-mediated PQC as a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Can Zhang
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | | | - Swathi Swaminathan
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Daniel R. Marenda
- Department of Biology, Drexel University, Philadelphia, PA, United States
- Division of Biological Infrastructure, National Science Foundation, Alexandria, VA, United States
| | - Aleister J. Saunders
- Department of Biology, Drexel University, Philadelphia, PA, United States
- *Correspondence: Aleister J. Saunders,
| |
Collapse
|
30
|
Differential gene expression by RNA-seq during Alzheimer’s disease-like progression in the Drosophila melanogaster model. Neurosci Res 2022; 180:1-12. [DOI: 10.1016/j.neures.2022.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 02/16/2022] [Accepted: 02/20/2022] [Indexed: 01/10/2023]
|
31
|
Smith C, Smith H, Roberts L, Coward L, Gorman G, Verma A, Li Q, Buford TW, Carter CS, Jumbo-Lucioni P. Probiotic Releasing Angiotensin (1-7) in a Drosophila Model of Alzheimer's Disease Produces Sex-Specific Effects on Cognitive Function. J Alzheimers Dis 2022; 85:1205-1217. [PMID: 34924372 PMCID: PMC9549527 DOI: 10.3233/jad-210464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND While extensive research on the brain has failed to identify effective therapies, using probiotics to target the gut microbiome has shown therapeutic potential in Alzheimer's disease (AD). Genetically modified probiotics (GMP) are a promising strategy to deliver key therapeutic peptides with high efficacy and tissue specificity. Angiotensin (Ang)-(1-7) levels inversely correlate to AD severity, but its administration is challenging. Our group has successfully established a GMP-based method of Ang-(1-7) delivery. OBJECTIVE Since Drosophila represents an excellent model to study the effect of probiotics on complex disorders in a high throughput manner, we tested whether oral supplementation with Lactobacillus paracasei releasing Ang-(1-7) (LP-A) delays memory loss in a Drosophila AD model. METHODS Flies overexpressing the human amyloid-β protein precursor and its β-site cleaving enzyme in neurons were randomized to receive four 24-h doses of Lactobacillus paracasei alone (LP), LP-A or sucrose over 14 days. Memory was assessed via an aversive phototaxic suppression assay. RESULTS Optimal dilution,1:2, was determined based on palatability. LP-A improved memory in trained AD males but worsened cognition in AD females. LP-supplementation experiments confirmed that Ang-(1-7) conferred additional cognitive benefits in males and was responsible for the deleterious cognitive effects in females. Sex-specific differences in the levels of angiotensin peptides and differential activation of the kynurenine pathway of tryptophan metabolism in response to supplementation may underlie this male-only therapeutic response. CONCLUSION In summary, LP-A ameliorated the memory deficits of a Drosophila AD model, but effects were sex-specific. Dosage optimization may be required to address this differential response.
Collapse
Affiliation(s)
- C.Aaron Smith
- McWhorter School of Pharmacy, Samford University, Birmingham, AL
| | - Haddon Smith
- McWhorter School of Pharmacy, Samford University, Birmingham, AL
| | - Lisa Roberts
- Department of Medicine; Division of Gerontology, Geriatrics, and Palliative Care, University of Alabama at Birmingham, Birmingham, AL
| | - Lori Coward
- Pharmaceutical Sciences Research Institute, Samford University, Birmingham, AL
| | - Gregory Gorman
- McWhorter School of Pharmacy, Samford University, Birmingham, AL,Pharmaceutical Sciences Research Institute, Samford University, Birmingham, AL
| | - Amrisha Verma
- Department of Ophthalmology, College of Medicine, University of Florida Gainesville, FL
| | - Qiuhong Li
- Department of Ophthalmology, College of Medicine, University of Florida Gainesville, FL
| | - Thomas W. Buford
- Department of Medicine; Division of Gerontology, Geriatrics, and Palliative Care, University of Alabama at Birmingham, Birmingham, AL,Geriatric Research Education and Clinical Center, Birmingham VA Medical Center, Birmingham, AL,Corresponding authors: Thomas W. Buford, Phone: (205) 975-9042; ; Patricia Jumbo-Lucioni, Phone: (205) 726-4170;
| | - Christy S. Carter
- Department of Medicine; Division of Gerontology, Geriatrics, and Palliative Care, University of Alabama at Birmingham, Birmingham, AL
| | - Patricia Jumbo-Lucioni
- McWhorter School of Pharmacy, Samford University, Birmingham, AL,Department of Biology, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL.,Corresponding authors: Thomas W. Buford, Phone: (205) 975-9042; ; Patricia Jumbo-Lucioni, Phone: (205) 726-4170;
| |
Collapse
|
32
|
Tan FHP, Hadri NAB, Najimudin N, Watanabe N, Azzam G. Ethyl caffeate ameliorated amyloid-beta42 protein-associated toxicity in PC12 cells and Drosophila melanogaster. Geriatr Gerontol Int 2021; 21:1125-1130. [PMID: 34699118 DOI: 10.1111/ggi.14296] [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: 06/29/2021] [Revised: 09/14/2021] [Accepted: 10/04/2021] [Indexed: 11/29/2022]
