1
|
Aparajita A, Jain U, Srivastava P. "Current and emerging drug therapies in Alzheimer's disease: A pathophysiological Perspective". Neuroscience 2025; 565:499-518. [PMID: 39662528 DOI: 10.1016/j.neuroscience.2024.11.078] [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: 08/05/2024] [Revised: 11/18/2024] [Accepted: 11/30/2024] [Indexed: 12/13/2024]
Abstract
The analytical and experimental investigation of several targets and biomarkers that help in explaining significant cognitive deficits, covering drug development and precision medicine aimed at different chronic neurodegenerative conditions such as Alzheimer's disease (AD), Parkinson's disease, synaptic dysfunction, brain damage from neuronal apoptosis, and other disease pathologies; this served as the foundation for all phase studies. The focus of current therapeutic approaches is on developing humanized antibodies, agonist and antagonist drugs, receptors, signaling molecules, major targeted drug-metabolizing enzymes, and other metabolites to treat neurodegeneration in the AD brain brought on by tau hyperphosphorylation, amyloid plagues, or other cholinergic effects. The five A's-amnesia, agnosia, aphasia, apraxia, and anomia-are the typical symptoms associated with AD. While the main goal of drug therapeutics studies is modified amino acids acting as pro-drugs, pharmacokinetics studies and trends in evaluating drug-drug interactions focus on interactions between drugs and antibodies, drugs and therapeutic biologics like metabolites, herbs, interleukin-based, and gene silencing mechanism-based. Studies on the biotransformation of xenobiotic compounds and the metabolism of exogenous and endogenous substances are conducted under Phase I, Phase II, and Phase III trials because the pivotal pharmacokinetic properties of drugs, such as absorption, distribution, metabolism, and excretion (ADME), aid in understanding variations in the crucial improvement of various target drugs. This review also highlights the developments in soon-to-be genetically created targeted medications that may serve as ground-breaking treatments for cholinergic illnesses in the brains of AD patients and other neurodegenerative conditions.
Collapse
Affiliation(s)
- Aparajita Aparajita
- Department of Biosciences, Institute of Management Studies Ghaziabad 9(University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India
| | - Unnati Jain
- Department of Biosciences, Institute of Management Studies Ghaziabad 9(University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India
| | - Priyanka Srivastava
- Department of Biosciences, Institute of Management Studies Ghaziabad 9(University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India.
| |
Collapse
|
2
|
Kraybill EP, Mojabi FS, Heath AM, Spikes CR, Beard C, McNerney MW. rTMS Modulation of Behavioral and Biological Measures in 3xTg-AD Mice. Brain Sci 2024; 14:1186. [PMID: 39766385 PMCID: PMC11674534 DOI: 10.3390/brainsci14121186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/20/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES The biological basis for behavioral manifestations of Alzheimer's disease remains unclear. Emotional and behavioral alterations of Alzheimer's disease can result in substantial caregiver burden and lack effective management. This study expands upon previous work investigating behavioral alterations in mice with Alzheimer's disease and a potential treatment of increasing brain-derived neurotrophic factor (BDNF) using repetitive transcranial magnetic stimulation (rTMS). METHODS A total of 47 3xTg-AD (Alzheimer's) and 53 B6 (wildtype) mice were administered ANA12 (an antagonist of TrkB receptor) or Vehicle (saline) and then rTMS or Sham treatment daily. After 14 days of treatments and injections, mouse behavior was assessed under various behavioral cognitive tests. Mice were then perfused, and brain samples were processed for histology and protein assays. Brain homogenates were analyzed for BDNF and its downstream signaling molecules. RESULTS Open field testing demonstrated that 3xTg-AD mice spent more time in the center than B6 mice. 3xTg-AD-Sham mice injected with ANA12 were the only group to travel significantly less distance than B6-ANA12-Sham or B6-Vehicle-Sham mice (p < 0.05), while 3xTg-AD-rTMS mice (irrespective of injection) were not significantly different from B6 mice. 3xTg-AD mice had significantly greater measured levels of BDNF and TrkB than the wild-type mice. CONCLUSIONS Treatment of Alzheimer's disease using rTMS positively affects elements of hypoactivity, but not all behavioral abnormalities. rTMS shifted 3xTg-AD open field behavioral test measures, generating significant differences between untreated 3xTg-AD and B6 genotypes. Despite its benefit, further investigation of rTMS as a treatment for Alzheimer's disease as well as its biological underpinnings are needed.
Collapse
Affiliation(s)
- Eric P. Kraybill
- Mental Illness Research Education and Clinical Center (MIRECC), Department of Veteran Affairs, 3801 Miranda Ave, Palo Alto, CA 94304, USA
| | - Fatemeh S. Mojabi
- Mental Illness Research Education and Clinical Center (MIRECC), Department of Veteran Affairs, 3801 Miranda Ave, Palo Alto, CA 94304, USA
| | - Alesha M. Heath
- Mental Illness Research Education and Clinical Center (MIRECC), Department of Veteran Affairs, 3801 Miranda Ave, Palo Alto, CA 94304, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Rd, Stanford, CA 94305, USA
| | - Cierra R. Spikes
- Mental Illness Research Education and Clinical Center (MIRECC), Department of Veteran Affairs, 3801 Miranda Ave, Palo Alto, CA 94304, USA
- Department of Psychology, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA
| | - Charlotte Beard
- Mental Illness Research Education and Clinical Center (MIRECC), Department of Veteran Affairs, 3801 Miranda Ave, Palo Alto, CA 94304, USA
| | - M. Windy McNerney
- Mental Illness Research Education and Clinical Center (MIRECC), Department of Veteran Affairs, 3801 Miranda Ave, Palo Alto, CA 94304, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Rd, Stanford, CA 94305, USA
| |
Collapse
|
3
|
Deceuninck P, Gastaldello A, Mennecozzi M, Pistollato F. Exploring the connection between EU-funded research and methodological approaches: insights from a retrospective analysis. J Transl Med 2024; 22:891. [PMID: 39363357 PMCID: PMC11447993 DOI: 10.1186/s12967-024-05557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 07/29/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Over the last two decades, substantial investments have been directed towards supporting fundamental and applied research in Alzheimer's disease (AD), breast cancer (BC), and prostate cancer (PC), which continue to pose significant health challenges. Recently, the Joint Research Centre (JRC) of the European Commission (EC) conducted a retrospective analysis to examine the major scientific advancements resulting from EU-funded research in these disease areas and their impact on society. METHODS Building upon this analysis, our subsequent investigation delves into the methodological approaches-both animal and non-animal models and methods-employed in AD, BC, and PC research funded under past EU framework programs (FP5, FP6, FP7, and H2020), and explored the notable research outputs associated with these approaches. RESULTS Our findings indicate a prevalent use of animal-based methodologies in AD research, particularly evident in projects funded under H2020. Notably, projects focused on drug development, testing, or repurposing heavily relied on animal models. Conversely, research aimed at clinical trial design, patient stratification, diagnosis and diagnostic tool development, lifestyle interventions, and prevention-outputs with potential societal impact-more frequently utilised non-animal methods. Advanced investigations leveraging imaging, computational tools, biomarker discovery and organ/tissue chip technologies predominantly favoured non-animal strategies. CONCLUSIONS These insights highlight a correlation between methodological choices and the translational potential of research outcomes, suggesting the need for a reconsideration of research strategy planning in future framework programs.
Collapse
|
4
|
Liu N, Liang X, Chen Y, Xie L. Recent trends in treatment strategies for Alzheimer 's disease and the challenges: A topical advancement. Ageing Res Rev 2024; 94:102199. [PMID: 38232903 DOI: 10.1016/j.arr.2024.102199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/12/2024] [Indexed: 01/19/2024]
Abstract
Alzheimer's Disease (AD) is an irreversible and progressive neurological disease that has affected at least 50 million people around the globe. Considering the severity of the disease and the continuous increase in the number of patients, the development of new effective drugs or intervention strategies for AD has become urgent. AD is caused by a combination of genetic, environmental, and lifestyle factors, but its exact cause has not yet been clarified. Given the current challenges being faced in the clinical treatment of AD, such as complex AD pathological network and insufficient early diagnosis, herein, we have focused on the three core pathological features of AD, including amyloid-β (Aβ) aggregation, tau phosphorylation and tangles, and activation of inflammatory factors. In this review, we have briefly underscored the primary evidence supporting each pathology and discuss AD pathological network among Aβ, tau, and inflammation. We have also comprehensively summarized the most instructive drugs and their treatment strategies against Aβ, tau, or neuroinflammation used in basic research and clinical trials. Finally, we have discussed and outlined the pros and cons of each pathological approach and looked forward to potential personalized diagnosis and treatment strategies that are beneficial to AD patients.
Collapse
Affiliation(s)
- Ni Liu
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Xiaohan Liang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Yu Chen
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Lihang Xie
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
5
|
Boyton I, Valenzuela SM, Collins-Praino LE, Care A. Neuronanomedicine for Alzheimer's and Parkinson's disease: Current progress and a guide to improve clinical translation. Brain Behav Immun 2024; 115:631-651. [PMID: 37967664 DOI: 10.1016/j.bbi.2023.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 09/19/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023] Open
Abstract
Neuronanomedicine is an emerging multidisciplinary field that aims to create innovative nanotechnologies to treat major neurodegenerative disorders, such as Alzheimer's (AD) and Parkinson's disease (PD). A key component of neuronanomedicine are nanoparticles, which can improve drug properties and demonstrate enhanced safety and delivery across the blood-brain barrier, a major improvement on existing therapeutic approaches. In this review, we critically analyze the latest nanoparticle-based strategies to modify underlying disease pathology to slow or halt AD/PD progression. We find that a major roadblock for neuronanomedicine translation to date is a poor understanding of how nanoparticles interact with biological systems (i.e., bio-nano interactions), which is partly due to inconsistent reporting in published works. Accordingly, this review makes a set of specific recommendations to help guide researchers to harness the unique properties of nanoparticles and thus realise breakthrough treatments for AD/PD.
Collapse
Affiliation(s)
- India Boyton
- School of Life Sciences, University of Technology Sydney, Gadigal Country, NSW 2007, Australia
| | - Stella M Valenzuela
- School of Life Sciences, University of Technology Sydney, Gadigal Country, NSW 2007, Australia
| | | | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Gadigal Country, NSW 2007, Australia.
| |
Collapse
|
6
|
Piotrowski SL, Tucker A, Jacobson S. The elusive role of herpesviruses in Alzheimer's disease: current evidence and future directions. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:253-266. [PMID: 38013835 PMCID: PMC10474380 DOI: 10.1515/nipt-2023-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/26/2023] [Indexed: 11/29/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. While pathologic hallmarks, such as extracellular beta-amyloid plaques, are well-characterized in affected individuals, the pathogenesis that causes plaque formation and eventual cognitive decline is not well understood. A recent resurgence of the decades-old "infectious hypothesis" has garnered increased attention on the potential role that microbes may play in AD. In this theory, it is thought that pathogens such as viruses may act as seeds for beta-amyloid aggregation, ultimately leading to plaques. Interest in the infectious hypothesis has also spurred further investigation into additional characteristics of viral infection that may play a role in AD progression, such as neuroinflammation, latency, and viral DNA integration. While a flurry of research in this area has been recently published, with herpesviruses being of particular interest, the role of pathogens in AD remains controversial. In this review, the insights gained thus far into the possible role of herpesviruses in AD are summarized. The challenges and potential future directions of herpesvirus research in AD and dementia are also discussed.
