1
|
Messedi M, Makni-Ayadi F. 24S-Hydroxycholesterol in Neuropsychiatric Diseases: Schizophrenia, Autism Spectrum Disorder, and Bipolar Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:293-304. [PMID: 38036886 DOI: 10.1007/978-3-031-43883-7_15] [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: 12/02/2023]
Abstract
Neuropsychiatric diseases (NPDs) are severe, debilitating psychiatric conditions that affect the nervous system. These are among the most challenging disorders in medicine. Some examples include Alzheimer's, anxiety disorders, autism spectrum disorder, bipolar disorder, and schizophrenia. NPDs represent an ever-increasing burden on public health and are prevalent throughout the world. For most of these diseases, the particular etiopathogeneses are still enigmatic. NPDs are also associated with structural and functional changes in the brain, along with altered neurotransmitter and neuroendocrine systems.Approximately 25% of the total human body cholesterol is located in the brain. Its involvement in neuronal functions starts in the early growth stages and remains important throughout adulthood. It is also an integral part of the neuronal membrane, ensuring membrane lipid organization and regulating membrane fluidity. The main mechanism for removing cholesterol from the brain is cholesterol 24-hydroxylation by cytochrome P450 46A1 (CYP46A1), an enzyme specifically found in the central nervous system. Although research on 24S-OHC and its role in neuropsychiatric diseases is still in its early stages, this oxidized cholesterol metabolite is thought to play a crucial role in the etiology of NPDs. 24S-OHC can affect neurons, astrocytes, oligodendrocytes, and vascular cells. In addition to regulating the homeostasis of cholesterol in the brain, this oxysterol is involved in neurotransmission, oxidative stress, and inflammation. The role of 24S-OHC in NPDs has been found to be controversial in terms of the findings so far. There are several intriguing discrepancies in the data gathered so far regarding 24S-OHC and NPDs. In fact, 24S-OHC levels were reported to have decreased in a number of NPDs and increased in others.Hence, in this chapter, we first summarize the available data regarding 24S-OHC as a biomarker in NPDs, including schizophrenia, autism spectrum disorder, and bipolar disorder. Then, we present a brief synopsis of the pharmacological targeting of 24S-OHC levels through the modulation of CYP46A1 activity.
Collapse
Affiliation(s)
- Meriam Messedi
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Sfax, Tunisia
| | - Fatma Makni-Ayadi
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Sfax, Tunisia
- Department of Clinical biochemistry, Habib Bourguiba Hospital, Sfax, Tunisia
| |
Collapse
|
2
|
Al-Kuraishy HM, Jabir MS, Al-Gareeb AI, Albuhadily AK, Albukhaty S, Sulaiman GM, Batiha GES. Evaluation and targeting of amyloid precursor protein (APP)/amyloid beta (Aβ) axis in amyloidogenic and non-amyloidogenic pathways: A time outside the tunnel. Ageing Res Rev 2023; 92:102119. [PMID: 37931848 DOI: 10.1016/j.arr.2023.102119] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
In Alzheimer disease (AD), amyloid precursor protein (APP) and production of amyloid beta (Aβ) which is generated by amyloidogenic pathway is implicated in neurotoxicity and neuronal cell deaths. However, physiological Aβ level is essential to improves neuronal survival, attenuates neuronal apoptosis and has neuroprotective effect. In addition, physiological APP level has neurotrophic effect on the central nervous system (CNS). APP has a critical role in the brain growth and development via activation of long-term potentiation (LTP) and acceleration of neurite outgrowth. Moreover, APP is cleaved by α secretase to form a neuroprotective soluble APP alpha (sAPPα) in non-amyloidogenic pathway. Consequently, this mini-review purposes to highlight the possible beneficial role of APP and Aβ. In addition, this mini-review discussed the modulation of APP processing and Aβ production.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Majid S Jabir
- Department of Applied science, University of Technology, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali K Albuhadily
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Salim Albukhaty
- Department of Chemistry, College of Science, University of Misan, Maysan 62001, Iraq
| | | | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511, Egypt
| |
Collapse
|
3
|
Sokol DK, Lahiri DK. Neurodevelopmental disorders and microcephaly: how apoptosis, the cell cycle, tau and amyloid-β precursor protein APPly. Front Mol Neurosci 2023; 16:1201723. [PMID: 37808474 PMCID: PMC10556256 DOI: 10.3389/fnmol.2023.1201723] [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: 04/07/2023] [Accepted: 08/08/2023] [Indexed: 10/10/2023] Open
Abstract
Recent studies promote new interest in the intersectionality between autism spectrum disorder (ASD) and Alzheimer's Disease. We have reported high levels of Amyloid-β Precursor Protein (APP) and secreted APP-alpha (sAPPa ) and low levels of amyloid-beta (Aβ) peptides 1-40 and 1-42 (Aβ40, Aβ42) in plasma and brain tissue from children with ASD. A higher incidence of microcephaly (head circumference less than the 3rd percentile) associates with ASD compared to head size in individuals with typical development. The role of Aβ peptides as contributors to acquired microcephaly in ASD is proposed. Aβ may lead to microcephaly via disruption of neurogenesis, elongation of the G1/S cell cycle, and arrested cell cycle promoting apoptosis. As the APP gene exists on Chromosome 21, excess Aβ peptides occur in Trisomy 21-T21 (Down's Syndrome). Microcephaly and some forms of ASD associate with T21, and therefore potential mechanisms underlying these associations will be examined in this review. Aβ peptides' role in other neurodevelopmental disorders that feature ASD and acquired microcephaly are reviewed, including dup 15q11.2-q13, Angelman and Rett syndrome.
Collapse
Affiliation(s)
- Deborah K. Sokol
- Section of Pediatrics, Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Debomoy K. Lahiri
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
4
|
Abu-Elfotuh K, Selim HMRM, Riad OKM, Hamdan AME, Hassanin SO, Sharif AF, Moustafa NM, Gowifel AM, Mohamed MYA, Atwa AM, Zaghlool SS, El-Din MN. The protective effects of sesamol and/or the probiotic, Lactobacillus rhamnosus, against aluminum chloride-induced neurotoxicity and hepatotoxicity in rats: Modulation of Wnt/β-catenin/GSK-3β, JAK-2/STAT-3, PPAR-γ, inflammatory, and apoptotic pathways. Front Pharmacol 2023; 14:1208252. [PMID: 37601053 PMCID: PMC10436218 DOI: 10.3389/fphar.2023.1208252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/03/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction: Aluminium (Al) is accumulated in the brain causing neurotoxicity and neurodegenerative disease like Alzheimer's disease (AD), multiple sclerosis, autism and epilepsy. Hence, attenuation of Al-induced neurotoxicity has become a "hot topic" in looking for an intervention that slow down the progression of neurodegenerative diseases. Objective: Our study aims to introduce a new strategy for hampering aluminum chloride (AlCl3)-induced neurotoxicity using a combination of sesamol with the probiotic bacteria; Lactobacillus rhamnosus (L. rhamnosus) and also to test their possible ameliorative effects on AlCl3-induced hepatotoxicity. Methods: Sprague-Dawley male rats were randomly divided into five groups (n = 10/group) which are control, AlCl3, AlCl3 + Sesamol, AlCl3 + L. rhamnosus and AlCl3 + Sesamol + L. rhamnosus. We surveilled the behavioral, biochemical, and histopathological alterations centrally in the brain and peripherally in liver. Results: This work revealed that the combined therapy of sesamol and L. rhamnosus produced marked reduction in brain amyloid-β, p-tau, GSK-3β, inflammatory and apoptotic biomarkers, along with marked elevation in brain free β-catenin and Wnt3a, compared to AlCl3-intoxicated rats. Also, the combined therapy exerted pronounced reduction in hepatic expressions of JAK-2/STAT-3, inflammatory (TNF-α, IL-6, NF-κB), fibrotic (MMP-2, TIMP-1, α-SMA) and apoptotic markers, (caspase-3), together with marked elevation in hepatic PPAR-γ expression, compared to AlCl3 -intoxicated rats. Behavioral and histopathological assessments substantiated the efficiency of this combined regimen in halting the effect of neurotoxicity. Discussion: Probiotics can be used as an add-on therapy with sesamol ameliorate AlCl3 -mediated neurotoxicity and hepatotoxicity.
