1
|
Wehrli JM, Xia Y, Meister L, Tursunova S, Kleim B, Bach DR, Quednow BB. Forget me not: The effect of doxycycline on human declarative memory. Eur Neuropsychopharmacol 2024; 89:1-9. [PMID: 39217739 DOI: 10.1016/j.euroneuro.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Investigations into neuroprotective drugs are in high demand for the treatment of neurodegenerative diseases, such as multiple sclerosis or Alzheimer's disease, but also psychiatric disorders, such as depression, trauma, and substance use. One potential drug class being investigated are tetracyclines impacting on a variety of neuroprotective mechanisms. At the same time, tetracyclines like doxycycline have been suggested to affect human fear and spatial memory as well as reducing declarative memory retention. Based on the assumed necessity for synaptic consolidation in hippocampus-dependent learning, we hypothesised declarative memory may be similarly impaired by doxycycline as fear and spatial memory. Therefore, in this study we investigate the potential diminishing effects of doxycycline on consolidation of declarative memory in healthy humans. Additionally, to test for effect specificity we assessed motor memory, sustained attention, and processing speed. We administered a neuropsychological test battery in three independent randomized placebo-controlled double-blind trials (RCTs), in which healthy young volunteers (total N = 252) either received a single oral dose doxycycline (200 mg, n = 126) or placebo (n = 126) in a between-subject design. We found no evidence for a detrimental effect of doxycycline on declarative memory; instead, doxycycline improved declarative learning (p-value=0.022, Cohen's d=0.15) and memory consolidation (p=0.040, d=0.26). Contrarily, doxycycline slightly reduced motor learning (p=0.001, d=0.10) but subtly strengthened long-term motor memory (p=0.001, d=0.10). These results suggest that doxycycline can improve declarative learning and memory without having long term negative effects on other cognitive domains in healthy humans. Our results give hope to further investigate doxycycline in neuroprotective treatment applications.
Collapse
Affiliation(s)
- Jelena M Wehrli
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland.
| | - Yanfang Xia
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland
| | - Laura Meister
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland
| | - Sarrina Tursunova
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland
| | - Birgit Kleim
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland
| | - Dominik R Bach
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland; University of Bonn, Transdisciplinary Research Area "Life and Health", Hertz Chair for Artificial Intelligence and Neuroscience, Bonn, Germany
| | - Boris B Quednow
- Experimental Pharmacopsychology and Psychological Addiction Research, Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, Joint Center of University of Zurich and Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Song Z, Ho CT, Zhang X. Gut Microbiota Mediate the Neuroprotective Effect of Oolong Tea Polyphenols in Cognitive Impairment Induced by Circadian Rhythm Disorder. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12184-12197. [PMID: 38745351 DOI: 10.1021/acs.jafc.4c01922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Oolong tea polyphenols (OTP) have attracted wide attention due to their ability to reduce inflammatory response, regulate gut microbiota, and improve cognitive function. However, exactly how the gut microbiota modulates nervous system activity is still an open question. We previously expounded that supplementing with OTP alleviated neuroinflammation in circadian rhythm disorder (CRD) mice. Here, we showed that OTP can relieve microglia activation by reducing harmful microbial metabolites lipopolysaccharide (LPS) that alleviate CRD-induced cognitive decline. Mechanistically, OTP suppressed the inflammation response by regulating the gut microbiota composition, including upregulating the relative abundance of Muribaculaceae and Clostridia_UCG-014 and downregulating Desulfovibrio, promoting the production of short-chain fatty acids (SCFAs). Moreover, the use of OTP alleviated intestinal barrier damage and decreased the LPS transport to the serum. These results further inhibited the activation of microglia, thus alleviating cognitive impairment by inhibiting neuroinflammation, neuron damage, and neurotoxicity metabolite glutamate elevation. Meanwhile, OTP upregulated the expression of synaptic plasticity-related protein postsynaptic density protein 95 (PSD-95) and synaptophysin (SYN) by elevating the brain-derived neurotrophic factor (BDNF) level. Taken together, our findings suggest that the OTP has the potential to prevent CRD-induced cognition decline by modulating gut microbiota and microbial metabolites.
