1
|
El Tabaa MM, Faheem H, Elballal MS, Rashad E, Mohsen M, El Tabaa MM. The PPAR-α agonist oleoyethanolamide (OEA) ameliorates valproic acid-induced steatohepatitis in rats via suppressing Wnt3a/β-catenin and activating PGC-1α: Involvement of network pharmacology and molecular docking. Eur J Pharmacol 2025; 991:177306. [PMID: 39880183 DOI: 10.1016/j.ejphar.2025.177306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/23/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
Liver damage is one of the most severe side effects of valproic acid (VPA) therapy. Research indicates that PPAR-α prevents Wnt3a/β-catenin-induced PGC-1α dysregulation, which is linked to liver injury. Although PPAR-α activation has hepatoprotective effects, its role in preventing VPA-induced liver injury remains unclear. Our research used network analysis, molecular docking, and in-vivo validation to predict and assess targets and pathways associated with the hepatoprotective effects of oleoylethanolamide (OEA), a PPAR-α agonist, on VPA-induced steatohepatitis. For in-vivo experiments, 24 rats were assigned to V, OEA, VPA, and OEA + VPA. Liver functions, TGs, cholesterol, and LDL were tested. Hepatic levels of PPAR-α, ACO, TNF-α, IL-1β, HO-1, MDA, and TAC, along with Wnt3a/β-catenin, PGC-1α, and Nrf2 expression were assessed. Further, NF-κB, Bax, Bcl-2, and caspase-3 expression were detected immunohistochemically. Network pharmacology identified 258 targets for OEA-steatohepatitis connection, including NFKB1, PPARA, and NFE2L2, in addition to TNF, non-alcoholic fatty liver, NF-κB, PPAR, and WNT signaling, as contributing to steatohepatitis pathogenesis. The docking revealed a strong affinity between OEA and Wnt3a, β-catenin, and PGC-1α. Therefore, we postulated that the hepatoprotective effect of OEA may be due to Wnt3a/β-catenin-mediated inactivation of PGC1-α pathway. In vivo, OEA inhibited Wnt3a/β-catenin and increased PGC1-α by activating PPAR-α. Hence, PGC1-α reduced fat cell β-oxidation and NF-κB-mediated inflammation. OEA lessened MDA and raised TAC to mitigate oxidative damage. OEA additionally reduced apoptosis by lowering Bax/Bcl-2 ratio and caspase-3. In summary, PPAR-α involvement in the protective effects of OEA against VPA-induced steatohepatitis can be confirmed by suppressing Wnt3a/β-catenin and activating PGC-1α signaling.
Collapse
Affiliation(s)
| | - Heba Faheem
- Physiology Department, Faculty of Medicine, Tanta University, Egypt.
| | - Mohammed Salah Elballal
- Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
| | - Eman Rashad
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| | - Mohamed Mohsen
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, 32897, Menoufia, Egypt.
| |
Collapse
|
2
|
Aboul-Fotouh S, Zohny SM, Elnahas EM, Habib MZ, Hassan GA. Can memantine treat autism? Answers from preclinical and clinical studies. Neurosci Biobehav Rev 2025; 169:106019. [PMID: 39826825 DOI: 10.1016/j.neubiorev.2025.106019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/02/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Autism Spectrum Disorder (ASD) represents a clinical challenge due to its diverse behavioral symptoms and complex neuro-pathophysiology. Finding effective treatments that target the fundamental mechanisms of ASD remains a top priority. This narrative review presents the potential of the NMDA-receptor blocker memantine in managing ASD symptoms. Preclinical studies indicate that memantine could abrogate excitotoxicity, GABA/glutamate imbalance, reduced levels of brain-derived neurotrophic factor (BDNF), blood-brain barrier (BBB) leakage, and neuroinflammation, offering hope for managing core deficits associated with ASD like impaired social interaction and repetitive behaviors. However, clinical trials yield conflicting results, with some showing slight improvements in symptom severity and cognitive function, while others demonstrate limited efficacy. Further exploration of memantine's neurobiological mechanisms and refinement of treatment approaches are crucial for comprehensively tackling ASD complexities. Drawing from both animal models and clinical data, this review examines memantine's impact on core ASD symptoms, cognitive function, and potential mechanisms of action. Lastly, it identifies research gaps and proposes avenues for future investigations to enhance our understanding and utilization of memantine in ASD management.
Collapse
Affiliation(s)
- Sawsan Aboul-Fotouh
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sohir M Zohny
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Esraa M Elnahas
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed Z Habib
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt; Basic Medical Sciences Department, Faculty of Medicine, King Salman International University, El Tor, South Sinai, Egypt.
| | - Ghada Am Hassan
- Neuropsychiatry Department, Faculty of Medicine, Galala University, Suez, Egypt; Neuropsychiatry Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
3
|
Williams OOF, Coppolino M, Micelli CB, McCallum RT, Henry-Duru PT, Manduca JD, Lalonde J, Perreault ML. Prenatal exposure to valproic acid induces sex-specific alterations in rat cortical and hippocampal neuronal structure and function in vitro. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111222. [PMID: 39701172 DOI: 10.1016/j.pnpbp.2024.111222] [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: 09/18/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
There are substantial differences in the characteristics of males and females with an autism spectrum disorder (ASD), yet there is little knowledge surrounding the mechanistic underpinnings of these differences. The valproic acid (VPA) rodent model is based upon the human fetal valproate spectrum disorder, which is associated with increased risk of developing ASD. This model, which displays significant social, learning, and memory alterations, has therefore been widely used to further our understanding of specific biological features of ASD. However, to date, almost all of the studies employing this model have used male rodents. To fill this knowledge gap, we evaluated sex differences for neuronal activity, morphology, and glycogen synthase kinase-3 (GSK-3) signaling in primary cortical (CTX) and hippocampal (HIP) neurons prepared from rats exposed to VPA in utero. In vivo, sex-specific VPA-induced alterations in the frontal CTX transcriptome at birth were also determined. Overall, VPA induced more robust changes in neuronal function and structure in the CTX than in the HIP. Male- and female-derived primary CTX neurons from rats exposed to prenatal VPA had elevated activity and showed more disorganized firing. In the HIP, only the female VPA neurons showed elevated firing, while the male VPA neurons exhibited disorganized activity. Dendritic arborization of CTX neurons from VPA rats was less complex in both sexes, though this was more pronounced in the females. Conversely, both female and male HIP neurons from VPA rats showed elevated complexity distal to the soma. Female VPA CTX neurons also had an elevated number of dendritic spines. The relative activity of the α and β isoforms of GSK-3 were suppressed in both female and male VPA CTX neurons, with no changes in the HIP neurons. On postnatal day 0, alterations in CTX genes associated with neuropeptides (e.g., penk, pdyn) and receptors (e.g., drd1, adora2a) were seen in both sexes, though they were downregulated in females and upregulated in males. Together these findings suggest that substantial sex differences in neuronal structure and function in the VPA model may have relevance to the reported sex differences in idiopathic ASD.