Collapse
Affiliation(s)
- Karema Abu-Elfotuh
- Clinical Pharmacy Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Heba Mohammed Refat M. Selim
- Pharmaceutical Sciences Department, Faculty of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
- Microbiology and Immunology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Omnia Karem M. Riad
- Microbiology and Immunology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Ahmed M. E. Hamdan
- Pharmacy Practice Department, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Soha Osama Hassanin
- Biochemistry Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Asmaa F. Sharif
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
- Clinical Medical Sciences Department, College of Medicine, Dar Al Uloom University, Riyadh, Saudi Arabia
| | - Nouran Magdy Moustafa
- Basic Medical Science Department, College of Medicine, Dar Al Uloom University, Riyadh, Saudi Arabia
- Medical Microbiology and Immunology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ayah M.H. Gowifel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Marwa Y. A. Mohamed
- Biology Department, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Ahmed M. Atwa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Sameh S. Zaghlool
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Mahmoud Nour El-Din
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City (USC), Menoufia, Egypt
| |
Collapse
|
5
|
Mohammadian Rasnani F, Zavieh A, Heidari A, Motamed M. From neurodevelopmental to neurodegenerative disorders: Investigating symptoms of attention deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) in patients with dementia. APPLIED NEUROPSYCHOLOGY. ADULT 2023:1-10. [PMID: 37410670 DOI: 10.1080/23279095.2023.2230507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Dementia is characterized by a progressive cognitive decline that could be caused by several disorders. Autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD) are two prevalent neurodevelopmental disorders that might overlap with dementia symptoms. Hence, this study aimed to evaluate the ASD and ADHD symptoms in dementia patients referred to a memory clinic in Iran. We recruited 65 dementia patients and instructed them to fill out the autism quotient (AQ) and the Conners' Adult ADHD Rating Scales (CAARS) questionnaires. Considering the cutoff points of AQ and CAARS questionnaires, 18.5% of participants were at higher risk of ASD, and 35.4% were at higher risk of ADHD. The results indicated that ADHD and ASD symptoms might be common comorbidities in patients with dementia which can increase the disease burden. Specialized ADHD and ASD screening tools in the elderly population with dementia are needed to prevent misdiagnoses due to symptom overlaps.
Collapse
Affiliation(s)
| | - Amir Zavieh
- Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Heidari
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahtab Motamed
- Department of Psychiatry, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Cencelli G, Pacini L, De Luca A, Messia I, Gentile A, Kang Y, Nobile V, Tabolacci E, Jin P, Farace MG, Bagni C. Age-Dependent Dysregulation of APP in Neuronal and Skin Cells from Fragile X Individuals. Cells 2023; 12:758. [PMID: 36899894 PMCID: PMC10000963 DOI: 10.3390/cells12050758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 03/04/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common form of monogenic intellectual disability and autism, caused by the absence of the functional fragile X messenger ribonucleoprotein 1 (FMRP). FXS features include increased and dysregulated protein synthesis, observed in both murine and human cells. Altered processing of the amyloid precursor protein (APP), consisting of an excess of soluble APPα (sAPPα), may contribute to this molecular phenotype in mice and human fibroblasts. Here we show an age-dependent dysregulation of APP processing in fibroblasts from FXS individuals, human neural precursor cells derived from induced pluripotent stem cells (iPSCs), and forebrain organoids. Moreover, FXS fibroblasts treated with a cell-permeable peptide that decreases the generation of sAPPα show restored levels of protein synthesis. Our findings suggest the possibility of using cell-based permeable peptides as a future therapeutic approach for FXS during a defined developmental window.
Collapse
Affiliation(s)
- Giulia Cencelli
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Catholic University, 00168 Rome, Italy
| | - Laura Pacini
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Faculty of Medicine, UniCamillus, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
| | - Anastasia De Luca
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ilenia Messia
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonietta Gentile
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Roma, 00166 Rome, Italy
| | - Yunhee Kang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Veronica Nobile
- Institute of Genomic Medicine, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Catholic University, 00168 Rome, Italy
| | - Elisabetta Tabolacci
- Institute of Genomic Medicine, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Catholic University, 00168 Rome, Italy
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Maria Giulia Farace
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
7
|
Kong L, Zhang D, Huang S, Lai J, Lu L, Zhang J, Hu S. Extracellular Vesicles in Mental Disorders: A State-of-art Review. Int J Biol Sci 2023; 19:1094-1109. [PMID: 36923936 PMCID: PMC10008693 DOI: 10.7150/ijbs.79666] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/26/2023] [Indexed: 03/13/2023] Open
Abstract
Extracellular vesicles (EVs) are nanoscale particles with various physiological functions including mediating cellular communication in the central nervous system (CNS), which indicates a linkage between these particles and mental disorders such as schizophrenia, bipolar disorder, major depressive disorder, etc. To date, known characteristics of mental disorders are mainly neuroinflammation and dysfunctions of homeostasis in the CNS, and EVs are proven to be able to regulate these pathological processes. In addition, studies have found that some cargo of EVs, especially miRNAs, were significantly up- or down-regulated in patients with mental disorders. For many years, interest has been generated in exploring new diagnostic and therapeutic methods for mental disorders, but scale assessment and routine drug intervention are still the first-line applications so far. Therefore, underlying the downstream functions of EVs and their cargo may help uncover the pathogenetic mechanisms of mental disorders as well as provide novel biomarkers and therapeutic candidates. This review aims to address the connection between EVs and mental disorders, and discuss the current strategies that focus on EVs-related psychiatric detection and therapy.