Collapse
Affiliation(s)
- Zheyi Song
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, P. R. China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, P. R. China
| |
Collapse
|
3
|
Kocanci FG, Goksu AY. Anti-inflammatory and antioxidative actions of tacrolimus (FK506) on human microglial HMC3 cell line. Scand J Immunol 2024; 99:e13339. [PMID: 38441214 DOI: 10.1111/sji.13339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/07/2023] [Accepted: 10/02/2023] [Indexed: 03/07/2024]
Abstract
Microglial cells are indispensable for the normal development and functioning of neurons in the central nervous system, where they play a crucial role in maintaining brain homeostasis by surveilling the microenvironment for signs of injury or stress and responding accordingly. However, in neurodegenerative diseases, the density and phenotypes of microglial cells undergo changes, leading to chronic activation and inflammation. Shifting the focus from neurons to microglia in drug discovery for neurodegenerative diseases has become an important therapeutic target. This study was aimed to investigate the potential of Tacrolimus (FK506) an FDA-approved calcineurin inhibitor, to modulate the pathology of neurodegenerative diseases through anti-inflammatory and antioxidative effects on microglial activation. The human microglia clone 3 (HMC3) cells were exposed to 1 μg/mL LPS in the presence and absence of doses of FK506. Survival rates of cells were determined using the 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) method. Morphological evaluation of cells showed that FK506 restored the normal morphology of activated microglia. Furthermore, FK506 treatment increases the total antioxidant capacity and reduces the total oxidative capacity, indicating its potential antioxidant effects. Data from ELISA and RT-PCR analyses showed that LPS abolished its promoting effects on the release of proinflammatory IL-1β and IL-6 cytokines in HMC3 cells, reflecting the anti-inflammatory effect of FK506. These findings support the idea that FK506 could be a promising therapeutic agent for neurodegenerative diseases by modulating microglial activation and reducing inflammation and oxidative stress.
Collapse
Affiliation(s)
- Fatma Gonca Kocanci
- Department of Medical Laboratory Techniques, Vocational High School of Health Services, Alanya Alaaddin Keykubat University, Alanya, Antalya, Turkey
| | - Azize Yasemin Goksu
- Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
4
|
Tang D, Sun C, Yang J, Fan L, Wang Y. Advances in the Study of the Pathology and Treatment of Alzheimer's Disease and Its Association with Periodontitis. Life (Basel) 2023; 13:2203. [PMID: 38004343 PMCID: PMC10672606 DOI: 10.3390/life13112203] [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: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's disease (AD) has become one of the leading causes of health problems in the elderly, and studying its causes and treatments remains a serious challenge for researchers worldwide. The two main pathological features of Alzheimer's disease are the extracellular deposition of β-amyloid (Aβ) to form senile plaques and the intracellular aggregation of hyperphosphorylated Tau protein to form neurofibrillary tangles (NFTs). Researchers have proposed several hypotheses to elucidate the pathogenesis of AD, but due to the complexity of the pathophysiologic factors involved in the development of AD, no effective drugs have been found to stop the progression of the disease. Currently, the mainstay drugs used to treat AD can only alleviate the patient's symptoms and do not have a therapeutic effect. As researchers explore interactions among diseases, much evidence suggests that there is a close link between periodontitis and AD, and that periodontal pathogenic bacteria can exacerbate Aβ deposition and Tau protein hyperphosphorylation through neuroinflammatory mechanisms, thereby advancing the pathogenesis of AD. This article reviews recent advances in the pathogenesis of AD, available therapeutic agents, the relevance of periodontitis to AD, and mechanisms of action.
Collapse
Affiliation(s)
- Dan Tang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China; (C.S.); (L.F.)
| | - Chang Sun
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China; (C.S.); (L.F.)
| | - Jumei Yang
- Lanzhou University Second Hospital, Lanzhou 730000, China;
| | - Lili Fan
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China; (C.S.); (L.F.)
| | - Yonggang Wang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China; (C.S.); (L.F.)