Collapse
Affiliation(s)
- Olivia O F Williams
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada.
| | - Madeleine Coppolino
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| | - Cecilia B Micelli
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| | - Ryan T McCallum
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| | - Paula T Henry-Duru
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada.
| | - Joshua D Manduca
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada.
| | - Jasmin Lalonde
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada.
| | - Melissa L Perreault
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
4
|
Liang Q, Zhang C, Lv P, Huang Y, Zhao H, Jiang S, Xu W. The important role of the Wnt/β-catenin signaling pathway in small molecules mediated gingival mesenchymal stem cells transdifferentiate into neuron-like cells. Arch Oral Biol 2025; 169:106115. [PMID: 39488928 DOI: 10.1016/j.archoralbio.2024.106115] [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: 06/22/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVE Given their neural crest origin, gingival mesenchymal stem cells (GMSCs) possess high neurogenic potential, which makes them suitable for cell replacement therapy against neurodegenerative diseases. This study investigated whether GMSCs can be transdifferentiated into neurons in vitro using a protocol involving small molecules VCRFY (VPA, CHIR99021, Repsox, Forskolin, and Y-27632). The regulatory mechanisms of key signaling pathways were also investigated. METHODS Neuronal induction of GMSCs was conducted using a small molecules-based protocol over 7 days, which included the evaluation of cell morphology, proliferation, expressions of neurogenic markers, and intracellular calcium oscillation. The activation of canonical the Wnt signaling pathway was assessed by examining the protein content and subcellular localization of β-catenin. RESULTS Small molecules-treated GMSCs displayed neuronal morphology and increased expression of neurogenic markers, including class III beta-tubulin (TUJ1), neuron-specific enolase (NSE), microtube-associated protein 2 (MAP2), and neurofilament medium (NFM), verified through RT-qPCR, western blotting, and immunocytochemistry. Based on the results of Fluo-4 AM calcium flux assay, small molecules-treated GMSCs exhibited enhanced electrophysiological activity. GMSC proliferation halted after 2 days of treatment. Among the small molecules, CHIR99021 exhibited the highest neuronal induction efficiency. Furthermore, activation of the Wnt/β-catenin signaling pathway augmented neuronal differentiation. CONCLUSIONS Small molecule-based cellular reprogramming can efficiently generate neurons from GMSCs, with Wnt/β-catenin signaling to play a critical role in neuronal induction.
Collapse
Affiliation(s)
- Qiuying Liang
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China
| | - Chuhan Zhang
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China
| | - Peiyi Lv
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China
| | - Yongmao Huang
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China
| | - Hang Zhao
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China
| | - Shan Jiang
- Department of Periodontics and Oral Medicine, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China.
| | - Wenan Xu
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China.
| |
Collapse
|
5
|
Sandhu A, Rawat K, Gautam V, Kumar A, Sharma A, Bhatia A, Grover S, Saini L, Saha L. Neuroprotective effect of PPAR gamma agonist in rat model of autism spectrum disorder: Role of Wnt/β-catenin pathway. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111126. [PMID: 39179196 DOI: 10.1016/j.pnpbp.2024.111126] [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: 05/02/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND The clinical manifestation of autism spectrum disorder (ASD) is linked to the disruption of fundamental neurodevelopmental pathways. Emerging evidences claim to have an upregulation of canonical Wnt/β-catenin pathway while downregulation of PPARγ pathway in ASD. This study aims to investigate the therapeutic potential of pioglitazone, a PPARγ agonist, in rat model of ASD. The study further explores the possible role of PPARγ and Wnt/β-catenin pathway and their interaction in ASD by using their modulators. MATERIAL AND METHODS Pregnant female Wistar rats received 600 mg/kg of valproic acid (VPA) to induce autistic symptoms in pups. Pioglitazone (10 mg/kg) was used to evaluate neurobehaviors, relative mRNA expression of inflammatory (IL-1β, IL-6, IL-10, TNF-α), apoptotic markers (Bcl-2, Bax, & Caspase-3) and histopathology (H&E, Nissl stain, Immunohistochemistry). Effect of pioglitazone was evaluated on Wnt pathway and 4 μg/kg dose of 6-BIO (Wnt modulator) was used to study the PPARγ pathway. RESULTS ASD model was established in pups as indicated by core autistic symptoms, increased neuroinflammation, apoptosis and histopathological neurodegeneration in cerebellum, hippocampus and amygdala. Pioglitazone significantly attenuated these alterations in VPA-exposed rats. The expression study results indicated an increase in key transcription factor, β-catenin in VPA-rats suggesting an upregulation of canonical Wnt pathway in them. Pioglitazone significantly downregulated the Wnt signaling by suppressing the expression of Wnt signaling-associated proteins. The inhibiting effect of Wnt pathway on PPARγ activity was indicated by downregulation of PPARγ-associated protein in VPA-exposed rats and those administered with 6-BIO. CONCLUSION In the present study, upregulation of canonical Wnt/β-catenin pathway was demonstrated in ASD rat model. Pioglitazone administration significantly ameliorated these symptoms potentially through its neuroprotective effect and its ability to downregulate the Wnt/β-catenin pathway. The antagonism between the PPARγ and Wnt pathway offers a promising therapeutic approach for addressing ASD.
Collapse
Affiliation(s)
- Arushi Sandhu
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India
| | - Kajal Rawat
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India
| | - Vipasha Gautam
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India
| | - Anil Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India
| | - Antika Sharma
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Sandeep Grover
- Department of Psychiatry, Post Graduate Institute ofMedical Education and Research (PGIMER), Chandigarh 160012, India
| | - Lokesh Saini
- Department of Paediatrics, All India Institute of Medical Sciences (AIIMS), Jodhpur, Rajasthan 342001, India
| | - Lekha Saha
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India.
| |
Collapse
|
6
|
Drehmer I, Santos-Terra J, Gottfried C, Deckmann I. mTOR signaling pathway as a pathophysiologic mechanism in preclinical models of autism spectrum disorder. Neuroscience 2024; 563:33-42. [PMID: 39481829 DOI: 10.1016/j.neuroscience.2024.10.050] [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: 09/05/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/03/2024]
Abstract
Autism spectrum disorder (ASD) is a highly prevalent multifactorial disorder characterized by social deficits and stereotypies. Despite extensive research efforts, the etiology of ASD remains poorly understood. However, studies using preclinical models have identified the mechanistic target of rapamycin kinase (mTOR) signaling pathway as a key player in ASD-related features. This review examines genetic and environmental models of ASD, focusing on their association with the mTOR pathway. We organize findings on alterations within this pathway, providing insights about the potential mechanisms involved in the onset and maintenance of ASD symptoms. Our analysis highlights the central role of mTOR hyperactivation in disrupting autophagic processes, neural organization, and neurotransmitter pathways, which collectively contribute to ASD phenotypes. The review also discusses the therapeutic potential of mTOR pathway inhibitors, such as rapamycin, in mitigating ASD characteristics. These insights underscore the importance of the mTOR pathway as a target for future research and therapeutic intervention in ASD. This review innovates by bringing the convergence of disrupted mTOR signaling in preclinical models and clinical data associated with ASD.