Collapse
Affiliation(s)
- Lingzhuo Kong
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Danhua Zhang
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shu Huang
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jianbo Lai
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.,The Key Laboratory of Mental Disorder's Management in Zhejiang Province, Hangzhou 310003, China.,Brain Research Institute of Zhejiang University, Hangzhou 310003, China.,Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou 310003, China.,Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Jing Zhang
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China.,National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Zhejiang, China
| | - Shaohua Hu
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.,The Key Laboratory of Mental Disorder's Management in Zhejiang Province, Hangzhou 310003, China.,Brain Research Institute of Zhejiang University, Hangzhou 310003, China.,Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou 310003, China.,Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
8
|
Wang Q, Song LJ, Ding ZB, Chai Z, Yu JZ, Xiao BG, Ma CG. Advantages of Rho-associated kinases and their inhibitor fasudil for the treatment of neurodegenerative diseases. Neural Regen Res 2022; 17:2623-2631. [PMID: 35662192 PMCID: PMC9165373 DOI: 10.4103/1673-5374.335827] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/01/2021] [Accepted: 11/06/2021] [Indexed: 11/20/2022] Open
Abstract
Ras homolog (Rho)-associated kinases (ROCKs) belong to the serine-threonine kinase family, which plays a pivotal role in regulating the damage, survival, axon guidance, and regeneration of neurons. ROCKs are also involved in the biological effects of immune cells and glial cells, as well as the development of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Previous studies by us and others confirmed that ROCKs inhibitors attenuated the symptoms and progression of experimental models of the abovementioned neurodegenerative diseases by inhibiting neuroinflammation, regulating immune imbalance, repairing the blood-brain barrier, and promoting nerve repair and myelin regeneration. Fasudil, the first ROCKs inhibitor to be used clinically, has a good therapeutic effect on neurodegenerative diseases. Fasudil increases the activity of neural stem cells and mesenchymal stem cells, thus optimizing cell therapy. This review will systematically describe, for the first time, the effects of abnormal activation of ROCKs on T cells, B cells, microglia, astrocytes, oligodendrocytes, and pericytes in neurodegenerative diseases of the central nervous system, summarize the therapeutic potential of fasudil in several experimental models of neurodegenerative diseases, and clarify the possible cellular and molecular mechanisms of ROCKs inhibition. This review also proposes that fasudil is a novel potential treatment, especially in combination with cell-based therapy. Findings from this review add support for further investigation of ROCKs and its inhibitor fasudil for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Qing Wang
- Research Center of Neurobiology, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
| | - Li-Juan Song
- Research Center of Neurobiology, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Zhi-Bin Ding
- Research Center of Neurobiology, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
| | - Zhi Chai
- Research Center of Neurobiology, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
| | - Jie-Zhong Yu
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Shanxi Datong University, Datong, Shanxi Province, China
- Department of Neurology, Datong Fifth People’s Hospital, Datong, Shanxi Province, China
| | - Bao-Guo Xiao
- Research Center of Neurobiology, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Cun-Gen Ma
- Research Center of Neurobiology, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Shanxi Datong University, Datong, Shanxi Province, China
| |
Collapse
|
9
|
Rhodus EK, Barber J, Kryscio RJ, Abner EL, Bahrani AA, Lewis KES, Carey B, Nelson PT, Van Eldik LJ, Jicha GA. Frontotemporal neurofibrillary tangles and cerebrovascular lesions are associated with autism spectrum behaviors in late-life dementia. J Neurol 2022; 269:5105-5113. [PMID: 35596794 PMCID: PMC9644295 DOI: 10.1007/s00415-022-11167-y] [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: 10/13/2021] [Revised: 04/29/2022] [Accepted: 05/01/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND OBJECTIVES The pathologic substrates or neuroanatomic regions responsible for similarities in behavioral features seen in autism spectrum disorder and late-life dementia remain unknown. The present study examined the neuropathologic features of late-life dementia in research volunteers with and without antemortem behaviors characteristic of autism spectrum disorders. METHODS Antemortem cross-sectional assessment of autistic spectrum behaviors proximal to death in persons with diagnosis of mild cognitive impairment or dementia was completed using the Gilliam Autism Rating Scale, 2nd edition (GARS-2), followed by postmortem quantitative and semiquantitative neuropathologic assessment. All individuals who completed the GARS-2 prior to autopsy were included (n = 56) and we note that no participants had known diagnosis of autism spectrum disorder. The GARS-2 was used as an antemortem screening tool to stratify participants into two groups: "Autism Possible/Very Likely" or "Autism Unlikely." Data were analyzed using nonparametric statistics comparing location and scale to evaluate between-group differences in pathologic features. RESULTS Neurofibrillary tangles (NFT; p = 0.028) density and tau burden (p = 0.032) in the frontal region, the NFT density (p = 0.048) and neuritic plaque burden (p = 0.042), and the tau burden (p = 0.032) of the temporal region, were significantly different in scale between groups. For measures with significant group differences, the medians of the Autism Possible/Very Likely group were roughly equal to the 75th percentile of the Autism Unlikely group (i.e., the distributions were shifted to the right). DISCUSSION This study links behaviors characteristic of autism to increased pathologic tau burden in the frontal and temporal lobes in persons with late-life dementia. Additional studies are needed to determine causal factors and treatment options for behaviors characteristic of autism behaviors in late-life dementias.
Collapse
Affiliation(s)
- Elizabeth K Rhodus
- Sanders-Brown Center on Aging, University of Kentucky, 1030 S. Broadway, Ste 5, Lexington, KY, 40536, USA.
- Department of Behavioral Science, University of Kentucky, Lexington, KY, USA.
| | - Justin Barber
- Sanders-Brown Center on Aging, University of Kentucky, 1030 S. Broadway, Ste 5, Lexington, KY, 40536, USA
| | - Richard J Kryscio
- Sanders-Brown Center on Aging, University of Kentucky, 1030 S. Broadway, Ste 5, Lexington, KY, 40536, USA
- Department of Statistics, University of Kentucky, Lexington, KY, USA
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, University of Kentucky, 1030 S. Broadway, Ste 5, Lexington, KY, 40536, USA
- Department of Epidemiology, University of Kentucky, Lexington, KY, USA
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Ahmed A Bahrani
- Sanders-Brown Center on Aging, University of Kentucky, 1030 S. Broadway, Ste 5, Lexington, KY, 40536, USA
| | - Kristine E Shady Lewis
- Sanders-Brown Center on Aging, University of Kentucky, 1030 S. Broadway, Ste 5, Lexington, KY, 40536, USA
| | - Brandi Carey
- Sanders-Brown Center on Aging, University of Kentucky, 1030 S. Broadway, Ste 5, Lexington, KY, 40536, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, 1030 S. Broadway, Ste 5, Lexington, KY, 40536, USA
- Department of Pathology and Division of Neuropathology, University of Kentucky, Lexington, KY, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, 1030 S. Broadway, Ste 5, Lexington, KY, 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, University of Kentucky, 1030 S. Broadway, Ste 5, Lexington, KY, 40536, USA
- Department of Behavioral Science, University of Kentucky, Lexington, KY, USA
- Department of Neurology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
10
|
Provasek VE, Mitra J, Malojirao VH, Hegde ML. DNA Double-Strand Breaks as Pathogenic Lesions in Neurological Disorders. Int J Mol Sci 2022; 23:ijms23094653. [PMID: 35563044 PMCID: PMC9099445 DOI: 10.3390/ijms23094653] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 02/07/2023] Open
Abstract
The damage and repair of DNA is a continuous process required to maintain genomic integrity. DNA double-strand breaks (DSBs) are the most lethal type of DNA damage and require timely repair by dedicated machinery. DSB repair is uniquely important to nondividing, post-mitotic cells of the central nervous system (CNS). These long-lived cells must rely on the intact genome for a lifetime while maintaining high metabolic activity. When these mechanisms fail, the loss of certain neuronal populations upset delicate neural networks required for higher cognition and disrupt vital motor functions. Mammalian cells engage with several different strategies to recognize and repair chromosomal DSBs based on the cellular context and cell cycle phase, including homologous recombination (HR)/homology-directed repair (HDR), microhomology-mediated end-joining (MMEJ), and the classic non-homologous end-joining (NHEJ). In addition to these repair pathways, a growing body of evidence has emphasized the importance of DNA damage response (DDR) signaling, and the involvement of heterogeneous nuclear ribonucleoprotein (hnRNP) family proteins in the repair of neuronal DSBs, many of which are linked to age-associated neurological disorders. In this review, we describe contemporary research characterizing the mechanistic roles of these non-canonical proteins in neuronal DSB repair, as well as their contributions to the etiopathogenesis of selected common neurological diseases.