| |
Collapse
|
5
|
Boylan MA, Pincetic A, Romano G, Tatton N, Kenkare-Mitra S, Rosenthal A. Targeting Progranulin as an Immuno-Neurology Therapeutic Approach. Int J Mol Sci 2023; 24:15946. [PMID: 37958929 PMCID: PMC10647331 DOI: 10.3390/ijms242115946] [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: 09/19/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Immuno-neurology is an emerging therapeutic strategy for dementia and neurodegeneration designed to address immune surveillance failure in the brain. Microglia, as central nervous system (CNS)-resident myeloid cells, routinely perform surveillance of the brain and support neuronal function. Loss-of-function (LOF) mutations causing decreased levels of progranulin (PGRN), an immune regulatory protein, lead to dysfunctional microglia and are associated with multiple neurodegenerative diseases, including frontotemporal dementia caused by the progranulin gene (GRN) mutation (FTD-GRN), Alzheimer's disease (AD), Parkinson's disease (PD), limbic-predominant age-related transactivation response deoxyribonucleic acid binding protein 43 (TDP-43) encephalopathy (LATE), and amyotrophic lateral sclerosis (ALS). Immuno-neurology targets immune checkpoint-like proteins, offering the potential to convert aging and dysfunctional microglia into disease-fighting cells that counteract multiple disease pathologies, clear misfolded proteins and debris, promote myelin and synapse repair, optimize neuronal function, support astrocytes and oligodendrocytes, and maintain brain vasculature. Several clinical trials are underway to elevate PGRN levels as one strategy to modulate the function of microglia and counteract neurodegenerative changes associated with various disease states. If successful, these and other immuno-neurology drugs have the potential to revolutionize the treatment of neurodegenerative disorders by harnessing the brain's immune system and shifting it from an inflammatory/pathological state to an enhanced physiological/homeostatic state.
Collapse
Affiliation(s)
| | | | | | | | | | - Arnon Rosenthal
- Alector, Inc., 131 Oyster Point Blvd, Suite 600, South San Francisco, CA 94080, USA
| |
Collapse
|
6
|
Mamsa R, Prabhavalkar KS, Bhatt LK. Crosstalk between NLRP3 inflammasome and calpain in Alzheimer's disease. Eur J Neurosci 2023; 58:3719-3731. [PMID: 37652164 DOI: 10.1111/ejn.16139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 08/12/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
Amyloid plaques are considered to be the pathological hallmark of Alzheimer's disease (AD). Neuroinflammation further aggravates the pathogenesis of Alzheimer's disease. Calpains and NOD-like receptor protein-3 (NLRP3) inflammasomes are involved in the neuroinflammatory pathway and affect the progression of Alzheimer's disease. Hyperactivation of calpains is responsible for the activation of NLRP3 inflammasome, thereby affecting each other's molecular mechanism and causing astrogliosis, microgliosis, and neuronal dysfunction. Further, calpain hyperactivation is also associated with calcium homeostasis that acts as one of the triggers in the activation of NLRP3 inflammasome. Calpain activity is required for the maturation of interleukin-1β, a key mediator of neuroinflammatory responses. The membrane potential/calcium/calpain/caspase-1 axis acts as an unconventional regulator of inflammasomes. The complex crosstalk between NLRP3 inflammasome and calpain leads to a series of events. Targeting the molecular mechanism associated with calpain-NLRP3 inflammasome activation and regulation can be a therapeutic and prophylactic perspective towards Alzheimer's disease. This review discusses calpains and NLRP3 inflammasome crosstalk in the pathogenesis of AD.
Collapse
Affiliation(s)
- Rumaiza Mamsa
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Kedar S Prabhavalkar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
7
|
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
|
8
|
Chadarevian JP, Lombroso SI, Peet GC, Hasselmann J, Tu C, Marzan DE, Capocchi J, Purnell FS, Nemec KM, Lahian A, Escobar A, England W, Chaluvadi S, O'Brien CA, Yaqoob F, Aisenberg WH, Porras-Paniagua M, Bennett ML, Davtyan H, Spitale RC, Blurton-Jones M, Bennett FC. Engineering an inhibitor-resistant human CSF1R variant for microglia replacement. J Exp Med 2023; 220:e20220857. [PMID: 36584406 DOI: 10.1084/jem.20220857] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/11/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) can replace endogenous microglia with circulation-derived macrophages but has high mortality. To mitigate the risks of HSCT and expand the potential for microglia replacement, we engineered an inhibitor-resistant CSF1R that enables robust microglia replacement. A glycine to alanine substitution at position 795 of human CSF1R (G795A) confers resistance to multiple CSF1R inhibitors, including PLX3397 and PLX5622. Biochemical and cell-based assays show no discernable gain or loss of function. G795A- but not wildtype-CSF1R expressing macrophages efficiently engraft the brain of PLX3397-treated mice and persist after cessation of inhibitor treatment. To gauge translational potential, we CRISPR engineered human-induced pluripotent stem cell-derived microglia (iMG) to express G795A. Xenotransplantation studies demonstrate that G795A-iMG exhibit nearly identical gene expression to wildtype iMG, respond to inflammatory stimuli, and progressively expand in the presence of PLX3397, replacing endogenous microglia to fully occupy the brain. In sum, we engineered a human CSF1R variant that enables nontoxic, cell type, and tissue-specific replacement of microglia.
