1
|
Dubin J, Vandenberghe R, Poesen K. Interval-specific likelihood ratios and probability-based models for interpreting combined CSF biomarkers for Alzheimer's disease. Clin Chim Acta 2025; 564:119941. [PMID: 39181294 DOI: 10.1016/j.cca.2024.119941] [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: 02/05/2024] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND In Alzheimer's disease (AD) diagnosis, a cerebrospinal fluid (CSF) biomarker panel is commonly interpreted with binary cutoff values. However, these values are not generic and do not reflect the disease continuum. We explored the use of interval-specific likelihood ratios (LRs) and probability-based models for AD using a CSF biomarker panel. METHODS CSF biomarker (Aβ1-42, tTau and pTau181) data for both a clinical discovery cohort of 241 patients (measured with INNOTEST) and a clinical validation cohort of 129 patients (measured with EUROIMMUN), both including AD and non-AD dementia/cognitive complaints were retrospectively retrieved in a single-center study. Interval-specific LRs for AD were calculated and validated for univariate and combined (Aβ1-42/tTau and pTau181) biomarkers, and a continuous bivariate probability-based model for AD, plotting Aβ1-42/tTau versus pTau181 was constructed and validated. RESULTS LR for AD increased as individual CSF biomarker values deviated from normal. Interval-specific LRs of a combined biomarker model showed that once one biomarker became abnormal, LRs increased even further when another biomarker largely deviated from normal, as replicated in the validation cohort. A bivariate probability-based model predicted AD with a validated accuracy of 88% on a continuous scale. CONCLUSIONS Interval-specific LRs in a combined biomarker model and prediction of AD using a continuous bivariate biomarker probability-based model, offer a more meaningful interpretation of CSF AD biomarkers on a (semi-)continuous scale with respect to the post-test probability of AD across different assays and cohorts.
Collapse
Affiliation(s)
- Jonas Dubin
- Laboratory for Molecular Neurobiomarker Research, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Laboratory Medicine, UZ Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Alzheimer Research Centre, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium; Neurology Department, UZ Leuven, Leuven, Belgium
| | - Koen Poesen
- Laboratory for Molecular Neurobiomarker Research, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Laboratory Medicine, UZ Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Chatterjee D, Bhattacharya S, Kumari L, Datta A. Aptamers: ushering in new hopes in targeted glioblastoma therapy. J Drug Target 2024; 32:1005-1028. [PMID: 38923419 DOI: 10.1080/1061186x.2024.2373306] [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: 04/16/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024]
Abstract
Glioblastoma, a formidable brain cancer, has remained a therapeutic challenge due to its aggressive nature and resistance to conventional treatments. Recent data indicate that aptamers, short synthetic DNA or RNA molecules can be used in anti-cancer therapy due to their better tumour penetration, specific binding affinity, longer retention in tumour sites and their ability to cross the blood-brain barrier. With the ability to modify these oligonucleotides through the selection process, and using rational design to modify them, post-SELEX aptamers offer several advantages in glioblastoma treatment, including precise targeting of cancer cells while sparing healthy tissue. This review discusses the pivotal role of aptamers in glioblastoma therapy and diagnosis, emphasising their potential to enhance treatment efficacy and also highlights recent advancements in aptamer-based therapies which can transform the landscape of glioblastoma treatment, offering renewed hope to patients and clinicians alike.
Collapse
Affiliation(s)
- Debarpan Chatterjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Srijan Bhattacharya
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Leena Kumari
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Aparna Datta
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| |
Collapse
|
3
|
Kuznetsov AV. The growth rate of senile plaques is determined by the competition between the rate of deposition of free Aβ aggregates into plaques and the autocatalytic production of free Aβ aggregates. J Theor Biol 2024; 593:111900. [PMID: 38992461 DOI: 10.1016/j.jtbi.2024.111900] [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: 04/07/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
The formation of amyloid beta (Aβ) deposits (senile plaques) is one of the hallmarks of Alzheimer's disease (AD). This study investigates what processes are primarily responsible for their formation. A model is developed to simulate the diffusion of amyloid beta (Aβ) monomers, the production of free Aβ aggregates through nucleation and autocatalytic processes, and the deposition of these aggregates into senile plaques. The model suggests that efficient degradation of Aβ monomers alone may suffice to prevent the growth of senile plaques, even without degrading Aβ aggregates and existing plaques. This is because the degradation of Aβ monomers interrupts the supply of reactants needed for plaque formation. The impact of Aβ monomer diffusivity is demonstrated to be small, enabling the application of the lumped capacitance approximation and the derivation of approximate analytical solutions for limiting cases with both small and large rates of Aβ aggregate deposition into plaques. It is found that the rate of plaque growth is governed by two competing processes. One is the deposition rate of free Aβ aggregates into senile plaques. If this rate is small, the plaque grows slowly. However, if the rate of deposition of Aβ aggregates into senile plaques is very large, the free Aβ aggregates are removed from the intracellular fluid by deposition into the plaques, leaving insufficient free Aβ aggregates to catalyze the production of new aggregates. This suggests that under certain conditions, Aβ plaques may offer neuroprotection and impede their own growth. Additionally, it indicates that there exists an optimal rate of deposition of free Aβ aggregates into the plaques, at which the plaques attain their maximum size.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA.
| |
Collapse
|
4
|
Bhadane P, Roul K, Belemkar S, Kumar D. Immunotherapeutic approaches for Alzheimer's disease: Exploring active and passive vaccine progress. Brain Res 2024; 1840:149018. [PMID: 38782231 DOI: 10.1016/j.brainres.2024.149018] [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: 01/12/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegeneration having non-effective treatments. Vaccines or monoclonal antibodies are two typical immunotherapies for AD. Due to Aβ neurotoxicity, most of the treatments target its generation and deposition. However, therapies that specifically target tau protein are also being investigated. UB311 vaccine generates N-terminal anti-Aβ antibodies, that neutralize Aβ toxicity and promote plaque clearance. It is designed to elicit specific B-cell and wide T-cell responses. ACC001 or PF05236806 vaccine has the same Aβ fragment and QS21 as an adjuvant. CAD106 stimulates response against Aβ1-6. However, Nasopharyngitis and injection site erythema are its side effects. AN1792, the first-generation vaccine was formulated in proinflammatory QS21 adjuvant. However, T-cell epitopes are omitted from the developed epitope AD vaccine with Aβ1-42B-cell epitopes. The first-generation vaccine immune response was immensely successful in clearing Aβ, but it was also sufficient to provoke meningoencephalitis. Immunotherapies have been at the forefront of these initiatives in recent years. The review covers the recent updates on active and passive immunotherapy for AD.
Collapse
Affiliation(s)
- Priyanshu Bhadane
- School of Pharmacy & Technology Management, SVKM's NMIMS University, Mukesh Patel Technology Park, Shirpur 425405, India
| | - Krishnashish Roul
- School of Pharmacy & Technology Management, SVKM's NMIMS University, Mukesh Patel Technology Park, Shirpur 425405, India
| | - Sateesh Belemkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed to be University, Vile Parle (W) Mumbai 400 056, India
| | - Devendra Kumar
- School of Pharmacy & Technology Management, SVKM's NMIMS University, Mukesh Patel Technology Park, Shirpur 425405, India.
| |
Collapse
|
5
|
Mahnashi MH, Ayaz M, Ghufran M, Almazni IA, Alqahtani O, Alyami BA, Alqahtani YS, Khan HA, Sadiq A, Waqas M. Phytochemicals-based β-amyloid cleaving enzyme-1 and MAO-B inhibitors for the treatment of Alzheimer's disease: molecular simulations-based predictions. J Biomol Struct Dyn 2024; 42:8359-8371. [PMID: 37815007 DOI: 10.1080/07391102.2023.2265494] [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: 02/27/2023] [Accepted: 08/01/2023] [Indexed: 10/11/2023]
Abstract
Alzheimer's disease (AD) is among the highly prevalent neurodegenerative disorder of the aging brain and is allied with cognitive and behavioral abnormalities. Unfortunately, there is very limited drug discovery for the effective management of AD, and the clinically approved drugs have limited efficacy. Consequently, there is an immediate demand for the development of new compounds that have the ability to act as multitarget-directed ligands (MTDLs). As major pathological targets of the disease, the current study aimed to investigate lead natural bioactive compounds including apigenin, epigallocatechin-3-gallate, berberine, curcumin, genistein, luteolin, quercetin, resveratrol for their inhibitory potentials against β-amyloid cleaving enzyme-1 (BACE1) and monoamine oxidase-B (MAO-B) enzymes. The study compounds were docked against the target enzymes (MAO-B and BACE1) using MOE software and subsequent molecular dynamics simulations (MDS) studies. The molecular docking analysis revealed that these phytochemicals (MTDLs) showed good interactions with the target enzymes as compared to the reference inhibitors. Among these eight phytocompounds, the epigallocatechin-3-gallate compound was an active inhibitor against both drug targets, with the highest docking scores and good interactions with the active residues of the enzymes. Furthermore, the docking result of the active one inhibitor in complex with the target enzymes (epigallocatechin-3-gallate/BACE1, epigallocatechin-3-gallate/MAO-B, reference/BACE1 and reference/MAO-B) were further validated by MDS. According to the findings of our study, epigallocatechin-3-gallate has the potential to be a candidate for use in the treatment of neurological illnesses like AD. This compound has MTDL potential and may be exploited to create new compounds with disease-modifying features.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mater H Mahnashi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Muhammad Ayaz
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara, Pakistan
| | - Mehreen Ghufran
- Department of Pathology, Medical Teaching Institution, Bacha Khan Medical College (BKMC), Mardan, Pakistan
| | - Ibrahim Abdullah Almazni
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran, Kingdom of Saudi Arabia
| | - Omaish Alqahtani
- Department of Pharmacognosy, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Bandar A Alyami
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Yahya S Alqahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Haider Ali Khan
- Department of Biochemistry, Abdul Wali Khan University, Mardan, Pakistan
| | - Abdul Sadiq
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Muhammad Waqas
- Department of Biotechnology and Genetic Engineering, Hazara University Mansehra, Dhodial, Pakistan
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| |
Collapse
|
6
|
Bhatt A, Bhardwaj H, Srivastava P. Mesenchymal stem cell therapy for Alzheimer's disease: A novel therapeutic approach for neurodegenerative diseases. Neuroscience 2024; 555:52-68. [PMID: 39032806 DOI: 10.1016/j.neuroscience.2024.07.019] [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: 04/22/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Alzheimer's disease (AD) is one of the most progressive and prevalent types of neurodegenerative diseases in the aging population (aged >65 years) and is considered a major factor for dementia, affecting 55 million people worldwide. In the current scenario, drug-based therapies have been employed for the treatment of Alzheimer's disease but are only able to provide symptomatic relief to patients rather than a permanent solution from Alzheimer's. Recent advancements in stem cell research unlock new horizons for developing effective and highly potential therapeutic approaches due to their self-renewal, self-replicating, regenerative, and high differentiation capabilities. Stem cells come in multiple lineages such as embryonic, neural, and induced pluripotent, among others. Among different kinds of stem cells, mesenchymal stem cells are the most investigated for Alzheimer's treatment due to their multipotent nature, low immunogenicity, ability to penetrate the blood-brain barrier, and low risk of tumorigenesis, immune & inflammatory modulation, etc. They have been seen to substantially promote neurogenesis, synaptogenesis by secreting neurotrophic growth factors, as well as in ameliorating the Aβ and tau-mediated toxicity. This review covers the pathophysiology of AD, new medications, and therapies. Further, it will focus on the advancements and benefits of Mesenchymal Stem Cell therapies, their administration methods, clinical trials concerning AD progression, along with their future prospective.
Collapse
Affiliation(s)
- Aditya Bhatt
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India
| | - Harshita Bhardwaj
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India
| | - Priyanka Srivastava
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India.
| |
Collapse
|
7
|
Aranda-Abreu GE, Rojas-Durán F, Hernández-Aguilar ME, Herrera-Covarrubias D, Chí-Castañeda LD, Toledo-Cárdenas MR, Suárez-Medellín JM. Alzheimer's Disease: Cellular and Pharmacological Aspects. Geriatrics (Basel) 2024; 9:86. [PMID: 39051250 PMCID: PMC11270425 DOI: 10.3390/geriatrics9040086] [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: 04/30/2024] [Revised: 05/23/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease was described more than 100 years ago and despite the fact that several molecules are being tested for its treatment, which are in phase III trials, the disease continues to progress. The main problem is that these molecules function properly in healthy neurons, while neuronal pathology includes plasma membrane disruption, malfunction of various organelles, and hyperphosphorylation of Tau and amyloid plaques. The main objective of this article is the discussion of a neuronal restoration therapy, where molecules designed for the treatment of Alzheimer's disease would probably be more effective, and the quality of life of people would be better.