Collapse
Affiliation(s)
- Isabela Drehmer
- Translational Research Group on Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Brazil; Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, Brazil; Psychiatry Molecular Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Júlio Santos-Terra
- Translational Research Group on Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Brazil; Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, Brazil; Psychiatry Molecular Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carmem Gottfried
- Translational Research Group on Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Brazil; Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, Brazil; Psychiatry Molecular Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Iohanna Deckmann
- Translational Research Group on Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Brazil; Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, Brazil; Psychiatry Molecular Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
| |
Collapse
|
7
|
Thomas SD, Jayaprakash P, Marwan NZHJ, Aziz EABA, Kuder K, Łażewska D, Kieć-Kononowicz K, Sadek B. Alleviation of Autophagic Deficits and Neuroinflammation by Histamine H3 Receptor Antagonist E159 Ameliorates Autism-Related Behaviors in BTBR Mice. Pharmaceuticals (Basel) 2024; 17:1293. [PMID: 39458934 PMCID: PMC11510413 DOI: 10.3390/ph17101293] [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: 08/09/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Autism spectrum disorder (ASD) is a neurodevelopmental condition marked by social interaction difficulties, repetitive behaviors, and immune dysregulation with elevated pro-inflammatory markers. Autophagic deficiency also contributes to social behavior deficits in ASD. Histamine H3 receptor (H3R) antagonism is a potential treatment strategy for brain disorders with features overlapping ASD, such as schizophrenia and Alzheimer's disease. METHODS This study investigated the effects of sub-chronic systemic treatment with the H3R antagonist E159 on social deficits, repetitive behaviors, neuroinflammation, and autophagic disruption in male BTBR mice. RESULTS E159 (2.5, 5, and 10 mg/kg, i.p.) improved stereotypic repetitive behavior by reducing self-grooming time and enhancing spontaneous alternation in addition to attenuating social deficits. It also decreased pro-inflammatory cytokines in the cerebellum and hippocampus of treated BTBR mice. In BTBR mice, reduced expression of autophagy-related proteins LC3A/B and Beclin 1 was observed, which was elevated following treatment with E159, attenuating the disruption in autophagy. The co-administration with the H3R agonist MHA (10 mg/kg, i.p.) reversed these effects, highlighting the role of histaminergic neurotransmission in observed behavioral improvements. CONCLUSIONS These preliminary findings suggest the therapeutic potential of H3R antagonists in targeting neuroinflammation and autophagic disruption to improve ASD-like behaviors.
Collapse
Affiliation(s)
- Shilu Deepa Thomas
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Petrilla Jayaprakash
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Nurfirzana Z. H. J. Marwan
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Ezzatul A. B. A. Aziz
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Kamil Kuder
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna Str. 9, 30-688 Kraków, Poland; (K.K.); (D.Ł.); (K.K.-K.)
| | - Dorota Łażewska
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna Str. 9, 30-688 Kraków, Poland; (K.K.); (D.Ł.); (K.K.-K.)
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna Str. 9, 30-688 Kraków, Poland; (K.K.); (D.Ł.); (K.K.-K.)
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
8
|
Karimi Z, Zarifkar A, Mirzaei E, Dianatpour M, Dara M, Aligholi H. Therapeutic effects of nanosilibinin in valproic acid-zebrafish model of autism spectrum disorder: Focusing on Wnt signaling pathway and autism spectrum disorder-related cytokines. Int J Dev Neurosci 2024; 84:454-468. [PMID: 38961588 DOI: 10.1002/jdn.10348] [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: 07/22/2023] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 07/05/2024] Open
Abstract
In this study, we delved into the intricate world of autism spectrum disorder (ASD) and its connection to the disturbance in the Wnt signaling pathway and immunological abnormalities. Our aim was to evaluate the impact of silibinin, a remarkable modulator of both the Wnt signaling pathway and the immune system, on the neurobehavioral and molecular patterns observed in a zebrafish model of ASD induced by valproic acid (VPA). Because silibinin is a hydrophobic molecule and highly insoluble in water, it was used in the form of silibinin nanoparticles (nanosilibinin, NS). After assessing survival, hatching rate, and morphology of zebrafish larvae exposed to different concentrations of NS, the appropriate concentrations were chosen. Then, zebrafish embryos were exposed to VPA (1 μM) and NS (100 and 200 μM) at the same time for 120 h. Next, anxiety and inattentive behaviors and the expression of CHD8, CTNNB, GSK3beta, LRP6, TNFalpha, IL1beta, and BDNF genes were assessed 7 days post fertilization. The results indicated that higher concentrations of NS had adverse effects on survival, hatching, and morphological development. The concentrations of 100 and 200 μM of NS could ameliorate the anxiety-like behavior and learning deficit and decrease ASD-related cytokines (IL1beta and TNFalpha) in VPA-treated larvae. In addition, only 100 μM of NS prevented raising the gene expression of Wnt signaling-related factors (CHD8, CTNNB, GSK3beta, and LRP6). In conclusion, NS treatment for the first 120 h showed therapeutic effect on an autism-like phenotype probably via reducing the expression of pro-inflammatory cytokines genes and changing the expression of Wnt signaling components genes.
Collapse
Affiliation(s)
- Zahra Karimi
- Department of Neuroscience, School of Advanced Medical Science and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asadollah Zarifkar
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Esmaeil Mirzaei
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahintaj Dara
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Aligholi
- Department of Neuroscience, School of Advanced Medical Science and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Yu L, Liu Y, Xia J, Feng S, Chen F. KCNH5 deletion increases autism susceptibility by regulating neuronal growth through Akt/mTOR signaling pathway. Behav Brain Res 2024; 470:115069. [PMID: 38797494 DOI: 10.1016/j.bbr.2024.115069] [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: 03/28/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Recent clinical studies have highlighted mutations in the voltage-gated potassium channel Kv10.2 encoded by the KCNH5 gene among individuals with autism spectrum disorder (ASD). Our preliminary study found that Kv10.2 was decreased in the hippocampus of valproic acid (VPA) - induced ASD rats. Nevertheless, it is currently unclear how KCNH5 regulates autism-like features, or becomes a new target for autism treatment. We employed KCNH5 knockout (KCNH5-/-) rats and VPA - induced ASD rats in this study. Then, we used behavioral assessments, combined with electrophysiological recordings and hippocampal brain slice, to elucidate the impact of KCNH5 deletion and environmental factors on neural development and function in rats. We found that KCNH5-/- rats showed early developmental delay, neuronal overdevelopment, and abnormal electroencephalogram (EEG) signals, but did not exhibit autism-like behavior. KCNH5-/- rats exposed to VPA (KCNH5-/--VPA) exhibit even more severe autism-like behaviors and abnormal neuronal development. The absence of KCNH5 excessively enhances the activity of the Protein Kinase B (Akt)/Mechanistic Target of Rapamycin (mTOR) signaling pathway in the hippocampus of rats after exposure to VPA. Overall, our findings underscore the deficiency of KCNH5 increases the susceptibility to autism under environmental exposures, suggesting its potential utility as a target for screening and diagnosis in ASD.
Collapse
Affiliation(s)
- Lele Yu
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, PR China.
| | - Yamei Liu
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, PR China.
| | - Junyu Xia
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, PR China.
| | - Shini Feng
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, PR China.
| | - Fuxue Chen
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, PR China.