Collapse
Affiliation(s)
- Jean Paul Chadarevian
- Department of Neurobiology & Behavior, University of California, Irvine , Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Sonia I Lombroso
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
- Pharmacology Graduate Group, Biomedical Graduate Studies Program, University of Pennsylvania , Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania , Philadelphia, PA, USA
| | - Graham C Peet
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
- Neuroscience Graduate Program and Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus , Aurora, CO, USA
| | - Jonathan Hasselmann
- Department of Neurobiology & Behavior, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Christina Tu
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Dave E Marzan
- Department of Biology, The College of New Jersey , Ewing, NJ, USA
| | - Joia Capocchi
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
| | - Freddy S Purnell
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
| | - Kelsey M Nemec
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine , Philadelphia, PA, USA
| | - Alina Lahian
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Adrian Escobar
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Whitney England
- Department of Pharmaceutical Sciences, University of California, Irvine , Irvine, CA, USA
| | - Sai Chaluvadi
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine , Philadelphia, PA, USA
| | - Carleigh A O'Brien
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
| | - Fazeela Yaqoob
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
| | - William H Aisenberg
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
| | | | - Mariko L Bennett
- Department of Neuroscience, Perelman School of Medicine , Philadelphia, PA, USA
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia, PA, USA
| | - Hayk Davtyan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Robert C Spitale
- Department of Pharmaceutical Sciences, University of California, Irvine , Irvine, CA, USA
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, University of California, Irvine , Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia, PA, USA
| |
Collapse
|
9
|
Dhami M, Raj K, Singh S. Relevance of Gut Microbiota to Alzheimer's Disease (AD): Potential Effects of Probiotic in Management of AD. AGING AND HEALTH RESEARCH 2023. [DOI: 10.1016/j.ahr.2023.100128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
|
10
|
Wu Q, Su S, Cai C, Xu L, Fan X, Ke H, Dai Z, Fang S, Zhuo Y, Wang Q, Pan H, Gu Y, Fang J. Network Proximity-based computational pipeline identifies drug candidates for different pathological stages of Alzheimer's disease. Comput Struct Biotechnol J 2023; 21:1907-1920. [PMID: 36936813 PMCID: PMC10015208 DOI: 10.1016/j.csbj.2023.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
Despite the massive investment in Alzheimer's disease (AD), there are still no disease-modifying treatments (DMTs) for AD. One major reason is attributed to the limitation of clinical "one-size-fits-all" approach, since the same AD treatment solely based on clinical diagnosis was unlikely to achieve good clinical efficacy. In recent years, computational approaches based on multiomics data have provided an unprecedented opportunity for drug discovery since they can substantially lower the costs and boost the efficiency. In this study, we intended to identify potential drug candidates for different pathological stages of AD by computationally repurposing Food and Drug Administration (FDA) approved drugs. First, we assembled gene expression data from three different AD pathological stages, which include mild cognitive impairment (MCI) and early and late stages of AD (EAD, LAD). We next quantified the network distances between drug target networks and AD modules by utilizing a network proximity approach, and identified 193 candidates that possessed significant associations with AD. After searching for previous literature evidence, 63 out of 193 (32.6%) predicted drugs were demonstrated to exert therapeutic effects on AD. We further explored the novel mechanism of action (MOA) for these drug candidates by determining the specific brain cells they might function on based on AD patient single cell transcriptomic data. Additionally, we selected several promising candidates that could cross the blood brain barrier together with confirmed neuroprotective effects, and subsequently determined the antioxidative activity of these compounds. Experimental results showed that azathioprine decreased the reactive oxygen species (ROS) and malondialdehyde (MDA) levels and improved the superoxide dismutase (SOD) activity in APP-SH-SY5Y cells. Finally, we deciphered the potential MOA of azathioprine against AD via network analysis and validated several apoptosis-related proteins (Caspase 3, Cleaved Caspase 3, Bax, Bcl2) through western blotting. In summary, this study presented an effective computational strategy utilizing omics data for AD drug repurposing, which provides a new perspective for drug discovery and development.