Collapse
Affiliation(s)
- Gonzalo Emiliano Aranda-Abreu
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91192, Mexico; (F.R.-D.); (M.E.H.-A.); (D.H.-C.); (L.D.C.-C.); (M.R.T.-C.); (J.M.S.-M.)
| | | | | | | | | | | | | |
Collapse
|
8
|
Kuznetsov AV. Numerical modeling of senile plaque development under conditions of limited diffusivity of amyloid-β monomers. J Theor Biol 2024; 587:111823. [PMID: 38608804 DOI: 10.1016/j.jtbi.2024.111823] [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: 02/24/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024]
Abstract
This paper introduces a new model to simulate the progression of senile plaques, focusing on scenarios where concentrations of amyloid beta (Aβ) monomers and aggregates vary between neurons. Extracellular variations in these concentrations may arise due to limited diffusivity of Aβ monomers and a high rate of Aβ monomer production at lipid membranes, requiring a substantial concentration gradient for diffusion-driven transport of Aβ monomers. The dimensionless formulation of the model is presented, which identifies four key dimensionless parameters governing the solutions for Aβ monomer and aggregate concentrations, as well as the radius of a growing Aβ plaque within the control volume. These parameters include the dimensionless diffusivity of Aβ monomers, the dimensionless rate of Aβ monomer production, and the dimensionless half-lives of Aβ monomers and aggregates. A dimensionless parameter is then introduced to evaluate the validity of the lumped capacitance approximation. An approximate solution is derived for the scenario involving large diffusivity of Aβ monomers and dysfunctional protein degradation machinery, resulting in infinitely long half-lives for Aβ monomers and aggregates. In this scenario, the concentrations of Aβ aggregates and the radius of the Aβ plaque depend solely on a single dimensionless parameter that characterizes the rate of Aβ monomer production. According to the approximate solution, the concentration of Aβ aggregates is linearly dependent on the rate of monomer production, and the radius of an Aβ plaque is directly proportional to the cube root of the rate of monomer production. However, when departing from the conditions of the approximate solution (e.g., finite half-lives), the concentrations of Aβ monomers and aggregates, along with the plaque radius, exhibit complex dependencies on all four dimensionless parameters. For instance, under physiological half-life conditions, the plaque radius reaches a maximum value and stabilizes thereafter.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA.
| |
Collapse
|
9
|
Lee B, Yu MS, Song JG, Lee HM, Kim HW, Na D. Corydalis ternata Nakai Alleviates Cognitive Decline in Alzheimer's Disease by Reducing β-Amyloid and Neuroinflammation. Rejuvenation Res 2024; 27:87-101. [PMID: 38545769 DOI: 10.1089/rej.2023.0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Recently, natural herbs have gained increasing attention owing to their comparatively low toxicity levels and the abundance of historical medical documentation regarding their use. Nevertheless, owing to a lack of knowledge regarding these herbs and their compounds, attempts to find those that could be beneficial for treating diseases have often been ad hoc; thus, there is now a growing demand for an in silico method to identify beneficial herbs. In this study, we present a computational approach for identifying natural herbs specifically effective in treating cognitive decline in Alzheimer's disease (AD) sufferers, which analyzes the similarities between herbal compounds and known drugs targeting AD-related proteins. Our in silico method suggests that Corydalis ternata can improve cognitive decline in AD sufferers. Behavioral tests with an AD mouse model for the confirmation of the in silico prediction reveals that C. ternata significantly alleviated the cognitive decline (memory and motor functions) caused by neurodegeneration. Further pathology analyses reveal that C. ternata decreases the level of Aβ plaques, reduces neuroinflammation, and promotes autophagy flux, and thus C. ternata can be clinically effective for preventing mild cognitive impairment during the early stages of AD. These findings highlight the potential utility of our in silico method and the potential clinical application of the identified natural herb in treating and preventing AD.
Collapse
Affiliation(s)
- Bomi Lee
- Department of Bio-Integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Myeong-Sang Yu
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Jae Gwang Song
- Department of Bio-Integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Hyang-Mi Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Hyung Wook Kim
- Department of Bio-Integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Dokyun Na
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Kuznetsov AV. Numerical and Analytical Simulation of the Growth of Amyloid-β Plaques. J Biomech Eng 2024; 146:061004. [PMID: 38421364 DOI: 10.1115/1.4064969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
Numerical and analytical solutions were employed to calculate the radius of an amyloid-β (Aβ) plaque over time. To the author's knowledge, this study presents the first model simulating the growth of Aβ plaques. Findings indicate that the plaque can attain a diameter of 50 μm after 20 years of growth, provided the Aβ monomer degradation machinery is malfunctioning. A mathematical model incorporates nucleation and autocatalytic growth processes using the Finke-Watzky model. The resulting system of ordinary differential equations was solved numerically, and for the simplified case of infinitely long Aβ monomer half-life, an analytical solution was found. Assuming that Aβ aggregates stick together and using the distance between the plaques as an input parameter of the model, it was possible to calculate the plaque radius from the concentration of Aβ aggregates. This led to the "cube root hypothesis," positing that Aβ plaque size increases proportionally to the cube root of time. This hypothesis helps explain why larger plaques grow more slowly. Furthermore, the obtained results suggest that the plaque size is independent of the kinetic constants governing Aβ plaque agglomeration, indicating that the kinetics of Aβ plaque agglomeration is not a limiting factor for plaque growth. Instead, the plaque growth rate is limited by the rates of Aβ monomer production and degradation.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910
| |
Collapse
|
11
|
Chen Y, Li Y, Wu M, Li Z. Electroacupuncture improves cognitive function in APP/PS1 mice by inhibiting oxidative stress related hippocampal neuronal ferroptosis. Brain Res 2024; 1831:148744. [PMID: 38163562 DOI: 10.1016/j.brainres.2023.148744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/03/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Electroacupuncture, recognized as a crucial non-pharmacological therapeutic approach, has demonstrated notable efficacy in enhancing cognitive function among Alzheimer's disease (AD) patients. This study aimed to investigate the neuroprotective properties of electroacupuncture in APP/PS1 mice with AD. METHODS A total of thirty APP/PS1 mice were randomly assigned to three groups: the Alzheimer's disease group (AD), the electroacupuncture treatment group (EA), and the ferroptosis inhibitor deferasirox treatment group (DFX). Additionally, ten C57BL/6 mice were included as a control group (Control). In the EA group, mice underwent flat needling at Baihui and Yintang, as well as point needling at Renzhong, once daily for 15 min each time. In the DFX group, mice received intraperitoneal injections of deferasirox at a dosage of 100 mg/kg/day. Following the 28-day treatment period, behavioral evaluation, morphological observation of neurons, and detection of neuronal ferroptosis were conducted. RESULTS The electroacupuncture treatment demonstrated a significant improvement in spatial learning, memory ability, and neuronal damage in mice with AD. Analysis of neuronal ferroptosis markers indicated that electroacupuncture interventions reduced the elevated levels of malondialdehyde, iron, and ptgs2 expression, while also increasing superoxide dismutase activity, Ferroportin 1 and glutathione peroxidase 4 expression. Moreover, the regulatory impact of electroacupuncture on ferroptosis may be attributed to its ability to enhance the expression and nuclear translocation of Nrf2. CONCLUSIONS This study suggested that electroacupuncture could inhibit the neuronal ferroptosis by activating the antioxidant function in neurons through p62/Keap1/Nrf2 signal pathway, thereby improve the cognitive function of AD mice by the neuronal protection effect.
Collapse
Affiliation(s)
- Yu Chen
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yitong Li
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Meng Wu
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Zhigang Li
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
12
|
Li J, Yang M, Dai Y, Guo X, Ding Y, Li X, Zhang S, Xu W, Chen L, Tao J, Liu W. Electroacupuncture regulates Rab5a-mediating NGF transduction to improve learning and memory ability in the early stage of AD mice. CNS Neurosci Ther 2024; 30:e14743. [PMID: 38780008 PMCID: PMC11112630 DOI: 10.1111/cns.14743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 03/22/2024] [Accepted: 04/11/2024] [Indexed: 05/25/2024] Open
Abstract
AIMS Nerve growth factor (NGF) loss is a potential factor for the degeneration of basal forebrain cholinergic neurons (BFCNs) in Alzheimer's disease (AD), and Rab5a is a key regulatory molecule of NGF signaling transduction. Here, we investigated the changes of Rab5a in 5 × FAD mice and further explored the mechanism of Electroacupuncture (EA) treatment in improving cognition in the early stage of AD. METHODS The total Rab5a and Rab5a-GTP in 5-month-old 5 × FAD mice and wild-type mice were detected using WB and IP technologies. 5 × FAD mice were treated with EA at the Bai hui (DU20) and Shen ting (DU24) acupoints for 4 weeks and CRE/LOXP technology was used to confirm the role of Rab5a in AD mediated by EA stimulation. The Novel Object Recognition and Morris water maze tests were used to evaluate the cognitive function of 5 × FAD mice. The Nissl, immunohistochemistry, and Thioflavin S staining were used to observe pathological morphological changes in the basal forebrain circuit. The Golgi staining was used to investigate the synaptic plasticity of the basal forebrain circuit and WB technology was used to detect the expression levels of cholinergic-related and NGF signal-related proteins. RESULTS The total Rab5a was unaltered, but Rab5a-GTP increased and the rab5a-positive early endosomes appeared enlarged in the hippocampus of 5 × FAD mice. Notably, EA reduced Rab5a-GTP in the hippocampus in the early stage of 5 × FAD mice. EA could improve object recognition memory and spatial learning memory by reducing Rab5a activity in the early stage of 5 × FAD mice. Moreover, EA could reduce Rab5a activity to increase NGF transduction and increase the levels of phosphorylated TrkA, AKT, and ERK in the basal forebrain and hippocampus, and increase the expression of cholinergic-related proteins, such as ChAT, vAchT, ChT1, m1AchR, and m2AchR in the basal forebrain and ChAT, m1AchR, and m2AchR in the hippocampus, improving synaptic plasticity in the basal forebrain hippocampal circuit in the early stage of 5 × FAD mice. CONCLUSIONS Rab5a hyperactivation is an early pathological manifestation of 5 × FAD mice. EA could suppress Rab5a-GTP to promote the transduction of NGF signaling, and enhance the synaptic plasticity of the basal forebrain hippocampal circuit improving cognitive impairment in the early stage of 5 × FAD mice.
Collapse
Affiliation(s)
- Jianhong Li
- The Institute of Rehabilitation IndustryFujian University of Traditional Chinese MedicineFuzhouChina
- Fujian Key Laboratory of Aptamers Technology900TH hospital of Joint Logistics Support ForceFuzhouChina
| | - Minguang Yang
- The Institute of Rehabilitation IndustryFujian University of Traditional Chinese MedicineFuzhouChina
| | - Yaling Dai
- National‐Local Joint Engineering Research Center of Rehabilitation Medicine TechnologyFujian University of Traditional Chinese MedicineFuzhouChina
| | - Xiaoqin Guo
- National‐Local Joint Engineering Research Center of Rehabilitation Medicine TechnologyFujian University of Traditional Chinese MedicineFuzhouChina
| | - Yanyi Ding
- National‐Local Joint Engineering Research Center of Rehabilitation Medicine TechnologyFujian University of Traditional Chinese MedicineFuzhouChina
| | - Xiaoling Li
- Provincial and Ministerial Co‐founded Collaborative Innovation Center of Rehabilitation TechnologyFujian University of Traditional Chinese MedicineFuzhouChina
| | - Shenghang Zhang
- Fujian Key Laboratory of Aptamers Technology900TH hospital of Joint Logistics Support ForceFuzhouChina
| | - Wenshan Xu
- Fujian Key Laboratory of Cognitive RehabilitationAffiliated Rehabilitation Hospital of Fujian University of Traditional Chinese MedicineFuzhouChina
| | - Lidian Chen
- Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese MedicineFujian University of Traditional Chinese MedicineFuzhouChina
| | - Jing Tao
- The Institute of Rehabilitation IndustryFujian University of Traditional Chinese MedicineFuzhouChina
| | - Weilin Liu
- The Institute of Rehabilitation IndustryFujian University of Traditional Chinese MedicineFuzhouChina
| |
Collapse
|
13
|
Wu X, Ze X, Qin S, Zhang B, Li X, Gong Q, Zhang H, Zhu Z, Xu J. Design, Synthesis, and Biological Evaluation of Novel Tetrahydroacridin Hybrids with Sulfur-Inserted Linkers as Potential Multitarget Agents for Alzheimer's Disease. Molecules 2024; 29:1782. [PMID: 38675602 PMCID: PMC11051924 DOI: 10.3390/molecules29081782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease that can lead to the loss of cognitive function. The progression of AD is regulated by multiple signaling pathways and their associated targets. Therefore, multitarget strategies theoretically have greater potential for treating AD. In this work, a series of new hybrids were designed and synthesized by the hybridization of tacrine (4, AChE: IC50 = 0.223 μM) with pyrimidone compound 5 (GSK-3β: IC50 = 3 μM) using the cysteamine or cystamine group as the connector. The biological evaluation results demonstrated that most of the compounds exhibited moderate to good inhibitory activities against acetylcholinesterase (AChE) and glycogen synthase kinase 3β (GSK-3β). The optimal compound 18a possessed potent dual AChE/GSK-3β inhibition (AChE: IC50 = 0.047 ± 0.002 μM, GSK-3β: IC50 = 0.930 ± 0.080 μM). Further molecular docking and enzymatic kinetic studies revealed that this compound could occupy both the catalytic anionic site and the peripheral anionic site of AChE. The results also showed a lack of toxicity to SH-SY5Y neuroblastoma cells at concentrations of up to 25 μM. Collectively, this work explored the structure-activity relationships of novel tetrahydroacridin hybrids with sulfur-inserted linkers, providing a reference for the further research and development of new multitarget anti-AD drugs.