Collapse
Affiliation(s)
- Vincent E. Provasek
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (V.E.P.); (V.H.M.)
- College of Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Joy Mitra
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (V.E.P.); (V.H.M.)
- Correspondence: (J.M.); (M.L.H.)
| | - Vikas H. Malojirao
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (V.E.P.); (V.H.M.)
| | - Muralidhar L. Hegde
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (V.E.P.); (V.H.M.)
- College of Medicine, Texas A&M University, College Station, TX 77843, USA
- Department of Neurosciences, Weill Cornell Medical College, New York, NY 11021, USA
- Correspondence: (J.M.); (M.L.H.)
| |
Collapse
|
11
|
Rhodus EK, Barber J, Abner EL, Bardach SH, Gibson A, Jicha GA. Comparison of behaviors characteristic of autism spectrum disorder behaviors and behavioral and psychiatric symptoms of dementia. Aging Ment Health 2022; 26:586-594. [PMID: 33222510 PMCID: PMC8212388 DOI: 10.1080/13607863.2020.1849025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Similarities exist in behavioral expression of autism spectrum disorder (ASD) and Alzheimer's disease and related dementias (ADRD). The purpose of this study was to assess presence of behavioral and psychiatric symptoms of dementia (BPSD) and ASD-like behaviors in adults with ADRD. METHODS Using a cross-sectional design, data from University of Kentucky Alzheimer's Disease Center participant cohort were used. Hierarchical linear regression was used to assess (1) the relationship between ASD-like behaviors (measured by the Gilliam Autism Rating Scale-Second Edition, GARS-2) and BPSD measured by the Neuropsychiatric Inventory (NPI), and (2) the relationship between ASD-like behaviors and dementia severity (measured by the Clinical Dementia Rating [CDR] sum of boxes), when controlling for BPSD. RESULTS Complete data were available for 142 participants. Using α of 0.05, analyses identified ASD behaviors were significantly associated with BPSD severity ratings (r = 0.47; p < 0.001) and dementia severity (r = 0.46; p < 0.001). GARS-2 explained 6.1% (p < 0.001) of variance in CDR sum of boxes when controlling for NPI and other covariates. DISCUSSION There is significant overlap in behaviors characteristic of ASD and BPSD as assessed by the NPI and GARS-2, despite the use of these instruments in disparate developmental vs. aging settings. ASD behaviors appear to not be solely present in early childhood as a manifestation of ASD but are also present in older adults with neurodegenerative cognitive impairment. Such associations warrant additional research into causation, assessment, and behavioral interventions to further enable new therapeutic approaches targeting ASD behaviors across the lifespan.
Collapse
Affiliation(s)
| | - Justin Barber
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
| | - Erin L. Abner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY,Department of Epidemiology, University of Kentucky, Lexington, KY
| | - Shoshana H. Bardach
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY,Graduate Center for Gerontology, University of Kentucky, Lexington, KY
| | - Allison Gibson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY,College of Social Work, University of Kentucky, Lexington, KY
| | - Gregory A. Jicha
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY,Department of Behavioral Science, University of Kentucky, Lexington, KY,Department of Neurology, University of Kentucky, Lexington, KY
| |
Collapse
|
12
|
Wang R, Lahiri DK. Effects of microRNA-298 on APP and BACE1 translation differ according to cell type and 3'-UTR variation. Sci Rep 2022; 12:3074. [PMID: 35197498 PMCID: PMC8866491 DOI: 10.1038/s41598-022-05164-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is marked by neurofibrillary tangles and senile plaques composed of amyloid β (Aβ) peptides. However, specific contributions of different cell types to Aβ deposition remain unknown. Non-coding microRNAs (miRNA) play important roles in AD by regulating translation of major associated proteins, such as Aβ precursor protein (APP) and β-site APP-cleaving enzyme (BACE1), two key proteins associated with Aβ biogenesis. MiRNAs typically silence protein expression via binding specific sites in mRNAs' 3'-untranslated regions (3'-UTR). MiRNAs regulate protein levels in a cell-type specific manner; however, mechanisms of the variation of miRNA activity remain unknown. We report that miR-298 treatment reduced native APP and BACE1 protein levels in an astrocytic but not in a neuron-like cell line. From miR-298's effects on APP-3'-UTR activity and native protein levels, we infer that differences in APP 3'-UTR length could explain differential miR-298 activity. Such varied or truncated, but natural, 3'-UTR specific to a given cell type provides an opportunity to regulate native protein levels by particular miRNA. Thus, miRNA's effect tailoring to a specific cell type, bypassing another undesired cell type with a truncated 3'-UTR would potentially advance clinically-relevant translational research.
Collapse
Affiliation(s)
- Ruizhi Wang
- Laboratory of Molecular Neurogenetics' Departments of Psychiatry and Medical & Molecular Genetics' Indiana University School of Medicine' Indiana Alzheimer's Disease Research Center, Stark Neuroscience Research Institute, Indianapolis, 320 West 15th Street, IN, 46202, USA
| | - Debomoy K Lahiri
- Laboratory of Molecular Neurogenetics' Departments of Psychiatry and Medical & Molecular Genetics' Indiana University School of Medicine' Indiana Alzheimer's Disease Research Center, Stark Neuroscience Research Institute, Indianapolis, 320 West 15th Street, IN, 46202, USA.
| |
Collapse
|
13
|
Wang R, Chopra N, Nho K, Maloney B, Obukhov AG, Nelson PT, Counts SE, Lahiri DK. Human microRNA (miR-20b-5p) modulates Alzheimer's disease pathways and neuronal function, and a specific polymorphism close to the MIR20B gene influences Alzheimer's biomarkers. Mol Psychiatry 2022; 27:1256-1273. [PMID: 35087196 PMCID: PMC9054681 DOI: 10.1038/s41380-021-01351-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with loss of cognitive, executive, and other mental functions, and is the most common form of age-related dementia. Amyloid-β peptide (Aβ) contributes to the etiology and progression of the disease. Aβ is derived from the amyloid-β precursor protein (APP). Multiple microRNA (miRNA) species are also implicated in AD. We report that human hsa-miR20b-5p (miR-20b), produced from the MIR20B gene on Chromosome X, may play complex roles in AD pathogenesis, including Aβ regulation. Specifically, miR-20b-5p miRNA levels were altered in association with disease progression in three regions of the human brain: temporal neocortex, cerebellum, and posterior cingulate cortex. In cultured human neuronal cells, miR-20b-5p treatment interfered with calcium homeostasis, neurite outgrowth, and branchpoints. A single-nucleotide polymorphism (SNP) upstream of the MIR20B gene (rs13897515) associated with differences in levels of cerebrospinal fluid (CSF) Aβ1-42 and thickness of the entorhinal cortex. We located a miR-20b-5p binding site in the APP mRNA 3'-untranslated region (UTR), and treatment with miR-20b-5p reduced APP mRNA and protein levels. Network analysis of protein-protein interactions and gene coexpression revealed other important potential miR-20b-5p targets among AD-related proteins/genes. MiR-20b-5p, a miRNA that downregulated APP, was paradoxically associated with an increased risk for AD. However, miR-20b-5p also reduced, and the blockade of APP by siRNA likewise reduced calcium influx. As APP plays vital roles in neuronal health and does not exist solely to be the source of "pathogenic" Aβ, the molecular etiology of AD is likely to not just be a disease of "excess" but a disruption of delicate homeostasis.