| |
Collapse
|
10
|
Le Belle JE, Condro M, Cepeda C, Oikonomou KD, Tessema K, Dudley L, Schoenfield J, Kawaguchi R, Geschwind D, Silva AJ, Zhang Z, Shokat K, Harris NG, Kornblum HI. Acute rapamycin treatment reveals novel mechanisms of behavioral, physiological, and functional dysfunction in a maternal inflammation mouse model of autism and sensory over-responsivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602602. [PMID: 39026891 PMCID: PMC11257517 DOI: 10.1101/2024.07.08.602602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Maternal inflammatory response (MIR) during early gestation in mice induces a cascade of physiological and behavioral changes that have been associated with autism spectrum disorder (ASD). In a prior study and the current one, we find that mild MIR results in chronic systemic and neuro-inflammation, mTOR pathway activation, mild brain overgrowth followed by regionally specific volumetric changes, sensory processing dysregulation, and social and repetitive behavior abnormalities. Prior studies of rapamycin treatment in autism models have focused on chronic treatments that might be expected to alter or prevent physical brain changes. Here, we have focused on the acute effects of rapamycin to uncover novel mechanisms of dysfunction and related to mTOR pathway signaling. We find that within 2 hours, rapamycin treatment could rapidly rescue neuronal hyper-excitability, seizure susceptibility, functional network connectivity and brain community structure, and repetitive behaviors and sensory over-responsivity in adult offspring with persistent brain overgrowth. These CNS-mediated effects are also associated with alteration of the expression of several ASD-,ion channel-, and epilepsy-associated genes, in the same time frame. Our findings suggest that mTOR dysregulation in MIR offspring is a key contributor to various levels of brain dysfunction, including neuronal excitability, altered gene expression in multiple cell types, sensory functional network connectivity, and modulation of information flow. However, we demonstrate that the adult MIR brain is also amenable to rapid normalization of these functional changes which results in the rescue of both core and comorbid ASD behaviors in adult animals without requiring long-term physical alterations to the brain. Thus, restoring excitatory/inhibitory imbalance and sensory functional network modularity may be important targets for therapeutically addressing both primary sensory and social behavior phenotypes, and compensatory repetitive behavior phenotypes.
Collapse
|
11
|
Mehra S, Ahsan AU, Sharma M, Budhwar M, Chopra M. Gestational Fisetin Exerts Neuroprotection by Regulating Mitochondria-Directed Canonical Wnt Signaling, BBB Integrity, and Apoptosis in Prenatal VPA-Induced Rodent Model of Autism. Mol Neurobiol 2024; 61:4001-4020. [PMID: 38048031 DOI: 10.1007/s12035-023-03826-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023]
Abstract
Embryonic valproic acid (VPA) has been considered a potential risk factor for autism. Majority of studies indicated that targeting autism-associated alterations in VPA-induced autistic model could be promising in defining and designing therapeutics for autism. Numerous investigations in this field investigated the role of canonical Wnt signaling cascade in regulating the pathophysiology of autism. The impaired blood-brain barrier (BBB) permeability and mitochondrial dysfunction are some key implied features of the autistic brain. So, the current study was conducted to target canonical Wnt signaling pathway with a natural polyphenolic modulator cum antioxidant namely fisetin. A single dose of intraperitoneal VPA sodium salt (400 mg/kg) at gestational day 12.5 induced developmental delays, social behaviour impairments (tube dominance test), and anxiety-like behaviour (sucrose preference test) similar to autism. VPA induced mitochondrial damage and over-activated the canonical Wnt signaling which further increased the blood-brain barrier (BBB) disruption, apoptosis, and neuronal damage. Our findings revealed that oral administration of 10 mg/kg gestational fisetin (GD 13-till parturition) improved social and anxiety-like behaviour by modulating the ROS-regulated mitochondrial-canonical Wnt signaling. Moreover, fisetin controls BBB permeability, apoptosis, and neuronal damage in autism model proving its neuroprotective efficacy. Collectively, our findings revealed that fisetin-evoked modulation of the Wnt signaling cascade successfully relieved the associated symptoms of autism along with developmental delays in the model and indicates its potential as a bioceutical against autism.
Collapse
Affiliation(s)
- Sweety Mehra
- Cell and Molecular Biology Lab, Department of Zoology, Panjab University, Chandigarh, 160014, India
| | - Aitizaz Ul Ahsan
- Cell and Molecular Biology Lab, Department of Zoology, Panjab University, Chandigarh, 160014, India
| | - Madhu Sharma
- Cell and Molecular Biology Lab, Department of Zoology, Panjab University, Chandigarh, 160014, India
| | - Muskan Budhwar
- Cell and Molecular Biology Lab, Department of Zoology, Panjab University, Chandigarh, 160014, India
| | - Mani Chopra
- Cell and Molecular Biology Lab, Department of Zoology, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
12
|
Flores-Prieto B, Caycho-Salazar F, Manzo J, Hernández-Aguilar ME, Coria-Avila AG, Herrera-Covarrubias D, Rojas-Dúran F, Aranda-Abreu GE, Pérez-Estudillo CA, Toledo-Cárdenas MR. Effect of Enriched Environment on Cerebellum and Social Behavior of Valproic Zebrafish. NEUROSCI 2024; 5:128-140. [PMID: 39483495 PMCID: PMC11477906 DOI: 10.3390/neurosci5020009] [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: 03/20/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 11/03/2024] Open
Abstract
The etiology of autism spectrum disorder (ASD) has been linked to both genetic and epigenetic factors. Among the epigenetic factors, exposure to valproic acid (VPA), an antiepileptic and mood-modulating drug, has been shown to induce characteristic traits of ASD when exposed to during embryogenesis. Conversely, in animal models, enriched environment (EE) has demonstrated positive behavioral and neural effects, suggesting its potential as a complementary treatment to pharmacological approaches in central nervous system disorders. In this study, we utilized zebrafish to model ASD characteristics induced by VPA and hypothesized that sensory stimulation through EE could ameliorate the behavioral and neuroanatomical features associated with ASD. To test this hypothesis, we assessed social behavior, cerebellar volume, and Purkinje cell populations via histology and immunohistochemistry after exposing the fish to EE. The results revealed that zebrafish exposed to VPA exhibited social deficits, reduced cerebellar cortex volume, and a decrease in c-Fos-positive cells in the Purkinje layer. In contrast, VPA-exposed fish treated with EE showed increased socialization, augmented cerebellar cortex volume, and an elevation in c-Fos-positive Purkinje cells. These findings suggest that alterations induced by VPA may be ameliorated through EE treatment, highlighting the potential therapeutic impact of sensory stimulation in conditions related to ASD.
Collapse
Affiliation(s)
| | - Flower Caycho-Salazar
- Doctorate in Brain Research, Universidad Veracruzana, Veracruz 91190, Mexico; (B.F.-P.)
| | - Jorge Manzo
- Institute of Brain Research, Universidad Veracruzana, Veracruz 91190, Mexico
| | | | | | | | - Fausto Rojas-Dúran
- Institute of Brain Research, Universidad Veracruzana, Veracruz 91190, Mexico
| | | | | | | |
Collapse
|
13
|
Sandhu A, Rawat K, Gautam V, Bhatia A, Grover S, Saini L, Saha L. Ameliorating effect of pioglitazone on prenatal valproic acid-induced behavioral and neurobiological abnormalities in autism spectrum disorder in rats. Pharmacol Biochem Behav 2024; 237:173721. [PMID: 38307465 DOI: 10.1016/j.pbb.2024.173721] [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: 11/30/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopment disorder that mainly arises due to abnormalities in different brain regions, resulting in behavioral deficits. Besides its diverse phenotypical features, ASD is associated with complex and varied etiology, presenting challenges in understanding its precise neuro-pathophysiology. Pioglitazone was reported to have a fundamental role in neuroprotection in various other neurological disorders. The present study aimed to investigate the therapeutic potential of pioglitazone in the prenatal valproic acid (VPA)-model of ASD in Wistar rats. Pregnant female Wistar rats received VPA on Embryonic day (E.D12.5) to induce autistic-like-behavioral and neurobiological alterations in their offspring. VPA-exposed rats presented core behavioral symptoms of ASD such as deficits in social interaction, poor spatial and learning behavior, increased anxiety, locomotory and repetitive activity, and decreased exploratory activity. Apart from these, VPA exposure also stimulated neurochemical and histopathological neurodegeneration in various brain regions. We administered three different doses of pioglitazone i.e., 2.5, 5, and 10 mg/kg in rats to assess various parameters. Of all the doses, our study highlighted that 10 mg/kg pioglitazone efficiently attenuated the autistic symptoms along with other neurochemical alterations such as oxidative stress, neuroinflammation, and apoptosis. Moreover, pioglitazone significantly attenuated the neurodegeneration by restoring the neuronal loss in the hippocampus and cerebellum. Taken together, our study suggests that pioglitazone exhibits therapeutic potential in alleviating behavioral abnormalities induced by prenatal VPA exposure in rats. However, further research is needed to fully understand and establish pioglitazone's effectiveness in treating ASD.