Collapse
Affiliation(s)
- Xiuyuan Wu
- State Key Laboratory of Natural Medicines, Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; (X.W.); (X.Z.); (S.Q.); (X.L.)
| | - Xiaotong Ze
- State Key Laboratory of Natural Medicines, Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; (X.W.); (X.Z.); (S.Q.); (X.L.)
| | - Shuai Qin
- State Key Laboratory of Natural Medicines, Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; (X.W.); (X.Z.); (S.Q.); (X.L.)
| | - Beiyu Zhang
- Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, UK;
| | - Xinnan Li
- State Key Laboratory of Natural Medicines, Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; (X.W.); (X.Z.); (S.Q.); (X.L.)
| | - Qi Gong
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; (Q.G.); (H.Z.)
| | - Haiyan Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; (Q.G.); (H.Z.)
| | - Zheying Zhu
- Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, UK;
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines, Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; (X.W.); (X.Z.); (S.Q.); (X.L.)
| |
Collapse
|
14
|
Adamu A, Li S, Gao F, Xue G. The role of neuroinflammation in neurodegenerative diseases: current understanding and future therapeutic targets. Front Aging Neurosci 2024; 16:1347987. [PMID: 38681666 PMCID: PMC11045904 DOI: 10.3389/fnagi.2024.1347987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/25/2024] [Indexed: 05/01/2024] Open
Abstract
Neuroinflammation refers to a highly complicated reaction of the central nervous system (CNS) to certain stimuli such as trauma, infection, and neurodegenerative diseases. This is a cellular immune response whereby glial cells are activated, inflammatory mediators are liberated and reactive oxygen and nitrogen species are synthesized. Neuroinflammation is a key process that helps protect the brain from pathogens, but inappropriate, or protracted inflammation yields pathological states such as Parkinson's disease, Alzheimer's, Multiple Sclerosis, and other neurodegenerative disorders that showcase various pathways of neurodegeneration distributed in various parts of the CNS. This review reveals the major neuroinflammatory signaling pathways associated with neurodegeneration. Additionally, it explores promising therapeutic avenues, such as stem cell therapy, genetic intervention, and nanoparticles, aiming to regulate neuroinflammation and potentially impede or decelerate the advancement of these conditions. A comprehensive understanding of the intricate connection between neuroinflammation and these diseases is pivotal for the development of future treatment strategies that can alleviate the burden imposed by these devastating disorders.
Collapse
Affiliation(s)
| | | | | | - Guofang Xue
- Department of Neurology, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
15
|
Akhtar A, Singh S, Kaushik R, Awasthi R, Behl T. Types of memory, dementia, Alzheimer's disease, and their various pathological cascades as targets for potential pharmacological drugs. Ageing Res Rev 2024; 96:102289. [PMID: 38582379 DOI: 10.1016/j.arr.2024.102289] [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: 07/22/2023] [Revised: 03/30/2024] [Accepted: 03/30/2024] [Indexed: 04/08/2024]
Abstract
Alzheimer's disease (AD) is the most common type of dementia accounting for 90% of cases; however, frontotemporal dementia, vascular dementia, etc. prevails only in a minority of populations. The term dementia is defined as loss of memory which further takes several other categories of memories like working memory, spatial memory, fear memory, and long-term, and short-term memory into consideration. In this review, these memories have critically been elaborated based on context, duration, events, appearance, intensity, etc. The most important part and purpose of the review is the various pathological cascades as well as molecular levels of targets of AD, which have extracellular amyloid plaques and intracellular hyperphosphorylated tau protein as major disease hallmarks. There is another phenomenon that either leads to or arises from the above-mentioned hallmarks, such as oxidative stress, mitochondrial dysfunction, neuroinflammation, cholinergic dysfunction, and insulin resistance. Several potential drugs like antioxidants, anti-inflammatory drugs, acetylcholinesterase inhibitors, insulin mimetics or sensitizers, etc. studied in various previous preclinical or clinical reports were put as having the capacity to act on these pathological targets. Additionally, agents directly or indirectly targeting amyloid and tau were also discussed. This could be further investigated in future research.
Collapse
Affiliation(s)
- Ansab Akhtar
- Louisiana State University Health Sciences Center, Neuroscience Center of Excellence, School of Medicine, New Orleans, LA 70112, USA.
| | - Siddharth Singh
- School of Health Sciences & Technology, UPES University, Bidholi, Dehradun, Uttarakhand 248007, India
| | - Ravinder Kaushik
- School of Health Sciences & Technology, UPES University, Bidholi, Dehradun, Uttarakhand 248007, India
| | - Rajendra Awasthi
- School of Health Sciences & Technology, UPES University, Bidholi, Dehradun, Uttarakhand 248007, India
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab 140306, India
| |
Collapse
|
16
|
Gu X, Qi L, Qi Q, Zhou J, Chen S, Wang L. Monoclonal antibody therapy for Alzheimer's disease focusing on intracerebral targets. Biosci Trends 2024; 18:49-65. [PMID: 38382942 DOI: 10.5582/bst.2023.01288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. Due to the complexity of the disorder and the presence of the blood-brain barrier (BBB), its drug discovery and development are facing enormous challenges, especially after several failures of monoclonal antibody (mAb) trials. Nevertheless, the Food and Drug Administration's approval of the mAb aducanumab has ushered in a new day. As we better understand the disease's pathogenesis and identify novel intracerebral therapeutic targets, antibody-based therapies have advanced over the past few years. The mAb drugs targeting β-amyloid or hyperphosphorylated tau protein are the focus of the current research. Massive neuronal loss and glial cell-mediated inflammation are also the vital pathological hallmarks of AD, signaling a new direction for research on mAb drugs. We have elucidated the mechanisms by which AD-specific mAbs cross the BBB to bind to targets. In order to investigate therapeutic approaches to treat AD, this review focuses on the promising mAbs targeting intracerebral dysfunction and related strategies to cross the BBB.
Collapse
Affiliation(s)
- Xiaolei Gu
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Long Qi
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Qing Qi
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Zhou
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Song Chen
- Postdoctoral Station of Xiamen University, Fujian, China
| | - Ling Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| |
Collapse
|
17
|
Chen Z, Wang X, Du S, Liu Q, Xu Z, Guo Y, Lin X. A review on traditional Chinese medicine natural products and acupuncture intervention for Alzheimer's disease based on the neuroinflammatory. Chin Med 2024; 19:35. [PMID: 38419106 PMCID: PMC10900670 DOI: 10.1186/s13020-024-00900-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with insidious onset and progressive development. It is clinically characterized by cognitive impairment, memory impairment and behavioral change. Chinese herbal medicine and acupuncture are important components of traditional Chinese medicine (TCM), and are commonly used in clinical treatment of AD. This paper systematically summarizes the research progress of traditional Chinese medicine natural products and acupuncture treatment of AD, which combined with existing clinical and preclinical evidence, based on a comprehensive review of neuroinflammation, and discusses the efficacy and potential mechanisms of traditional Chinese medicine natural products and acupuncture treatment of AD. Resveratrol, curcumin, kaempferol and other Chinese herbal medicine components can significantly inhibit the neuroinflammation of AD in vivo and in vitro, and are candidates for the treatment of AD. Acupuncture can alleviate the memory and cognitive impairment of AD by improving neuroinflammation, synaptic plasticity, nerve cell apoptosis and reducing the production and aggregation of amyloid β protein (Aβ) in the brain. It has the characteristics of early, safe, effective and benign bidirectional adjustment. The purpose of this paper is to provide a basis for improving the clinical strategies of TCM for the treatment of AD.
Collapse
Affiliation(s)
- Zhihan Chen
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Xinrui Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Simin Du
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Qi Liu
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Zhifang Xu
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin, 301617, People's Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People's Republic of China
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin, 301617, People's Republic of China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People's Republic of China.
| | - Xiaowei Lin
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin, 301617, People's Republic of China.
| |
Collapse
|
18
|
Karimi Tari P, Parsons CG, Collingridge GL, Rammes G. Memantine: Updating a rare success story in pro-cognitive therapeutics. Neuropharmacology 2024; 244:109737. [PMID: 37832633 DOI: 10.1016/j.neuropharm.2023.109737] [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: 02/16/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023]
Abstract
The great potential for NMDA receptor modulators as druggable targets in neurodegenerative disorders has been met with limited success. Considered one of the rare exceptions, memantine has consistently demonstrated restorative and prophylactic properties in many AD models. In clinical trials memantine slows the decline in cognitive performance associated with AD. Here, we provide an overview of the basic properties including pharmacological targets, toxicology and cellular effects of memantine. Evidence demonstrating reductions in molecular, physiological and behavioural indices of AD-like impairments associated with memantine treatment are also discussed. This represents both an extension and homage to Dr. Chris Parson's considerable contributions to our fundamental understanding of a success story in the AD treatment landscape.
Collapse
Affiliation(s)
- Parisa Karimi Tari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Chris G Parsons
- Galimedix Therapeutics, Inc., 2704 Calvend Lane, Kensington, 20895, MD, USA
| | - Graham L Collingridge
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada; Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada; TANZ Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care Medicine of the Technical University of Munich, School of Medicine, 22, 81675, Munich, Germany.
| |
Collapse
|
19
|
Liu Y, Ma C, Li Y, Li M, Cui T, Zhao X, Li Z, Jia H, Wang H, Xiu X, Hu D, Zhang R, Wang N, Liu P, Yang H, Cheng M. Design, synthesis and biological evaluation of carbamate derivatives incorporating multifunctional carrier scaffolds as pseudo-irreversible cholinesterase inhibitors for the treatment of Alzheimer's disease. Eur J Med Chem 2024; 265:116071. [PMID: 38157596 DOI: 10.1016/j.ejmech.2023.116071] [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: 10/10/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
In this study, a series of carbamate derivatives incorporating multifunctional carrier scaffolds were designed, synthesized, and evaluated as potential therapeutic agents for Alzheimer's disease (AD). We used tacrine to modify the aliphatic substituent, and employed rivastigmine, indole and sibiriline fragments as carrier scaffolds. The majority of compounds exhibited good inhibitory activity for cholinesterase. Notably, compound C7 with sibiriline fragment exhibited potent inhibitory activities against human acetylcholinesterase (hAChE, IC50 = 30.35 ± 2.07 nM) and human butyrylcholinesterase (hBuChE, IC50 = 48.03 ± 6.41 nM) with minimal neurotoxicity. Further investigations have demonstrated that C7 exhibited a remarkable capacity to safeguard PC12 cells against H2O2-induced apoptosis and effectively suppressed the production of reactive oxygen species (ROS). Moreover, in an inflammation model of BV2 cells induced by lipopolysaccharide (LPS), C7 effectively attenuated the levels of pro-inflammatory cytokines. After 12 h of dialysis, C7 continued to exhibit an inhibitory effect on cholinesterase activity. An acute toxicity test in vivo demonstrated that C7 exhibited a superior safety profile and no hepatotoxicity compared to the parent nucleus tacrine. In the scopolamine-induced AD mouse model, C7 (20 mg/kg) significantly reduced cholinesterase activity in the brain of the mice. C7 was tested in a pharmacological AD mouse model induced by Aβ1-42 and attenuated memory deficits at doses as low as 5 mg/kg. The pseudo-irreversible cholinesterase inhibitory properties and multifunctional therapeutic attributes of C7 render it a promising candidate for further investigation in the treatment of AD.
Collapse
Affiliation(s)
- Yaoyang Liu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Chao Ma
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Yingbo Li
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Mengzhen Li
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Tao Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Xueqi Zhao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Zhenli Li
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Hongwei Jia
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Hanxun Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Xiaomeng Xiu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Dexiang Hu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Ruiwen Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Ningwei Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Peng Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Huali Yang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China.
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China.
| |
Collapse
|
20
|
Nasb M, Tao W, Chen N. Alzheimer's Disease Puzzle: Delving into Pathogenesis Hypotheses. Aging Dis 2024; 15:43-73. [PMID: 37450931 PMCID: PMC10796101 DOI: 10.14336/ad.2023.0608] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disease characterized by both amnestic and non-amnestic clinical manifestations. It accounts for approximately 60-70% of all dementia cases worldwide. With the increasing number of AD patients, elucidating underlying mechanisms and developing corresponding interventional strategies are necessary. Hypotheses about AD such as amyloid cascade, Tau hyper-phosphorylation, neuroinflammation, oxidative stress, mitochondrial dysfunction, cholinergic, and vascular hypotheses are not mutually exclusive, and all of them play a certain role in the development of AD. The amyloid cascade hypothesis is currently the most widely studied; however, other hypotheses are also gaining support. This article summarizes the recent evidence regarding major pathological hypotheses of AD and their potential interplay, as well as the strengths and weaknesses of each hypothesis and their implications for the development of effective treatments. This could stimulate further studies and promote the development of more effective therapeutic strategies for AD.
Collapse
Affiliation(s)
| | | | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| |
Collapse
|
21
|
Liu Y, Tan Y, Zhang Z, Yi M, Zhu L, Peng W. The interaction between ageing and Alzheimer's disease: insights from the hallmarks of ageing. Transl Neurodegener 2024; 13:7. [PMID: 38254235 PMCID: PMC10804662 DOI: 10.1186/s40035-024-00397-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Ageing is a crucial risk factor for Alzheimer's disease (AD) and is characterised by systemic changes in both intracellular and extracellular microenvironments that affect the entire body instead of a single organ. Understanding the specific mechanisms underlying the role of ageing in disease development can facilitate the treatment of ageing-related diseases, such as AD. Signs of brain ageing have been observed in both AD patients and animal models. Alleviating the pathological changes caused by brain ageing can dramatically ameliorate the amyloid beta- and tau-induced neuropathological and memory impairments, indicating that ageing plays a crucial role in the pathophysiological process of AD. In this review, we summarize the impact of several age-related factors on AD and propose that preventing pathological changes caused by brain ageing is a promising strategy for improving cognitive health.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Yejun Tan
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Min Yi
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Weijun Peng
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China.