Collapse
Affiliation(s)
- Stacey L. Piotrowski
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Comparative Biomedical Scientist Training Program, National Institutes of Health, Bethesda, MD, USA
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Allison Tucker
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Steven Jacobson
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
7
|
Hu L, Tao Y, Jiang Y, Qin F. Recent progress of nanomedicine in the treatment of Alzheimer's disease. Front Cell Dev Biol 2023; 11:1228679. [PMID: 37457297 PMCID: PMC10340527 DOI: 10.3389/fcell.2023.1228679] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of memory disruption in elderly subjects, with the prevalence continuing to rise mainly because of the aging world population. Unfortunately, no efficient therapy is currently available for the AD treatment, due to low drug potency and several challenges to delivery, including low bioavailability and the impediments of the blood-brain barrier. Recently, nanomedicine has gained considerable attention among researchers all over the world and shown promising developments in AD treatment. A wide range of nano-carriers, such as polymer nanoparticles, liposomes, solid lipid nanoparticles, dendritic nanoparticles, biomimetic nanoparticles, magnetic nanoparticles, etc., have been adapted to develop successful new treatment strategies. This review comprehensively summarizes the recent advances of different nanomedicine for their efficacy in pre-clinical studies. Finally, some insights and future research directions are proposed. This review can provide useful information to guide the future design and evaluation of nanomedicine in AD treatment.
Collapse
Affiliation(s)
- Liqiang Hu
- Mental Health Center and West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yiran Tao
- Mental Health Center and West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yanjiao Jiang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Feng Qin
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Wang Z, Gao C, Zhang L, Sui R. Hesperidin methylchalcone (HMC) hinders amyloid-β induced Alzheimer's disease by attenuating cholinesterase activity, macromolecular damages, oxidative stress and apoptosis via regulating NF-κB and Nrf2/HO-1 pathways. Int J Biol Macromol 2023; 233:123169. [PMID: 36623626 DOI: 10.1016/j.ijbiomac.2023.123169] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/04/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Phytocompounds therapy has recently emerged as an effective strategy to treat Alzheimer's disease. Herein, the protective effect of hesperidin methylchalcone (HMC) was evaluated through Alzheimer's disease models of Neuro-2a cells and Wistar rats. The in vitro results showed that HMC possesses significant ability to inhibit the acetylcholinesterase enzyme and exhibiting anti-aggregation and disaggregation properties. Furthermore, HMC could protect the Neuro-2a cells against Aβ-induced neurotoxicity. Simultaneously, HMC treatment significantly improved the cognitive deficits caused by Aβ-peptide on spatial memory in Wistar rats. HMC significantly enhanced the cholinergic effects by inhibiting AChE, BuChE, β-secretase activity, caspase-3 activity, and attenuating macromolecular damages and apoptosis. Notably, HMC reduced the Aβ-induced oxidative stress by activating the antioxidative defence enzymes. In addition, the HMC treatment suppressed the expression of immunocytokines such as p-NF-κB p65, p-IκBα, induced by Aβ; whereas upregulating Nrf2, HO-1 in brain homogenate. These results suggest that HMC could attenuate Aβ-induced neuroinflammation in brain via suppressing NF-κB signalling pathway and activating the Nrf2/HO-1 pathway, thereby improving memory and cognitive impairments in Wistar rats. Overall, the present study reports that HMC can act as a potent candidate with multi-faceted neuroprotective potential against Aβ-induced memory dysfunction in Wistar rats for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Zhuo Wang
- School of Nursing, Jinzhou Medical University, Jinzhou 121099, China
| | - Chao Gao
- School of Nursing, Jinzhou Medical University, Jinzhou 121099, China
| | - Lei Zhang
- School of Nursing, Jinzhou Medical University, Jinzhou 121099, China
| | - Rubo Sui
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121099, China.
| |
Collapse
|
9
|
Torok J, Anand C, Verma P, Raj A. Connectome-based biophysics models of Alzheimer's disease diagnosis and prognosis. Transl Res 2023; 254:13-23. [PMID: 36031051 PMCID: PMC11019890 DOI: 10.1016/j.trsl.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022]
Abstract
With the increasing prevalence of Alzheimer's disease (AD) among aging populations and the limited therapeutic options available to slow or reverse its progression, the need has never been greater for improved diagnostic tools for identifying patients in the preclinical and prodomal phases of AD. Biophysics models of the connectome-based spread of amyloid-beta (Aβ) and microtubule-associated protein tau (τ) have enjoyed recent success as tools for predicting the time course of AD-related pathological changes. However, given the complex etiology of AD, which involves not only connectome-based spread of protein pathology but also the interactions of many molecular and cellular players over multiple spatiotemporal scales, more robust, complete biophysics models are needed to better understand AD pathophysiology and ultimately provide accurate patient-specific diagnoses and prognoses. Here we discuss several areas of active research in AD whose insights can be used to enhance the mathematical modeling of AD pathology as well as recent attempts at developing improved connectome-based biophysics models. These efforts toward a comprehensive yet parsimonious mathematical description of AD hold great promise for improving both the diagnosis of patients at risk for AD and our mechanistic understanding of how AD progresses.
Collapse
Affiliation(s)
- Justin Torok
- Department of Radiology, University of California, San Francisco, San Francisco, California.
| | - Chaitali Anand
- Department of Radiology, University of California, San Francisco, San Francisco, California
| | - Parul Verma
- Department of Radiology, University of California, San Francisco, San Francisco, California
| | - Ashish Raj
- Department of Radiology, University of California, San Francisco, San Francisco, California; Department of Bioengineering, University of California, Berkeley and University of California, San Francisco, Berkeley, California; Department of Radiology, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
10
|
Minh Quang N, Tran Thai H, Le Thi H, Duc Cuong N, Hien NQ, Hoang D, Ngoc VTB, Ky Minh V, Van Tat P. Novel Thiosemicarbazone Quantum Dots in the Treatment of Alzheimer's Disease Combining In Silico Models Using Fingerprints and Physicochemical Descriptors. ACS OMEGA 2023; 8:11076-11099. [PMID: 37008140 PMCID: PMC10061515 DOI: 10.1021/acsomega.2c07934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/07/2023] [Indexed: 06/19/2023]
Abstract
Searching for thiosemicarbazone derivatives with the potential to inhibit acetylcholinesterase for the treatment of Alzheimer's disease (AD) is an important current goal. The QSARKPLS, QSARANN, and QSARSVR models were constructed using binary fingerprints and physicochemical (PC) descriptors of 129 thiosemicarbazone compounds screened from a database of 3791 derivatives. The R 2 and Q 2 values for the QSARKPLS, QSARANN, and QSARSVR models are greater than 0.925 and 0.713 using dendritic fingerprint (DF) and PC descriptors, respectively. The in vitro pIC50 activities of four new design-oriented compounds N1, N2, N3, and N4, from the QSARKPLS model using DFs, are consistent with the experimental results and those from the QSARANN and QSARSVR models. The designed compounds N1, N2, N3, and N4 do not violate Lipinski-5 and Veber rules using the ADME and BoiLED-Egg methods. The binding energy, kcal mol-1, of the novel compounds to the 1ACJ-PDB protein receptor of the AChE enzyme was also obtained by molecular docking and dynamics simulations consistent with those predicted from the QSARANN and QSARSVR models. New compounds N1, N2, N3, and N4 were synthesized, and the experimental in vitro pIC50 activity was determined in agreement with those obtained from in silico models. The newly synthesized thiosemicarbazones N1, N2, N3, and N4 can inhibit 1ACJ-PDB, which is predicted to be able to cross the barrier. The DFT B3LYP/def-SV(P)-ECP quantization calculation method was used to calculate E HOMO and E LUMO to account for the activities of compounds N1, N2, N3, and N4. The quantum calculation results explained are consistent with those obtained in in silico models. The successful results here may contribute to the search for new drugs for the treatment of AD.
Collapse
Affiliation(s)
- Nguyen Minh Quang
- Faculty
of Chemical Engineering, Industrial University
of Ho Chi Minh City, 12 Nguyen Van Bao, Dist. Go Vap, Ho Chi Minh 700000, Viet Nam
| | - Hoa Tran Thai
- Faculty
of Chemistry, Hue University of Sciences, Hue University, 77 Nguyen Hue, Hue City 530000, Viet Nam
| | - Hoa Le Thi
- Faculty
of Chemistry, Hue University of Sciences, Hue University, 77 Nguyen Hue, Hue City 530000, Viet Nam
| | - Nguyen Duc Cuong
- Faculty
of Chemistry, Hue University of Sciences, Hue University, 77 Nguyen Hue, Hue City 530000, Viet Nam
- School
of Hospitality and Tourism, Hue University, 22 Lam Hoang, Hue City 530000, Viet
Nam
| | - Nguyen Quoc Hien
- Vietnam
Atomic Energy Institute, 59 Ly Thuong Kiet, Dist. Hoan Kiem, Hanoi
City 100000, Viet Nam
| | - DongQuy Hoang
- Faculty
of
Materials Science and Technology, University of Science, Vietnam National University, Ho Chi Minh 700000, Viet Nam
- Vietnam
National University, Ho Chi Minh
City 700000, Viet Nam
| | - Vu Thi Bao Ngoc
- Faculty
of Chemistry and Environment, University
of Dalat, 01 Phu Dong Thien Vuong, Dalat City 660000, Viet Nam
| | - Vo Ky Minh
- Franklin
High School, 6400 Whitelock Pkwy, Elk Grove, California 95757, United States
| | - Pham Van Tat
- Department
of Sciences and Journal Management, Hoa
Sen University, 08 Nguyen Van Trang, Dist. 01, Ho Chi Minh 700000, Viet Nam
| |
Collapse
|
11
|
Charpignon ML, Vakulenko-Lagun B, Zheng B, Magdamo C, Su B, Evans K, Rodriguez S, Sokolov A, Boswell S, Sheu YH, Somai M, Middleton L, Hyman BT, Betensky RA, Finkelstein SN, Welsch RE, Tzoulaki I, Blacker D, Das S, Albers MW. Causal inference in medical records and complementary systems pharmacology for metformin drug repurposing towards dementia. Nat Commun 2022; 13:7652. [PMID: 36496454 PMCID: PMC9741618 DOI: 10.1038/s41467-022-35157-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Metformin, a diabetes drug with anti-aging cellular responses, has complex actions that may alter dementia onset. Mixed results are emerging from prior observational studies. To address this complexity, we deploy a causal inference approach accounting for the competing risk of death in emulated clinical trials using two distinct electronic health record systems. In intention-to-treat analyses, metformin use associates with lower hazard of all-cause mortality and lower cause-specific hazard of dementia onset, after accounting for prolonged survival, relative to sulfonylureas. In parallel systems pharmacology studies, the expression of two AD-related proteins, APOE and SPP1, was suppressed by pharmacologic concentrations of metformin in differentiated human neural cells, relative to a sulfonylurea. Together, our findings suggest that metformin might reduce the risk of dementia in diabetes patients through mechanisms beyond glycemic control, and that SPP1 is a candidate biomarker for metformin's action in the brain.
Collapse
Affiliation(s)
- Marie-Laure Charpignon
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Bang Zheng
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - Colin Magdamo
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Bowen Su
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Kyle Evans
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Steve Rodriguez
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Artem Sokolov
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Sarah Boswell
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Yi-Han Sheu
- Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Melek Somai
- Inception Labs, Collaborative for Health Delivery Sciences, Medical College of Wisconsin, Wauwatosa, WI, USA
| | - Lefkos Middleton
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
- Public Health Directorate, Imperial College London NHS Healthcare Trust, London, UK
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Rebecca A Betensky
- Department of Biostatistics, School of Global Public Health, New York University, New York, NY, USA
| | - Stan N Finkelstein
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Clinical Informatics, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Roy E Welsch
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA, USA
- Sloan School of Management, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
- Dementia Research Institute, Imperial College London, London, UK.
- Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece.
| | - Deborah Blacker
- Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
| | - Mark W Albers
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Ko H, Yoon SP. Optogenetic neuromodulation with gamma oscillation as a new strategy for Alzheimer disease: a narrative review. JOURNAL OF YEUNGNAM MEDICAL SCIENCE 2022; 39:269-277. [PMID: 35152662 PMCID: PMC9580057 DOI: 10.12701/jyms.2021.01683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/27/2022] [Accepted: 02/04/2022] [Indexed: 12/31/2022]
Abstract
The amyloid hypothesis has been considered a major explanation of the pathogenesis of Alzheimer disease. However, failure of phase III clinical trials with anti-amyloid-beta monoclonal antibodies reveals the need for other therapeutic approaches to treat Alzheimer disease. Compared to its relatively short history, optogenetics has developed considerably. The expression of microbial opsins in cells using genetic engineering allows specific control of cell signals or molecules. The application of optogenetics to Alzheimer disease research or clinical approaches is increasing. When applied with gamma entrainment, optogenetic neuromodulation can improve Alzheimer disease symptoms. Although safety problems exist with optogenetics such as the use of viral vectors, this technique has great potential for use in Alzheimer disease. In this paper, we review the historical applications of optogenetic neuromodulation with gamma entrainment to investigate the mechanisms involved in Alzheimer disease and potential therapeutic strategies.
Collapse
Affiliation(s)
- Haneol Ko
- Medical Course, Jeju National University School of Medicine, Jeju, Korea
| | - Sang-Pil Yoon
- Department of Anatomy, Jeju National University College of Medicine, Jeju, Korea
| |
Collapse
|
13
|
Li C, Xiang Y, Wang Y, Li P. Study on Nano Drug Particles in the Diagnosis and Treatment of Alzheimer's Disease in the Elderly. Bioinorg Chem Appl 2022; 2022:3335581. [PMID: 36212985 PMCID: PMC9536957 DOI: 10.1155/2022/3335581] [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: 08/08/2022] [Revised: 08/22/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
Objective To study the clinical effect of nano drug particles in the diagnosis and treatment of Alzheimer's disease in the elderly, we promote the research on the treatment of Alzheimer's disease and provide basis. Methods 80 patients with Alzheimer's disease treated in our hospital from April 2021 to April 2022 were selected as the research objects and were divided into the reference group and the observation group by random number table method. The reference group was given traditional treatment drugs, and the observation group was given nano drug particles. The treatment effect, blood lipid level, cognitive function status, and comprehensive effective rate of the two groups were compared, and the treatment data of the patients were scored by ADL and MMSE scale to understand the intelligence level and daily living ability of the patients. Results From the comparison results, it is found that the blood lipid level of the control group was in the normal range. The cognitive function of the control group was also better than that of the reference group. The scores of ADL and MMSE in the control group were higher than those in the reference group. The effective rate of the control group was also higher than that of the reference group; P < 0.05, with statistical significance. Conclusion In the diagnosis and treatment of Alzheimer's disease in the elderly, nano drug particles have good stability and less toxic and side effects, improve the ability of daily living of patients, and have good clinical treatment effect, which is worthy of clinical application.
Collapse
Affiliation(s)
- ChangQing Li
- Department of Laboratory Medicine, Chengdu Eighth People's Hospital (Geriatric Hospital of Chengdu Medical College), Chengdu 610000, Sichuan, China
| | - Yu Xiang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunqian Wang
- Department of Laboratory Medicine, Chengdu Eighth People's Hospital (Geriatric Hospital of Chengdu Medical College), Chengdu 610000, Sichuan, China
| | - Ping Li
- Department of Laboratory Medicine, Chengdu Eighth People's Hospital (Geriatric Hospital of Chengdu Medical College), Chengdu 610000, Sichuan, China
| |
Collapse
|
14
|
Needham H, Torpey G, Flores CC, Davis CJ, Vanderheyden WM, Gerstner JR. A Dichotomous Role for FABP7 in Sleep and Alzheimer's Disease Pathogenesis: A Hypothesis. Front Neurosci 2022; 16:798994. [PMID: 35844236 PMCID: PMC9280343 DOI: 10.3389/fnins.2022.798994] [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: 10/20/2021] [Accepted: 05/10/2022] [Indexed: 11/15/2022] Open
Abstract
Fatty acid binding proteins (FABPs) are a family of intracellular lipid chaperone proteins known to play critical roles in the regulation of fatty acid uptake and transport as well as gene expression. Brain-type fatty acid binding protein (FABP7) is enriched in astrocytes and has been implicated in sleep/wake regulation and neurodegenerative diseases; however, the precise mechanisms underlying the role of FABP7 in these biological processes remain unclear. FABP7 binds to both arachidonic acid (AA) and docosahexaenoic acid (DHA), resulting in discrete physiological responses. Here, we propose a dichotomous role for FABP7 in which ligand type determines the subcellular translocation of fatty acids, either promoting wakefulness aligned with Alzheimer's pathogenesis or promoting sleep with concomitant activation of anti-inflammatory pathways and neuroprotection. We hypothesize that FABP7-mediated translocation of AA to the endoplasmic reticulum of astrocytes increases astrogliosis, impedes glutamatergic uptake, and enhances wakefulness and inflammatory pathways via COX-2 dependent generation of pro-inflammatory prostaglandins. Conversely, we propose that FABP7-mediated translocation of DHA to the nucleus stabilizes astrocyte-neuron lactate shuttle dynamics, preserves glutamatergic uptake, and promotes sleep by activating anti-inflammatory pathways through the peroxisome proliferator-activated receptor-γ transcriptional cascade. Importantly, this model generates several testable hypotheses applicable to other neurodegenerative diseases, including amyotrophic lateral sclerosis and Parkinson's disease.
Collapse
Affiliation(s)
- Hope Needham
- Department of Biology, Gonzaga University, Spokane, WA, United States
| | - Grace Torpey
- Department of Biology, Gonzaga University, Spokane, WA, United States
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - William M. Vanderheyden
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
15
|
Cummings J, Montes A, Kamboj S, Cacho JF. The role of basket trials in drug development for neurodegenerative disorders. Alzheimers Res Ther 2022; 14:73. [PMID: 35614479 PMCID: PMC9131689 DOI: 10.1186/s13195-022-01015-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/09/2022] [Indexed: 11/23/2022]
Abstract
Background Drug development for neurodegenerative disorders (NDDs) is a long, complex, and expensive enterprise. Methods to optimize drug development for NDDs are needed. Basket trials have been widely used in oncology and have been promoted by the Food and Drug Administration as a means of enhancing the efficiency of drug development. Discussion We reviewed clinical trials for NDDs registered on clinicaltrials.gov in the past 10 years. We identified 59 basket trials assessing the impact of treatment on more than one NDD in the trial. Forty-one of the trials were for 25 agents addressing symptoms of NDD such as motor impairment, hypotension, or psychosis. Eighteen of the trials assessed 14 disease-modifying therapies; the principal targets were mitochondrial function, tau biology, or alpha-synuclein aggregation. Basket trials are most common in phase 2 but have been conducted in phase 1, phase 3, and phase 4. The duration and size of the basket trials are highly variable depending on their developmental phase and the intent of the trial. Parkinson’s disease was the most common disorder included in basket trials of symptomatic agents, and Alzheimer’s disease was the most common disorder included in basket trials of disease-modifying therapies. Most of the basket trials of symptomatic agents were sponsored by pharmaceutical companies (29 of 41 trials); similarly, most of the basket trials investigating DMTs in basket trials were sponsored by the biopharmaceutical industry (11/17 trials). Conclusions Basket trials may increase drug development efficiency by reducing redundancy in trial implementation, enhancing recruitment, sharing placebo groups, and using biomarkers relevant to the mechanism of action of the treatment across NDDs. There have been relatively few basket trials including multiple NDDs in the same trial conducted over the past 10 years. The use of the basket trial strategy may represent an opportunity to increase the efficiency of development programs for agents to treat NDDs.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA. .,, Henderson, NV, 89052, USA.
| | - Arturo Montes
- Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Sana Kamboj
- Department of Neurosurgery, John Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jorge Fonseca Cacho
- Department of Computer Science, Howard Hughes School of Engineering, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| |
Collapse
|
16
|
Ye C, Cheng M, Ma L, Zhang T, Sun Z, Yu C, Wang J, Dou Y. Oxytocin Nanogels Inhibit Innate Inflammatory Response for Early Intervention in Alzheimer's Disease. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21822-21835. [PMID: 35510352 DOI: 10.1021/acsami.2c00007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Prevention of Alzheimer's disease (AD) is a global imperative, but reliable early interventions are currently lacking. Microglia-mediated chronic neuroinflammation is thought to occur in the early stage of AD and plays a critical role in AD pathogenesis. Here, oxytocin (OT)-loaded angiopep-2-modified chitosan nanogels (AOC NGs) were designed for early treatment of AD via inhibiting innate inflammatory response. Through the effective transcytosis of angiopep-2, AOC NGs were driven intravenously to cross the blood-brain barrier, enter the brain, and enrich in brain areas affected by AD. A large amount of OT was then released and specifically bound to the pathological upregulated OT receptor, thus effectively inhibiting microglial activation and reducing inflammatory cytokine levels through blocking the ERK/p38 MAPK and COX-2/iNOS NF-κB signaling pathways. Consecutive weekly intravenous administration of AOC NGs into 12-week-old young APP/PS1 mice, representing the early stage of AD, remarkably slowed the progression of Aβ deposition and neuronal apoptosis in the APP/PS1 mice as they aged and ultimately prevented cognitive impairment and delayed hippocampal atrophy. Together, the findings suggest that AOC NGs, which show good biosafety, can serve as a promising therapeutic candidate to combat neuroinflammation for early prevention of AD.
Collapse
Affiliation(s)
- Caihua Ye
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Meng Cheng
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Lin Ma
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Tianzhu Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Junping Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| |
Collapse
|
17
|
Cuní-López C, Stewart R, Quek H, White AR. Recent Advances in Microglia Modelling to Address Translational Outcomes in Neurodegenerative Diseases. Cells 2022; 11:cells11101662. [PMID: 35626698 PMCID: PMC9140031 DOI: 10.3390/cells11101662] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are deteriorating conditions of the nervous system that are rapidly increasing in the aging population. Increasing evidence suggests that neuroinflammation, largely mediated by microglia, the resident immune cells of the brain, contributes to the onset and progression of neurodegenerative diseases. Hence, microglia are considered a major therapeutic target that could potentially yield effective disease-modifying treatments for neurodegenerative diseases. Despite the interest in studying microglia as drug targets, the availability of cost-effective, flexible, and patient-specific microglia cellular models is limited. Importantly, the current model systems do not accurately recapitulate important pathological features or disease processes, leading to the failure of many therapeutic drugs. Here, we review the key roles of microglia in neurodegenerative diseases and provide an update on the current microglia platforms utilised in neurodegenerative diseases, with a focus on human microglia-like cells derived from peripheral blood mononuclear cells as well as human-induced pluripotent stem cells. The described microglial platforms can serve as tools for investigating disease biomarkers and improving the clinical translatability of the drug development process in neurodegenerative diseases.