Collapse
Affiliation(s)
- Ruizhi Wang
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Nipun Chopra
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- DePauw University, Greencastle, IN, 46135, USA
| | - Kwangsik Nho
- Radiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bryan Maloney
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alexander G Obukhov
- Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Kentucky Alzheimer's Disease Research Center, Lexington, KY, 40536, USA
| | - Scott E Counts
- Departments of Translational Neuroscience & Family Medicine, Michigan State University, Grand Rapids, and Michigan Alzheimer's Disease Research Center, Ann Arbor, MI, USA
| | - Debomoy K Lahiri
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
14
|
Nadeem MS, Hosawi S, Alshehri S, Ghoneim MM, Imam SS, Murtaza BN, Kazmi I. Symptomatic, Genetic, and Mechanistic Overlaps between Autism and Alzheimer's Disease. Biomolecules 2021; 11:1635. [PMID: 34827633 PMCID: PMC8615882 DOI: 10.3390/biom11111635] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 02/02/2023] Open
Abstract
Autism spectrum disorder (ASD) and Alzheimer's disease (AD) are neurodevelopmental and neurodegenerative disorders affecting two opposite ends of life span, i.e., childhood and old age. Both disorders pose a cumulative threat to human health, with the rate of incidences increasing considerably worldwide. In the context of recent developments, we aimed to review correlated symptoms and genetics, and overlapping aspects in the mechanisms of the pathogenesis of ASD and AD. Dementia, insomnia, and weak neuromuscular interaction, as well as communicative and cognitive impairments, are shared symptoms. A number of genes and proteins linked with both disorders have been tabulated, including MECP2, ADNP, SCN2A, NLGN, SHANK, PTEN, RELN, and FMR1. Theories about the role of neuron development, processing, connectivity, and levels of neurotransmitters in both disorders have been discussed. Based on the recent literature, the roles of FMRP (Fragile X mental retardation protein), hnRNPC (heterogeneous ribonucleoprotein-C), IRP (Iron regulatory proteins), miRNAs (MicroRNAs), and α-, β0, and γ-secretases in the posttranscriptional regulation of cellular synthesis and processing of APP (amyloid-β precursor protein) have been elaborated to describe the parallel and overlapping routes and mechanisms of ASD and AD pathogenesis. However, the interactive role of genetic and environmental factors, oxidative and metal ion stress, mutations in the associated genes, and alterations in the related cellular pathways in the development of ASD and AD needs further investigation.
Collapse
Affiliation(s)
- Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.S.N.); (S.H.)
| | - Salman Hosawi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.S.N.); (S.H.)
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Bibi Nazia Murtaza
- Department of Zoology, Abbottabad University of Science and Technology (AUST), Abbottabad 22310, Pakistan;
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.S.N.); (S.H.)
| |
Collapse
|
15
|
Vivanti G, Tao S, Lyall K, Robins DL, Shea LL. The prevalence and incidence of early-onset dementia among adults with autism spectrum disorder. Autism Res 2021; 14:2189-2199. [PMID: 34378867 PMCID: PMC8487995 DOI: 10.1002/aur.2590] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/06/2021] [Accepted: 07/23/2021] [Indexed: 12/18/2022]
Abstract
The prevalence and incidence of early-onset dementia among adults with autism spectrum disorder (ASD) is currently unknown. In this case-control study, the prevalence and incidence of early-onset dementia in individuals with ASD was examined during 2008-2012 using Medicaid Analytic eXtract files. Participants were 30-64 year-old adults who were Medicaid beneficiaries and had either a diagnosis of ASD only (n = 12,648), a diagnosis of ASD with co-occurring intellectual disability (ID) (n = 26,168), a diagnosis of ID without ASD (n = 406,570), or no ASD nor ID diagnoses (n = 798,828). The 5-year prevalence of dementia was 4.04% among adults with ASD only, and 5.22% for those with ASD and co-occurring ID. This prevalence was higher compared to the prevalence of dementia in individuals with no ASD and no ID (0.97%), but lower compared to individuals with ID only (7.10%). Risk factors associated with the increased prevalence in the general population were similarly associated with the increased risk of dementia in individuals with ASD. Even after adjusting for these risk factors, compared to the general population, dementia was found to occur more frequently in individuals with ASD only (adjusted hazard ratio, 1.96; 95% CI, 1.69-2.28), as well as individuals with ASD and co-occurring ID (adjusted hazard ratio, 2.89; 95% CI, 2.62-3.17). In conclusion, adults with ASD under the age of 65 were approximately 2.6 times more likely to be diagnosed with dementia compared to the general population in our study. LAY SUMMARY: It is unclear whether adults diagnosed with autism spectrum disorder (ASD) are at higher risk of being diagnosed with early-onset dementia compared to those who are not on the autism spectrum. In this study we examined for the first time the nationwide prevalence and incidence of Alzheimer's Disease and other types of dementia in ASD in a sample of adults with ASD aged 30-64 years who were enrolled in Medicaid, the largest insurer of behavioral health services in the US. Medicaid claims data, which include information on the diagnoses that beneficiaries receive, suggested that the adults with ASD were approximately 2.6 times more likely to be diagnosed with early-onset Alzheimer's disease and related dementias compared to the general population.
Collapse
Affiliation(s)
- Giacomo Vivanti
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA 19104, USA
| | - Sha Tao
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA 19104, USA
| | - Kristen Lyall
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA 19104, USA
| | - Diana L. Robins
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA 19104, USA
| | - Lindsay L. Shea
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA 19104, USA
| |
Collapse
|
16
|
Mencer S, Kartawy M, Lendenfeld F, Soluh H, Tripathi MK, Khaliulin I, Amal H. Proteomics of autism and Alzheimer's mouse models reveal common alterations in mTOR signaling pathway. Transl Psychiatry 2021; 11:480. [PMID: 34535637 PMCID: PMC8448888 DOI: 10.1038/s41398-021-01578-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/03/2021] [Accepted: 08/20/2021] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) and Alzheimer's disease (AD) are two different neurological disorders that share common clinical features, such as language impairment, executive functions, and motor problems. A genetic convergence has been proposed as well. However, the molecular mechanisms of these pathologies are still not well understood. Protein S-nitrosylation (SNO), the nitric oxide (NO)-mediated posttranslational modification, targets key proteins implicated in synaptic and neuronal functions. Previously, we have shown that NO and SNO are involved in the InsG3680(+/+) ASD and P301S AD mouse models. Here, we performed large-scale computational biology analysis of the SNO-proteome followed by biochemical validation to decipher the shared mechanisms between the pathologies. This analysis pointed to the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway as one of the shared molecular mechanisms. Activation of mTOR in the cortex of both mouse models was confirmed by western blots that showed increased phosphorylation of RPS6, a major substrate of mTORC1. Other molecular alterations affected by SNO and shared between the two mouse models, such as synaptic-associated processes, PKA signaling, and cytoskeleton-related processes were also detected. This is the first study to decipher the SNO-related shared mechanisms between SHANK3 and MAPT mutations. Understanding the involvement of SNO in neurological disorders and its intersection between ASD and AD might help developing an effective novel therapy for both neuropathologies.