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China.
| |
Collapse
|
22
|
Zhang M, Wang W, Ye Q, Fu Y, Li X, Yang K, Gao F, Zhou A, Wei Y, Tian S, Li S, Wei F, Shi W, Li WD. Histone deacetylase inhibitors VPA and WT161 ameliorate the pathological features and cognitive impairments of the APP/PS1 Alzheimer's disease mouse model by regulating the expression of APP secretases. Alzheimers Res Ther 2024; 16:15. [PMID: 38245771 PMCID: PMC10799458 DOI: 10.1186/s13195-024-01384-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 01/03/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a degenerative neurological disorder. Recent studies have indicated that histone deacetylases (HDACs) are among the most prominent epigenetic therapy targets and that HDAC inhibitors have therapeutic effects on AD. Here, we identified sodium valproate (VPA), a pan-HDAC inhibitor, and WT161, a novel HDAC6 selective inhibitor, as potential therapeutic agents for AD. Underlying molecular mechanisms were investigated. METHODS A cellular model, N2a-APPswe, was established via lentiviral infection, and the APPswe/PSEN1dE9 transgenic mouse model was employed in the study. LC-MS/MS was applied to quantify the concentration of WT161 in the mouse brain. Western blotting, immunohistochemical staining, thioflavin-S staining and ELISA were applied to detect protein expression in cells, tissues, or serum. RNA interference was utilized to knockdown the expression of specific genes in cells. The cognitive function of mice was assessed via the nest-building test, novel object recognition test and Morris water maze test. RESULTS Previous studies have focused mainly on the impact of HDAC inhibitors on histone deacetylase activity. Our study discovered that VPA and WT161 can downregulate the expression of multiple HDACs, such as HDAC1 and HDAC6, in both AD cell and mouse models. Moreover, they also affect the expression of APP and APP secretases (BACE1, PSEN1, ADAM10). RNA interference and subsequent vitamin C induction further confirmed that the expression of APP and APP secretases is indeed regulated by HDAC1 and HDAC6, with the JNK pathway being the intermediate link in this regulatory process. Through the above pathways, VPA and WT161 effectively reduced Aβ deposition in both AD cell and mouse models and significantly improved cognitive function in AD mice. CONCLUSIONS In general, we have discovered that the HDAC6-JNK-APP secretases cascade is an important pathway for VPA and WT161 to exert their therapeutic effects on AD. Investigations into the safety and efficacy of VPA and WT161 were also conducted, providing essential preclinical evidence for assessing these two epigenetic drugs for the treatment of AD.
Collapse
Affiliation(s)
- Miaomiao Zhang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
- Prenatal Diagnostic Center, Yiwu Maternity and Children Hospital, Yiwu, 322000, China
| | - Wanyao Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Qun Ye
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yun Fu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
- College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Xuemin Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ke Yang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Fan Gao
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - An Zhou
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yonghui Wei
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Shuang Tian
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Shen Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Fengjiang Wei
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Wentao Shi
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Wei-Dong Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
23
|
Aljassabi A, Zieneldien T, Kim J, Regmi D, Cao C. Alzheimer's Disease Immunotherapy: Current Strategies and Future Prospects. J Alzheimers Dis 2024; 98:755-772. [PMID: 38489183 DOI: 10.3233/jad-231163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Alzheimer's disease (AD) is an extremely complex and heterogeneous pathology influenced by many factors contributing to its onset and progression, including aging, amyloid-beta (Aβ) plaques, tau fibril accumulation, inflammation, etc. Despite promising advances in drug development, there is no cure for AD. Although there have been substantial advancements in understanding the pathogenesis of AD, there have been over 200 unsuccessful clinical trials in the past decade. In recent years, immunotherapies have been at the forefront of these efforts. Immunotherapy alludes to the immunological field that strives to identify disease treatments via the enhancement, suppression, or induction of immune responses. Interestingly, immunotherapy in AD is a relatively new approach for non-infectious disease. At present, antibody therapy (passive immunotherapy) that targets anti-Aβ aimed to prevent the fibrillization of Aβ peptides and disrupt pre-existing fibrils is a predominant AD immunotherapy due to the continuous failure of active immunotherapy for AD. The most rational and safe strategies will be those targeting the toxic molecule without triggering an abnormal immune response, offering therapeutic advantages, thus making clinical trial design more efficient. This review offers a concise overview of immunotherapeutic strategies, including active and passive immunotherapy for AD. Our review encompasses approved methods and those presently under investigation in clinical trials, while elucidating the recent challenges, complications, successes, and potential treatments. Thus, immunotherapies targeting Aβ throughout the disease progression using a mutant oligomer-Aβ stimulated dendritic cell vaccine may offer a promising therapy in AD.
Collapse
Affiliation(s)
- Ali Aljassabi
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Tarek Zieneldien
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Janice Kim
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Deepika Regmi
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Chuanhai Cao
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| |
Collapse
|
24
|
Lee EH, Kim GH, Park HK, Kang HJ, Park YK, Lee HA, Hong CH, Moon SY, Kang W, Oh HS, Yoon HJ, Choi SH, Jeong JH. Effects of the multidomain intervention with nutritional supplements on cognition and gut microbiome in early symptomatic Alzheimer's disease: a randomized controlled trial. Front Aging Neurosci 2023; 15:1266955. [PMID: 38020771 PMCID: PMC10652389 DOI: 10.3389/fnagi.2023.1266955] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
Background The SoUth Korean study to PrEvent cognitive impaiRment and protect BRAIN health through lifestyle intervention in at-risk elderly people (SUPERBRAIN) is a part of the World-Wide Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (WW-FINGERS) network. This study aimed to demonstrate the effects of the SUPERBRAIN-based multidomain intervention with nutritional supplements in amyloid positive emission tomography (PET) proven early symptomatic Alzheimer's disease patients. Methods Forty-six participants who were diagnosed with mild cognitive impairment or mild dementia and were positive in the amyloid PET study randomized into three groups: group A, the multidomain intervention with nutritional supplements; group B, nutritional supplements only; and a control group. The primary outcome was a change in the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) total scale index score after an 8-week intervention. Secondary outcomes, including gut microbiome data, were also analyzed. Results The RBANS total scale index score improved significantly in group A compared with group B (p < 0.032) and compared with the control group (p < 0.001). After intervention, beta diversity of the gut microbiome between group A and the control group increased, and patients in group A were more enriched with Bifidobacterium. Conclusion SUPERBRAIN-based multidomain intervention with nutritional supplements improves cognition and gut microbiota in patients with early symptomatic Alzheimer's disease who were amyloid-positive by PET.
Collapse
Affiliation(s)
- Eun Hye Lee
- Department of Neurology, Ewha Womans University School of Medicine, Seoul, Republic of Korea
- Department of Neurology, Ewha Womans University Seoul Hospital, Seoul, Republic of Korea
| | - Geon Ha Kim
- Department of Neurology, Ewha Womans University School of Medicine, Seoul, Republic of Korea
- Department of Neurology, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Hee Kyung Park
- Division of Psychiatry, Department of Mental Health Care of Older People, University College London, London, United Kingdom
| | - Hae Jin Kang
- Department of Medical Nutrition (AgeTech-Service Convergence Major), Kyung Hee University, Yongin, Republic of Korea
| | - Yoo Kyoung Park
- Department of Medical Nutrition (AgeTech-Service Convergence Major), Kyung Hee University, Yongin, Republic of Korea
| | - Hye Ah Lee
- Clinical Trial Center, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Chang Hyung Hong
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - So Young Moon
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Woorim Kang
- CJ Bioscience Inc., Seoul, Republic of Korea
| | | | - Hai-Jeon Yoon
- Department of Nuclear Medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Seong Hye Choi
- Department of Neurology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jee Hyang Jeong
- Department of Neurology, Ewha Womans University School of Medicine, Seoul, Republic of Korea
- Department of Neurology, Ewha Womans University Seoul Hospital, Seoul, Republic of Korea
| |
Collapse
|
25
|
Zhou JC, Li HL, Zhou Y, Li XT, Yang ZY, Tohda C, Komatsu K, Piao XH, Ge YW. The roles of natural triterpenoid saponins against Alzheimer's disease. Phytother Res 2023; 37:5017-5040. [PMID: 37491018 DOI: 10.1002/ptr.7967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/23/2023] [Accepted: 07/07/2023] [Indexed: 07/27/2023]
Abstract
The aging of the world population and increasing stress levels in life are the major cause of the increased incidence of neurological disorders. Alzheimer's disease (AD) creates a huge burden on the lives and health of individuals and has become a big concern for society. Triterpenoid saponins (TS), representative natural product components, have a wide range of pharmacological bioactivities such as anti-inflammation, antioxidation, antiapoptosis, hormone-like, and gut microbiota regulation. Notably, some natural TS exhibited promising neuroprotective activity that can intervene in AD progress, especially in the early stage. Recently, studies have indicated that TS play a pronounced positive role in the prevention and treatment of AD. This review discusses the recent research on the neuroprotection of TS and proceeds to detail the action mechanisms of TS against AD, hoping to provide a reference for drug development for anti-AD.
Collapse
Affiliation(s)
- Jie-Chun Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative team of research on effective substances of traditional Chinese medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hui-Lin Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative team of research on effective substances of traditional Chinese medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yu Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative team of research on effective substances of traditional Chinese medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xi-Tao Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative team of research on effective substances of traditional Chinese medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhi-You Yang
- College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Institute of Nutrition and Marine Drugs, Guangdong Ocean University, Zhanjiang, China
| | - Chihiro Tohda
- Division of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Katsuko Komatsu
- Division of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Xiu-Hong Piao
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yue-Wei Ge
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative team of research on effective substances of traditional Chinese medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
26
|
Perxés Perich M, Palma-Florez S, Solé C, Goberna-Ferrón S, Samitier J, Gómez-Romero P, Mir M, Lagunas A. Polyoxometalate-Decorated Gold Nanoparticles Inhibit β-Amyloid Aggregation and Cross the Blood-Brain Barrier in a µphysiological Model. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2697. [PMID: 37836338 PMCID: PMC10574493 DOI: 10.3390/nano13192697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023]
Abstract
Alzheimer's disease is characterized by a combination of several neuropathological hallmarks, such as extracellular aggregates of beta amyloid (Aβ). Numerous alternatives have been studied for inhibiting Aβ aggregation but, at this time, there are no effective treatments available. Here, we developed the tri-component nanohybrid system AuNPs@POM@PEG based on gold nanoparticles (AuNPs) covered with polyoxometalates (POMs) and polyethylene glycol (PEG). In this work, AuNPs@POM@PEG demonstrated the inhibition of the formation of amyloid fibrils, showing a 75% decrease in Aβ aggregation in vitro. As it is a potential candidate for the treatment of Alzheimer's disease, we evaluated the cytotoxicity of AuNPs@POM@PEG and its ability to cross the blood-brain barrier (BBB). We achieved a stable nanosystem that is non-cytotoxic below 2.5 nM to human neurovascular cells. The brain permeability of AuNPs@POM@PEG was analyzed in an in vitro microphysiological model of the BBB (BBB-on-a-chip), containing 3D human neurovascular cell co-cultures and microfluidics. The results show that AuNPs@POM@PEG was able to cross the brain endothelial barrier in the chip and demonstrated that POM does not affect the barrier integrity, giving the green light to further studies into this system as a nanotherapeutic.
Collapse
Affiliation(s)
- Marta Perxés Perich
- Catalan Institute of Nanoscience and Nanotechnology(ICN2) CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Materials Chemistry and Catalysis, Debye Institute for Nanomaterials Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Sujey Palma-Florez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Clara Solé
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Sara Goberna-Ferrón
- Catalan Institute of Nanoscience and Nanotechnology(ICN2) CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Instituto Universitario de Tecnología Química (CSIC-UPV), Universitat Politècnica de València, Avda. De los Naranjos s/n, 46022 Valencia, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN) Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Pedro Gómez-Romero
- Catalan Institute of Nanoscience and Nanotechnology(ICN2) CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Mònica Mir
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN) Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Anna Lagunas
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN) Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
| |
Collapse
|
27
|
Liu J, Li T, Zhong G, Pan Y, Gao M, Su S, Liang Y, Ma C, Liu Y, Wang Q, Shi Q. Exploring the therapeutic potential of natural compounds for Alzheimer's disease: Mechanisms of action and pharmacological properties. Biomed Pharmacother 2023; 166:115406. [PMID: 37659206 DOI: 10.1016/j.biopha.2023.115406] [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: 07/18/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023] Open
Abstract
Alzheimer's Disease (AD) is a global public health priority characterized by high mortality rates in adults and an increasing prevalence in aging populations worldwide. Despite significant advancements in comprehending the pathogenesis of AD since its initial report in 1907, there remains a lack of effective curative or preventive measures for the disease. In recent years, natural compounds sourced from diverse origins have garnered considerable attention as potential therapeutic agents for AD, owing to their anti-inflammatory, antioxidant, and neuroprotective properties. This review aims to consolidate the therapeutic effects of natural compounds on AD, specifically targeting the reduction of β-amyloid (Aβ) overproduction, anti-apoptosis, autophagy, neuroinflammation, oxidative stress, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction. Notably, the identified compounds exhibiting these effects predominantly originate from plants. This review provides valuable insights into the potential of natural compounds as a reservoir of novel therapeutic agents for AD, thereby stimulating further research and contributing to the development of efficacious treatments for this devastating disease.