Collapse
Affiliation(s)
- Arushi Sandhu
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), 4th Floor, Research Block B, Chandigarh 160012, India
| | - Kajal Rawat
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), 4th Floor, Research Block B, Chandigarh 160012, India
| | - Vipasha Gautam
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), 4th Floor, Research Block B, Chandigarh 160012, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education andResearch (PGIMER), 2nd Floor, Research Block B, Chandigarh 160012, India
| | - Sandeep Grover
- Department of Psychiatry, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Lokesh Saini
- Department of Paediatrics, All India Institute of Medical Sciences (AIIMS), Jodhpur 342001, Rajasthan, India
| | - Lekha Saha
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), 4th Floor, Research Block B, Chandigarh 160012, India.
| |
Collapse
|
14
|
Li H, Cui J, Hu C, Li H, Luo X, Hao Y. Identification and Analysis of ZIC-Related Genes in Cerebellum of Autism Spectrum Disorders. Neuropsychiatr Dis Treat 2024; 20:325-339. [PMID: 38410689 PMCID: PMC10895985 DOI: 10.2147/ndt.s444138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/09/2024] [Indexed: 02/28/2024] Open
Abstract
Objective Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with significant genetic heterogeneity. The ZIC gene family can regulate neurodevelopment, especially in the cerebellum, and has been implicated in ASD-like behaviors in mice. We performed bioinformatic analysis to identify the ZIC gene family in the ASD cerebellum. Methods We explored the roles of ZIC family genes in ASD by investigating (i) the association of ZIC genes with ASD risk genes from the Simons Foundation Autism Research Initiative (SFARI) database and ZIC genes in the brain regions of the Human Protein Atlas (HPA) database; (ii) co-expressed gene networks of genes positively and negatively correlated with ZIC1, ZIC2, and ZIC3, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and receiver operating characteristic (ROC) curve analysis of genes in these networks; and (iii) the relationship between ZIC1, ZIC2, ZIC3, and their related genes with cerebellar immune cells and stromal cells in ASD patients. Results (i) ZIC1, ZIC2, and ZIC3 were associated with neurodevelopmental disorders and risk genes related to ASD in the human cerebellum and (ii) ZIC1, ZIC2, and ZIC3 were highly expressed in the cerebellum, which may play a pathogenic role by affecting neuronal development and the cerebellar internal environment in patients with ASD, including immune cells, astrocytes, and endothelial cells. (iii) OLFM3, SLC27A4, GRB2, TMED1, NR2F1, and STRBP are closely related to ZIC1, ZIC2, and ZIC3 in ASD cerebellum and have good diagnostic accuracy. (iv) ASD mice in the maternal immune activation model demonstrated that Zic3 and Nr2f1 levels were decreased in the immune-activated cerebellum. Conclusion Our study supports the role of ZIC1, ZIC2, and ZIC3 in ASD pathogenesis and provides potential targets for early and accurate prediction of ASD.
Collapse
Affiliation(s)
- Heli Li
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jinru Cui
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Cong Hu
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Hao Li
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Yan Hao
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| |
Collapse
|
15
|
Zarate-Lopez D, Torres-Chávez AL, Gálvez-Contreras AY, Gonzalez-Perez O. Three Decades of Valproate: A Current Model for Studying Autism Spectrum Disorder. Curr Neuropharmacol 2024; 22:260-289. [PMID: 37873949 PMCID: PMC10788883 DOI: 10.2174/1570159x22666231003121513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 10/25/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder with increased prevalence and incidence in recent decades. Its etiology remains largely unclear, but it seems to involve a strong genetic component and environmental factors that, in turn, induce epigenetic changes during embryonic and postnatal brain development. In recent decades, clinical studies have shown that inutero exposure to valproic acid (VPA), a commonly prescribed antiepileptic drug, is an environmental factor associated with an increased risk of ASD. Subsequently, prenatal VPA exposure in rodents has been established as a reliable translational model to study the pathophysiology of ASD, which has helped demonstrate neurobiological changes in rodents, non-human primates, and brain organoids from human pluripotent stem cells. This evidence supports the notion that prenatal VPA exposure is a valid and current model to replicate an idiopathic ASD-like disorder in experimental animals. This review summarizes and describes the current features reported with this animal model of autism and the main neurobiological findings and correlates that help elucidate the pathophysiology of ASD. Finally, we discuss the general framework of the VPA model in comparison to other environmental and genetic ASD models.
Collapse
Affiliation(s)
- David Zarate-Lopez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
- Physiological Science Ph.D. Program, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Ana Laura Torres-Chávez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
- Physiological Science Ph.D. Program, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Alma Yadira Gálvez-Contreras
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, University of Guadalajara, Guadalajara 44340, México
| | - Oscar Gonzalez-Perez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
| |
Collapse
|
16
|
Park G, Jang WE, Kim S, Gonzales EL, Ji J, Choi S, Kim Y, Park JH, Mohammad HB, Bang G, Kang M, Kim S, Jeon SJ, Kim JY, Kim KP, Shin CY, An JY, Kim MS, Lee YS. Dysregulation of the Wnt/β-catenin signaling pathway via Rnf146 upregulation in a VPA-induced mouse model of autism spectrum disorder. Exp Mol Med 2023; 55:1783-1794. [PMID: 37524878 PMCID: PMC10474298 DOI: 10.1038/s12276-023-01065-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/15/2023] [Accepted: 05/29/2023] [Indexed: 08/02/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder associated with impaired social behavior and communication, repetitive behaviors, and restricted interests. In addition to genetic factors, environmental factors such as prenatal drug exposure contribute to the development of ASD. However, how those prenatal factors induce behavioral deficits in the adult stage is not clear. To elucidate ASD pathogenesis at the molecular level, we performed a high-resolution mass spectrometry-based quantitative proteomic analysis on the prefrontal cortex (PFC) of mice exposed to valproic acid (VPA) in utero, a widely used animal model of ASD. Differentially expressed proteins (DEPs) in VPA-exposed mice showed significant overlap with ASD risk genes, including differentially expressed genes from the postmortem cortex of ASD patients. Functional annotations of the DEPs revealed significant enrichment in the Wnt/β-catenin signaling pathway, which is dysregulated by the upregulation of Rnf146 in VPA-exposed mice. Consistently, overexpressing Rnf146 in the PFC impaired social behaviors and altered the Wnt signaling pathway in adult mice. Furthermore, Rnf146-overexpressing PFC neurons showed increased excitatory synaptic transmission, which may underlie impaired social behavior. These results demonstrate that Rnf146 is critical for social behavior and that dysregulation of Rnf146 underlies social deficits in VPA-exposed mice.