Collapse
Affiliation(s)
- Carla Cuní-López
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- Faculty of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Romal Stewart
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- UQ Centre for Clinical Research, The University of Queensland, Royal Brisbane & Women’s Hospital, Brisbane, QLD 4006, Australia
| | - Hazel Quek
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- Correspondence: (H.Q.); (A.R.W.)
| | - Anthony R. White
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- Correspondence: (H.Q.); (A.R.W.)
| |
Collapse
|
18
|
Abdi S, Javanmehr N, Ghasemi-Kasman M, Bali HY, Pirzadeh M. Stem Cell-based Therapeutic and Diagnostic Approaches in Alzheimer's Disease. Curr Neuropharmacol 2022; 20:1093-1115. [PMID: 34970956 PMCID: PMC9886816 DOI: 10.2174/1570159x20666211231090659] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/24/2021] [Accepted: 09/21/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative impairment mainly recognized by memory loss and cognitive deficits. However, the current therapies against AD are mostly limited to palliative medications, prompting researchers to investigate more efficient therapeutic approaches for AD, such as stem cell therapy. Recent evidence has proposed that extensive neuronal and synaptic loss and altered adult neurogenesis, which is perceived pivotal in terms of plasticity and network maintenance, occurs early in the course of AD, which exacerbates neuronal vulnerability to AD. Thus, regeneration and replenishing the depleted neuronal networks by strengthening the endogenous repair mechanisms or exogenous stem cells and their cargoes is a rational therapeutic approach. Currently, several stem cell-based therapies as well as stem cell products like exosomes, have shown promising results in the early diagnosis of AD. OBJECTIVE This review begins with a comparison between AD and normal aging pathophysiology and a discussion on open questions in the field. Next, summarizing the current stem cell-based therapeutic and diagnostic approaches, we declare the advantages and disadvantages of each method. Also, we comprehensively evaluate the human clinical trials of stem cell therapies for AD. METHODOLOGY Peer-reviewed reports were extracted through Embase, PubMed, and Google Scholar until 2021. RESULTS With several ongoing clinical trials, stem cells and their derivatives (e.g., exosomes) are an emerging and encouraging field in diagnosing and treating neurodegenerative diseases. Although stem cell therapies have been successful in animal models, numerous clinical trials in AD patients have yielded unpromising results, which we will further discuss.
Collapse
Affiliation(s)
- Sadaf Abdi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Nima Javanmehr
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran;,Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran,Address correspondence to this author at the Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, P.O. Box 4136747176, Babol, Iran; Tel/Fax: +98-11-32190557; E-mail:
| | | | - Marzieh Pirzadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
19
|
TARBP2-stablized SNHG7 regulates blood-brain barrier permeability by acting as a competing endogenous RNA to miR-17-5p/NFATC3 in Aβ-microenvironment. Cell Death Dis 2022; 13:457. [PMID: 35562351 PMCID: PMC9106673 DOI: 10.1038/s41419-022-04920-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 12/14/2022]
Abstract
Breakdown of blood-brain barrier (BBB) is recognized as serious pathological marker of Alzheimer's disease development. Studies confirmed that β-amyloid (Aβ) deposition induced high BBB permeability by disrupting tight junction (TJ) proteins formed from endothelial cells (ECs). Here, we found TARBP2, SNHG7 and NFATC3 in expressions were increased and miR-17-5p expression was decreased in Aβ(1-42)-incubated ECs. Overexpression of TARBP2, SNHG7 and NFATC3 elevated BBB permeability and knockdown of them had converse results. Agomir-17-5p decreased BBB permeability and antagomir-17-5p increased BBB permeability. TARBP2 as a RNA-binding protein (RBP) bound to SNHG7 and resulted in longer half-life of SNHG7. The decreased expression of miR-17-5p had a negative post-transcriptional regulation to NFATC3, leading to the increased expression of NFATC3. In addition, SNHG7 regulated NFATC3 expression by acting as a molecule sponge targeting to miR-17-5p. NFATC3 inhibited TJ proteins expression by functioning as a transcription factor. TARBP2/SNHG7/miR-17-5p/NFATC3 pathway implied a potential mechanism in studies of BBB changes in AD pathological progression.
Collapse
|
20
|
Menden A, Hall D, Hahn-Townsend C, Broedlow CA, Joshi U, Pearson A, Crawford F, Evans JE, Klatt N, Crynen S, Mullan M, Ait-Ghezala G. Exogenous lipase administration alters gut microbiota composition and ameliorates Alzheimer's disease-like pathology in APP/PS1 mice. Sci Rep 2022; 12:4797. [PMID: 35314754 PMCID: PMC8938460 DOI: 10.1038/s41598-022-08840-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 03/03/2022] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) represents the most common form of dementia in the elderly with no available disease modifying treatments. Altered gut microbial composition has been widely acknowledged as a common feature of AD, which potentially contributes to progression or onset of AD. To assess the hypothesis that Candida rugosa lipase (CRL), which has been shown to enhance gut microbiome and metabolite composition, can rebalance the gut microbiome composition and reduce AD pathology, the treatment effects in APPswe/PS1de9 (APP/PS1) mice were investigated. The analysis revealed an increased abundance of Acetatifactor and Clostridiales vadin BB60 genera in the gut; increased lipid hydrolysis in the gut lumen, normalization of peripheral unsaturated fatty acids, and reduction of neuroinflammation and memory deficits post treatment. Finally, we demonstrated that the evoked benefits on memory could be transferred via fecal matter transplant (FMT) into antibiotic-induced microbiome-depleted (AIMD) wildtype mice, ameliorating their memory deficits. The findings herein contributed to improve our understanding of the role of the gut microbiome in AD's complex networks and suggested that targeted modification of the gut could contribute to amelioration of AD neuropathology.
Collapse
Affiliation(s)
- Ariane Menden
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK.
| | - Davane Hall
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | | | - Courtney A Broedlow
- Division of Surgical Outcomes and Precision Medicine Research, Department of Surgery, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Utsav Joshi
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Andrew Pearson
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK
| | - Fiona Crawford
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK
- James A. Haley Veterans' Hospital, 13000 Bruce B. Downs Boulevard, Tampa, FL, 33612, USA
| | - James E Evans
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Nichole Klatt
- Division of Surgical Outcomes and Precision Medicine Research, Department of Surgery, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Stefan Crynen
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK
| | - Michael Mullan
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK
| | - Ghania Ait-Ghezala
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK
| |
Collapse
|
21
|
The Multifaceted Role of Neuroprotective Plants in Alzheimer’s Disease Treatment. Geriatrics (Basel) 2022; 7:geriatrics7020024. [PMID: 35314596 PMCID: PMC8938774 DOI: 10.3390/geriatrics7020024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is an age-related, progressive neurodegenerative disorder characterized by impaired cognition, memory loss, and altered personality. Many of the available pharmaceutical treatments do not alter the onset of disease progression. Recently, alternatives to developed drug candidates have been explored including medicinal plants and herbal treatments for the treatment of AD. This article examines the role of herbal plant extracts and the neuroprotective effects as alternative modes of intervention for AD progression. These extracts contain key metabolites that culminate alterations in AD progression. The traditional plant extracts explored in this article induce a variety of beneficial properties, including antioxidants, anti-inflammatory, and enhanced cognition, while also inducing activity on AD drug targets such as Aβ degradation. While these neuroprotective aspects for AD are relatively recent, there is great potential in the drug discovery aspect of these plant extracts for future use in AD treatment.
Collapse
|
22
|
Jayatunga DPW, Hone E, Fernando WMADB, Garg ML, Verdile G, Martins RN. Mitoprotective Effects of a Synergistic Nutraceutical Combination: Basis for a Prevention Strategy Against Alzheimer’s Disease. Front Aging Neurosci 2022; 13:781468. [PMID: 35264941 PMCID: PMC8899513 DOI: 10.3389/fnagi.2021.781468] [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: 09/22/2021] [Accepted: 12/21/2021] [Indexed: 11/29/2022] Open
Abstract
Evidence to date suggests the consumption of food rich in bioactive compounds, such as polyphenols, flavonoids, omega-3 fatty acids may potentially minimize age-related cognitive decline. For neurodegenerative diseases, such as Alzheimer’s disease (AD), which do not yet have definitive treatments, the focus has shifted toward using alternative approaches, including prevention strategies rather than disease reversal. In this aspect, certain nutraceuticals have become promising compounds due to their neuroprotective properties. Moreover, the multifaceted AD pathophysiology encourages the use of multiple bioactive components that may be synergistic in their protective roles when combined. The objective of the present study was to determine mechanisms of action underlying the inhibition of Aβ1–42-induced toxicity by a previously determined, three-compound nutraceutical combination D5L5U5 for AD. In vitro experiments were carried out in human neuroblastoma BE(2)-M17 cells for levels of ROS, ATP mitophagy, and mitobiogenesis. The component compounds luteolin (LUT), DHA, and urolithin A (UA) were independently protective of mitochondria; however, the D5L5U5 preceded its single constituents in all assays used. Overall, it indicated that D5L5U5 had potent inhibitory effects against Aβ1–42-induced toxicity through protecting mitochondria. These mitoprotective activities included minimizing oxidative stress, increasing ATP and inducing mitophagy and mitobiogenesis. However, this synergistic nutraceutical combination warrants further investigations in other in vitro and in vivo AD models to confirm its potential to be used as a preventative therapy for AD.
Collapse
Affiliation(s)
- Dona P. W. Jayatunga
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Eugene Hone
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Cooperative Research Centre for Mental Health, Carlton, VIC, Australia
| | - W. M. A. D. Binosha Fernando
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Manohar L. Garg
- Faculty of Health and Medicine, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Giuseppe Verdile
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Faculty of Health Sciences, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Ralph N. Martins
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Australian Alzheimer’s Research Foundation, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
- *Correspondence: Ralph N. Martins,
| |
Collapse
|
23
|
Varesi A, Pierella E, Romeo M, Piccini GB, Alfano C, Bjørklund G, Oppong A, Ricevuti G, Esposito C, Chirumbolo S, Pascale A. The Potential Role of Gut Microbiota in Alzheimer’s Disease: from Diagnosis to Treatment. Nutrients 2022; 14:nu14030668. [PMID: 35277027 PMCID: PMC8840394 DOI: 10.3390/nu14030668] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 12/04/2022] Open
Abstract
Gut microbiota is emerging as a key regulator of many disease conditions and its dysregulation is implicated in the pathogenesis of several gastrointestinal and extraintestinal disorders. More recently, gut microbiome alterations have been linked to neurodegeneration through the increasingly defined gut microbiota brain axis, opening the possibility for new microbiota-based therapeutic options. Although several studies have been conducted to unravel the possible relationship between Alzheimer’s Disease (AD) pathogenesis and progression, the diagnostic and therapeutic potential of approaches aiming at restoring gut microbiota eubiosis remain to be fully addressed. In this narrative review, we briefly summarize the role of gut microbiota homeostasis in brain health and disease, and we present evidence for its dysregulation in AD patients. Based on these observations, we then discuss how dysbiosis might be exploited as a new diagnostic tool in early and advanced disease stages, and we examine the potential of prebiotics, probiotics, fecal microbiota transplantation, and diets as complementary therapeutic interventions on disease pathogenesis and progression, thus offering new insights into the diagnosis and treatment of this devastating and progressive disease.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy;
- Almo Collegio Borromeo, 27100 Pavia, Italy
- Correspondence: (A.V.); (G.R.)
| | - Elisa Pierella
- School of Medicine, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK; (E.P.); (A.O.)
| | - Marcello Romeo
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy;
| | | | - Claudia Alfano
- Department of Emergency Medicine and Surgery, IRCCS Fondazione Policlinico San Matteo, 27100 Pavia, Italy;
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), 8610 Mo i Rana, Norway;
| | - Abigail Oppong
- School of Medicine, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK; (E.P.); (A.O.)
| | - Giovanni Ricevuti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
- Correspondence: (A.V.); (G.R.)
| | - Ciro Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri, University of Pavia, 27100 Pavia, Italy;
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37121 Verona, Italy;
| | - Alessia Pascale
- Section of Pharmacology, Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
24
|
McKee CA, Lee J, Cai Y, Saito T, Saido T, Musiek ES. Astrocytes deficient in circadian clock gene Bmal1 show enhanced activation responses to amyloid-beta pathology without changing plaque burden. Sci Rep 2022; 12:1796. [PMID: 35110643 PMCID: PMC8810760 DOI: 10.1038/s41598-022-05862-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
An emerging link between circadian clock function and neurodegeneration has indicated a critical role for the molecular clock in brain health. We previously reported that deletion of the core circadian clock gene Bmal1 abrogates clock function and induces cell-autonomous astrocyte activation. Regulation of astrocyte activation has important implications for protein aggregation, inflammation, and neuronal survival in neurodegenerative conditions such as Alzheimer's disease (AD). Here, we investigated how astrocyte activation induced by Bmal1 deletion regulates astrocyte gene expression, amyloid-beta (Aβ) plaque-associated activation, and plaque deposition. To address these questions, we crossed astrocyte-specific Bmal1 knockout mice (Aldh1l1-CreERT2;Bmal1fl/fl, termed BMAL1 aKO), to the APP/PS1-21 and the APPNL-G-F models of Aβ accumulation. Transcriptomic profiling showed that BMAL1 aKO induced a unique transcriptional profile affecting genes involved in both the generation and elimination of Aβ. BMAL1 aKO mice showed exacerbated astrocyte activation around Aβ plaques and altered gene expression. However, this astrogliosis did not affect plaque accumulation or neuronal dystrophy in either model. Our results demonstrate that the striking astrocyte activation induced by Bmal1 knockout does not influence Aβ deposition, which indicates that the effect of astrocyte activation on plaque pathology in general is highly dependent on the molecular mechanism of activation.