Abstract
AIM Alzheimer's disease (AD) is the most pervasive neurodegenerative disorder in societies globally. Till now, the mechanism behind this disease is still equivocal. Amyloid-beta42 protein (Aβ42), the most toxic and aggressive Aβ species, is the main focus of this study. The naturally occurring ethyl caffeate (EC) is associated with various medicinal properties. Here, EC was tested for its protective properties against Aβ42's toxic effects. METHODS As treatment of Aβ42 has been shown to cause neuronal cell death, EC was first screened with Aβ42-incubated PC12 neuronal cells. Next, the compound was tested on the Drosophila melanogaster AD model using the rough eye phenotype assay, lifespan assay and negative geotaxis assay. RESULTS EC ameliorated PC12 cells from cell death linked to Aβ42 exposure. Using Drosophila expressing human Aβ42, feeding of EC was able to partially rescue the rough eye phenotype, lengthen the lifespan of AD Drosophila and enhanced the mobility of middle-aged AD Drosophila. CONCLUSION Overall, the results of this study showed that EC might possess therapeutic properties for AD. Geriatr Gerontol Int 2021; ••: ••-••.
Collapse
Affiliation(s)
- Florence Hui Ping Tan
- School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia.,USM-RIKEN International Center for Aging Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia
| | | | - Nazalan Najimudin
- School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia.,USM-RIKEN International Center for Aging Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia
| | - Nobumoto Watanabe
- USM-RIKEN International Center for Aging Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia.,Bioprobe Application Research Unit, RIKEN Center for Sustainable Resource Science, RIKEN, Wako, Japan
| | - Ghows Azzam
- School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia.,USM-RIKEN International Center for Aging Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
33
|
Elovsson G, Bergkvist L, Brorsson AC. Exploring Aβ Proteotoxicity and Therapeutic Candidates Using Drosophila melanogaster. Int J Mol Sci 2021; 22:ijms221910448. [PMID: 34638786 PMCID: PMC8508956 DOI: 10.3390/ijms221910448] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease is a widespread and devastating neurological disorder associated with proteotoxic events caused by the misfolding and aggregation of the amyloid-β peptide. To find therapeutic strategies to combat this disease, Drosophila melanogaster has proved to be an excellent model organism that is able to uncover anti-proteotoxic candidates due to its outstanding genetic toolbox and resemblance to human disease genes. In this review, we highlight the use of Drosophila melanogaster to both study the proteotoxicity of the amyloid-β peptide and to screen for drug candidates. Expanding the knowledge of how the etiology of Alzheimer’s disease is related to proteotoxicity and how drugs can be used to block disease progression will hopefully shed further light on the field in the search for disease-modifying treatments.
Collapse
Affiliation(s)
- Greta Elovsson
- Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, 58183 Linköping, Sweden;
| | - Liza Bergkvist
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, 17164 Solna, Sweden;
| | - Ann-Christin Brorsson
- Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, 58183 Linköping, Sweden;
- Correspondence:
| |
Collapse
|
34
|
Beaver M, Karisetty BC, Zhang H, Bhatnagar A, Armour E, Parmar V, Brown R, Xiang M, Elefant F. Chromatin and transcriptomic profiling uncover dysregulation of the Tip60 HAT/HDAC2 epigenomic landscape in the neurodegenerative brain. Epigenetics 2021; 17:786-807. [PMID: 34369292 PMCID: PMC9336495 DOI: 10.1080/15592294.2021.1959742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Disruption of histone acetylation-mediated gene control is a critical step in Alzheimer’s Disease (AD), yet chromatin analysis of antagonistic histone acetyltransferases (HATs) and histone deacetylases (HDACs) causing these alterations remains uncharacterized. We report the first Tip60 HAT versus HDAC2 chromatin (ChIP-seq) and transcriptional (RNA-seq) profiling study in Drosophila melanogaster brains that model early human AD. We find Tip60 and HDAC2 predominantly recruited to identical neuronal genes. Moreover, AD brains exhibit robust genome-wide early alterations that include enhanced HDAC2 and reduced Tip60 binding and transcriptional dysregulation. Orthologous human genes to co-Tip60/HDAC2 D. melanogaster neural targets exhibit conserved disruption patterns in AD patient hippocampi. Notably, we discovered distinct transcription factor binding sites close or within Tip60/HDAC2 co-peaks in neuronal genes, implicating them in coenzyme recruitment. Increased Tip60 protects against transcriptional dysregulation and enhanced HDAC2 enrichment genome-wide. We advocate Tip60 HAT/HDAC2 mediated epigenetic neuronal gene disruption as a genome-wide initial causal event in AD.