Collapse
Affiliation(s)
- Gaeun Park
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Wooyoung Eric Jang
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Seoyeon Kim
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, Republic of Korea
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841, Republic of Korea
| | - Edson Luck Gonzales
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jungeun Ji
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, Republic of Korea
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841, Republic of Korea
| | - Seunghwan Choi
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, 02841, Republic of Korea
| | - Yujin Kim
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, Republic of Korea
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841, Republic of Korea
| | - Ji Hwan Park
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | | | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Minkyung Kang
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Soobin Kim
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Se Jin Jeon
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, 17104, Republic of Korea
- Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Chan Young Shin
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea.
| | - Joon-Yong An
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, Republic of Korea.
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841, Republic of Korea.
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, 02841, Republic of Korea.
| | - Min-Sik Kim
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea.
- New Biology Research Center, DGIST, Daegu, 42988, Republic of Korea.
- Center for Cell Fate Reprogramming and Control, DGIST, Daegu, 42988, Republic of Korea.
| | - Yong-Seok Lee
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Wide River Institute of Immunology, Seoul National University, Hongcheon, 25159, Republic of Korea.
| |
Collapse
|
17
|
Atia AA, Ashour RH, Zaki MM, Rahman KM, Ramadan NM. The comparative effectiveness of metformin and risperidone in a rat model of valproic acid-induced autism, Potential role for enhanced autophagy. Psychopharmacology (Berl) 2023; 240:1313-1332. [PMID: 37133558 PMCID: PMC10172247 DOI: 10.1007/s00213-023-06371-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/25/2023] [Indexed: 05/04/2023]
Abstract
RATIONALE Risperidone is the first antipsychotic to be approved by Food and Drug Administration (FDA) for treating autism spectrum disorder (ASD). The potential efficacy of metformin in preventing and/or controlling ASD behavioral deficits was also recently reported. Suppression of hippocampus autophagy was suggested as a potential pathologic mechanism in ASD. OBJECTIVES Is metformin's ability to improve ASD clinical phenotype driven by its autophagy-enhancing properties? And does hippocampus autophagy enhancement underlie risperidone's efficacy as well? Both questions are yet to be answered. METHODS The effectiveness of metformin on alleviation of ASD-like behavioral deficits in adolescent rats exposed prenatally to valproic acid (VPA) was compared to that of risperidone. The potential modulatory effects of risperidone on hippocampal autophagic activity were also assessed and compared to those of metformin. RESULTS Male offspring exposed to VPA during gestation exhibited marked anxiety, social impairment and aggravation of stereotyped grooming; such deficits were efficiently rescued by postnatal risperidone or metformin therapy. This autistic phenotype was associated with suppressed hippocampal autophagy; as evidenced by reduced gene/dendritic protein expression of LC3B (microtubule-associated proteins 1 light chain 3B) and increased somatic P62 (Sequestosome 1) protein aggregates. Interestingly, compared to risperidone, the effectiveness of metformin in controlling ASD symptoms and improving hippocampal neuronal survival was well correlated to its ability to markedly induce pyramidal neuronal LC3B expression while lowering P62 accumulation. CONCLUSIONS Our work highlights, for the first time, positive modulation of hippocampus autophagy as potential mechanism underlying improvements in autistic behaviors, observed with metformin, as well as risperidone, therapy.
Collapse
Affiliation(s)
- Amany Aa Atia
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, 60 El-Gomhoria Street, Mansoura, Al-Dakahlia, 35516, Egypt
| | - Rehab H Ashour
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, 60 El-Gomhoria Street, Mansoura, Al-Dakahlia, 35516, Egypt
| | - Marwa Maf Zaki
- Department of Pathology, Faculty of Medicine, Mansoura University, 60 El-Gomhoria Street, Mansoura, Al-Dakahlia, 35516, Egypt
| | - Karawan Ma Rahman
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, 60 El-Gomhoria Street, Mansoura, Al-Dakahlia, 35516, Egypt
| | - Nehal M Ramadan
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, 60 El-Gomhoria Street, Mansoura, Al-Dakahlia, 35516, Egypt.
| |
Collapse
|
18
|
Zhuang W, Ye T, Wang W, Song W, Tan T. CTNNB1 in neurodevelopmental disorders. Front Psychiatry 2023; 14:1143328. [PMID: 37009120 PMCID: PMC10061110 DOI: 10.3389/fpsyt.2023.1143328] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
CTNNB1 is the gene that encodes β-catenin which acts as a key player in the Wnt signaling pathway and regulates cellular homeostasis. Most CTNNB1-related studies have been mainly focused on its role in cancer. Recently, CTNNB1 has also been found involved in neurodevelopmental disorders (NDDs), such as intellectual disability, autism, and schizophrenia. Mutations of CTNNB1 lead to the dysfunction of the Wnt signaling pathway that regulates gene transcription and further disturbs synaptic plasticity, neuronal apoptosis, and neurogenesis. In this review, we discuss a wide range of aspects of CTNNB1 and its physiological and pathological functions in the brain. We also provide an overview of the most recent research regarding CTNNB1 expression and its function in NDDs. We propose that CTNNB1 would be one of the top high-risk genes for NDDs. It could also be a potential therapeutic target for the treatment of NDDs.
Collapse
Affiliation(s)
- Wenting Zhuang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| | - Tong Ye
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| | - Wei Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Weihong Song
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Weihong Song,
| | - Tao Tan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Tao Tan,
| |
Collapse
|
19
|
Peng Q, Liu Y, Yu L, Shen Y, Li F, Feng S, Chen F. Deletion of Arrb2 Down-regulates Autophagy in the Mouse Hippocampus via Akt-mTOR Pathway Activation. Neuroscience 2023; 519:120-130. [PMID: 36796753 DOI: 10.1016/j.neuroscience.2023.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 02/16/2023]
Abstract
The cytoplasmic multifunctional adaptor protein β-arrestin 2 (Arrb2) is involved in the occurrence of various nervous system diseases, such as Alzheimer's disease and Parkinson's disease. Previous laboratory studies have shown that the expression and function of the Arrb2 gene was increased in valproic acid-induced autistic mice models. However, few reports have examined the possible role of Arrb2 in the pathogenesis of autism spectrum disorder. Therefore, Arrb2-deficient (Arrb2-/-) mice were further studied to uncover the physiological function of Arrb2 in the nervous system. In this study, we found that Arrb2-/- mice had normal behavioral characteristics compared with wild-type mice. The autophagy marker protein LC3B was decreased in the hippocampus of Arrb2-/- mice compared to wild-type mice. Western blot analysis revealed that deletion of Arrb2 caused hyperactivation of Akt-mTOR signaling in the hippocampus. In addition, abnormal mitochondrial dysfunction was observed in Arrb2-/- hippocampal neurons, which was characterized by a reduction in mitochondrial membrane potential and adenosine triphosphate production and an increase in reactive oxygen species levels. Therefore, this study elucidates the interaction between Arrb2 and the Akt-mTOR signaling pathway and provides insights into the role of Arrb2 in hippocampal neuron autophagy.