Collapse
Affiliation(s)
- Celia A McKee
- Department of Neurology and Center On Biological Rhythms And Sleep, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jiyeon Lee
- Department of Neurology and Center On Biological Rhythms And Sleep, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Yuqi Cai
- Department of Neurology and Center On Biological Rhythms And Sleep, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan
| | - Erik S Musiek
- Department of Neurology and Center On Biological Rhythms And Sleep, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
25
|
Aillaud I, Kaniyappan S, Chandupatla RR, Ramirez LM, Alkhashrom S, Eichler J, Horn AHC, Zweckstetter M, Mandelkow E, Sticht H, Funke SA. A novel D-amino acid peptide with therapeutic potential (ISAD1) inhibits aggregation of neurotoxic disease-relevant mutant Tau and prevents Tau toxicity in vitro. Alzheimers Res Ther 2022; 14:15. [PMID: 35063014 PMCID: PMC8783508 DOI: 10.1186/s13195-022-00959-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/06/2022] [Indexed: 12/19/2022]
Abstract
Background Alzheimer’s disease (AD), the most common form of dementia, is a progressive neurodegenerative disorder that mainly affects older adults. One of the pathological hallmarks of AD is abnormally aggregated Tau protein that forms fibrillar deposits in the brain. In AD, Tau pathology correlates strongly with clinical symptoms, cognitive dysfunction, and neuronal death. Methods We aimed to develop novel therapeutic D-amino acid peptides as Tau fibrillization inhibitors. It has been previously demonstrated that D-amino acid peptides are protease stable and less immunogenic than L-peptides, and these characteristics may render them suitable for in vivo applications. Using a phage display procedure against wild type full-length Tau (TauFL), we selected a novel Tau binding L-peptide and synthesized its D-amino acid version ISAD1 and its retro inversed form, ISAD1rev, respectively. Results While ISAD1rev inhibited Tau aggregation only moderately, ISAD1 bound to Tau in the aggregation-prone PHF6 region and inhibited fibrillization of TauFL, disease-associated mutant full-length Tau (TauFLΔK, TauFL-A152T, TauFL-P301L), and pro-aggregant repeat domain Tau mutant (TauRDΔK). ISAD1 and ISAD1rev induced the formation of large high molecular weight TauFL and TauRDΔK oligomers that lack proper Thioflavin-positive β-sheet conformation even at lower concentrations. In silico modeling of ISAD1 Tau interaction at the PHF6 site revealed a binding mode similar to those known for other PHF6 binding peptides. Cell culture experiments demonstrated that ISAD1 and its inverse form are taken up by N2a-TauRDΔK cells efficiently and prevent cytotoxicity of externally added Tau fibrils as well as of internally expressed TauRDΔK. Conclusions ISAD1 and related peptides may be suitable for therapy development of AD by promoting off-pathway assembly of Tau, thus preventing its toxicity. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-00959-z.
Collapse
Affiliation(s)
- Isabelle Aillaud
- Institute of Bioanalysis, Coburg University of Applied Sciences, Coburg, Germany
| | - Senthilvelrajan Kaniyappan
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | | | - Lisa Marie Ramirez
- Forschungsgruppe Translationale Strukturbiologie, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Sewar Alkhashrom
- Institut für Chemie und Pharmazie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jutta Eichler
- Institut für Chemie und Pharmazie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anselm H C Horn
- Bioinformatik, Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Institut für Medizinische Genetik, Universität Zürich, Zürich, Switzerland
| | - Markus Zweckstetter
- Forschungsgruppe Translationale Strukturbiologie, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany.,Abteilung für NMR-basierte Strukturbiologie, Max-Planck-Institut für Biophysikalische Chemie, Göttingen, Germany
| | - Eckhard Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany.,CAESAR Research Center, Bonn, Germany
| | - Heinrich Sticht
- Bioinformatik, Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Susanne Aileen Funke
- Institute of Bioanalysis, Coburg University of Applied Sciences, Coburg, Germany.
| |
Collapse
|
26
|
Raihan SMS, Ahmed M, Sharma A, Hossain MS, Islam RU, Andersson K. A Belief Rule Based Expert System to Diagnose Alzheimer’s Disease Using Whole Blood Gene Expression Data. Brain Inform 2022. [DOI: 10.1007/978-3-031-15037-1_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
27
|
Zhang Y, Chen H, Long X, Xu T. Three-dimensional-engineered bioprinted in vitro human neural stem cell self-assembling culture model constructs of Alzheimer's disease. Bioact Mater 2021; 11:192-205. [PMID: 34938923 PMCID: PMC8665263 DOI: 10.1016/j.bioactmat.2021.09.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/26/2021] [Accepted: 09/16/2021] [Indexed: 12/31/2022] Open
Abstract
The pathogenic cascade of Alzheimer's disease (AD) characterized by amyloid-β protein accumulation is still poorly understood, partially owing to the limitations of relevant models without in vivo neural tissue microenvironment to recapitulate cell-cell interactions. To better mimic neural tissue microenvironment, three-dimensional (3D) core-shell AD model constructs containing human neural progenitor cells (NSCs) with 2% matrigel as core bioink and 2% alginate as shell bioink have been bioprinted by a co-axial bioprinter, with a suitable shell thickness for nutrient exchange and barrier-free cell interaction cores. These constructs exhibit cell self-clustering and -assembling properties and engineered reproducibility with long-term cell viability and self-renewal, and a higher differentiation level compared to 2D and 3D MIX models. The different effects of 3D bioprinted, 2D, and MIX microenvironments on the growth of NSCs are mainly related to biosynthesis of amino acids and glyoxylate and dicarboxylate metabolism on day 2 and ribosome, biosynthesis of amino acids and proteasome on day 14. Particularly, the model constructs demonstrated Aβ aggregation and higher expression of Aβ and tau isoform genes compared to 2D and MIX controls. AD model constructs will provide a promising strategy to facilitate the development of a 3D in vitro AD model for neurodegeneration research.
Collapse
Affiliation(s)
- Yi Zhang
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Haiyan Chen
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Xiaoyan Long
- East China Institute of Digital Medical Engineering, Shangrao, 334000, China
| | - Tao Xu
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| |
Collapse
|
28
|
Lou C, Habes M, Illenberger NA, Ezzati A, Lipton RB, Shaw PA, Stephens-Shields AJ, Akbari H, Doshi J, Davatzikos C, Shinohara RT. Leveraging machine learning predictive biomarkers to augment the statistical power of clinical trials with baseline magnetic resonance imaging. Brain Commun 2021; 3:fcab264. [PMID: 34806001 PMCID: PMC8600962 DOI: 10.1093/braincomms/fcab264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/03/2021] [Accepted: 09/17/2021] [Indexed: 11/12/2022] Open
Abstract
A key factor in designing randomized clinical trials is the sample size required to achieve a particular level of power to detect the benefit of a treatment. Sample size calculations depend upon the expected benefits of a treatment (effect size), the accuracy of measurement of the primary outcome, and the level of power specified by the investigators. In this study, we show that radiomic models, which leverage complex brain MRI patterns and machine learning, can be utilized in clinical trials with protocols that incorporate baseline MR imaging to significantly increase statistical power to detect treatment effects. Akin to the historical control paradigm, we propose to utilize a radiomic prediction model to generate a pseudo-control sample for each individual in the trial of interest. Because the variability of expected outcome across patients can mask our ability to detect treatment effects, we can increase the power to detect a treatment effect in a clinical trial by reducing that variability through using radiomic predictors as surrogates. We illustrate this method with simulations based on data from two cohorts in different neurologic diseases, Alzheimer's disease and glioblastoma multiforme. We present sample size requirements across a range of effect sizes using conventional analysis and models that include a radiomic predictor. For our Alzheimer's disease cohort, at an effect size of 0.35, total sample size requirements for 80% power declined from 246 to 212 for the endpoint cognitive decline. For our glioblastoma multiforme cohort, at an effect size of 1.65 with the endpoint survival time, total sample size requirements declined from 128 to 74. This methodology can decrease the required sample sizes by as much as 50%, depending on the strength of the radiomic predictor. The power of this method grows with increased accuracy of radiomic prediction, and furthermore, this method is most helpful when treatment effect sizes are small. Neuroimaging biomarkers are a powerful and increasingly common suite of tools that are, in many cases, highly predictive of disease outcomes. Here, we explore the possibility of using MRI-based radiomic biomarkers for the purpose of improving statistical power in clinical trials in the contexts of brain cancer and prodromal Alzheimer's disease. These methods can be applied to a broad range of neurologic diseases using a broad range of predictors of outcome to make clinical trials more efficient.
Collapse
Affiliation(s)
- Carolyn Lou
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.,Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Mohamad Habes
- Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Nicholas A Illenberger
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Ali Ezzati
- Department of Neurology, Albert Einstein College of Medicine, New York City, New York, 10461, USA
| | - Richard B Lipton
- Department of Neurology, Albert Einstein College of Medicine, New York City, New York, 10461, USA
| | - Pamela A Shaw
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Alisa J Stephens-Shields
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Hamed Akbari
- Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Jimit Doshi
- Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Russell T Shinohara
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.,Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.,Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| |
Collapse
|
29
|
Prasanna P, Rathee S, Rahul V, Mandal D, Chandra Goud MS, Yadav P, Hawthorne S, Sharma A, Gupta PK, Ojha S, Jha NK, Villa C, Jha SK. Microfluidic Platforms to Unravel Mysteries of Alzheimer's Disease: How Far Have We Come? Life (Basel) 2021; 11:life11101022. [PMID: 34685393 PMCID: PMC8537508 DOI: 10.3390/life11101022] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is a significant health concern with enormous social and economic impact globally. The gradual deterioration of cognitive functions and irreversible neuronal losses are primary features of the disease. Even after decades of research, most therapeutic options are merely symptomatic, and drugs in clinical practice present numerous side effects. Lack of effective diagnostic techniques prevents the early prognosis of disease, resulting in a gradual deterioration in the quality of life. Furthermore, the mechanism of cognitive impairment and AD pathophysiology is poorly understood. Microfluidics exploits different microscale properties of fluids to mimic environments on microfluidic chip-like devices. These miniature multichambered devices can be used to grow cells and 3D tissues in vitro, analyze cell-to-cell communication, decipher the roles of neural cells such as microglia, and gain insights into AD pathophysiology. This review focuses on the applications and impact of microfluidics on AD research. We discuss the technical challenges and possible solutions provided by this new cutting-edge technique to understand disease-associated pathways and mechanisms.