Collapse
Affiliation(s)
- Shira Mencer
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Felix Lendenfeld
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Huda Soluh
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
17
|
Boksha IS, Prokhorova TA, Tereshkina EB, Savushkina OK, Burbaeva GS. Protein Phosphorylation Signaling Cascades in Autism: The Role of mTOR Pathway. BIOCHEMISTRY (MOSCOW) 2021; 86:577-596. [PMID: 33993859 DOI: 10.1134/s0006297921050072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway is a central regulator of cell metabolism, growth, and survival in response to hormones, growth factors, nutrients, and stress-induced signals. In this review, we analyzed the studies on the molecular abnormalities of the mTOR-associated signaling cascades in autism spectrum disorders (ASDs) and outlined the prospects for the pathogenicity-targeting pharmacotherapeutic approaches to ASDs, in particular syndromic ASDs. Based on available experimental and clinical data, we suggest that very early detection of molecular abnormalities in the ASD risk groups can be facilitated by using peripheral blood platelets. Also, identification of the time window of critical dysregulations in the described pathways in the ASD risk groups might suggest further research directions leading to more efficacious pharmacotherapeutic interventions in ASDs.
Collapse
Affiliation(s)
- Irina S Boksha
- Mental Health Research Center, Moscow, 115522, Russia. .,Gamaleya Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, 123098, Russia
| | | | | | | | | |
Collapse
|
18
|
Habif M, Do Carmo S, Báez MV, Colettis NC, Cercato MC, Salas DA, Acutain MF, Sister CL, Berkowicz VL, Canal MP, González Garello T, Cuello AC, Jerusalinsky DA. Early Long-Term Memory Impairment and Changes in the Expression of Synaptic Plasticity-Associated Genes, in the McGill-R-Thy1-APP Rat Model of Alzheimer's-Like Brain Amyloidosis. Front Aging Neurosci 2021; 12:585873. [PMID: 33551786 PMCID: PMC7862771 DOI: 10.3389/fnagi.2020.585873] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/23/2020] [Indexed: 12/27/2022] Open
Abstract
Accruing evidence supports the hypothesis that memory deficits in early Alzheimer Disease (AD) might be due to synaptic failure caused by accumulation of intracellular amyloid beta (Aβ) oligomers, then secreted to the extracellular media. Transgenic mouse AD models provide valuable information on AD pathology. However, the failure to translate these findings to humans calls for models that better recapitulate the human pathology. McGill-R-Thy1-APP transgenic (Tg) rat expresses the human amyloid precursor protein (APP751) with the Swedish and Indiana mutations (of familial AD), leading to an AD-like slow-progressing brain amyloid pathology. Therefore, it offers a unique opportunity to investigate learning and memory abilities at early stages of AD, when Aβ accumulation is restricted to the intracellular compartment, prior to plaque deposition. Our goal was to further investigate early deficits in memory, particularly long-term memory in McGill-R-Thy1-APP heterozygous (Tg+/–) rats. Short-term- and long-term habituation to an open field were preserved in 3-, 4-, and 6-month-old (Tg+/–). However, long-term memory of inhibitory avoidance to a foot-shock, novel object-recognition and social approaching behavior were seriously impaired in 4-month-old (Tg+/–) male rats, suggesting that they are unable to either consolidate and/or evoke such associative and discriminative memories with aversive, emotional and spatial components. The long-term memory deficits were accompanied by increased transcript levels of genes relevant to synaptic plasticity, learning and memory processing in the hippocampus, such as Grin2b, Dlg4, Camk2b, and Syn1. Our findings indicate that in addition to the previously well-documented deficits in learning and memory, McGill-R-Thy1-APP rats display particular long-term-memory deficits and deep social behavior alterations at pre-plaque early stages of the pathology. This highlights the importance of Aβ oligomers and emphasizes the validity of the model to study AD-like early processes, with potentially predictive value.
Collapse
Affiliation(s)
- Martín Habif
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - María Verónica Báez
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| | - Natalia Claudia Colettis
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| | - Magalí Cecilia Cercato
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| | - Daniela Alejandra Salas
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| | - María Florencia Acutain
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| | - Caterina Laura Sister
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| | - Valeria Laura Berkowicz
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| | - María Pilar Canal
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| | - Tomás González Garello
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Diana Alicia Jerusalinsky
- Laboratory of Neuroplasticity and Neurotoxins (LaN&N), Facultad de Medicina, Instituto de Biología Celular y Neurociencia (IBCN) "Prof. Eduardo De Robertis" (Universidad de Buenos Aires - CONICET), Buenos Aires, Argentina
| |
Collapse
|
19
|
Gandawijaya J, Bamford RA, Burbach JPH, Oguro-Ando A. Cell Adhesion Molecules Involved in Neurodevelopmental Pathways Implicated in 3p-Deletion Syndrome and Autism Spectrum Disorder. Front Cell Neurosci 2021; 14:611379. [PMID: 33519384 PMCID: PMC7838543 DOI: 10.3389/fncel.2020.611379] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/15/2020] [Indexed: 01/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is characterized by impaired social interaction, language delay and repetitive or restrictive behaviors. With increasing prevalence, ASD is currently estimated to affect 0.5–2.0% of the global population. However, its etiology remains unclear due to high genetic and phenotypic heterogeneity. Copy number variations (CNVs) are implicated in several forms of syndromic ASD and have been demonstrated to contribute toward ASD development by altering gene dosage and expression. Increasing evidence points toward the p-arm of chromosome 3 (chromosome 3p) as an ASD risk locus. Deletions occurring at chromosome 3p result in 3p-deletion syndrome (Del3p), a rare genetic disorder characterized by developmental delay, intellectual disability, facial dysmorphisms and often, ASD or ASD-associated behaviors. Therefore, we hypothesize that overlapping molecular mechanisms underlie the pathogenesis of Del3p and ASD. To investigate which genes encoded in chromosome 3p could contribute toward Del3p and ASD, we performed a comprehensive literature review and collated reports investigating the phenotypes of individuals with chromosome 3p CNVs. We observe that high frequencies of CNVs occur in the 3p26.3 region, the terminal cytoband of chromosome 3p. This suggests that CNVs disrupting genes encoded within the 3p26.3 region are likely to contribute toward the neurodevelopmental phenotypes observed in individuals affected by Del3p. The 3p26.3 region contains three consecutive genes encoding closely related neuronal immunoglobulin cell adhesion molecules (IgCAMs): Close Homolog of L1 (CHL1), Contactin-6 (CNTN6), and Contactin-4 (CNTN4). CNVs disrupting these neuronal IgCAMs may contribute toward ASD phenotypes as they have been associated with key roles in neurodevelopment. CHL1, CNTN6, and CNTN4 have been observed to promote neurogenesis and neuronal survival, and regulate neuritogenesis and synaptic function. Furthermore, there is evidence that these neuronal IgCAMs possess overlapping interactomes and participate in common signaling pathways regulating axon guidance. Notably, mouse models deficient for these neuronal IgCAMs do not display strong deficits in axonal migration or behavioral phenotypes, which is in contrast to the pronounced defects in neuritogenesis and axon guidance observed in vitro. This suggests that when CHL1, CNTN6, or CNTN4 function is disrupted by CNVs, other neuronal IgCAMs may suppress behavioral phenotypes by compensating for the loss of function.