Collapse
Affiliation(s)
- Qingyu Peng
- School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Yamei Liu
- School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Lele Yu
- School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Yizhe Shen
- School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Feng Li
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, PR China
| | - Shini Feng
- School of Life Sciences, Shanghai University, Shanghai 200444, PR China.
| | - Fuxue Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
20
|
Iezzi D, Curti L, Ranieri G, Gerace E, Costa A, Ilari A, La Rocca A, Luceri C, D'Ambrosio M, Silvestri L, Scardigli M, Mannaioni G, Masi A. Acute rapamycin rescues the hyperexcitable phenotype of accumbal medium spiny neurons in the valproic acid rat model of autism spectrum disorder. Pharmacol Res 2022; 183:106401. [PMID: 35987482 DOI: 10.1016/j.phrs.2022.106401] [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: 06/06/2022] [Revised: 08/04/2022] [Accepted: 08/14/2022] [Indexed: 11/17/2022]
Abstract
We previously demonstrated that prenatal exposure to valproic acid (VPA), an environmental model of autism spectrum disorder (ASD), leads to a hyperexcitable phenotype associated with downregulation of inward-rectifying potassium currents in nucleus accumbens (NAc) medium spiny neurons (MSNs) of adolescent rats. Aberrant mTOR pathway function has been associated with autistic-like phenotypes in multiple animal models, including gestational exposure to VPA. The purpose of this work was to probe the involvement of the mTOR pathway in VPA-induced alterations of striatal excitability. Adolescent male Wistar rats prenatally exposed to VPA were treated acutely with the mTOR inhibitor rapamycin and used for behavioral tests, ex vivo brain slice electrophysiology, single-neuron morphometric analysis, synaptic protein quantification and gene expression analysis in the NAc. We report that postnatal rapamycin ameliorates the social deficit and reverts the abnormal excitability, but not the inward-rectifying potassium current defect, of accumbal MSNs. Synaptic transmission and neuronal morphology were largely unaffected by prenatal VPA exposure or postnatal rapamycin treatment. Transcriptome analysis revealed extensive deregulation of genes implied in neurodevelopmental disorders and ionic mechanisms exerted by prenatal VPA, which was partially reverted by postnatal rapamycin. The results of this work support the existence of antagonistic interaction between mTOR and VPA-induced pathways on social behavior, neurophysiological phenotype and gene expression profile, thus prompting further investigation of the mTOR pathway in the quest for specific therapeutic targets in ASD.
Collapse
Affiliation(s)
- D Iezzi
- Università degli Studi di Firenze, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino - NEUROFARBA, viale Pieraccini 6, 50139, Firenze, Italy; Institut de Neurobiologie de la MEDiterranée - INMED, 163, Avenue de Luminy - Parc Scientifique, 13009, Marseille, France
| | - L Curti
- Università degli Studi di Firenze, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino - NEUROFARBA, viale Pieraccini 6, 50139, Firenze, Italy
| | - G Ranieri
- Università degli Studi di Firenze, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino - NEUROFARBA, viale Pieraccini 6, 50139, Firenze, Italy
| | - E Gerace
- Università degli Studi di Firenze, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino - NEUROFARBA, viale Pieraccini 6, 50139, Firenze, Italy
| | - A Costa
- Università degli Studi di Firenze, Dipartimento di Scienze della Salute, viale Pieraccini 6, 50139, Firenze, Italy
| | - A Ilari
- Università degli Studi di Firenze, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino - NEUROFARBA, viale Pieraccini 6, 50139, Firenze, Italy
| | - A La Rocca
- Università degli Studi di Firenze, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino - NEUROFARBA, viale Pieraccini 6, 50139, Firenze, Italy
| | - C Luceri
- Università degli Studi di Firenze, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino - NEUROFARBA, viale Pieraccini 6, 50139, Firenze, Italy
| | - M D'Ambrosio
- Università degli Studi di Firenze, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino - NEUROFARBA, viale Pieraccini 6, 50139, Firenze, Italy
| | - L Silvestri
- Università degli Studi di Firenze, Dipartimento di Fisica e Astronomia, Via Sansone 1, 50019, Sesto Fiorentino, Italy; European Laboratory for Non-Linear Spectroscopy, Via Nello Carrara 1, 50019, Sesto Fiorentino, Italy
| | - M Scardigli
- Università degli Studi di Firenze, Dipartimento di Fisica e Astronomia, Via Sansone 1, 50019, Sesto Fiorentino, Italy; European Laboratory for Non-Linear Spectroscopy, Via Nello Carrara 1, 50019, Sesto Fiorentino, Italy
| | - G Mannaioni
- Università degli Studi di Firenze, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino - NEUROFARBA, viale Pieraccini 6, 50139, Firenze, Italy
| | - A Masi
- Università degli Studi di Firenze, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino - NEUROFARBA, viale Pieraccini 6, 50139, Firenze, Italy.
| |
Collapse
|
21
|
Wang J, Chen S, Pan C, Li G, Tang Z. Application of Small Molecules in the Central Nervous System Direct Neuronal Reprogramming. Front Bioeng Biotechnol 2022; 10:799152. [PMID: 35875485 PMCID: PMC9301571 DOI: 10.3389/fbioe.2022.799152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
The lack of regenerative capacity of neurons leads to poor prognoses for some neurological disorders. The use of small molecules to directly reprogram somatic cells into neurons provides a new therapeutic strategy for neurological diseases. In this review, the mechanisms of action of different small molecules, the approaches to screening small molecule cocktails, and the methods employed to detect their reprogramming efficiency are discussed, and the studies, focusing on neuronal reprogramming using small molecules in neurological disease models, are collected. Future research efforts are needed to investigate the in vivo mechanisms of small molecule-mediated neuronal reprogramming under pathophysiological states, optimize screening cocktails and dosing regimens, and identify safe and effective delivery routes to promote neural regeneration in different neurological diseases.
Collapse
Affiliation(s)
| | | | | | - Gaigai Li
- *Correspondence: Gaigai Li, ; Zhouping Tang,
| | | |
Collapse
|
22
|
Li Y, Zhao Y, Lu Y, Lu X, Hu Y, Li Q, Shuai M, Li R. Autism spectrum disorder-like behavior induced in rat offspring by perinatal exposure to di-(2-ethylhexyl) phthalate. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:52083-52097. [PMID: 35254616 DOI: 10.1007/s11356-022-19531-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 02/26/2022] [Indexed: 06/14/2023]
Abstract
Autism spectrum disorders (ASD), also known as childhood autism, is a common neurological developmental disorder. Although it is generally believed that genetic factors are a primary cause for ASD development, more and more studies show that an increasing number of ASD diagnoses are related to environmental exposure. Epidemiological studies indicated that perinatal exposure to endocrine disruptors might cause neurodevelopmental disorders in children. Di-(2-ethylhexyl) phthalate (DEHP) is widely used as a plasticizer in many products. To explore the neurodevelopmental effect induced by perinatal exposure to DEHP on rat offspring, and the potential mechanisms, female Wistar rats were exposed to 1, 10, and 100 mg/kg/day DEHP during pregnancy and lactation, while valproic acid (VPA) was used as a positive control. The behavior tests showed that rat pups exposed to VPA and 100 mg/kg/day DEHP were not good as those from the control group in both their socialability and social novelty. Expression of mTOR pathway-related components increased while the number of autophagosomes decreased in the brain tissue of the rat offspring exposed to 100 mg/kg/day DEHP. In addition, perinatal exposure to DEHP at all dosages decreased the level of autophagy proteins LC3II and Beclin1 in the brain tissue of rat pups. Our results indicated that perinatal DEHP exposure would induce ASD-like behavioral changes in rat offspring, which might be mediated by activation of the mTOR signaling pathway, and inhibition of autophagy in the brain.