Collapse
Affiliation(s)
- Qihui Wu
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Hainan Clinical Center for Encephalopathy of Chinese Medicine, Haikou, China
- Hainan Clinical Research Center for Preventive Treatment of Diseases, Haikou, China
| | - Shijie Su
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuipu Cai
- Department of Computer Science, Key Laboratory of Intelligent Manufacturing Technology of Ministry of Education, Shantou University, Shantou, China
| | - Lina Xu
- Department of Cardiac Surgery, Qingdao Fuwai Cardiovascular Hospital, Qingdao, China
| | - Xiude Fan
- Department of Infectious Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hanzhong Ke
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Zhao Dai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuhuan Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue Zhuo
- 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
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Gu
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, China
- Hainan Clinical Center for Encephalopathy of Chinese Medicine, Haikou, China
- Hainan Clinical Research Center for Preventive Treatment of Diseases, Haikou, China
- Corresponding author at: Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, China.
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Corresponding author.
| |
Collapse
|
11
|
Honkisz-Orzechowska E, Popiołek-Barczyk K, Linart Z, Filipek-Gorzała J, Rudnicka A, Siwek A, Werner T, Stark H, Chwastek J, Starowicz K, Kieć-Kononowicz K, Łażewska D. Anti-inflammatory effects of new human histamine H 3 receptor ligands with flavonoid structure on BV-2 neuroinflammation. Inflamm Res 2023; 72:181-194. [PMID: 36370200 PMCID: PMC9925557 DOI: 10.1007/s00011-022-01658-z] [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: 02/07/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Microglia play an important role in the neuroinflammation developed in response to various pathologies. In this study, we examined the anti-inflammatory effect of the new human histamine H3 receptor (H3R) ligands with flavonoid structure in murine microglial BV-2 cells. MATERIAL AND METHODS The affinity of flavonoids (E243 -flavone and IIIa-IIIc-chalcones) for human H3R was evaluated in the radioligand binding assay. The cytotoxicity on BV-2 cell viability was investigated with the MTS assay. Preliminary evaluation of anti-inflammatory properties was screened by the Griess assay in an in vitro neuroinflammation model of LPS-treated BV-2 cells. The expression and secretion of pro-inflammatory cytokines were evaluated by real-time qPCR and ELISA, respectively. The expression of microglial cell markers were determined by immunocytochemistry. RESULTS Chalcone derivatives showed high affinity at human H3R with Ki values < 25 nM. At the highest nontoxic concentration (6.25 μM) compound IIIc was the most active in reducing the level of nitrite in Griess assay. Additionally, IIIc treatment attenuated inflammatory process in murine microglia cells by down-regulating pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) at both the level of mRNA and protein level. Our immunocytochemistry studies revealed expression of microglial markers (Iba1, CD68, CD206) in BV-2 cell line. CONCLUSIONS These results emphasize the importance of further research to accurately identify the anti-inflammatory mechanism of action of chalcones.
Collapse
Affiliation(s)
- Ewelina Honkisz-Orzechowska
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College in Kraków, Medyczna 9, 30-688, Kraków, Poland.
| | - Katarzyna Popiołek-Barczyk
- grid.418903.70000 0001 2227 8271Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Zuzanna Linart
- grid.5522.00000 0001 2162 9631Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College in Kraków, Medyczna 9, 30-688 Kraków, Poland
| | - Jadwiga Filipek-Gorzała
- grid.5522.00000 0001 2162 9631Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College in Kraków, Medyczna 9, 30-688 Kraków, Poland
| | - Anna Rudnicka
- grid.5522.00000 0001 2162 9631Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College in Kraków, Medyczna 9, 30-688 Kraków, Poland
| | - Agata Siwek
- grid.5522.00000 0001 2162 9631Faculty of Pharmacy, Department of Pharmacobiology, Jagiellonian University Medical College in Kraków, Medyczna 9, 30-688 Kraków, Poland
| | - Tobias Werner
- grid.411327.20000 0001 2176 9917Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Duesseldorf, Universitaetsstr. 1, 40225 Duesseldorf, Germany
| | - Holger Stark
- grid.411327.20000 0001 2176 9917Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Duesseldorf, Universitaetsstr. 1, 40225 Duesseldorf, Germany
| | - Jakub Chwastek
- grid.418903.70000 0001 2227 8271Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Katarzyna Starowicz
- grid.418903.70000 0001 2227 8271Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Katarzyna Kieć-Kononowicz
- grid.5522.00000 0001 2162 9631Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College in Kraków, Medyczna 9, 30-688 Kraków, Poland
| | - Dorota Łażewska
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College in Kraków, Medyczna 9, 30-688, Kraków, Poland.