Collapse
Affiliation(s)
- Josan Gandawijaya
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Rosemary A Bamford
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - J Peter H Burbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Asami Oguro-Ando
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
20
|
Zhou B, Zhu Z, Ransom BR, Tong X. Oligodendrocyte lineage cells and depression. Mol Psychiatry 2021; 26:103-117. [PMID: 33144710 PMCID: PMC7815509 DOI: 10.1038/s41380-020-00930-0] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/01/2020] [Accepted: 10/22/2020] [Indexed: 12/25/2022]
Abstract
Depression is a common mental illness, affecting more than 300 million people worldwide. Decades of investigation have yielded symptomatic therapies for this disabling condition but have not led to a consensus about its pathogenesis. There are data to support several different theories of causation, including the monoamine hypothesis, hypothalamic-pituitary-adrenal axis changes, inflammation and immune system alterations, abnormalities of neurogenesis and a conducive environmental milieu. Research in these areas and others has greatly advanced the current understanding of depression; however, there are other, less widely known theories of pathogenesis. Oligodendrocyte lineage cells, including oligodendrocyte progenitor cells and mature oligodendrocytes, have numerous important functions, which include forming myelin sheaths that enwrap central nervous system axons, supporting axons metabolically, and mediating certain forms of neuroplasticity. These specialized glial cells have been implicated in psychiatric disorders such as depression. In this review, we summarize recent findings that shed light on how oligodendrocyte lineage cells might participate in the pathogenesis of depression, and we discuss new approaches for targeting these cells as a novel strategy to treat depression.
Collapse
Affiliation(s)
- Butian Zhou
- Center for Brain Science, Shanghai Children's Medical Center; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongqun Zhu
- Department of Cardiothoracic Surgery, Center for Brain Science, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bruce R Ransom
- Neuroscience Department, City University of Hong Kong, Hong Kong, China.
| | - Xiaoping Tong
- Center for Brain Science, Shanghai Children's Medical Center; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
21
|
Xu JL, Guo Y. Identification of Gene Loci That Overlap Between Mental Disorders and Poor Prognosis of Cancers. Front Psychiatry 2021; 12:678943. [PMID: 34262492 PMCID: PMC8273260 DOI: 10.3389/fpsyt.2021.678943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Co-morbid psychiatric disorders are common in patients with cancers, which make the treatment more difficult. Studying the connection between mental disease-related genes and the prognosis of cancers may potentially lead to novel therapeutic methods. Method: All mental disorders genes were selected from published articles. The correlations between the expression of these genes and the prognosis of different cancers were analyzed by starBase v2.0 and TIMER. The molecular functions, reactome pathways, and interactions among diverse genes were explored via the STRING tool. Results: 239 genes were identified for further survival analysis, 5 of which were overlapping genes across at least five cancer types, including RHEBL1, PDE4B, ANKRD55, EPHB2, and GIMAP7. 146 high-expression and 157 low-expression genes were found to be correlated with the unfavorable prognosis of diverse cancer types. Tight links existed among various mental disease genes. Besides, risk genes were mostly related to the dismal outcome of low-grade glioma (LGG) and kidney renal clear cell carcinoma (KIRC) patients. Gene Ontology (GO) and reactome pathway analysis revealed that most genes involved in various critical molecular functions and primarily related to metabolism, signal transduction, and hemostasis. Conclusions: To explore co-expression genes between mental illnesses and cancers may aid in finding preventive strategies and therapeutic methods for high-risk populations and patients with one or more diseases.
Collapse
Affiliation(s)
- Ji-Li Xu
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yong Guo
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
22
|
Yang CC, Hsiao LD, Yang CM. Galangin Inhibits LPS-Induced MMP-9 Expression via Suppressing Protein Kinase-Dependent AP-1 and FoxO1 Activation in Rat Brain Astrocytes. J Inflamm Res 2020; 13:945-960. [PMID: 33244253 PMCID: PMC7685391 DOI: 10.2147/jir.s276925] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022] Open
Abstract
Purpose Neuroinflammation, characterized by the increased expression of inflammatory proteins such as matrix metalloproteinases (MMPs), plays a critical role in neurodegenerative disorders. Lipopolysaccharide (LPS) has been shown to upregulate MMP-9 expression through the activation of various transcription factors, including activator protein 1 (AP-1) and forkhead box protein O1 (FoxO1). The flavonoid 3,5,7-trihydroxy-2-phenyl-4H-1-benzopyran-4-one (galangin) has been demonstrated to possess antioxidant and anti-inflammatory properties in various types of cells. Here, we investigated the mechanisms underlying the inhibitory effect of galangin on LPS-induced MMP-9 expression in rat brain astrocytes (RBA-1 cells). Methods Pharmacological inhibitors and siRNAs were employed to explore the effects of galangin on LPS-challenged RBA-1 cells. Gelatin zymography, Western blotting, real-time PCR, and a luciferase reporter assay were used to detect MMP-9 activity, protein expression, mRNA levels, and promoter activity, respectively. The protein kinases involved in the LPS-induced MMP-9 expression were determined by Western blot. A chromatin immunoprecipitation (ChIP) assay was employed to evaluate the activity of c-Jun at the MMP-9 promoter. Results Galangin treatment attenuated the LPS-mediated induction of MMP-9 protein and mRNA expression, as well as the activity at the MMP-9 promoter. In addition, galangin exerted its inhibitory effects on MMP-9 expression through suppressing the LPS-stimulated activation of proline-rich tyrosine kinase (Pyk2), platelet-derived growth factor receptor beta (PDGFRβ), phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), mammalian target of rapamycin (mTOR), and mitogen-activated protein kinases (MAPKs). Pretreatment with galangin attenuated the LPS-induced phosphorylation of c-Jun and FoxO1. LPS-induced cell migration was also suppressed by galangin pretreatment. Conclusion Galangin attenuates the LPS-induced inflammatory responses, including the induction of MMP-9 expression and cell migration, via inhibiting Pyk2/PDGFRβ/PI3K/Akt/mTOR/JNK1/JNK2 and p44/p42 MAPK cascade-dependent AP-1 and FoxO1 activities. These results provide new insights into the mechanisms through which galangin mitigates LPS-induced inflammatory responses, and suggest novel strategies for the management of LPS-related brain diseases.
Collapse
Affiliation(s)
- Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Tao-Yuan, Kwei-San, Tao-Yuan 33302, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan 33302, Taiwan
| | - Li-Der Hsiao
- Department of Pharmacology, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Chuen-Mao Yang
- Department of Pharmacology, College of Medicine, China Medical University, Taichung 40402, Taiwan.,Program for Biotch Pharmaceutical Industry, China Medical University, Taichung 40402, Taiwan.,Department of Post-Baccalaureate Veterinary Medicine, College of Medical and Health Science, Asia University, Wufeng, Taichung 41354, Taiwan
| |
Collapse
|
23
|
Jęśko H, Cieślik M, Gromadzka G, Adamczyk A. Dysfunctional proteins in neuropsychiatric disorders: From neurodegeneration to autism spectrum disorders. Neurochem Int 2020; 141:104853. [PMID: 32980494 DOI: 10.1016/j.neuint.2020.104853] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/05/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
Despite fundamental differences in disease course and outcomes, neurodevelopmental (autism spectrum disorders - ASD) and neurodegenerative disorders (Alzheimer's disease - AD and Parkinson's disease - PD) present surprising, common traits in their molecular pathomechanisms. Uncontrolled oligomerization and aggregation of amyloid β (Aβ), microtubule-associated protein (MAP) tau, or α-synuclein (α-syn) contribute to synaptic impairment and the ensuing neuronal death in both AD and PD. Likewise, the pathogenesis of ASD may be attributed, at least in part, to synaptic dysfunction; attention has also been recently paid to irregularities in the metabolism and function of the Aβ precursor protein (APP), tau, or α-syn. Commonly affected elements include signaling pathways that regulate cellular metabolism and survival such as insulin/insulin-like growth factor (IGF) - PI3 kinase - Akt - mammalian target of rapamycin (mTOR), and a number of key synaptic proteins critically involved in neuronal communication. Understanding how these shared pathomechanism elements operate in different conditions may help identify common targets and therapeutic approaches.