Collapse
Affiliation(s)
- Pragya Prasanna
- School of Applied Sciences, KK University, Nalanda 803115, Bihar, India;
- Correspondence: or (P.P.); (S.K.J.)
| | - Shweta Rathee
- Department of Food Science and Technology, National Institute of Food Technology, Entrepreneurship and Management, Sonipat 131028, Haryana, India;
| | - Vedanabhatla Rahul
- Department of Mechanical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India;
| | - Debabrata Mandal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur 844101, Bihar, India;
| | | | - Pardeep Yadav
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida 201310, Uttar Pradesh, India; (P.Y.); (N.K.J.)
| | - Susan Hawthorne
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Cromore Road, Coleraine, Co., Londonderry BT52 1SA, UK;
| | - Ankur Sharma
- Department of Life Sciences, School of Basic Science and Research (SBSR), Sharda University, Greater Noida 201310, Uttar Pradesh, India; (A.S.); (P.K.G.)
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Science and Research (SBSR), Sharda University, Greater Noida 201310, Uttar Pradesh, India; (A.S.); (P.K.G.)
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, P.O. Box 17666, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida 201310, Uttar Pradesh, India; (P.Y.); (N.K.J.)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida 201310, Uttar Pradesh, India; (P.Y.); (N.K.J.)
- Correspondence: or (P.P.); (S.K.J.)
| |
Collapse
|
30
|
Vitali F, Branigan GL, Brinton RD. Preventing Alzheimer's disease within reach by 2025: Targeted-risk-AD-prevention (TRAP) strategy. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12190. [PMID: 34584937 PMCID: PMC8451031 DOI: 10.1002/trc2.12190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/20/2021] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a progressive neurodegenerative disease that currently affects 6.2 million people in the United States and is projected to impact 12.7 million worldwide in 2050 with no effective disease-modifying therapeutic or cure. In 2011 as part of the National Alzheimer's Project Act, the National Plan to Address Alzheimer's Disease was signed into law which proposed to effectively prevent AD by 2025, which is rapidly approaching. The preclinical phase of AD can begin 20 years prior to diagnosis, which provides an extended window for preventive measures that would exert a transformative impact on incidence and prevalence of AD. METHODS A novel combination of text-mining and natural language processing strategies to identify (1) AD risk factors, (2) therapeutics that can target risk factor pathways, and (3) studies supporting therapeutics in the PubMed database was conducted. To classify the literature relevant to AD preventive strategies, a relevance score (RS) based on STRING (search tool for the retrieval of interacting genes/proteins) score for protein-protein interactions and a confidence score (CS) on Bayesian inference were developed. To address mechanism of action, network analysis of protein targets for effective drugs was conducted. Collectively, the analytic approach, referred to as a targeted-risk-AD-prevention (TRAP) strategy, led to a ranked list of candidate therapeutics to reduce AD risk. RESULTS Based on TRAP mining of 9625 publications, 364 AD risk factors were identified. Based on risk factor indications, 629 Food and Drug Administration-approved drugs were identified. Computation of ranking scores enabled identification of 46 relevant high confidence (RS & CS > 0.7) drugs associated with reduced AD risk. Within these candidate therapeutics, 16 had more than one clinical study supporting AD risk reduction. Top-ranked therapeutics with high confidence emerged within lipid-lowering, anti-inflammatory, hormone, and metabolic-related drug classes. DISCUSSION Outcomes of our novel bioinformatic strategy support therapeutic targeting of biological mechanisms and pathways underlying relevant AD risk factors with high confidence. Early interventions that target pathways associated with increased risk of AD have the potential to support the goal of effectively preventing AD by 2025.
Collapse
Affiliation(s)
- Francesca Vitali
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of Neurology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Center for Biomedical Informatics and Biostatistics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Gregory L. Branigan
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- MD‐PhD training program, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of Neurology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
31
|
Yong SJ, Yong MH, Teoh SL, Soga T, Parhar I, Chew J, Lim WL. The Hippocampal Vulnerability to Herpes Simplex Virus Type I Infection: Relevance to Alzheimer's Disease and Memory Impairment. Front Cell Neurosci 2021; 15:695738. [PMID: 34483839 PMCID: PMC8414573 DOI: 10.3389/fncel.2021.695738] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) as a possible infectious etiology in Alzheimer’s disease (AD) has been proposed since the 1980s. The accumulating research thus far continues to support the association and a possible causal role of HSV-1 in the development of AD. HSV-1 has been shown to induce neuropathological and behavioral changes of AD, such as amyloid-beta accumulation, tau hyperphosphorylation, as well as memory and learning impairments in experimental settings. However, a neuroanatomical standpoint of HSV-1 tropism in the brain has not been emphasized in detail. In this review, we propose that the hippocampal vulnerability to HSV-1 infection plays a part in the development of AD and amnestic mild cognitive impairment (aMCI). Henceforth, this review draws on human studies to bridge HSV-1 to hippocampal-related brain disorders, namely AD and aMCI/MCI. Next, experimental models and clinical observations supporting the neurotropism or predilection of HSV-1 to infect the hippocampus are examined. Following this, factors and mechanisms predisposing the hippocampus to HSV-1 infection are discussed. In brief, the hippocampus has high levels of viral cellular receptors, neural stem or progenitor cells (NSCs/NPCs), glucocorticoid receptors (GRs) and amyloid precursor protein (APP) that support HSV-1 infectivity, as well as inadequate antiviral immunity against HSV-1. Currently, the established diseases HSV-1 causes are mucocutaneous lesions and encephalitis; however, this review revises that HSV-1 may also induce and/or contribute to hippocampal-related brain disorders, especially AD and aMCI/MCI.
Collapse
Affiliation(s)
- Shin Jie Yong
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Min Hooi Yong
- Department of Psychology, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia.,Aging Health and Well-being Research Centre, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Tomoko Soga
- Jeffrey Cheah School of Medicine and Health Sciences, Brain Research Institute Monash Sunway, Monash University Malaysia, Subang Jaya, Malaysia
| | - Ishwar Parhar
- Jeffrey Cheah School of Medicine and Health Sciences, Brain Research Institute Monash Sunway, Monash University Malaysia, Subang Jaya, Malaysia
| | - Jactty Chew
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia.,Aging Health and Well-being Research Centre, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| |
Collapse
|
32
|
Rybak-Wolf A, Plass M. RNA Dynamics in Alzheimer's Disease. Molecules 2021; 26:5113. [PMID: 34500547 PMCID: PMC8433936 DOI: 10.3390/molecules26175113] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/09/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder that heavily burdens healthcare systems worldwide. There is a significant requirement to understand the still unknown molecular mechanisms underlying AD. Current evidence shows that two of the major features of AD are transcriptome dysregulation and altered function of RNA binding proteins (RBPs), both of which lead to changes in the expression of different RNA species, including microRNAs (miRNAs), circular RNAs (circRNAs), long non-coding RNAs (lncRNAs), and messenger RNAs (mRNAs). In this review, we will conduct a comprehensive overview of how RNA dynamics are altered in AD and how this leads to the differential expression of both short and long RNA species. We will describe how RBP expression and function are altered in AD and how this impacts the expression of different RNA species. Furthermore, we will also show how changes in the abundance of specific RNA species are linked to the pathology of AD.
Collapse
Affiliation(s)
- Agnieszka Rybak-Wolf
- Max Delbrück Center for Molecular Medicine (MDC), Berlin Institute for Medical Systems Biology (BIMSB), 10115 Berlin, Germany
| | - Mireya Plass
- Gene Regulation of Cell Identity, Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, 08908 Barcelona, Spain
- Program for Advancing Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L'Hospitalet del Llobregat, 08908 Barcelona, Spain
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
33
|
Rastegari A, Safavi M, Vafadarnejad F, Najafi Z, Hariri R, Bukhari SNA, Iraji A, Edraki N, Firuzi O, Saeedi M, Mahdavi M, Akbarzadeh T. Synthesis and evaluation of novel arylisoxazoles linked to tacrine moiety: in vitro and in vivo biological activities against Alzheimer's disease. Mol Divers 2021; 26:409-428. [PMID: 34273065 DOI: 10.1007/s11030-021-10248-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/05/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is now ranked as the third leading cause of death after heart disease and cancer. There is no definite cure for AD due to the multi-factorial nature of the disease, hence, multi-target-directed ligands (MTDLs) have attracted lots of attention. In this work, focusing on the efficient cholinesterase inhibitory activity of tacrine, design and synthesis of novel arylisoxazole-tacrine analogues was developed. In vitro acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibition assay confirmed high potency of the title compounds. Among them, compounds 7l and 7b demonstrated high activity toward AChE and BChE with IC50 values of 0.050 and 0.039 μM, respectively. Both compounds showed very good self-induced Aβ aggregation and AChE-induced inhibitory activity (79.4 and 71.4% for compound 7l and 61.8 and 58.6% for compound 7b, respectively). Also, 7l showed good anti-BACE1 activity with IC50 value of 1.65 µM. The metal chelation test indicated the ability of compounds 7l and 7b to chelate biometals (Zn2+, Cu2+, and Fe2+). However, they showed no significant neuroprotectivity against Aβ-induced damage in PC12 cells. Evaluation of in vitro hepatotoxicity revealed comparable toxicity of compounds 7l and 7b with tacrine. In vivo studies by Morris water maze (MWM) task demonstrated that compound 7l significantly reversed scopolamine-induced memory deficit in rats. Finally, molecular docking studies of compounds 7l and 7b confirmed establishment of desired interactions with the AChE, BChE, and BACE1 active sites.
Collapse
Affiliation(s)
- Arezoo Rastegari
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Fahimeh Vafadarnejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Najafi
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roshanak Hariri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Aljouf, 2014, Sakaka, Saudi Arabia
| | - Aida Iraji
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Central Research laboratory, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Saeedi
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Akbarzadeh
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Understanding Amyloid Structures and Disease: A Continuing Challenge in Health Research. Int J Mol Sci 2021; 22:ijms22126620. [PMID: 34205606 PMCID: PMC8234419 DOI: 10.3390/ijms22126620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/17/2022] Open
Abstract
Neurodegenerative disorders (NDDs), including Alzheimer's, Parkinson's, and Huntington's diseases, are a highly prevalent class of disorders that share the presence of aberrant aggregates called amyloids in the nervous system [...].
Collapse
|
35
|
Hakim AM. A Proposed Hypothesis on Dementia: Inflammation, Small Vessel Disease, and Hypoperfusion Is the Sequence That Links All Harmful Lifestyles to Cognitive Impairment. Front Aging Neurosci 2021; 13:679837. [PMID: 33994998 PMCID: PMC8116506 DOI: 10.3389/fnagi.2021.679837] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022] Open
Abstract
There is growing consensus that certain lifestyles can contribute to cognitive impairment and dementia, but the physiological steps that link a harmful lifestyle to its negative impact are not always evident. It is also unclear whether all lifestyles that contribute to dementia do so through the same intermediary steps. This article will focus on three lifestyles known to be risk factors for dementia, namely obesity, sedentary behavior, and insufficient sleep, and offer a unifying hypothesis proposing that lifestyles that negatively impact cognition do so through the same sequence of events: inflammation, small vessel disease, decline in cerebral perfusion, and brain atrophy. The hypothesis will then be tested in a recently identified risk factor for dementia, namely hearing deficit. If further studies confirm this sequence of events leading to dementia, a significant change in our approach to this debilitating and costly condition may be necessary, possible, and beneficial.