Collapse
Affiliation(s)
- Mariah Beaver
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | | | - Haolin Zhang
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Akanksha Bhatnagar
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Ellen Armour
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Visha Parmar
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Reshma Brown
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Merry Xiang
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Felice Elefant
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
35
|
Cheng KC, Chen YH, Wu CL, Lee WP, Cheung CHA, Chiang HC. Rac1 and Akt Exhibit Distinct Roles in Mediating Aβ-Induced Memory Damage and Learning Impairment. Mol Neurobiol 2021; 58:5224-5238. [PMID: 34273104 DOI: 10.1007/s12035-021-02471-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/24/2021] [Indexed: 11/25/2022]
Abstract
Accumulated beta-amyloid (Aβ) in the brain is the hallmark of Alzheimer's disease (AD). Despite Aβ accumulation is known to trigger cellular dysfunctions and learning and memory damage, the detailed molecular mechanism remains elusive. Recent studies have shown that the onset of memory impairment and learning damage in the AD animal is different, suggesting that the underlying mechanism of the development of memory impairment and learning damage may not be the same. In the current study, with the use of Aβ42 transgenic flies as models, we found that Aβ induces memory damage and learning impairment via differential molecular signaling pathways. In early stage, Aβ activates both Ras and PI3K to regulate Rac1 activity, which affects mostly on memory performance. In later stage, PI3K-Akt is strongly activated by Aβ, which leads to learning damage. Moreover, reduced Akt, but not Rac1, activity promotes cell viability in the Aβ42 transgenic flies, indicating that Akt and Rac1 exhibit differential roles in Aβ regulating toxicity. Taken together, different molecular and cellular mechanisms are involved in Aβ-induced learning damage and memory decline; thus, caution should be taken during the development of therapeutic intervention in the future.
Collapse
Affiliation(s)
- Kuan-Chung Cheng
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
| | - Ying-Hao Chen
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Lin Wu
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Wang-Pao Lee
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun Hei Antonio Cheung
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
| | - Hsueh-Cheng Chiang
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan, Taiwan.
| |
Collapse
|
36
|
Tsakiri EN, Gumeni S, Manola MS, Trougakos IP. Amyloid toxicity in a Drosophila Alzheimer's model is ameliorated by autophagy activation. Neurobiol Aging 2021; 105:137-147. [PMID: 34062489 DOI: 10.1016/j.neurobiolaging.2021.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) is the prevailing form of dementia. Protein degradation and antioxidant pathways have a critical role in preventing the accumulation of protein aggregation; thus, failure of proteostasis in neurons along with redox imbalance mark AD. Herein, we exploited an AD Drosophila model expressing human amyloid precursor (hAPP) and beta-secretase 1 (hBACE1) proteins, to better understand the role of proteostatic or antioxidant pathways in AD. Ubiquitous expression of hAPP, hBACE1 in flies caused more severe degenerative phenotypes versus neuronal targeted expression; it also, suppressed proteasome activity, increased oxidative stress and significantly enhanced stress-sensitivity. Overexpression of Prosβ5 proteasomal subunit or Nrf2 transcription factor in AD Drosophila flies partially restored proteasomal activity but did not rescue hAPP, hBACE1 induced neurodegeneration. On the other hand, expression of autophagy-related Atg8a in AD flies decelerated neurodegeneration, increased stress-resistance, and improved flies' health-/lifespan. Overall, our data suggest that the noxious effects of amyloid-beta aggregates can be alleviated by enhanced autophagy, thus dietary or pharmacological interventions that target autophagy should be considered in AD therapeutic approaches.
Collapse
Affiliation(s)
- Eleni N Tsakiri
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| | - Maria S Manola
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece.