Collapse
Affiliation(s)
- Jinman Liu
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen 529099, China
| | - Tianyao Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yaru Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Minghuang Gao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shijie Su
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yong Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Cuiru Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yuanyue Liu
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210017, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Qing Shi
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen 529099, China.
| |
Collapse
|
28
|
Luo B. Insights into the advances in therapeutic drugs for neuroinflammation-related diseases. Int J Neurosci 2023:1-26. [PMID: 37722706 DOI: 10.1080/00207454.2023.2260088] [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: 08/11/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Studies have shown that neurodegenerative diseases such as AD and PD are related to neuroinflammation. Neuroinflammation is a common inflammatory condition that can lead to a variety of dysfunction in the body. At present, it is no medications specifically approved to prevent or cure neuroinflammation, so even though many drugs can temporarily control the neurological symptoms of neuroinflammation, but no one can reverse the progress of neuroinflammation, let al.one completely cure neuroinflammation. Therefore, it is urgent to develop new drug development for neuroinflammation treatment. In this review, we highlight the therapeutic advancement in the field of neurodegenerative disorders, by focusing on the impact of neuroinflammation treatment has on these conditions, and the effective drugs for the treatment of neuroinflammation and neurodegenerative diseases and their latest research progress are reviewed according to the related signaling pathway, as well as the prospect of their clinical application is also discussed. The purpose of this review is to enable specialists to better understand the mechanisms underlying neuroinflammation and anti-inflammatory drugs, promote the development of therapeutic drugs for neuroinflammation and neurodegenerative diseases, and further provide therapeutic references for clinical neurologists.
Collapse
Affiliation(s)
- Bozhi Luo
- School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
29
|
Ren Y, Qu S. Constituent isoflavones of Puerariae radix as a potential neuroprotector in cognitive impairment: Evidence from preclinical studies. Ageing Res Rev 2023; 90:102040. [PMID: 37619620 DOI: 10.1016/j.arr.2023.102040] [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: 08/03/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
With the increasing aging population worldwide, the incidence of senile cognitive impairment (CI) is increasing, posing a serious threat to the health of elderly persons. Despite developing new drugs aimed at improving CI, progress in this regard has been insufficient. Natural preparations derived from plants have become an unparalleled resource for developing new drugs. Puerariae radix (PR) has a long history as Chinese herbal medicine. PR is rich in various chemical components such as isoflavones, triterpenes, and saponins. The isoflavones (puerarin, daidzein, formononetin, and genistein) exhibit potential therapeutic effects on CI through multiple mechanisms. Relevant literature was organized from major scientific databases such as PubMed, Elsevier, SpringerLink, ScienceDirect, and Web of Science. Using "Puerariae radix," "Pueraria lobata," "isoflavones," "puerarin," "antioxidant," "daidzein," "formononetin," "genistein," "Alzheimer"s disease," and "vascular cognitive impairment" as keywords, the relevant literature was extracted from the databases mentioned above. We found that isoflavones from PR have neuroprotective effects on multiple models of CI via multiple targets and mechanisms. These isoflavones prevent Aβ aggregation, inhibit tau hyperphosphorylation, increase cholinergic neurotransmitter levels, reduce neuroinflammation and oxidative stress, improve synaptic plasticity, promote nerve regeneration, and prevent apoptosis. PR has been used as traditional Chinese herbal medicine for a long time, and its constituent isoflavones exert significant therapeutic effects on CI through various neuroprotective mechanisms. This review will contribute to the future development of isoflavones present in PR as novel drug candidates for the clinical treatment of CI.
Collapse
Affiliation(s)
- Yaoyao Ren
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, 110004 Shenyang, Liaoning, PR China
| | - Shengtao Qu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, No. 36 Sanhao St, Shenyang 110004, PR China.
| |
Collapse
|
30
|
Mani V, Rashed Almutairi S. Impact of levetiracetam on cognitive impairment, neuroinflammation, oxidative stress, and neuronal apoptosis caused by lipopolysaccharides in rats. Saudi Pharm J 2023; 31:101728. [PMID: 37583755 PMCID: PMC10424214 DOI: 10.1016/j.jsps.2023.101728] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/28/2023] [Indexed: 08/17/2023] Open
Abstract
Introduction Neuroinflammation is associated with the elevation of toxic proinflammatory mediators that promote neurodegeneration and subsequently affect cognition. Causes of inflammation in the neuronal cells are believed to initiate various neurodegenerative disorders, mainly Alzheimer's disease. Levetiracetam is a second-generation antiepileptic drug. There is evidence supporting the memory-enhancing effect of levetiracetam from numerous experimental and clinical studies. Therefore, this research focused on finding its protective effects against lipopolysaccharides prompted cognitive impairment and exploring possible mechanisms underlining their neuroprotection. Methodology Two doses (100 or 200 mg/kg) of levetiracetam were administrated orally for 30 days. Additionally, four doses (250 µg/kg) of lipopolysaccharide were injected peripherally to induce neurotoxicity. Behavioral tests were carried out using various maze models. At the end of the tests, brain tissues were collected for biochemical evaluations. Cholinergic, neuroinflammatory, apoptosis, and oxidative-related parameters were analyzed in the brain homogenate to explore the possible mechanisms of action of levetiracetam. Results In lipopolysaccharide-induced rats, levetiracetam indicated a reduction (p < 0.01) in transfer latency using the elevated plus-maze. An improvement (p < 0.01) in novel and familiar objects exploration time using novel object recognition test. A rise (p < 0.05) in novel arm entries and extended time spent in the novel arm using the Y-maze test. In extension, the levels of acetylcholine (p < 0.001), anti-inflammatory factors (transforming growth factor-β1; p < 0.01 and interleukin-10; p < 0.05), and an antioxidant (catalase; p < 0.01) were elevated in lipopolysaccharide-induced rats after the administration of levetiracetam. In contrast, inflammatory factors (cyclooxygenase-2; p < 0.05, nuclear factor kappa B; p < 0.05, tumor necrosis factor-α; p < 0.01, and interleukin-6 (p < 0.01), apoptosis inducers (BCL2-associated X protein; p < 0.05 and Caspase-3 (p < 0.001), and oxidative stress (malondialdehyde; p < 0.05) were considerably reduced with levetiracetam in lipopolysaccharide-induced rats. Conclusion The collective results suggested that levetiracetam may be able to treat neuroinflammatory-related memory loss by enhancing cholinergic activity while reducing neuroinflammation, cellular apoptosis, and oxidative stress.
Collapse
Affiliation(s)
- Vasudevan Mani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Salem Rashed Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
31
|
Zhao X, Ge P, Lei S, Guo S, Zhou P, Zhao L, Qi Y, Wei X, Wu W, Wang N, Guo R, Yang N, Xiao Q, Zhang Q, Zhu H. An Exosome-Based Therapeutic Strategy Targeting Neuroinflammation in Alzheimer's Disease with Berberine and Palmatine. Drug Des Devel Ther 2023; 17:2401-2420. [PMID: 37609432 PMCID: PMC10441573 DOI: 10.2147/dddt.s417465] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/12/2023] [Indexed: 08/24/2023] Open
Abstract
Introduction Neuroinflammation is one of the major pathogeneses in Alzheimer's disease (AD) and mainly involves abnormal inflammatory activation of microglia by multiple pathological stimuli. The treatment of AD remains a major challenge due to the multifactorial characterization of AD and the inefficient ability of therapeutic drugs to permeate through the blood‒brain barrier (BBB). Accordingly, drug combination treatment and drug carrier delivery have become important therapeutic tools for the treatment of multifactorial diseases, especially AD. Methods Inflammatory cytokine levels in microglia, including NO, TNF-α, IL-1β, IL-4, and IL-10, were detected. The Morris water maze and object location task were used to investigate the learning and memory functions of APP/PS1 mice in different treatment groups. The number of neurons and plasticity of synapses were evaluated by immunofluorescence double labelling. Additionally, the ratio of β-amyloid plaques and the number of activated microglia were evaluated by immunofluorescence staining. The concentrations of β-amyloid plaques and inflammatory factors in the hippocampus were determined by ELISA. Microglia-derived exosomes (Exos) were extracted and purified by size exclusion chromatography. The distribution of exosomes and drugs was investigated in vitro and in vivo. Results Compared to single drug interventions, the combination of Ber and Pal (Ber/Pal) modulated microglial inflammatory cytokine levels. Ber/Pal promoted the recovery of learning and memory impairment in APP/PS1 mice. Immunofluorescence staining indicated that Ber/Pal restored neurons, inhibited Aβ plaque formation and microglial activation, and regulated the secretion of inflammatory factors. Exos promoted the accumulation of drugs in cells and tissues and improved the targeting of drugs across the BBB. Conclusion Ber/Pal could offer a synergistic and more comprehensive therapeutic effect in AD. Additionally, the microglia-derived Exos-Ber/Pal delivery system promoted the targeting and permeation of drugs into the brain, suggesting a creative strategy for targeting AD therapy by regulating neuroinflammation in microglial cells.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Pingyuan Ge
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Shaohua Lei
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Siqi Guo
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Peng Zhou
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Li Zhao
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yiyu Qi
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Xiaotong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Weizhen Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Ningjing Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Rui Guo
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Nianyun Yang
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Qingqing Xiao
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Qichun Zhang
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Huaxu Zhu
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
32
|
Zhang L, Xu J, Guo J, Wang Y, Wang Q. Elucidation of Pharmacological Mechanism Underlying the Anti-Alzheimer's Disease Effects of Evodia rutaecarpa and Discovery of Novel Lead Molecules: An In Silico Study. Molecules 2023; 28:5846. [PMID: 37570816 PMCID: PMC10421504 DOI: 10.3390/molecules28155846] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/29/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD) is a brain disease with a peculiarity of multiformity and an insidious onset. Multiple-target drugs, especially Chinese traditional medicine, have achieved a measure of success in AD treatment. Evodia rutaecarpa (Juss.) Benth. (Wuzhuyu, WZY, i.e., E. rutaecarpa), a traditional Chinese herb, has been identified as an effective drug to cure migraines. To our surprise, our in silico study showed that rather than migraines, Alzheimer's disease was the primary disease to which the E. rutaecarpa active compounds were targeted. Correspondingly, a behavioral experiment showed that E. rutaecarpa extract could improve impairments in learning and memory in AD model mice. However, the mechanism underlying the way that E. rutaecarpa compounds target AD is still not clear. For this purpose, we employed methods of pharmacology networking and molecular docking to explore this mechanism. We found that E. rutaecarpa showed significant AD-targeting characteristics, and alkaloids of E. rutaecarpa played the main role in binding to the key nodes of AD. Our research detected that E. rutaecarpa affects the pathologic development of AD through the serotonergic synapse signaling pathway (SLC6A4), hormones (PTGS2, ESR1, AR), anti-neuroinflammation (SRC, TNF, NOS3), transcription regulation (NR3C1), and molecular chaperones (HSP90AA1), especially in the key nodes of PTGS2, AR, SLCA64, and SRC. Graveoline, 5-methoxy-N, N-dimethyltryptamine, dehydroevodiamine, and goshuyuamide II in E. rutaecarpa show stronger binding affinities to these key proteins than currently known preclinical and clinical drugs, showing a great potential to be developed as lead molecules for treating AD.
Collapse
Affiliation(s)
- Lulu Zhang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
| | - Jia Xu
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China; (J.X.); (J.G.)
| | - Jiejie Guo
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China; (J.X.); (J.G.)
| | - Yun Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
| | - Qinwen Wang
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China; (J.X.); (J.G.)
| |
Collapse
|
33
|
Graham AS, Ben-Azu B, Tremblay MÈ, Torre P, Senekal M, Laughton B, van der Kouwe A, Jankiewicz M, Kaba M, Holmes MJ. A review of the auditory-gut-brain axis. Front Neurosci 2023; 17:1183694. [PMID: 37600010 PMCID: PMC10435389 DOI: 10.3389/fnins.2023.1183694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Hearing loss places a substantial burden on medical resources across the world and impacts quality of life for those affected. Further, it can occur peripherally and/or centrally. With many possible causes of hearing loss, there is scope for investigating the underlying mechanisms involved. Various signaling pathways connecting gut microbes and the brain (the gut-brain axis) have been identified and well established in a variety of diseases and disorders. However, the role of these pathways in providing links to other parts of the body has not been explored in much depth. Therefore, the aim of this review is to explore potential underlying mechanisms that connect the auditory system to the gut-brain axis. Using select keywords in PubMed, and additional hand-searching in google scholar, relevant studies were identified. In this review we summarize the key players in the auditory-gut-brain axis under four subheadings: anatomical, extracellular, immune and dietary. Firstly, we identify important anatomical structures in the auditory-gut-brain axis, particularly highlighting a direct connection provided by the vagus nerve. Leading on from this we discuss several extracellular signaling pathways which might connect the ear, gut and brain. A link is established between inflammatory responses in the ear and gut microbiome-altering interventions, highlighting a contribution of the immune system. Finally, we discuss the contribution of diet to the auditory-gut-brain axis. Based on the reviewed literature, we propose numerous possible key players connecting the auditory system to the gut-brain axis. In the future, a more thorough investigation of these key players in animal models and human research may provide insight and assist in developing effective interventions for treating hearing loss.