Collapse
Affiliation(s)
- Antoine M. Hakim
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
- Division of Neurology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
36
|
Palmer D, Dumont JR, Dexter TD, Prado MAM, Finger E, Bussey TJ, Saksida LM. Touchscreen cognitive testing: Cross-species translation and co-clinical trials in neurodegenerative and neuropsychiatric disease. Neurobiol Learn Mem 2021; 182:107443. [PMID: 33895351 DOI: 10.1016/j.nlm.2021.107443] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 02/06/2021] [Accepted: 02/26/2021] [Indexed: 01/06/2023]
Abstract
Translating results from pre-clinical animal studies to successful human clinical trials in neurodegenerative and neuropsychiatric disease presents a significant challenge. While this issue is clearly multifaceted, the lack of reproducibility and poor translational validity of many paradigms used to assess cognition in animal models are central contributors to this challenge. Computer-automated cognitive test batteries have the potential to substantially improve translation between pre-clinical studies and clinical trials by increasing both reproducibility and translational validity. Given the structured nature of data output, computer-automated tests also lend themselves to increased data sharing and other open science good practices. Over the past two decades, computer automated, touchscreen-based cognitive testing methods have been developed for non-human primate and rodent models. These automated methods lend themselves to increased standardization, hence reproducibility, and have become increasingly important for the elucidation of the neurobiological basis of cognition in animal models. More recently, there have been increased efforts to use these methods to enhance translational validity by developing task batteries that are nearly identical across different species via forward (i.e., translating animal tasks to humans) and reverse (i.e., translating human tasks to animals) translation. An additional benefit of the touchscreen approach is that a cross-species cognitive test battery makes it possible to implement co-clinical trials-an approach developed initially in cancer research-for novel treatments for neurodegenerative disorders. Co-clinical trials bring together pre-clinical and early clinical studies, which facilitates testing of novel treatments in mouse models with underlying genetic or other changes, and can help to stratify patients on the basis of genetic, molecular, or cognitive criteria. This approach can help to determine which patients should be enrolled in specific clinical trials and can facilitate repositioning and/or repurposing of previously approved drugs. This has the potential to mitigate the resources required to study treatment responses in large numbers of human patients.
Collapse
Affiliation(s)
- Daniel Palmer
- Robarts Research Institute, The University of Western Ontario, Ontario, Canada; Department of Physiology and Pharmacology, The University of Western Ontario, Ontario, Canada.
| | - Julie R Dumont
- Robarts Research Institute, The University of Western Ontario, Ontario, Canada; BrainsCAN, The University of Western Ontario, Ontario, Canada
| | - Tyler D Dexter
- Department of Physiology and Pharmacology, The University of Western Ontario, Ontario, Canada; Graduate Program in Neuroscience, The University of Western Ontario, Ontario, Canada
| | - Marco A M Prado
- Robarts Research Institute, The University of Western Ontario, Ontario, Canada; Department of Physiology and Pharmacology, The University of Western Ontario, Ontario, Canada; Graduate Program in Neuroscience, The University of Western Ontario, Ontario, Canada; Department of Anatomy and Cell Biology, The University of Western Ontario, Ontario, Canada
| | - Elizabeth Finger
- Robarts Research Institute, The University of Western Ontario, Ontario, Canada; Department of Clinical Neurological Sciences, The University of Western Ontario, Ontario, Canada; Lawson Health Research Institute, Ontario, Canada; Parkwood Institute, St. Josephs Health Care, Ontario, Canada
| | - Timothy J Bussey
- Robarts Research Institute, The University of Western Ontario, Ontario, Canada; Department of Physiology and Pharmacology, The University of Western Ontario, Ontario, Canada; Brain and Mind Institute, The University of Western Ontario, Ontario, Canada
| | - Lisa M Saksida
- Robarts Research Institute, The University of Western Ontario, Ontario, Canada; Department of Physiology and Pharmacology, The University of Western Ontario, Ontario, Canada; Brain and Mind Institute, The University of Western Ontario, Ontario, Canada
| |
Collapse
|
37
|
Pasieka A, Panek D, Jończyk J, Godyń J, Szałaj N, Latacz G, Tabor J, Mezeiova E, Chantegreil F, Dias J, Knez D, Lu J, Pi R, Korabecny J, Brazzolotto X, Gobec S, Höfner G, Wanner K, Więckowska A, Malawska B. Discovery of multifunctional anti-Alzheimer's agents with a unique mechanism of action including inhibition of the enzyme butyrylcholinesterase and γ-aminobutyric acid transporters. Eur J Med Chem 2021; 218:113397. [PMID: 33838585 DOI: 10.1016/j.ejmech.2021.113397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/31/2022]
Abstract
Looking for an effective anti-Alzheimer's agent is very challenging; however, a multifunctional ligand strategy may be a promising solution for the treatment of this complex disease. We herein present the design, synthesis and biological evaluation of novel hydroxyethylamine derivatives displaying unique, multiple properties that have not been previously reported. The original mechanism of action combines inhibitory activity against disease-modifying targets: β-secretase enzyme (BACE1) and amyloid β (Aβ) aggregation, along with an effect on targets associated with symptom relief - inhibition of butyrylcholinesterase (BuChE) and γ-aminobutyric acid transporters (GATs). Among the obtained molecules, compound 36 exhibited the most balanced and broad activity profile (eeAChE IC50 = 2.86 μM; eqBuChE IC50 = 60 nM; hBuChE IC50 = 20 nM; hBACE1 IC50 = 5.9 μM; inhibition of Aβ aggregation = 57.9% at 10 μM; mGAT1 IC50 = 10.96 μM; and mGAT2 IC50 = 19.05 μM). Moreover, we also identified 31 as the most potent mGAT4 and hGAT3 inhibitor (IC50 = 5.01 μM and IC50 = 2.95 μM, respectively), with high selectivity over other subtypes. Compounds 36 and 31 represent new anti-Alzheimer agents that can ameliorate cognitive decline and modify the progress of disease.
Collapse
Affiliation(s)
- Anna Pasieka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Jakub Jończyk
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Justyna Godyń
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Natalia Szałaj
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Julia Tabor
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Eva Mezeiova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Fabien Chantegreil
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - José Dias
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Damijan Knez
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Junfeng Lu
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Rongbiao Pi
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jan Korabecny
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Xavier Brazzolotto
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Stanislav Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Georg Höfner
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377, Munich, Germany
| | - Klaus Wanner
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377, Munich, Germany
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| |
Collapse
|
38
|
Too LK, Hunt N, Simunovic MP. The Role of Inflammation and Infection in Age-Related Neurodegenerative Diseases: Lessons From Bacterial Meningitis Applied to Alzheimer Disease and Age-Related Macular Degeneration. Front Cell Neurosci 2021; 15:635486. [PMID: 33867940 PMCID: PMC8044768 DOI: 10.3389/fncel.2021.635486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/05/2021] [Indexed: 12/21/2022] Open
Abstract
Age-related neurodegenerative diseases, such as Alzheimer disease (AD) and age-related macular degeneration (AMD), are multifactorial and have diverse genetic and environmental risk factors. Despite the complex nature of the diseases, there is long-standing, and growing, evidence linking microbial infection to the development of AD dementia, which we summarize in this article. Also, we highlight emerging research findings that support a role for parainfection in the pathophysiology of AMD, a disease of the neurosensory retina that has been shown to share risk factors and pathological features with AD. Acute neurological infections, such as Bacterial Meningitis (BM), trigger inflammatory events that permanently change how the brain functions, leading to lasting cognitive impairment. Neuroinflammation likewise is a known pathological event that occurs in the early stages of chronic age-related neurodegenerative diseases AD and AMD and might be triggered as a parainfectious event. To date, at least 16 microbial pathogens have been linked to the development of AD; on the other hand, investigation of a microbe-AMD relationship is in its infancy. This mini-review article provides a synthesis of existing evidence indicating a contribution of parainfection in the aetiology of AD and of emerging findings that support a similar process in AMD. Subsequently, it describes the major immunopathological mechanisms that are common to BM and AD/AMD. Together, this evidence leads to our proposal that both AD and AMD may have an infectious aetiology that operates through a dysregulated inflammatory response, leading to deleterious outcomes. Last, it draws fresh insights from the existing literature about potential therapeutic options for BM that might alleviate neurological disruption associated with infections, and which could, by extension, be explored in the context of AD and AMD.
Collapse
Affiliation(s)
- Lay Khoon Too
- Faculty of Medicine and Health, Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pathology, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas Hunt
- Faculty of Medicine and Health, Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Matthew P. Simunovic
- Faculty of Medicine and Health, Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
- Sydney Eye Hospital, Sydney, NSW, Australia
| |
Collapse
|
39
|
Ernest James Phillips T, Maguire E. Phosphoinositides: Roles in the Development of Microglial-Mediated Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2021; 15:652593. [PMID: 33841102 PMCID: PMC8032904 DOI: 10.3389/fncel.2021.652593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are increasingly recognized as vital players in the pathology of a variety of neurodegenerative conditions including Alzheimer’s (AD) and Parkinson’s (PD) disease. While microglia have a protective role in the brain, their dysfunction can lead to neuroinflammation and contributes to disease progression. Also, a growing body of literature highlights the seven phosphoinositides, or PIPs, as key players in the regulation of microglial-mediated neuroinflammation. These small signaling lipids are phosphorylated derivates of phosphatidylinositol, are enriched in the brain, and have well-established roles in both homeostasis and disease.Disrupted PIP levels and signaling has been detected in a variety of dementias. Moreover, many known AD disease modifiers identified via genetic studies are expressed in microglia and are involved in phospholipid metabolism. One of these, the enzyme PLCγ2 that hydrolyzes the PIP species PI(4,5)P2, displays altered expression in AD and PD and is currently being investigated as a potential therapeutic target.Perhaps unsurprisingly, neurodegenerative conditions exhibiting PIP dyshomeostasis also tend to show alterations in aspects of microglial function regulated by these lipids. In particular, phosphoinositides regulate the activities of proteins and enzymes required for endocytosis, toll-like receptor signaling, purinergic signaling, chemotaxis, and migration, all of which are affected in a variety of neurodegenerative conditions. These functions are crucial to allow microglia to adequately survey the brain and respond appropriately to invading pathogens and other abnormalities, including misfolded proteins. AD and PD therapies are being developed to target many of the above pathways, and although not yet investigated, simultaneous PIP manipulation might enhance the beneficial effects observed. Currently, only limited therapeutics are available for dementia, and although these show some benefits for symptom severity and progression, they are far from curative. Given the importance of microglia and PIPs in dementia development, this review summarizes current research and asks whether we can exploit this information to design more targeted, or perhaps combined, dementia therapeutics. More work is needed to fully characterize the pathways discussed in this review, but given the strength of the current literature, insights in this area could be invaluable for the future of neurodegenerative disease research.
Collapse
Affiliation(s)
| | - Emily Maguire
- UK Dementia Research Institute at Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
40
|
Gaurav I, Thakur A, Iyaswamy A, Wang X, Chen X, Yang Z. Factors Affecting Extracellular Vesicles Based Drug Delivery Systems. Molecules 2021; 26:molecules26061544. [PMID: 33799765 PMCID: PMC7999478 DOI: 10.3390/molecules26061544] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) play major roles in intracellular communication and participate in several biological functions in both normal and pathological conditions. Surface modification of EVs via various ligands, such as proteins, peptides, or aptamers, offers great potential as a means to achieve targeted delivery of therapeutic cargo, i.e., in drug delivery systems (DDS). This review summarizes recent studies pertaining to the development of EV-based DDS and its advantages compared to conventional nano drug delivery systems (NDDS). First, we compare liposomes and exosomes in terms of their distinct benefits in DDS. Second, we analyze what to consider for achieving better isolation, yield, and characterization of EVs for DDS. Third, we summarize different methods for the modification of surface of EVs, followed by discussion about different origins of EVs and their role in developing DDS. Next, several major methods for encapsulating therapeutic cargos in EVs have been summarized. Finally, we discuss key challenges and pose important open questions which warrant further investigation to develop more effective EV-based DDS.