| |
Collapse
|
37
|
Goel P, Dickman D. Synaptic homeostats: latent plasticity revealed at the Drosophila neuromuscular junction. Cell Mol Life Sci 2021; 78:3159-3179. [PMID: 33449150 PMCID: PMC8044042 DOI: 10.1007/s00018-020-03732-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/19/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022]
Abstract
Homeostatic signaling systems are fundamental forms of biological regulation that maintain stable functionality in a changing environment. In the nervous system, synapses are crucial substrates for homeostatic modulation, serving to establish, maintain, and modify the balance of excitation and inhibition. Synapses must be sufficiently flexible to enable the plasticity required for learning and memory but also endowed with the stability to last a lifetime. In response to the processes of development, growth, remodeling, aging, and disease that challenge synapses, latent forms of adaptive plasticity become activated to maintain synaptic stability. In recent years, new insights into the homeostatic control of synaptic function have been achieved using the powerful Drosophila neuromuscular junction (NMJ). This review will focus on work over the past 10 years that has illuminated the cellular and molecular mechanisms of five homeostats that operate at the fly NMJ. These homeostats adapt to loss of postsynaptic neurotransmitter receptor functionality, glutamate imbalance, axonal injury, as well as aberrant synaptic growth and target innervation. These diverse homeostats work independently yet can be simultaneously expressed to balance neurotransmission. Growing evidence from this model glutamatergic synapse suggests these ancient homeostatic signaling systems emerged early in evolution and are fundamental forms of plasticity that also function to stabilize mammalian cholinergic NMJs and glutamatergic central synapses.
Collapse
Affiliation(s)
- Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
38
|
Koch SC, Nelson A, Hartenstein V. Structural aspects of the aging invertebrate brain. Cell Tissue Res 2021; 383:931-947. [PMID: 33409654 PMCID: PMC7965346 DOI: 10.1007/s00441-020-03314-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/28/2020] [Indexed: 11/26/2022]
Abstract
Aging is characterized by a decline in neuronal function in all animal species investigated so far. Functional changes are accompanied by and may be in part caused by, structurally visible degenerative changes in neurons. In the mammalian brain, normal aging shows abnormalities in dendrites and axons, as well as ultrastructural changes in synapses, rather than global neuron loss. The analysis of the structural features of aging neurons, as well as their causal link to molecular mechanisms on the one hand, and the functional decline on the other hand is crucial in order to understand the aging process in the brain. Invertebrate model organisms like Drosophila and C. elegans offer the opportunity to apply a forward genetic approach to the analysis of aging. In the present review, we aim to summarize findings concerning abnormalities in morphology and ultrastructure in invertebrate brains during normal aging and compare them to what is known for the mammalian brain. It becomes clear that despite of their considerably shorter life span, invertebrates display several age-related changes very similar to the mammalian condition, including the retraction of dendritic and axonal branches at specific locations, changes in synaptic density and increased accumulation of presynaptic protein complexes. We anticipate that continued research efforts in invertebrate systems will significantly contribute to reveal (and possibly manipulate) the molecular/cellular pathways leading to neuronal aging in the mammalian brain.
Collapse
Affiliation(s)
- Sandra C Koch
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Annie Nelson
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Volker Hartenstein
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA.
| |
Collapse
|
39
|
Abstract
Drosophila melanogaster males reduce courtship behaviour after mating failure. In the lab, such conditioned courtship suppression, aka 'courtship conditioning', serves as a complex learning and memory assay. Interestingly, variations in the courtship conditioning assay can establish different types of memory. Here, we review research investigating the underlying cellular and molecular mechanisms that allow male flies to form memories of previous mating failures.
Collapse
Affiliation(s)
- Nicholas Raun
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Spencer Jones
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jamie M Kramer
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
40
|
Wang X, Davis RL. Early Mitochondrial Fragmentation and Dysfunction in a Drosophila Model for Alzheimer's Disease. Mol Neurobiol 2021; 58:143-155. [PMID: 32909149 PMCID: PMC7704861 DOI: 10.1007/s12035-020-02107-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/27/2020] [Indexed: 12/24/2022]
Abstract
Many different cellular systems and molecular processes become compromised in Alzheimer's disease (AD) including proteostasis, autophagy, inflammatory responses, synapse and neuronal circuitry, and mitochondrial function. We focused in this study on mitochondrial dysfunction owing to the toxic neuronal environment produced by expression of Aβ42, and its relationship to other pathologies found in AD including increased neuronal apoptosis, plaque deposition, and memory impairment. Using super-resolution microscopy, we have assayed mitochondrial status in the three distinct neuronal compartments (somatic, dendritic, axonal) of mushroom body neurons of Drosophila expressing Aβ42. The mushroom body neurons comprise a major center for olfactory memory formation in insects. We employed calcium imaging to measure mitochondrial function, immunohistochemical and staining techniques to measure apoptosis and plaque formation, and olfactory classical conditioning to measure learning. We found that mitochondria become fragmented at a very early age along with decreased function measured by mitochondrial calcium entry. Increased apoptosis and plaque deposition also occur early, yet interestingly, a learning impairment was found only after a much longer period of time-10 days, which is a large fraction of the fly's lifespan. This is similar to the pronounced delay between cellular pathologies and the emergence of a memory dysfunction in humans. Our studies are consistent with the model that mitochondrial dysfunction and/or other cellular pathologies emerge at an early age and lead to much later learning impairments. The results obtained further develop this Drosophila model as a useful in vivo system for probing the mechanisms by which Aβ42 produces mitochondrial and other cellular toxicities that produce memory dysfunction.