Collapse
Affiliation(s)
- Amy S. Graham
- Imaging Sciences, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Peter Torre
- School of Speech, Language, and Hearing Sciences, San Diego State University, San Diego, CA, United States
| | - Marjanne Senekal
- Department of Human Biology, Division of Physiological Sciences, University of Cape Town, Cape Town, South Africa
| | - Barbara Laughton
- Family Clinical Research Unit, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Andre van der Kouwe
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
- Department of Radiology, Harvard Medical School, Boston, MA, United States
| | - Marcin Jankiewicz
- Imaging Sciences, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa
| | - Mamadou Kaba
- Department of Pathology, Division of Medical Microbiology, University of Cape Town, Cape Town, South Africa
| | - Martha J. Holmes
- Imaging Sciences, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- ImageTech, Simon Fraser University, Surrey, BC, Canada
| |
Collapse
|
34
|
Luo D, Li J, Liu H, Wang J, Xia Y, Qiu W, Wang N, Wang X, Wang X, Ma C, Ge W. Integrative Transcriptomic Analyses of Hippocampal-Entorhinal System Subfields Identify Key Regulators in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300876. [PMID: 37232225 PMCID: PMC10401097 DOI: 10.1002/advs.202300876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/15/2023] [Indexed: 05/27/2023]
Abstract
The hippocampal-entorhinal system supports cognitive function and is selectively vulnerable to Alzheimer's disease (AD). Little is known about global transcriptomic changes in the hippocampal-entorhinal subfields during AD. Herein, large-scale transcriptomic analysis is performed in five hippocampal-entorhinal subfields of postmortem brain tissues (262 unique samples). Differentially expressed genes are assessed across subfields and disease states, and integrated genotype data from an AD genome-wide association study. An integrative gene network analysis of bulk and single-nucleus RNA sequencing (snRNA-Seq) data identifies genes with causative roles in AD progression. Using a system-biology approach, pathology-specific expression patterns for cell types are demonstrated, notably upregulation of the A1-reactive astrocyte signature in the entorhinal cortex (EC) during AD. SnRNA-Seq data show that PSAP signaling is involved in alterations of cell- communications in the EC during AD. Further experiments validate the key role of PSAP in inducing astrogliosis and an A1-like reactive astrocyte phenotype. In summary, this study reveals subfield-, cell type-, and AD pathology-specific changes and demonstrates PSAP as a potential therapeutic target in AD.
Collapse
Affiliation(s)
- Dan Luo
- Department of ImmunologyState Key Laboratory of Complex Severe and Rare DiseasesInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
- Department of Human AnatomyHistology and EmbryologyNeuroscience CenterNational Human Brain Bank for Development and FunctionInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| | - Jingying Li
- Department of ImmunologyState Key Laboratory of Complex Severe and Rare DiseasesInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| | - Hanyou Liu
- Department of ImmunologyState Key Laboratory of Complex Severe and Rare DiseasesInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| | - Jiayu Wang
- Department of ImmunologyState Key Laboratory of Complex Severe and Rare DiseasesInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| | - Yu Xia
- Department of Human AnatomyHistology and EmbryologyNeuroscience CenterNational Human Brain Bank for Development and FunctionInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| | - Wenying Qiu
- Department of Human AnatomyHistology and EmbryologyNeuroscience CenterNational Human Brain Bank for Development and FunctionInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| | - Naili Wang
- Department of Human AnatomyHistology and EmbryologyNeuroscience CenterNational Human Brain Bank for Development and FunctionInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| | - Xue Wang
- Department of Human AnatomyHistology and EmbryologyNeuroscience CenterNational Human Brain Bank for Development and FunctionInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| | - Xia Wang
- Department of ImmunologyState Key Laboratory of Complex Severe and Rare DiseasesInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| | - Chao Ma
- Department of Human AnatomyHistology and EmbryologyNeuroscience CenterNational Human Brain Bank for Development and FunctionInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| | - Wei Ge
- Department of ImmunologyState Key Laboratory of Complex Severe and Rare DiseasesInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijing100005China
| |
Collapse
|
35
|
Liu J, Yu W, Ma C, Li T, Liang Y, Su S, Zhong G, Xie Z, Wu Q, Chen J, Wang Q. Network Pharmacology and Mechanism Studies of the Protective Effect of Ginseng against Alzheimer's Disease Based on Aβ Pathogenesis. PLANTA MEDICA 2023; 89:990-1000. [PMID: 36649733 DOI: 10.1055/a-2014-6061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Alzheimer's disease (AD) is a critical neurodegenerative disease that manifests as progressive intellectual decline and is pathologically characterized by a progressive loss of neurons in the brain. Despite extensive research on this topic, the pathogenesis of AD is not fully understood, while the beta-amyloid (Aβ) hypothesis remains the dominant one and only a few symptomatic drugs are approved for the treatment of AD. Ginseng has been widely reported as an effective herbal medicine for the treatment of neurodegenerative diseases such as dementia. Therefore, we explore the protective effects of ginseng in AD by a network pharmacological approach based on the pathogenesis of Aβ. Twenty-one major ginsenosides are screened based on ultraperformance liquid chromatography-mass spectrometry/mass spectrometry (HPLC-MS/MS) data. Among them, MAPK8, MAPK9, BACE1, FLT1, CDK2, and CCR5 are the core targets. By molecular docking and validation with the in vitro cell model APPswe-SH-SY5Y, we find that ginsenosides Rg3 and Ro have good neuroprotective effects and can reduce the expression of Aβ 1 - 42 in APPswe-SH-SY5Y. Finally, through RT-qPCR experiment, we find that ginsenoside Rg3 targeted MAPK8, FLT1, and CCR5, while ginsenoside Ro targeted MAPK8, MAPK9, FLT1, and CCR5 for its potential anti-AD efficacy.
Collapse
Affiliation(s)
- Jinman Liu
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, China
| | - Wenqian Yu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cuiru Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianyao Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shijie Su
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhouyuan Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiqing Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaxin Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
36
|
Kuo YY, Chen WT, Lin GB, Chen YM, Liu HH, Chao CY. Thermal cycling-hyperthermia ameliorates Aβ 25-35-induced cognitive impairment in C57BL/6 mice. Neurosci Lett 2023; 810:137337. [PMID: 37315732 DOI: 10.1016/j.neulet.2023.137337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023]
Abstract
Despite continuation of some controversies, Alzheimer's disease (AD), the most common cause of dementia nowadays, has been widely believed to derive mainly from excessive β-amyloid (Aβ) aggregation, that would increase reactive oxygen species (ROS) and induce neuroinflammation, leading to neuron loss and cognitive impairment. Existing drugs on Aβ have been ineffective or offer only temporary relief at best, due to blood-brain barrier or severe side effects. The study employed thermal cycling-hyperthermia (TC-HT) to ease the Aβ-induced cognitive impairments and compared its effect with continuous hyperthermia (HT) in vivo. It established an AD mice model via intracerebroventricular (i.c.v.) injection of Aβ25-35, proving that TC-HT is much more effective in alleviating its performance decline in Y-maze and novel object recognition (NOR) tests, in comparison with HT. In addition, TC-HT also exhibits a better performance in decreasing the hippocampal Aβ and β-secretase (BACE1) expressions as well as the neuroinflammation markers-ionized calcium-binding adapter molecule 1 (Iba-1) and glial fibrillary acidic protein (GFAP) levels. Furthermore, the study finds that TC-HT can elevate more protein expressions of insulin degrading enzyme (IDE) and antioxidative enzyme superoxide dismutase 2 (SOD2) than HT. In sum, the study proves the potential of TC-HT in AD treatment, which can be put into application with the use of focused ultrasound (FUS).
Collapse
Affiliation(s)
- Yu-Yi Kuo
- Department of Physics, Lab for Medical Physics & Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan; Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 10051, Taiwan
| | - Wei-Ting Chen
- Department of Physics, Lab for Medical Physics & Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan; Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 10051, Taiwan
| | - Guan-Bo Lin
- Department of Physics, Lab for Medical Physics & Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan; Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 10051, Taiwan
| | - You-Ming Chen
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 10051, Taiwan; Graduate Institute of Applied Physics, Biophysics Division, National Taiwan University, Taipei 10617, Taiwan
| | - Hsu-Hsiang Liu
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 10051, Taiwan; Graduate Institute of Applied Physics, Biophysics Division, National Taiwan University, Taipei 10617, Taiwan
| | - Chih-Yu Chao
- Department of Physics, Lab for Medical Physics & Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan; Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 10051, Taiwan; Graduate Institute of Applied Physics, Biophysics Division, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
37
|
Mantik KEK, Kim S, Gu B, Moon S, Kwak HB, Park DH, Kang JH. Repositioning of Anti-Diabetic Drugs against Dementia: Insight from Molecular Perspectives to Clinical Trials. Int J Mol Sci 2023; 24:11450. [PMID: 37511207 PMCID: PMC10380685 DOI: 10.3390/ijms241411450] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Insulin resistance as a hallmark of type 2 DM (T2DM) plays a role in dementia by promoting pathological lesions or enhancing the vulnerability of the brain. Numerous studies related to insulin/insulin-like growth factor 1 (IGF-1) signaling are linked with various types of dementia. Brain insulin resistance in dementia is linked to disturbances in Aβ production and clearance, Tau hyperphosphorylation, microglial activation causing increased neuroinflammation, and the breakdown of tight junctions in the blood-brain barrier (BBB). These mechanisms have been studied primarily in Alzheimer's disease (AD), but research on other forms of dementia like vascular dementia (VaD), Lewy body dementia (LBD), and frontotemporal dementia (FTD) has also explored overlapping mechanisms. Researchers are currently trying to repurpose anti-diabetic drugs to treat dementia, which are dominated by insulin sensitizers and insulin substrates. Although it seems promising and feasible, none of the trials have succeeded in ameliorating cognitive decline in late-onset dementia. We highlight the possibility of repositioning anti-diabetic drugs as a strategy for dementia therapy by reflecting on current and previous clinical trials. We also describe the molecular perspectives of various types of dementia through the insulin/IGF-1 signaling pathway.
Collapse
Affiliation(s)
- Keren Esther Kristina Mantik
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Sujin Kim
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Bonsang Gu
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Sohee Moon
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Hyo-Bum Kwak
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
- Department of Kinesiology, College of Arts and Sports, Inha University, Incheon 22212, Republic of Korea
| | - Dong-Ho Park
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
- Department of Kinesiology, College of Arts and Sports, Inha University, Incheon 22212, Republic of Korea
| | - Ju-Hee Kang
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
38
|
Yang G, Hu Y, Qin X, Sun J, Miao Z, Wang L, Ke Z, Zheng Y. Micheliolide attenuates neuroinflammation to improve cognitive impairment of Alzheimer's disease by inhibiting NF-κB and PI3K/Akt signaling pathways. Heliyon 2023; 9:e17848. [PMID: 37456020 PMCID: PMC10344752 DOI: 10.1016/j.heliyon.2023.e17848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 05/29/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023] Open
Abstract
Inflammatory reaction in the brain activates glial cells, and over-activated glial cells secrete inflammatory mediators, which aggravates the inflammatory response in the brain and accelerates the development of Alzheimer's disease (AD) in turn. Numerous natural compounds from herbs can alleviate inflammation, and it is very promising to find anti-neuroinflammatory natural compounds. Micheliolide (MCL) is an asesquiterpene lactone. Studies have proved that MCL showed an obvious anti-inflammatory property. Nevertheless, whether MCL can treat AD has not been determined. In this research, AD model mice were fed with a diet supplemented MCL for 3 months, the cognitive ability and inflammatory state of mice were detected. We found that MCL raised the frequency of touching novel objects, cut down the escape latency, raised the number of crossing platform, inhibited the infiltration of inflammatory cells and the secretion of interleukin-1α (IL-1α), IL-12p40, IL-13, IL-17A, tumor necrosis factor-α (TNF-α), granulocyte colony stimulating factor (G-CSF), macrophage inflammatory protein-1α (MIP-1α) and monocyte chemotactic protein-1 (MCP-1) in peripheral blood samples, inhibited the hyperplasia of glial cells and the production of IL-1α, IL-4, G-CSF, granulocyte-macrophage colony stimulating factor (GM-CSF), MIP-1α and MIP-1β, and reduced the deposition of Aβ peptides in the brain of AD mice. We also concluded that MCL dropped the expression of IL-1β, TNF-α, cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and the phosphorylation of IκB, p65 and Akt in BV-2 cells. In conclusion, MCL alleviates the intensity of systemic inflammatory reaction via inhibiting nuclear transcription factor κ gene binding (NF-κB) and phosphoinositide-3-kinase/serine/threonine kinase (PI3K/Akt) pathways in glial cells, and improves the cognitive impairment of AD mice. Therefore, MCL could be a therapeutic candidate for AD.