Collapse
Affiliation(s)
- Isha Gaurav
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (A.I.); (X.W.); (X.C.)
| | - Abhimanyu Thakur
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation-CAS Limited, Hong Kong, China;
| | - Ashok Iyaswamy
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (A.I.); (X.W.); (X.C.)
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xuehan Wang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (A.I.); (X.W.); (X.C.)
| | - Xiaoyu Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (A.I.); (X.W.); (X.C.)
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (A.I.); (X.W.); (X.C.)
- Changshu Research Institute, Hong Kong Baptist University, Changshu Economic and Technological Development (CETD) Zone, Changshu 215500, Jiangsu Province, China
- Correspondence: ; Tel.: +852-3411-2961
| |
Collapse
|
41
|
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and accounts for approximately 50% to 80% of all cases of dementia. The diagnosis of probable AD is based on clinical criteria and overlapping clinical features pose a challenge to accurate diagnosis. However, neuroimaging has been included as a biomarker in various published criteria for the diagnosis of probable AD, in the absence of a confirmatory diagnostic test during life. Advances in neuroimaging techniques and their inclusion in diagnostic and research criteria for the diagnosis of AD includes the use of positron emission tomography (PET) imaging as a biomarker in various therapeutic and prognostic studies in AD. The development and application of a range of PET tracers will allow more detailed assessment of people with AD and will improve diagnostic specificity and targeted therapy of AD. The aim of this review is to summarize current evidence on PET imaging using the non-specific tracer [18F]fluorodeoxyglucose and specific tracers that target amyloid and tau pathology in people with AD.
Collapse
Affiliation(s)
- Shailendra Mohan Tripathi
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Alison D Murray
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| |
Collapse
|
42
|
MacDougall G, Brown LY, Kantor B, Chiba-Falek O. The Path to Progress Preclinical Studies of Age-Related Neurodegenerative Diseases: A Perspective on Rodent and hiPSC-Derived Models. Mol Ther 2021; 29:949-972. [PMID: 33429080 PMCID: PMC7934639 DOI: 10.1016/j.ymthe.2021.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/03/2020] [Accepted: 01/01/2021] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the two most prevalent age-related neurodegenerative diseases, and currently no effective clinical treatments exist for either, despite decades of clinical trials. The failure to translate preclinical findings into effective treatments is indicative of a problem in the current evaluation pipeline for potential therapeutics. At present, there are no useful animal models for AD and PD research that reflect the entire biology of the diseases, specifically, the more common non-Mendelian forms. Whereas the field continues to seek suitable rodent models for investigating potential therapeutics for these diseases, rodent models have still been used primarily for preclinical studies. Here, we advocate for a paradigm shift toward the application of human-induced pluripotent stem cell (hiPSC)-derived systems for PD and AD modeling and the development of improved human-based models in a dish for drug discovery and preclinical assessment of therapeutic targets.
Collapse
Affiliation(s)
- Gabriella MacDougall
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Logan Y Brown
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University Medical Center, Durham, NC 27710, USA; Viral Vector Core, Duke University Medical Center, Durham, NC 27710, USA
| | - Boris Kantor
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University Medical Center, Durham, NC 27710, USA; Viral Vector Core, Duke University Medical Center, Durham, NC 27710, USA.
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
43
|
Gklinos P, Papadopoulou M, Stanulovic V, Mitsikostas DD, Papadopoulos D. Monoclonal Antibodies as Neurological Therapeutics. Pharmaceuticals (Basel) 2021; 14:ph14020092. [PMID: 33530460 PMCID: PMC7912592 DOI: 10.3390/ph14020092] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 02/08/2023] Open
Abstract
Over the last 30 years the role of monoclonal antibodies in therapeutics has increased enormously, revolutionizing treatment in most medical specialties, including neurology. Monoclonal antibodies are key therapeutic agents for several neurological conditions with diverse pathophysiological mechanisms, including multiple sclerosis, migraines and neuromuscular disease. In addition, a great number of monoclonal antibodies against several targets are being investigated for many more neurological diseases, which reflects our advances in understanding the pathogenesis of these diseases. Untangling the molecular mechanisms of disease allows monoclonal antibodies to block disease pathways accurately and efficiently with exceptional target specificity, minimizing non-specific effects. On the other hand, accumulating experience shows that monoclonal antibodies may carry class-specific and target-associated risks. This article provides an overview of different types of monoclonal antibodies and their characteristics and reviews monoclonal antibodies currently in use or under development for neurological disease.
Collapse
Affiliation(s)
- Panagiotis Gklinos
- Department of Neurology, KAT General Hospital of Attica, 14561 Athens, Greece;
| | - Miranta Papadopoulou
- Center for Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Vid Stanulovic
- Global Pharmacovigilance, R&D Sanofi, 91385 Chilly-Mazarin, France;
| | - Dimos D. Mitsikostas
- 1st Neurology Department, Aeginition Hospital, National and Kapodistrian University of Athens, 11521 Athens, Greece;
| | - Dimitrios Papadopoulos
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, 129 Vasilissis Sophias Avenue, 11521 Athens, Greece
- Salpetriere Neuropsychiatric Clinic, 149 Papandreou Street, Metamorphosi, 14452 Athens, Greece
- Correspondence:
| |
Collapse
|
44
|
Camarillo-López RH, Hernández Rodríguez M, Torres-Ramos MA, Arciniega-Martínez IM, García-Marín ID, Correa Basurto J, Méndez Méndez JV, Rosales-Hernández MC. Tert-butyl-(4-hydroxy-3-((3-(2-methylpiperidin-yl)propyl)carbamoyl)phenyl)carbamate Has Moderated Protective Activity in Astrocytes Stimulated with Amyloid Beta 1-42 and in a Scopolamine Model. Molecules 2020; 25:molecules25215009. [PMID: 33137907 PMCID: PMC7672627 DOI: 10.3390/molecules25215009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with no cure nowadays; there is no treatment either to prevent or to stop its progression. In vitro studies suggested that tert-butyl-(4-hydroxy-3-((3-(2-methylpiperidin-yl)propyl)carbamoyl)phenyl) carbamate named the M4 compound can act as both β-secretase and an acetylcholinesterase inhibitor, preventing the amyloid beta peptide (Aβ) aggregation and the formation of fibrils (fAβ) from Aβ1-42. This work first aimed to assess in in vitro studies to see whether the death of astrocyte cells promoted by Aβ1-42 could be prevented. Second, our work investigated the ability of the M4 compound to inhibit amyloidogenesis using an in vivo model after scopolamine administration. The results showed that M4 possesses a moderate protective effect in astrocytes against Aβ1-42 due to a reduction in the TNF-α and free radicals observed in cell cultures. In the in vivo studies, however, no significant effect of M4 was observed in comparison with a galantamine model employed in rats, in which case this outcome was attributed to the bioavailability of M4 in the brain of the rats.
Collapse
Affiliation(s)
- Raúl Horacio Camarillo-López
- Laboratorio de Biofísica y biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, 11340 Ciudad de México, Mexico; (R.H.C.-L.); (M.H.R.); (I.D.G.-M.)
| | - Maricarmen Hernández Rodríguez
- Laboratorio de Biofísica y biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, 11340 Ciudad de México, Mexico; (R.H.C.-L.); (M.H.R.); (I.D.G.-M.)
| | - Mónica Adriana Torres-Ramos
- Unidad Periférica de Neurociencias, Facultad de Medicina UNAM-Instituto Nacional de Neurología y Neurocirugía, MVS-SSA, Insurgentes sur 3877, La Fama, Tlalpan, 14269 Ciudad de México, Mexico;
| | - Ivonne Maciel Arciniega-Martínez
- Laboratorio de Inmunidad de Mucosas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 Ciudad de México, Mexico;
| | - Iohanan Daniel García-Marín
- Laboratorio de Biofísica y biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, 11340 Ciudad de México, Mexico; (R.H.C.-L.); (M.H.R.); (I.D.G.-M.)
| | - José Correa Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340 Ciudad de México, Mexico;
| | - Juan Vicente Méndez Méndez
- Centro de Nanociencias y Micro y Nanotecnologías, Instituto Politécnico Nacional. Av. Luis Enrique Erro s/n, Nueva Industrial Vallejo, Gustavo A. Madero, 07738 Ciudad de México, Mexico;
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, 11340 Ciudad de México, Mexico; (R.H.C.-L.); (M.H.R.); (I.D.G.-M.)
- Correspondence:
| |
Collapse
|
45
|
Bae M, Yi HG, Jang J, Cho DW. Microphysiological Systems for Neurodegenerative Diseases in Central Nervous System. MICROMACHINES 2020; 11:E855. [PMID: 32947879 PMCID: PMC7570039 DOI: 10.3390/mi11090855] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022]
Abstract
Neurodegenerative diseases are among the most severe problems in aging societies. Various conventional experimental models, including 2D and animal models, have been used to investigate the pathogenesis of (and therapeutic mechanisms for) neurodegenerative diseases. However, the physiological gap between humans and the current models remains a hurdle to determining the complexity of an irreversible dysfunction in a neurodegenerative disease. Therefore, preclinical research requires advanced experimental models, i.e., those more physiologically relevant to the native nervous system, to bridge the gap between preclinical stages and patients. The neural microphysiological system (neural MPS) has emerged as an approach to summarizing the anatomical, biochemical, and pathological physiology of the nervous system for investigation of neurodegenerative diseases. This review introduces the components (such as cells and materials) and fabrication methods for designing a neural MPS. Moreover, the review discusses future perspectives for improving the physiological relevance to native neural systems.
Collapse
Affiliation(s)
- Mihyeon Bae
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
| | - Hee-Gyeong Yi
- Department of Rural and Biosystems Engineering, College of Agricultural Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| |
Collapse
|
46
|
Ahmed T, Zulfiqar A, Arguelles S, Rasekhian M, Nabavi SF, Silva AS, Nabavi SM. Map kinase signaling as therapeutic target for neurodegeneration. Pharmacol Res 2020; 160:105090. [PMID: 32707231 DOI: 10.1016/j.phrs.2020.105090] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/14/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022]
Abstract
Aging is known to be one of the major risk factors in many neurodegenerative diseases (ND) whose prevalence is estimated to rise in the coming years due to the increase in life expectancy. Examples of neurodegenerative diseases include Huntington's, Parkinson's, and Alzheimer's diseases, along with Amyotrophic Lateral Sclerosis, Spinocerebellar ataxias and Frontotemporal Dementia. Given that so far these ND do not have effective pharmacological therapies, a better understanding of the molecular and cellular mechanisms can contribute to development of effective treatments. During the previous decade, the data indicated that dysregulation of MAP kinases [which included c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase 1 and 2 (ERK1/2), and p38] are associated with several stages of the inflammatory process which in turn contributes to age-related neurodegenerative diseases. This evidence suggests that control of inflammation through regulation of MAP kinase could be a worthwhile approach against neurodegenerative diseases. In this review we summarize the pathways of MAP kinase signal transduction and different pharmacological inhibitors that can be used in its modulation against ND.
Collapse
Affiliation(s)
- Touqeer Ahmed
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan.
| | - Abida Zulfiqar
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Sandro Arguelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| | - Mahsa Rasekhian
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran; Division of Translational Medicine, Baqiyatallah Hospital, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ana Sanches Silva
- National Institute for Agricultural and Veterinary Research (INIAV), Vila Do Conde, Portugal; Center for Study in Animal Science (CECA), ICETA, University of Oporto, Oporto, Portugal
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran; Division of Translational Medicine, Baqiyatallah Hospital, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|