Collapse
Affiliation(s)
- Xingjun Wang
- Department of Neuroscience, Scripps Research Institute Florida, Jupiter, Florida, 33458, USA
| | - Ronald L Davis
- Department of Neuroscience, Scripps Research Institute Florida, Jupiter, Florida, 33458, USA.
| |
Collapse
|
41
|
Angiotensin Converting Enzyme Inhibitors and Angiotensin Receptor Blockers Rescue Memory Defects in Drosophila-Expressing Alzheimer's Disease-Related Transgenes Independently of the Canonical Renin Angiotensin System. eNeuro 2020; 7:ENEURO.0235-20.2020. [PMID: 33060184 PMCID: PMC7768280 DOI: 10.1523/eneuro.0235-20.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 01/09/2023] Open
Abstract
Alzheimer's disease (AD) is a degenerative disorder that causes progressive memory and cognitive decline. Recently, studies have reported that inhibitors of the mammalian renin angiotensin system (RAS) result in a significant reduction in the incidence and progression of AD by unknown mechanisms. Here, we used a genetic and pharmacological approach to evaluate the beneficial effects of angiotensin converting enzyme inhibitors (ACE-Is) and angiotensin receptor blockers (ARBs) in Drosophila expressing AD-related transgenes. Importantly, while ACE orthologs have been identified in Drosophila, other RAS components are not conserved. We show that captopril, an ACE-I, and losartan, an ARB, can suppress a rough eye phenotype and brain cell death in flies expressing a mutant human C99 transgene. Captopril also significantly rescues memory defects in these flies. Similarly, both drugs reduce cell death in Drosophila expressing human Aβ42 and losartan significantly rescues memory deficits. However, neither drug affects production, accumulation or clearance of Aβ42 Importantly, neither drug rescued brain cell death in Drosophila expressing human Tau, suggesting that RAS inhibitors specifically target the amyloid pathway. Of note, we also observed reduced cell death and a complete rescue of memory deficits when we crossed a null mutation in Drosophila Acer into each transgenic line demonstrating that the target of captopril in Drosophila is Acer. Together, these studies demonstrate that captopril and losartan are able to modulate AD related phenotypes in the absence of the canonical RAS pathway and suggest that both drugs have additional targets that can be identified in Drosophila.
Collapse
|
42
|
Aqsa, Sarkar S. Age dependent trans-cellular propagation of human tau aggregates in Drosophila disease models. Brain Res 2020; 1751:147207. [PMID: 33212022 DOI: 10.1016/j.brainres.2020.147207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 11/26/2022]
Abstract
Tauopathies is a class of neurodegenerative disorders which involves the transformation of physiological tau into pathogenic tau. One of the prime causes reported to drive this conversion is tau hyperphosphorylation and the subsequent propagation of pathogenic protein aggregates across the nervous system. Although past attempts have been made to deduce the details of tau propagation, yet not much is known about its mechanism. A better understanding of this aspect of disease pathology can prove to be beneficial for the development of diagnostic and therapeutic approaches. For the first time, we demonstrate that the human tau possesses an intrinsic property to spread trans-cellularly in the fly nervous system irrespective of the tau allele or the neuronal tissue type. Aggregate migration restricted by targeted down-regulation of a specific kinase, elucidates the role of hyper-phosphorylation in its movement. On the contrary to the previous models, our study delivers an easy and rapid in-vivo model for comprehensive examination of tau migration pathology. Henceforth, the developed model would not only be immensely helpful in uncovering the mechanistic in-depths of tau propagation pathology but also aid in modifier and/or drug screening for amelioration of tauopathies.
Collapse
Affiliation(s)
- Aqsa
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi 110 021, India
| | - Surajit Sarkar
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi 110 021, India.