Collapse
Affiliation(s)
- Guizhen Yang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosafety, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Immunology and Microbiology, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - You Hu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosafety, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiangyang Qin
- Department of Chemistry, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Jinxia Sun
- Department of Immunology and Microbiology, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhulei Miao
- Department of Immunology and Microbiology, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lixin Wang
- Department of Immunology and Microbiology, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zunji Ke
- Institute of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosafety, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
39
|
Wu X, Li Z, Chen G, Yin Y, Chen CYC. Hybrid neural network approaches to predict drug-target binding affinity for drug repurposing: screening for potential leads for Alzheimer's disease. Front Mol Biosci 2023; 10:1227371. [PMID: 37441162 PMCID: PMC10334190 DOI: 10.3389/fmolb.2023.1227371] [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: 05/29/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that primarily affects elderly individuals. Recent studies have found that sigma-1 receptor (S1R) agonists can maintain endoplasmic reticulum stress homeostasis, reduce neuronal apoptosis, and enhance mitochondrial function and autophagy, making S1R a target for AD therapy. Traditional experimental methods are costly and inefficient, and rapid and accurate prediction methods need to be developed, while drug repurposing provides new ways and options for AD treatment. In this paper, we propose HNNDTA, a hybrid neural network for drug-target affinity (DTA) prediction, to facilitate drug repurposing for AD treatment. The study combines protein-protein interaction (PPI) network analysis, the HNNDTA model, and molecular docking to identify potential leads for AD. The HNNDTA model was constructed using 13 drug encoding networks and 9 target encoding networks with 2506 FDA-approved drugs as the candidate drug library for S1R and related proteins. Seven potential drugs were identified using network pharmacology and DTA prediction results of the HNNDTA model. Molecular docking simulations were further performed using the AutoDock Vina tool to screen haloperidol and bromperidol as lead compounds for AD treatment. Absorption, distribution, metabolism, excretion, and toxicity (ADMET) evaluation results indicated that both compounds had good pharmacokinetic properties and were virtually non-toxic. The study proposes a new approach to computer-aided drug design that is faster and more economical, and can improve hit rates for new drug compounds. The results of this study provide new lead compounds for AD treatment, which may be effective due to their multi-target action. HNNDTA is freely available at https://github.com/lizhj39/HNNDTA.
Collapse
Affiliation(s)
- Xialin Wu
- School of Computer Science and Technology, Guangdong University of Technology, Guangzhou, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhuojian Li
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Guanxing Chen
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Yiyang Yin
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Calvin Yu-Chian Chen
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| |
Collapse
|
40
|
Hu E, Li TS, Wineinger NE, Su AI. Association study between drug prescriptions and Alzheimer's disease claims in a commercial insurance database. Alzheimers Res Ther 2023; 15:118. [PMID: 37355615 PMCID: PMC10290352 DOI: 10.1186/s13195-023-01255-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 06/01/2023] [Indexed: 06/26/2023]
Abstract
In the ongoing effort to discover treatments for Alzheimer's disease (AD), there has been considerable focus on investigating the use of repurposed drug candidates. Mining of electronic health record data has the potential to identify novel correlated effects between commonly used drugs and AD. In this study, claims from members with commercial health insurance coverage were analyzed to determine the correlation between the use of various drugs on AD incidence and claim frequency. We found that, within the insured population, several medications for psychotic and mental illnesses were associated with higher disease incidence and frequency, while, to a lesser extent, antibiotics and anti-inflammatory drugs were associated with lower AD incidence rates. The observations thus provide a general overview of the prescription and claim relationships between various drug types and Alzheimer's disease, with insights into which drugs have possible implications on resulting AD diagnosis.
Collapse
Affiliation(s)
- Eric Hu
- Integrative Structural and Computational Biology, Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA 92037 USA
| | - Tong Shu Li
- Integrative Structural and Computational Biology, Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA 92037 USA
| | | | - Andrew I. Su
- Integrative Structural and Computational Biology, Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA 92037 USA
- Present Address: Scripps Research Translational Institute, La Jolla, CA 92037 USA
| |
Collapse
|
41
|
Boldin R, Zychar BC, Gonçalves LRC, Sciani JM. Design, in silico and pharmacological evaluation of a peptide inhibitor of BACE-1. Front Pharmacol 2023; 14:1184006. [PMID: 37397495 PMCID: PMC10313070 DOI: 10.3389/fphar.2023.1184006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction: Alzheimer's disease (AD) is the main type of dementia, caused by the accumulation of amyloid plaques, formed by amyloid peptides after being processed from amyloid precursor protein (APP) by γ- and ß-secretases (BACE-1). Although amyloid peptides have been well established for AD, they have been found in other neurodegenerative diseases, such as Parkinson's disease, Lewy body dementia, and amyotrophic lateral sclerosis. Inhibitors of BACE-1 have been searched and developed, but clinical trials failed due to lack of efficacy or toxicity. Nevertheless, it is still considered a good therapeutic target, as it was proven to remove amyloid peptides and improve memory. Methods: In this work, we designed a peptide based on a sequence obtained from the marine fish Merluccius productus and evaluated it by molecular docking to verify its binding to BACE-1, which was tested experimentally by enzymatic kinetics and cell culture assays. The peptide was injected in healthy mice to study its pharmacokinetics and toxicity. Results: We could obtain a new sequence in which the first N-terminal amino acids and the last one bound to the catalytic site of BACE-1 and showed high stability and hydrophobicity. The synthetic peptide showed a competitive inhibition of BACE-1 and Ki = 94 nM, and when injected in differentiated neurons, it could reduce Aβ42o production. In plasma, its half-life is ∼1 h, clearance is 0.0015 μg/L/h, and Vss is 0.0015 μg/L/h. The peptide was found in the spleen and liver 30 min after injection and reduced its level after that, when it was quantified in the kidneys, indicating its fast distribution and urinary excretion. Interestingly, the peptide was found in the brain 2 h after its administration. Histological analysis showed no morphological alteration in any organ, as well as the absence of inflammatory cells, indicating a lack of toxicity. Discussion: We obtained a new BACE-1 inhibitor peptide with fast distribution to the tissues, without accumulation in any organ, but found in the brain, with the possibility to reach its molecular target, BACE-1, contributing to the reduction in the amyloid peptide, which causes amyloid-linked neurodegenerative diseases.
Collapse
Affiliation(s)
- Renata Boldin
- Unidade de Farmacologia e Gastroenterologia (UNIFAG), Universidade São Francisco, Bragança Paulista, São Paulo, Brazil
- Laboratório de Farmacologia Molecular e Compostos Bioativos, Universidade São Francisco, Bragança Paulista, São Paulo, Brazil
| | | | | | - Juliana Mozer Sciani
- Laboratório de Farmacologia Molecular e Compostos Bioativos, Universidade São Francisco, Bragança Paulista, São Paulo, Brazil
| |
Collapse
|
42
|
Tang B, Wang Y, Ren J. Basic information about memantine and its treatment of Alzheimer's disease and other clinical applications. IBRAIN 2023; 9:340-348. [PMID: 37786758 PMCID: PMC10527776 DOI: 10.1002/ibra.12098] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 10/04/2023]
Abstract
Memantine is a noncompetitive moderate-affinity strong voltage-dependent N-methyl-D-aspartate receptor antagonist. It has been used to treat Alzheimer's disease (AD) since 1989. In 2018, it became the second most commonly used drug for the treatment of dementia in the world. AD is nonreversible, and memantine can only relieve the symptoms of AD but not cure it. Over the past half-century, memantine's research and clinical application have been extensively developed. In this review, the basic composition of memantine, the mechanism and limitations of memantine in the treatment of AD, memantine combination therapy, comparison of memantine with other drugs for AD, and clinical studies of memantine in other diseases are reviewed to provide a valuable reference for further research and application of memantine for the treatment of AD.
Collapse
Affiliation(s)
- Bin‐Can Tang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Ya‐Ting Wang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Jie Ren
- Department of NeuroscienceThe University of SheffieldSheffieldUK
| |
Collapse
|
43
|
Yang X, Tohda C. Diosgenin restores memory function via SPARC-driven axonal growth from the hippocampus to the PFC in Alzheimer's disease model mice. Mol Psychiatry 2023; 28:2398-2411. [PMID: 37085711 PMCID: PMC10611574 DOI: 10.1038/s41380-023-02052-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/23/2023]
Abstract
Central nervous system axons have minimal capacity to regenerate in adult brains, hindering memory recovery in Alzheimer's disease (AD). Although recent studies have shown that damaged axons sprouted in adult and AD mouse brains, long-distance axonal re-innervation to their targets has not been achieved. We selectively visualized axon-growing neurons in the neural circuit for memory formation, from the hippocampus to the prefrontal cortex, and showed that damaged axons successfully extended to their native projecting area in mouse models of AD (5XFAD) by administration of an axonal regenerative agent, diosgenin. In vivo transcriptome analysis detected the expression profile of axon-growing neurons directly isolated from the hippocampus of 5XFAD mice. Secreted protein acidic and rich in cysteine (SPARC) was the most expressed gene in axon-growing neurons. Neuron-specific overexpression of SPARC via adeno-associated virus serotype 9 delivery in the hippocampus recovered memory deficits and axonal projection to the prefrontal cortex in 5XFAD mice. DREADDs (Designer receptors exclusively activated by designer drugs) analyses revealed that SPARC overexpression-induced axonal growth in the 5XFAD mouse brain directly contributes to memory recovery. Elevated levels of SPARC on axonal membranes interact with extracellular rail-like collagen type I to promote axonal remodeling along their original tracings in primary cultured hippocampal neurons. These findings suggest that SPARC-driven axonal growth in the brain may be a promising therapeutic strategy for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ximeng Yang
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Chihiro Tohda
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
44
|
Pang B, Zhang LL, Li B, Sun FX, Wang ZD. The sodium glucose co-transporter 2 inhibitor ertugliflozin for Alzheimer's disease: Inhibition of brain insulin signaling disruption-induced tau hyperphosphorylation. Physiol Behav 2023; 263:114134. [PMID: 36809844 DOI: 10.1016/j.physbeh.2023.114134] [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: 12/07/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023]
Abstract
An antidiabetic agent sodium glucose co-transporter 2 (SGLT2) inhibitor ertugliflozin has been revealed to bind to catalytic anionic site of acetylcholinesterase (AChE), which is considered to be associated with the cognitive decline in neurodegenerative diseases, such as Alzheimer's disease (AD). The aim of the present study was thus to probe the effect of ertugliflozin on AD. Intracerebroventricular injection of streptozotocin (STZ/i.c.v) (3 mg/kg) was done bilaterally in male Wistar rats at 7-8 weeks of age. Two treatment doses (5 mg/kg and 10 mg/kg) of ertugliflozin were given intragastrically to STZ/i.c.v-induced rats for 20 days daily for behavioral assessment. Biochemical estimations of cholinergic activity, neuronal apoptosis, mitochondrial function and synaptic plasticity were performed. Behavioral results with ertugliflozin treatment revealed attenuation of cognitive deficit. Ertugliflozin also inhibited hippocampal AChE activity, downregulated pro-apoptotic marker expression, as well as mitigated mitochondrial dysfunction and synaptic damage in STZ/i.c.v rats. Importantly, we found that the hyperphosphorylation of tau in the hippocampus of STZ/i.c.v rats was decreased after oral administration of ertugliflozin, which was accompanied by decreased Phospho.IRS-1Ser307/Total.IRS-1 ratio and increased Phospho.AktSer473/Total.Akt and Phospho.GSK3βSer9/Total.GSK3β ratios. Our results indicated that treatment with ertugliflozin reversed AD pathology, which may be associated with inhibition of insulin signaling disruption-induced tau hyperphosphorylation.
Collapse
Affiliation(s)
- Bo Pang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China; Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300134, China
| | - Lu-Lu Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Bin Li
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China; Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300134, China
| | - Feng-Xian Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhi-Da Wang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China; Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300134, China.
| |
Collapse
|
45
|
Colin IM, Szczepanski LW, Gérard AC, Elosegi JA. Emerging Evidence for the Use of Antidiabetic Drugs, Glucagon-like Peptide 1 Receptor Agonists, for the Treatment of Alzheimer's Disease. TOUCHREVIEWS IN ENDOCRINOLOGY 2023; 19:16-24. [PMID: 37313236 PMCID: PMC10258618 DOI: 10.17925/ee.2023.19.1.16] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/05/2023] [Indexed: 06/15/2023]
Abstract
From an epidemiological and pathophysiological point of view, Alzheimer's disease (AD) and type 2 diabetes (T2DM) should be considered 'sister' diseases. T2DM significantly increases the risk of developing AD, and the mechanisms of neuronal degeneration themselves worsen peripheral glucose metabolism in multiple ways. The pathophysiological links between the two diseases, particularly cerebral insulin resistance, which causes neuronal degeneration, are so close that AD is sometimes referred to as 'type 3 diabetes'. Although the latest news on the therapeutic front for AD is encouraging, no treatment has been shown to halt disease progression permanently. At best, the treatments slow down the progression; at worst, they are inactive, or cause worrying side effects, preventing their use on a larger scale. Therefore, it appears logical that optimizing the metabolic milieu through preventive or curative measures can also slow down the cerebral degeneration that characterizes AD. Among the different classes of hypoglycaemic drugs, glucagon-like peptide 1 receptor agonists, which are widely used in the treatment of T2DM, were shown to slow down, or even prevent, neuronal degeneration. Data from animal, preclinical, clinical phase II, cohort and large cardiovascular outcomes studies are encouraging. Of course, randomized clinical phase III studies, which are on-going, will be essential to verify this hypothesis. Thus, for once, there is hope for slowing down the neurodegenerative processes associated with diabetes, and that hope is the focus of this review.