Collapse
Affiliation(s)
- Henryk Jęśko
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Magdalena Cieślik
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Grażyna Gromadzka
- Cardinal Stefan Wyszynski University, Faculty of Medicine. Collegium Medicum, Wóycickiego 1/3, 01-938, Warsaw, Poland.
| | - Agata Adamczyk
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| |
Collapse
|
24
|
Eissa N, Sadeq A, Sasse A, Sadek B. Role of Neuroinflammation in Autism Spectrum Disorder and the Emergence of Brain Histaminergic System. Lessons Also for BPSD? Front Pharmacol 2020; 11:886. [PMID: 32612529 PMCID: PMC7309953 DOI: 10.3389/fphar.2020.00886] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/29/2020] [Indexed: 12/27/2022] Open
Abstract
Many behavioral and psychological symptoms of dementia (BPSD) share similarities in executive functioning and communication deficits with those described in several neuropsychiatric disorders, including Alzheimer's disease (AD), epilepsy, schizophrenia (SCH), and autism spectrum disorder (ASD). Numerous studies over the last four decades have documented altered neuroinflammation among individuals diagnosed with ASD. The purpose of this review is to examine the hypothesis that central histamine (HA) plays a significant role in the regulation of neuroinflammatory processes of microglia functions in numerous neuropsychiatric diseases, i.e., ASD, AD, SCH, and BPSD. In addition, this review summarizes the latest preclinical and clinical results that support the relevance of histamine H1-, H2-, and H3-receptor antagonists for the potential clinical use in ASD, SCH, AD, epilepsy, and BPSD, based on the substantial symptomatic overlap between these disorders with regards to cognitive dysfunction. The review focuses on the histaminergic neurotransmission as relevant in these brain disorders, as well as the effects of a variety of H3R antagonists in animal models and in clinical studies.
Collapse
Affiliation(s)
- Nermin Eissa
- Department of Pharmacology and Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Adel Sadeq
- College of Pharmacy, Al Ain University of Science and Technology, Al Ain, United Arab Emirates
| | - Astrid Sasse
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Bassem Sadek
- Department of Pharmacology and Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
25
|
Exley C, Mold MJ. Imaging of aluminium and amyloid β in neurodegenerative disease. Heliyon 2020; 6:e03839. [PMID: 32368656 PMCID: PMC7184253 DOI: 10.1016/j.heliyon.2020.e03839] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/16/2020] [Accepted: 04/21/2020] [Indexed: 12/28/2022] Open
Abstract
Objectives Recent research has confirmed the presence of aluminium in human brain tissue. Quantitative analyses suggest increased brain aluminium content in a number of neurodegenerative diseases including familial Alzheimer's disease, congophilic amyloid angiopathy, epilepsy and autism. Complementary aluminium-specific fluorescence microscopy identifies the location of aluminium in human brain tissue and demonstrates significant differences in distribution between diseases. Herein we combine these approaches in investigating associations between aluminium in human brain tissue and specific disease-associated neuropathologies. Methods We have used aluminium-specific fluorescence microscopy, Congo red staining using light and polarised light and thioflavin S fluorescence microscopy on serial sections of brain tissues to identify co-localisation of aluminium and amyloid β and tau neuropathology. Results A combination of light, polarised and fluorescence microscopy demonstrates an intimate relationship between aluminium and amyloid β in familial Alzheimer's disease but not in other conditions and diseases, such as congophilic amyloid angiopathy and autism. We demonstrate preliminary evidence of amyloid β pathology, including associations with vasculature and parenchymal tissues, in autism in tissues heavily loaded with aluminium. Conclusion We suggest that complementary aluminium-specific fluorescence microscopy may reveal important information about the putative toxicity of aluminium in neurodegenerative and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, United Kingdom
| | - Matthew J Mold
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, United Kingdom
| |
Collapse
|
26
|
Abdelzaher LA, Hussein OA, Ashry IEM. The Novel Potential Therapeutic Utility of Montelukast in Alleviating Autistic Behavior Induced by Early Postnatal Administration of Thimerosal in Mice. Cell Mol Neurobiol 2020; 41:129-150. [PMID: 32303879 DOI: 10.1007/s10571-020-00841-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/01/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIM: Thimerosal (THIM) is a mercury-containing preservative widely used in many biological and medical products including many vaccines. It has been accused of being a possible etiological factor for some neurodevelopmental disorders such as autistic spectrum disorders (ASDs). In our study, the potential therapeutic effect of montelukast, a leukotriene receptor antagonist used to treat seasonal allergies and asthma, on THIM mice model (ASDs model) was examined. METHODOLOGY Newborn mice were randomly distributed into three groups: (Group 1) Control (Cont.) group received saline injections. (Group 2) THIM-treated (THIM) group received THIM intramuscular (IM) at a dose of 3000 μg Hg/kg on postnatal days 7, 9, 11, and 15. (Group 3) Montelukast-treated (Monte) group received THIM followed by montelukast sodium (10 mg/kg/day) intraperitoneal (IP) for 3 weeks. Mice were evaluated for growth development, social interactions, anxiety, locomotor activity, and cognitive function. Brain histopathology, alpha 7 nicotinic acetylcholine receptors (α7nAChRs), nuclear factor kappa B p65 (NF-κB p65), apoptotic factor (Bax), and brain injury markers were evaluated as well. RESULTS THIIM significantly impaired social activity and growth development. Montelukast mitigated THIM-induced social deficit probably through α7nAChRs upregulation, NF-κB p65, Bax, and brain injury markers downregulation, thus suppressing THIM-induced neuronal toxicity and inflammation. CONCLUSION Neonatal exposure to THIM can induce growth retardation and abnormal social interactions similar to those observed in ASDs. Some of these abnormalities could be ameliorated by montelukast via upregulation of α7nAChRs that inhibited NF-κB activation and significant suppression of neuronal injury and the associated apoptosis.
Collapse
Affiliation(s)
- Lobna A Abdelzaher
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Ola A Hussein
- Department of Histology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - I E M Ashry
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
27
|
Shen L, Liu X, Zhang H, Lin J, Feng C, Iqbal J. Biomarkers in autism spectrum disorders: Current progress. Clin Chim Acta 2020; 502:41-54. [DOI: 10.1016/j.cca.2019.12.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
|
28
|
Neurofibromatosis Type 1 Implicates Ras Pathways in the Genetic Architecture of Neurodevelopmental Disorders. Behav Genet 2020; 50:191-202. [DOI: 10.1007/s10519-020-09991-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 01/04/2020] [Indexed: 01/12/2023]
|