| |
Collapse
|
43
|
Abstract
Aβ plaques are one of the two lesions in the brain that define the neuropathological diagnosis of Alzheimer's disease. Plaques are highly diverse structures; many of them include massed, fibrillar polymers of the Aβ protein referred to as Aβ-amyloid, but some lack the defining features of amyloid. Cellular elements in 'classical' plaques include abnormal neuronal processes and reactive glial cells, but these are not present in all plaques. Plaques have been given various names since their discovery in 1892, including senile plaques, amyloid plaques, and neuritic plaques. However, with the identification in the 1980s of Aβ as the obligatory and universal component of plaques, the term 'Aβ plaques' has become a unifying term for these heterogeneous formations. Tauopathy, the second essential lesion of the Alzheimer's disease diagnostic dyad, is downstream of Aβ-proteopathy, but it is critically important for the manifestation of dementia. The etiologic link between Aβ-proteopathy and tauopathy in Alzheimer's disease remains largely undefined. Aβ plaques develop and propagate via the misfolding, self-assembly and spread of Aβ by the prion-like mechanism of seeded protein aggregation. Partially overlapping sets of risk factors and sequelae, including inflammation, genetic variations, and various environmental triggers have been linked to plaque development and idiopathic Alzheimer's disease, but no single factor has emerged as a requisite cause. The value of Aβ plaques per se as therapeutic targets is uncertain; although some plaques are sites of focal gliosis and inflammation, the complexity of inflammatory biology presents challenges to glia-directed intervention. Small, soluble, oligomeric assemblies of Aβ are enriched in the vicinity of plaques, and these probably contribute to the toxic impact of Aβ aggregation on the brain. Measures designed to reduce the production or seeded self-assembly of Aβ can impede the formation of Aβ plaques and oligomers, along with their accompanying abnormalities; given the apparent long timecourse of the emergence, maturation and proliferation of Aβ plaques in humans, such therapies are likely to be most effective when begun early in the pathogenic process, before significant damage has been done to the brain. Since their discovery in the late 19th century, Aβ plaques have, time and again, illuminated fundamental mechanisms driving neurodegeneration, and they should remain at the forefront of efforts to understand, and therefore treat, Alzheimer's disease.
Collapse
Affiliation(s)
- Lary C. Walker
- Department of Neurology and Yerkes National Primate Research Center, Emory University
| |
Collapse
|
44
|
Shaposhnikov MV, Zemskaya NV, Koval LА, Minnikhanova NR, Kechko OI, Mitkevich VA, Makarov AA, Moskalev AА. Amyloid-β peptides slightly affect lifespan or antimicrobial peptide gene expression in Drosophila melanogaster. BMC Genet 2020; 21:65. [PMID: 33092519 PMCID: PMC7583308 DOI: 10.1186/s12863-020-00866-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 11/23/2022] Open
Abstract
Background Beta-amyloid peptide (Aβ) is the key protein in the pathogenesis of Alzheimer’s disease, the most common age-related neurodegenerative disorder in humans. Aβ peptide induced pathological phenotypes in different model organisms include neurodegeneration and lifespan decrease. However, recent experimental evidence suggests that Aβ may utilize oligomerization and fibrillization to function as an antimicrobial peptide (AMP), and protect the host from infections. We used the power of Drosophila model to study mechanisms underlying a dual role for Aβ peptides. Results We investigated the effects of Drosophila treatment with three Aβ42 peptide isoforms, which differ in their ability to form oligomers and aggregates on the lifespan, locomotor activity and AMP genes expression. Aβ42 slightly decreased female’s median lifespan (by 4.5%), but the effect was not related to the toxicity of peptide isoform. The lifespan and relative levels of AMP gene expression in male flies as well as locomotor activity in both sexes were largely unaffected by Aβ42 peptide treatment. Regardless of the effects on lifespan, Aβ42 peptide treatment induced decrease in AMP genes expression in females, but the effects were not robust. Conclusions The results demonstrate that chronic treatment with Aβ42 peptides does not drastically affect fly aging or immunity.
Collapse
Affiliation(s)
- Mikhail V Shaposhnikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia.,Institute of Biology of Komi Science Center of Ural Branch of RAS, 167982, Syktyvkar, Russia
| | - Nadezhda V Zemskaya
- Institute of Biology of Komi Science Center of Ural Branch of RAS, 167982, Syktyvkar, Russia
| | - Lyubov А Koval
- Institute of Biology of Komi Science Center of Ural Branch of RAS, 167982, Syktyvkar, Russia
| | - Natalya R Minnikhanova
- Institute of Biology of Komi Science Center of Ural Branch of RAS, 167982, Syktyvkar, Russia
| | - Olga I Kechko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Alexey А Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia. .,Institute of Biology of Komi Science Center of Ural Branch of RAS, 167982, Syktyvkar, Russia.