| |
Collapse
|
12
|
Wu YG, Song LJ, Yin LJ, Yin JJ, Wang Q, Yu JZ, Xiao BG, Ma CG. The effects and potential of microglial polarization and crosstalk with other cells of the central nervous system in the treatment of Alzheimer's disease. Neural Regen Res 2022; 18:947-954. [PMID: 36254973 PMCID: PMC9827789 DOI: 10.4103/1673-5374.355747] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Microglia are resident immune cells in the central nervous system. During the pathogenesis of Alzheimer's disease, stimulatory factors continuously act on the microglia causing abnormal activation and unbalanced phenotypic changes; these events have become a significant and promising area of research. In this review, we summarize the effects of microglial polarization and crosstalk with other cells in the central nervous system in the treatment of Alzheimer's disease. Our literature search found that phenotypic changes occur continuously in Alzheimer's disease and that microglia exhibit extensive crosstalk with astrocytes, oligodendrocytes, neurons, and penetrated peripheral innate immune cells via specific signaling pathways and cytokines. Collectively, unlike previous efforts to modulate microglial phenotypes at a single level, targeting the phenotypes of microglia and the crosstalk with other cells in the central nervous system may be more effective in reducing inflammation in the central nervous system in Alzheimer's disease. This would establish a theoretical basis for reducing neuronal death from central nervous system inflammation and provide an appropriate environment to promote neuronal regeneration in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Yi-Ge Wu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China,Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Li-Jun Yin
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
| | - Jun-Jun Yin
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China,Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
| | - Jie-Zhong Yu
- Institute of Brain Science/Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases/Medical School, Shanxi Datong University, Datong, Shanxi Province, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Cun-Gen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China,Institute of Brain Science/Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases/Medical School, Shanxi Datong University, Datong, Shanxi Province, China,Correspondence to: Cun-Gen Ma, .
| |
Collapse
|
13
|
Liu Q, Zhang J, Xiao C, Su D, Li L, Yang C, Zhao Z, Jiang W, You Z, Zhou T. Akebia saponin D protects hippocampal neurogenesis from microglia-mediated inflammation and ameliorates depressive-like behaviors and cognitive impairment in mice through the PI3K-Akt pathway. Front Pharmacol 2022; 13:927419. [PMID: 36110522 PMCID: PMC9468712 DOI: 10.3389/fphar.2022.927419] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Given the ability of akebia saponin D (ASD) to protect various types of stem cells, in the present study, we hypothesized that ASD could promote the proliferation, differentiation, and survival of neural stem/precursor cells (NSPCs), even in a microglia-mediated inflammatory environment, thereby mitigating inflammation-related neuropsychopathology. We established a mouse model of chronic neuroinflammation by exposing animals to low-dose lipopolysaccharide (LPS, 0.25 mg/kg/d) for 14 days. The results showed that chronic exposure to LPS strikingly reduced hippocampal levels of PI3K and pAkt and neurogenesis in mice. In the presen of a microglia-mediated inflammatory niche, the PI3K-Akt signaling in cultured NSPCs was inhibited, promoting their apoptosis and differentiation into astrocytes, while decreasing neurogenesis. Conversely, ASD strongly increased the levels of PI3K and pAkt and stimulated NSPC proliferation, survival and neuronal differentiation in the microglia-mediated inflammatory niche in vitro and in vivo. ASD also restored the synaptic function of hippocampal neurons and ameliorated depressive- and anxiety-like behaviors and cognitive impairment in mice chronically exposed to LPS. The results from network pharmacology analysis showed that the PI3K-AKT pathway is one of the targets of ASD to against major depressive disorder (MDD), anxiety and Alzheimer’s disease (AD). And the results from molecular docking based on computer modeling showed that ASD is bound to the interaction interface of the PI3K and AKT. The PI3K-Akt inhibitor LY294002 blocked the therapeutic effects of ASD in vitro and in vivo. These results suggested that ASD protects NSPCs from the microglia-mediated inflammatory niche, promoting their proliferation, survival and neuronal differentiation, as well as ameliorating depressive- and anxiety-like behaviors and cognitive impairment by activating the PI3K-AKT pathway. Our work suggests the potential of ASD for treating Alzheimer’s disease, depression and other cognitive disorders involving impaired neurogenesis by microglia-mediated inflammation.