Collapse
Affiliation(s)
- Ides M Colin
- Endocrino-Diabetology Research Unit, Department of Internal Medicine, Centre Hospitalier Régional Mons-Hainaut/Groupe Jolimont, Mons Belgium/Groupe Helora, Mons, Belgium
| | - Lidia W Szczepanski
- Endocrino-Diabetology Research Unit, Department of Internal Medicine, Centre Hospitalier Régional Mons-Hainaut/Groupe Jolimont, Mons Belgium/Groupe Helora, Mons, Belgium
| | - Anne-Catherine Gérard
- Endocrino-Diabetology Research Unit, Department of Internal Medicine, Centre Hospitalier Régional Mons-Hainaut/Groupe Jolimont, Mons Belgium/Groupe Helora, Mons, Belgium
- Group of Animal Molecular and Cellular Biology, Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-La-Neuve, Belgium
| | - Jose-Antonio Elosegi
- Neurology Unit, Centre Hospitalier Universitaire Ambroise Paré, Mons Belgium/Groupe Helora, Mons, Belgium
| |
Collapse
|
46
|
Bhatti JS, Khullar N, Mishra J, Kaur S, Sehrawat A, Sharma E, Bhatti GK, Selman A, Reddy PH. Stem cells in the treatment of Alzheimer's disease - Promises and pitfalls. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166712. [PMID: 37030521 DOI: 10.1016/j.bbadis.2023.166712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/31/2023] [Indexed: 04/10/2023]
Abstract
Alzheimer's disease (AD) is the most widespread form of neurodegenerative disorder that causes memory loss and multiple cognitive issues. The underlying mechanisms of AD include the build-up of amyloid-β and phosphorylated tau, synaptic damage, elevated levels of microglia and astrocytes, abnormal microRNAs, mitochondrial dysfunction, hormonal imbalance, and age-related neuronal loss. However, the etiology of AD is complex and involves a multitude of environmental and genetic factors. Currently, available AD medications only alleviate symptoms and do not provide a permanent cure. Therefore, there is a need for therapies that can prevent or reverse cognitive decline, brain tissue loss, and neural instability. Stem cell therapy is a promising treatment for AD because stem cells possess the unique ability to differentiate into any type of cell and maintain their self-renewal. This article provides an overview of the pathophysiology of AD and existing pharmacological treatments. This review article focuses on the role of various types of stem cells in neuroregeneration, the potential challenges, and the future of stem cell-based therapies for AD, including nano delivery and gaps in stem cell technology.
Collapse
Affiliation(s)
- Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India
| | - Jayapriya Mishra
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Satinder Kaur
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Abhishek Sehrawat
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Eva Sharma
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - Ashley Selman
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| |
Collapse
|
47
|
Palma-Florez S, López-Canosa A, Moralez-Zavala F, Castaño O, Kogan MJ, Samitier J, Lagunas A, Mir M. BBB-on-a-chip with integrated micro-TEER for permeability evaluation of multi-functionalized gold nanorods against Alzheimer's disease. J Nanobiotechnology 2023; 21:115. [PMID: 36978078 PMCID: PMC10053726 DOI: 10.1186/s12951-023-01798-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/27/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND The lack of predictive models that mimic the blood-brain barrier (BBB) hinders the development of effective drugs for neurodegenerative diseases. Animal models behave differently from humans, are expensive and have ethical constraints. Organ-on-a-chip (OoC) platforms offer several advantages to resembling physiological and pathological conditions in a versatile, reproducible, and animal-free manner. In addition, OoC give us the possibility to incorporate sensors to determine cell culture features such as trans-endothelial electrical resistance (TEER). Here, we developed a BBB-on-a-chip (BBB-oC) platform with a TEER measurement system in close distance to the barrier used for the first time for the evaluation of the permeability performance of targeted gold nanorods for theranostics of Alzheimer's disease. GNR-PEG-Ang2/D1 is a therapeutic nanosystem previously developed by us consisting of gold nanorods (GNR) functionalized with polyethylene glycol (PEG), angiopep-2 peptide (Ang2) to overcome the BBB and the D1 peptide as beta amyloid fibrillation inhibitor, finally obtaining GNR-PEG-Ang2/D1 which showed to be useful for disaggregation of the amyloid in in vitro and in vivo models. In this work, we evaluated its cytotoxicity, permeability, and some indications of its impact on the brain endothelium by employing an animal-free device based on neurovascular human cells. RESULTS In this work, we fabricated a BBB-oC with human astrocytes, pericytes and endothelial cells and a TEER measuring system (TEER-BBB-oC) integrated at a micrometric distance of the endothelial barrier. The characterization displayed a neurovascular network and the expression of tight junctions in the endothelium. We produced GNR-PEG-Ang2/D1 and determined its non-cytotoxic range (0.05-0.4 nM) for plated cells included in the BBB-oC and confirmed its harmless effect at the highest concentration (0.4 nM) in the microfluidic device. The permeability assays revealed that GNR-PEG-Ang2/D1 cross the BBB and this entry is facilitated by Ang2 peptide. Parallel to the permeability analysis of GNR-PEG-Ang2/D1, an interesting behavior of the TJs expression was observed after its administration probably related to the ligands on the nanoparticle surface. CONCLUSIONS BBB-oC with a novel TEER integrated setup which allow a correct read-out and cell imaging monitoring was proven as a functional and throughput platform to evaluate the brain permeability performance of nanotherapeutics in a physiological environment with human cells, putting forward a viable alternative to animal experimentation.
Collapse
Affiliation(s)
- Sujey Palma-Florez
- Nanobioengineering group, Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Science and Technology (BIST), 12 Baldiri Reixac 15-21, 08028, Barcelona, Spain
| | - Adrián López-Canosa
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Science and Technology (BIST), 12 Baldiri Reixac 15-21, 08028, Barcelona, Spain
| | - Francisco Moralez-Zavala
- Department of Pharmacology and Toxicology, Faculty of Chemistry and Pharmaceutical Sciences, University of Chile, Santos Dumont 964, 8380494, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Oscar Castaño
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Science and Technology (BIST), 12 Baldiri Reixac 15-21, 08028, Barcelona, Spain
| | - Marcelo J Kogan
- Department of Pharmacology and Toxicology, Faculty of Chemistry and Pharmaceutical Sciences, University of Chile, Santos Dumont 964, 8380494, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Josep Samitier
- Nanobioengineering group, Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Science and Technology (BIST), 12 Baldiri Reixac 15-21, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
| | - Anna Lagunas
- Nanobioengineering group, Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Science and Technology (BIST), 12 Baldiri Reixac 15-21, 08028, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona, Spain.
| | - Mònica Mir
- Nanobioengineering group, Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Science and Technology (BIST), 12 Baldiri Reixac 15-21, 08028, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona, Spain.
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain.
| |
Collapse
|
48
|
Tomihari A, Kiyota M, Matsuura A, Itakura E. Alpha 2-macroglobulin acts as a clearance factor in the lysosomal degradation of extracellular misfolded proteins. Sci Rep 2023; 13:4680. [PMID: 36977730 PMCID: PMC10050189 DOI: 10.1038/s41598-023-31104-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Proteostasis regulates protein folding and degradation; its maintenance is essential for resistance to stress and aging. The loss of proteostasis is associated with many age-related diseases. Within the cell, molecular chaperones facilitate the refolding of misfolded proteins into their bioactive forms, thus preventing undesirable interactions and aggregation. Although the mechanisms of intracellular protein degradation pathways for intracellular misfolded proteins have been extensively studied, the protein degradation pathway for extracellular proteins remain poorly understood. In this study, we identified several misfolded proteins that are substrates for alpha 2-macroglobulin (α2M), an extracellular chaperone. We also established a lysosomal internalization assay for α2M, which revealed that α2M mediates the lysosomal degradation of extracellular misfolded proteins. Comparative analyses of α2M and clusterin, another extracellular chaperone, indicated that α2M preferentially targets aggregation-prone proteins. Thus, we present the degradation pathway of α2M, which interacts with aggregation-prone proteins for lysosomal degradation via selective internalization.
Collapse
Affiliation(s)
- Ayaka Tomihari
- Department of Biology, Graduate School of Science and Engineering, Chiba University, Inage-Ku, Chiba, 263-8522, Japan
| | - Mako Kiyota
- Department of Biology, Graduate School of Science and Engineering, Chiba University, Inage-Ku, Chiba, 263-8522, Japan
| | - Akira Matsuura
- Department of Biology, Graduate School of Science, Chiba University, Inage-Ku, Chiba, 263-8522, Japan
| | - Eisuke Itakura
- Department of Biology, Graduate School of Science, Chiba University, Inage-Ku, Chiba, 263-8522, Japan.
| |
Collapse
|
49
|
The Involvement of Post-Translational Modifications in Regulating the Development and Progression of Alzheimer's Disease. Mol Neurobiol 2023; 60:3617-3632. [PMID: 36877359 DOI: 10.1007/s12035-023-03277-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/16/2023] [Indexed: 03/07/2023]
Abstract
Post-translational modifications (PTMs) have been recently reported to be involved in the development and progression of Alzheimer's disease (AD). In detail, PTMs include phosphorylation, glycation, acetylation, sumoylation, ubiquitination, methylation, nitration, and truncation, which are associated with pathological functions of AD-related proteins, such as β-amyloid (Aβ), β-site APP-cleavage enzyme 1 (BACE1), and tau protein. In particular, the roles of aberrant PTMs in the trafficking, cleavage, and degradation of AD-associated proteins, leading to the cognitive decline of the disease, are summarized under AD conditions. By summarizing these research progress, the gaps will be filled between PMTs and AD, which will facilitate the discovery of potential biomarkers, leading to the establishment of novel clinical intervention methods against AD.
Collapse
|
50
|
Vieira ADC, Medeiros EB, Zabot GC, Pereira NDS, do Nascimento NB, Lidio AV, Scheffer ÂK, Rempel LCT, Macarini BMN, Costa MDA, Gonçalves CL, Kucharska E, Rodrigues MS, Moreira JCF, de Oliveira J, Budni J. Neuroprotective effects of combined therapy with memantine, donepezil, and vitamin D in ovariectomized female mice subjected to dementia model. Prog Neuropsychopharmacol Biol Psychiatry 2023; 122:110653. [PMID: 36195205 DOI: 10.1016/j.pnpbp.2022.110653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/24/2022]
Abstract
Women older than 60 have a higher risk of dementia, aging-related cognitive decline, and Alzheimer's Disease (AD) than the rest of the population. The main reason is hormonal senescence after menopause, a period characterized by a decline in estrogen levels. Since the effectiveness of drugs currently approved for the treatment of AD is limited, it is necessary to seek the development of new therapeutic strategies. Vitamin D deficiency is prevalent in AD patients and individuals with dementia in general. The supplementation of this vitamin in dementia patients might be an interesting approach for increasing the effectiveness of pre-existing medications for dementia treatment. Thus, the present study aims to investigate the effect of vitamin D treatment associated with memantine and donepezil in female mice submitted to ovariectomy (OVX) for five months and subjected to a dementia animal model induced by intracerebroventricular injection of aggregated amyloid βeta (Aβ1-42). For this purpose, Balb/c mice were divided into five experimental groups, which received 17 days of combined therapy with vitamin D, donepezil, and memantine. Then, animals were subjected to behavioral tests. OVX groups exhibited reduced levels of estradiol (E2) in serum, which was not altered by the combined therapy. Higher levels of vitamin D3 were found in the OVX animals submitted to the triple-association treatment. Mice exposed to both OVX and the dementia animal model presented impairment in short and long-term spatial and habituation memories. Also, female mice exposed to Aβ and OVX exhibited a reduction in brain-derived neurotrophic factor (BDNF) and interleukin-4 (IL-4) levels, and an increase in tumor necrose factor-α (TNFα) levels in the hippocampus. Besides, increased levels of IL-1β in the hippocampus and cerebral cortex were observed, as well as a significant increase in immunoreactivity for glial fibrillary acidic protein (GFAP), an astrocytes marker, in the hippocampus. Notably, triple-association treatment reversed the effects of the exposition of mice to Aβ and OVX in the long-term spatial and habituation memories impairment, as well as reversed changes in TNFα, IL-1β, IL-4, and GFAP immunoreactivity levels in the hippocampus of treated animals. Our results indicate that the therapeutic association of vitamin D, memantine, and donepezil has beneficial effects on memory performance and attenuated the neuroinflammatory response in female mice subjected to OVX associated with a dementia animal model.
Collapse
Affiliation(s)
- Ana Daniela Coutinho Vieira
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Eduarda Behenck Medeiros
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Gabriel Casagrande Zabot
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Nathalia de Souza Pereira
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Natália Baltazar do Nascimento
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Adrielly Vargas Lidio
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Ândrea Kohlrausch Scheffer
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Lisienny Campoli Tono Rempel
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | | | - Maiara de Aguiar Costa
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Cinara Ludvig Gonçalves
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Ewa Kucharska
- Akademia Ignatianum w Krakowie Wydział Pedagogiczny Instytut Nauk o Wychowaniu, Krakow, Poland
| | - Matheus Scarpatto Rodrigues
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - José Cláudio Fonseca Moreira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Jade de Oliveira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Josiane Budni
- Graduate Program in Health Sciences, Academic Unit of Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil.
| |
Collapse
|