| |
Collapse
|
45
|
Mirzaei N, Shi H, Oviatt M, Doustar J, Rentsendorj A, Fuchs DT, Sheyn J, Black KL, Koronyo Y, Koronyo-Hamaoui M. Alzheimer's Retinopathy: Seeing Disease in the Eyes. Front Neurosci 2020; 14:921. [PMID: 33041751 PMCID: PMC7523471 DOI: 10.3389/fnins.2020.00921] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/10/2020] [Indexed: 01/18/2023] Open
Abstract
The neurosensory retina emerges as a prominent site of Alzheimer's disease (AD) pathology. As a CNS extension of the brain, the neuro retina is easily accessible for noninvasive, high-resolution imaging. Studies have shown that along with cognitive decline, patients with mild cognitive impairment (MCI) and AD often suffer from visual impairments, abnormal electroretinogram patterns, and circadian rhythm disturbances that can, at least in part, be attributed to retinal damage. Over a decade ago, our group identified the main pathological hallmark of AD, amyloid β-protein (Aβ) plaques, in the retina of patients including early-stage clinical cases. Subsequent histological, biochemical and in vivo retinal imaging studies in animal models and in humans corroborated these findings and further revealed other signs of AD neuropathology in the retina. Among these signs, hyperphosphorylated tau, neuronal degeneration, retinal thinning, vascular abnormalities and gliosis were documented. Further, linear correlations between the severity of retinal and brain Aβ concentrations and plaque pathology were described. More recently, extensive retinal pericyte loss along with vascular platelet-derived growth factor receptor-β deficiency were discovered in postmortem retinas of MCI and AD patients. This progressive loss was closely associated with increased retinal vascular amyloidosis and predicted cerebral amyloid angiopathy scores. These studies brought excitement to the field of retinal exploration in AD. Indeed, many questions still remain open, such as queries related to the temporal progression of AD-related pathology in the retina compared to the brain, the relations between retinal and cerebral changes and whether retinal signs can predict cognitive decline. The extent to which AD affects the retina, including the susceptibility of certain topographical regions and cell types, is currently under intense investigation. Advances in retinal amyloid imaging, hyperspectral imaging, optical coherence tomography, and OCT-angiography encourage the use of such modalities to achieve more accurate, patient- and user-friendly, noninvasive detection and monitoring of AD. In this review, we summarize the current status in the field while addressing the many unknowns regarding Alzheimer's retinopathy.
Collapse
Affiliation(s)
- Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Mia Oviatt
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Keith L. Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
46
|
Dcf1 alleviates C99-mediated deficits in drosophila by reducing the cleavage of C99. Biochem Biophys Res Commun 2020; 530:410-417. [DOI: 10.1016/j.bbrc.2020.05.063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/07/2020] [Indexed: 12/23/2022]
|
47
|
Ruiz-Arias Á, Paredes JM, Di Biase C, Cuerva JM, Giron MD, Salto R, González-Vera JA, Orte A. Seeding and Growth of β-Amyloid Aggregates upon Interaction with Neuronal Cell Membranes. Int J Mol Sci 2020; 21:ijms21145035. [PMID: 32708806 PMCID: PMC7404110 DOI: 10.3390/ijms21145035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/22/2022] Open
Abstract
In recent years, the prevalence of amyloid neurodegenerative diseases such as Alzheimer's disease (AD) has significantly increased in developed countries due to increased life expectancy. This amyloid disease is characterized by the presence of accumulations and deposits of β-amyloid peptide (Aβ) in neuronal tissue, leading to the formation of oligomers, fibers, and plaques. First, oligomeric intermediates that arise during the aggregation process are currently thought to be primarily responsible for cytotoxicity in cells. This work aims to provide further insights into the mechanisms of cytotoxicity by studying the interaction of Aβ aggregates with Neuro-2a (N2a) neuronal cells and the effects caused by this interaction. For this purpose, we have exploited the advantages of advanced, multidimensional fluorescence microscopy techniques to determine whether different types of Aβ are involved in higher rates of cellular toxicity, and we measured the cellular stress caused by such aggregates by using a fluorogenic intracellular biothiol sensor. Stress provoked by the peptide is evident by N2a cells generating high levels of biothiols as a defense mechanism. In our study, we demonstrate that Aβ aggregates act as seeds for aggregate growth upon interacting with the cellular membrane, which results in cell permeability and damage and induces lysis. In parallel, these damaged cells undergo a significant increase in intracellular biothiol levels.
Collapse
Affiliation(s)
- Álvaro Ruiz-Arias
- Departamento de Fisicoquímica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain; (Á.R.-A.); (J.M.P.); (C.D.B.); (J.A.G.-V.)
| | - Jose M. Paredes
- Departamento de Fisicoquímica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain; (Á.R.-A.); (J.M.P.); (C.D.B.); (J.A.G.-V.)
| | - Chiara Di Biase
- Departamento de Fisicoquímica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain; (Á.R.-A.); (J.M.P.); (C.D.B.); (J.A.G.-V.)
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Juan M. Cuerva
- Departamento de Química Orgánica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Ciencias, Universidad de Granada, Campus Fuentenueva, 18071 Granada, Spain;
| | - María D. Giron
- Departamento de Bioquímica y Biología Molecular II, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain; (M.D.G.); (R.S.)
| | - Rafael Salto
- Departamento de Bioquímica y Biología Molecular II, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain; (M.D.G.); (R.S.)
| | - Juan A. González-Vera
- Departamento de Fisicoquímica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain; (Á.R.-A.); (J.M.P.); (C.D.B.); (J.A.G.-V.)
| | - Angel Orte
- Departamento de Fisicoquímica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain; (Á.R.-A.); (J.M.P.); (C.D.B.); (J.A.G.-V.)