Collapse
Affiliation(s)
- Qin Liu
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jinqiang Zhang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- *Correspondence: Jinqiang Zhang, Tao Zhou,
| | - Chenghong Xiao
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dapeng Su
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Liangyuan Li
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Changgui Yang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhihuang Zhao
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Weike Jiang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zili You
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Zhou
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- *Correspondence: Jinqiang Zhang, Tao Zhou,
| |
Collapse
|
14
|
Abu-Elfotuh K, Hussein FH, Abbas AN, Al-Rekabi MD, Barghash SS, Zaghlool SS, El-Emam SZ. Melatonin and zinc supplements with physical and mental activities subside neurodegeneration and hepatorenal injury induced by aluminum chloride in rats: Inclusion of GSK-3β-Wnt/β-catenin signaling pathway. Neurotoxicology 2022; 91:69-83. [DOI: 10.1016/j.neuro.2022.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/17/2022] [Accepted: 05/02/2022] [Indexed: 12/14/2022]
|
15
|
Biosensor based on coupled enzyme reactions for determination of arginase activity. Bioelectrochemistry 2022; 146:108137. [DOI: 10.1016/j.bioelechem.2022.108137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/04/2022] [Accepted: 04/18/2022] [Indexed: 11/21/2022]
|
16
|
Rapaka D, Bitra VR, Challa SR, Adiukwu PC. Potentiation of microglial endocannabinoid signaling alleviates neuroinflammation in Alzheimer's disease. Neuropeptides 2021; 90:102196. [PMID: 34508923 DOI: 10.1016/j.npep.2021.102196] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) isaprogressive neurodegenerative disorder characterized by chronic inflammation due to the presence of neurotoxic Aβ and tau proteins. Increased microglial activation and inflated immune response are the other factors to be considered in AD pathology. Microglial cells own biochemical machinery that synthesizes and release endocannabinoids. The exploitation of therapeutic actions of endocannabinoid system has newly emerged in the field of Alzheimer's disease. The activation of cannabinoid receptors/ cannabinoid system modulates inflammatory responses. This review assesses the association between the microglial endocannabinoid system and neuroinflammation in AD. The data supporting the anti-inflammatory role of pharmacological agents modulating cannabinoid system are also reviewed.
Collapse
Affiliation(s)
- Deepthi Rapaka
- A.U. College of Pharmaceutical Sciences, Andhra University, Visakhapatnam 530003, India.
| | | | - Siva Reddy Challa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL 61614, USA
| | - Paul C Adiukwu
- School of Pharmacy, University of Botswana, P/Bag-0022, Gaborone, Botswana
| |
Collapse
|
17
|
Md S, Alhakamy NA, Alfaleh MA, Afzal O, Altamimi ASA, Iqubal A, Shaik RA. Mechanisms Involved in Microglial-Interceded Alzheimer's Disease and Nanocarrier-Based Treatment Approaches. J Pers Med 2021; 11:1116. [PMID: 34834468 PMCID: PMC8619529 DOI: 10.3390/jpm11111116] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder accountable for dementia and cognitive dysfunction. The etiology of AD is complex and multifactorial in origin. The formation and deposition of amyloid-beta (Aβ), hyperphosphorylated tau protein, neuroinflammation, persistent oxidative stress, and alteration in signaling pathways have been extensively explored among the various etiological hallmarks. However, more recently, the immunogenic regulation of AD has been identified, and macroglial activation is considered a limiting factor in its etiological cascade. Macroglial activation causes neuroinflammation via modulation of the NLRP3/NF-kB/p38 MAPKs pathway and is also involved in tau pathology via modulation of the GSK-3β/p38 MAPK pathways. Additionally, microglial activation contributes to the discrete release of neurotransmitters and an altered neuronal synaptic plasticity. Therefore, activated microglial cells appear to be an emerging target for managing and treating AD. This review article discussed the pathology of microglial activation in AD and the role of various nanocarrier-based anti-Alzeihmenr's therapeutic approaches that can either reverse or inhibit this activation. Thus, as a targeted drug delivery system, nanocarrier approaches could emerge as a novel means to overcome existing AD therapy limitations.
Collapse
Affiliation(s)
- Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed A. Alfaleh
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.)
| | - Abdulmalik S. A. Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.)
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
| | - Rasheed A. Shaik
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| |
Collapse
|