- Correspondence: ; Tel.: +34-958243825
| |
Collapse
|
48
|
Pharmacological Treatment of Alzheimer's Disease: Insights from Drosophila melanogaster. Int J Mol Sci 2020; 21:ijms21134621. [PMID: 32610577 PMCID: PMC7370071 DOI: 10.3390/ijms21134621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 01/01/2023] Open
Abstract
Aging is an ineluctable law of life. During the process of aging, the occurrence of neurodegenerative disorders is prevalent in the elderly population and the predominant type of dementia is Alzheimer’s disease (AD). The clinical symptoms of AD include progressive memory loss and impairment of cognitive functions that interfere with daily life activities. The predominant neuropathological features in AD are extracellular β-amyloid (Aβ) plaque deposition and intracellular neurofibrillary tangles (NFTs) of hyperphosphorylated Tau. Because of its complex pathobiology, some tangible treatment can only ameliorate the symptoms, but not prevent the disease altogether. Numerous drugs during pre-clinical or clinical studies have shown no positive effect on the disease outcome. Therefore, understanding the basic pathophysiological mechanism of AD is imperative for the rational design of drugs that can be used to prevent this disease. Drosophilamelanogaster has emerged as a highly efficient model system to explore the pathogenesis and treatment of AD. In this review we have summarized recent advancements in the pharmacological research on AD using Drosophila as a model species, discussed feasible treatment strategies and provided further reference for the mechanistic study and treatment of age-related AD.
Collapse
|
49
|
Biophysical studies of protein misfolding and aggregation in in vivo models of Alzheimer's and Parkinson's diseases. Q Rev Biophys 2020; 49:e22. [PMID: 32493529 DOI: 10.1017/s0033583520000025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurodegenerative disorders, including Alzheimer's (AD) and Parkinson's diseases (PD), are characterised by the formation of aberrant assemblies of misfolded proteins. The discovery of disease-modifying drugs for these disorders is challenging, in part because we still have a limited understanding of their molecular origins. In this review, we discuss how biophysical approaches can help explain the formation of the aberrant conformational states of proteins whose neurotoxic effects underlie these diseases. We discuss in particular models based on the transgenic expression of amyloid-β (Aβ) and tau in AD, and α-synuclein in PD. Because biophysical methods have enabled an accurate quantification and a detailed understanding of the molecular mechanisms underlying protein misfolding and aggregation in vitro, we expect that the further development of these methods to probe directly the corresponding mechanisms in vivo will open effective routes for diagnostic and therapeutic interventions.
Collapse
|
50
|
Wahid M, Ali A, Saqib F, Aleem A, Bibi S, Afzal K, Ali A, Baig A, Khan SA, Bin Asad MHH. Pharmacological exploration of traditional plants for the treatment of neurodegenerative disorders. Phytother Res 2020; 34:3089-3112. [DOI: 10.1002/ptr.6742] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Muqeet Wahid
- Faculty of Pharmacy, Department of Pharmacology Bahauddin Zakariya University Multan Pakistan
- Institute of Food Science and Nutrition Bahauddin Zakariya University Multan Pakistan
| | - Anam Ali
- Faculty of Pharmacy, Department of Pharmacology Bahauddin Zakariya University Multan Pakistan
| | - Fatima Saqib
- Faculty of Pharmacy, Department of Pharmacology Bahauddin Zakariya University Multan Pakistan
| | - Ambreen Aleem
- Faculty of Pharmacy, Department of Pharmacology Bahauddin Zakariya University Multan Pakistan
| | - Sumbal Bibi
- Department of Pharmacy COMSATS University Islamabad Abbottabad Pakistan
| | - Khurram Afzal
- Institute of Food Science and Nutrition Bahauddin Zakariya University Multan Pakistan
| | - Atif Ali
- Department of Pharmacy COMSATS University Islamabad Abbottabad Pakistan
| | - Ayesha Baig
- Department of Biotechnology COMSATS University Islamabad Abbottabad Pakistan
| | - Shujaat Ali Khan
- Department of Pharmacy COMSATS University Islamabad Abbottabad Pakistan
| | - Muhammad Hassham Hassan Bin Asad
- Department of Pharmacy COMSATS University Islamabad Abbottabad Pakistan
- Department of Genetics, Institute of Fundamental Medicine and Biology Kazan Federal University Kazan Russia
| |
Collapse
|