Collapse
Affiliation(s)
- Yao Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
- Office of the Youth League Committee, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yun Zhao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Yu Lu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xianxian Lu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Yingdan Hu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Qiulin Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Menglei Shuai
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Rui Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China.
| |
Collapse
|
23
|
Mehra S, Ul Ahsan A, Seth E, Chopra M. Critical Evaluation of Valproic Acid-Induced Rodent Models of Autism: Current and Future Perspectives. J Mol Neurosci 2022; 72:1259-1273. [DOI: 10.1007/s12031-022-02033-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/21/2022] [Indexed: 11/29/2022]
|
24
|
Montanari M, Martella G, Bonsi P, Meringolo M. Autism Spectrum Disorder: Focus on Glutamatergic Neurotransmission. Int J Mol Sci 2022; 23:ijms23073861. [PMID: 35409220 PMCID: PMC8998955 DOI: 10.3390/ijms23073861] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022] Open
Abstract
Disturbances in the glutamatergic system have been increasingly documented in several neuropsychiatric disorders, including autism spectrum disorder (ASD). Glutamate-centered theories of ASD are based on evidence from patient samples and postmortem studies, as well as from studies documenting abnormalities in glutamatergic gene expression and metabolic pathways, including changes in the gut microbiota glutamate metabolism in patients with ASD. In addition, preclinical studies on animal models have demonstrated glutamatergic neurotransmission deficits and altered expression of glutamate synaptic proteins. At present, there are no approved glutamatergic drugs for ASD, but several ongoing clinical trials are currently focusing on evaluating in autistic patients glutamatergic pharmaceuticals already approved for other conditions. In this review, we provide an overview of the literature concerning the role of glutamatergic neurotransmission in the pathophysiology of ASD and as a potential target for novel treatments.
Collapse
Affiliation(s)
- Martina Montanari
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Department of Systems Neuroscience, University Tor Vergata, 00133 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| |
Collapse
|
25
|
Matsuo K, Shinoda Y, Abolhassani N, Nakabeppu Y, Fukunaga K. Transcriptome Analysis in Hippocampus of Rats Prenatally Exposed to Valproic Acid and Effects of Intranasal Treatment of Oxytocin. Front Psychiatry 2022; 13:859198. [PMID: 35432011 PMCID: PMC9005872 DOI: 10.3389/fpsyt.2022.859198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/04/2022] [Indexed: 11/19/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous disorder characterized by repetitive behaviors and social impairments, often accompanied by learning disabilities. It has been documented that the neuropeptide oxytocin (OXT) ameliorates core symptoms in patients with ASD. We recently reported that chronic administration of intranasal OXT reversed social and learning impairments in prenatally valproic acid (VPA)-exposed rats. However, the underlying molecular mechanisms remain unclear. Here, we explored molecular alterations in the hippocampus of rats and the effects of chronic administration of intranasal OXT (12 μg/kg/d). Microarray analyses revealed that prenatal VPA exposure altered gene expression, a part of which is suggested as a candidate in ASD and is involved in key features including memory, developmental processes, and epilepsy. OXT partly improved the expression of these genes, which were predicted to interact with those involved in social behaviors and hippocampal-dependent memory. Collectively, the present study documented molecular profiling in the hippocampus related to ASD and improvement by chronic treatment with OXT.
Collapse
Affiliation(s)
- Kazuya Matsuo
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Nona Abolhassani
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
26
|
Zhou B, Zheng X, Chen Y, Yan X, Peng J, Liu Y, Zhang Y, Tang L, Wen M. The Changes of Amygdala Transcriptome in Autism Rat Model After Arginine Vasopressin Treatment. Front Neurosci 2022; 16:838942. [PMID: 35401102 PMCID: PMC8990166 DOI: 10.3389/fnins.2022.838942] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Background Some studies have shown that arginine vasopressin (AVP) can significantly improve the social interaction disorder of autism, but the mechanism remains unclear. Methods Female Wistar rats were intraperitoneally injected with VPA or normal saline at embryonic day 12.5 to establish an autism model or normal control in their offspring. Male offspring prenatally exposed to VPA were randomly assigned to two groups: the VPA-induced autism model group and the AVP group. The rats in the AVP group were treated with intranasal AVP at postnatal day (PND) 21 and for 3 weeks. The VPA-induced autism model group was given the same dose of normal saline in the same way. Behavioral responses were evaluated in the open field and three-chambered social test apparatus; the expression levels of AVP in serum were detected by enzyme-linked immunosorbent assay kit, and the gene expression levels on the amygdala were measured by RNA-seq at PND42. Results Intranasal administration of AVP can significantly improve the social interaction disorder and elevate the levels of AVP in serum. Transcriptome sequencing results showed that 518 differently expressed genes (DEGs) were identified in the VPA-induced autism model group compared with the control in this study. Gene Ontology biological process enrichment analysis of DEGs showed that the VPA-induced autism model group had significant nervous system developmental impairments compared with the normal group, particularly in gliogenesis, glial cell differentiation, and oligodendrocyte differentiation. Gene Set Enrichment Analysis (GSEA) enrichment analysis also showed that biological process of oligodendrocyte differentiation, axoneme assembly, and axon ensheathment were inhibited in the VPA-induced autism model group. Pathway enrichment analysis of DEGs between the control and VPA-induced autism model group showed that the PI3K/AKT and Wnt pathways were significantly dysregulated in the VPA-induced autism model group. Few DEGs were found when compared with the transcriptome between the VPA-induced autism model group and the AVP treatment group. GSEA enrichment analysis showed deficits in oligodendrocyte development and function were significantly improved after AVP treatment; the pathways were mainly enriched in the NOTCH, mitogen-activated protein kinase, and focal adhesion signaling pathways, but not in the PI3K/AKT and Wnt pathways. The expression patterns analysis also showed the same results. Conclusion AVP can significantly improve the social interaction disorder of VPA-induced autism model, and AVP may target behavioral symptoms in autism by modulating the vasopressin pathways, rather than primary disease mechanisms.
Collapse
Affiliation(s)
- Bo Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- College of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Xiaoli Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- College of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yunhua Chen
- College of Basic Medical, Guizhou Medical University, Guiyang, China
| | - Xuehui Yan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- College of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Jinggang Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- College of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yibu Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- College of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yi Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- College of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Lei Tang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- College of Pharmacy, Guizhou Medical University, Guiyang, China
- *Correspondence: Lei Tang,
| | - Min Wen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- College of Pharmacy, Guizhou Medical University, Guiyang, China
- Min Wen,
| |
Collapse
|
27
|
Sato A, Kotajima-Murakami H, Tanaka M, Katoh Y, Ikeda K. Influence of Prenatal Drug Exposure, Maternal Inflammation, and Parental Aging on the Development of Autism Spectrum Disorder. Front Psychiatry 2022; 13:821455. [PMID: 35222122 PMCID: PMC8863673 DOI: 10.3389/fpsyt.2022.821455] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/12/2022] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorder (ASD) affects reciprocal social interaction and produces abnormal repetitive, restrictive behaviors and interests. The diverse causes of ASD are divided into genetic alterations and environmental risks. The prevalence of ASD has been rising for several decades, which might be related to environmental risks as it is difficult to consider that the prevalence of genetic disorders related to ASD would increase suddenly. The latter includes (1) exposure to medications, such as valproic acid (VPA) and selective serotonin reuptake inhibitors (SSRIs) (2), maternal complications during pregnancy, including infection and hypertensive disorders of pregnancy, and (3) high parental age. Epidemiological studies have indicated a pathogenetic role of prenatal exposure to VPA and maternal inflammation in the development of ASD. VPA is considered to exert its deleterious effects on the fetal brain through several distinct mechanisms, such as alterations of γ-aminobutyric acid signaling, the inhibition of histone deacetylase, the disruption of folic acid metabolism, and the activation of mammalian target of rapamycin. Maternal inflammation that is caused by different stimuli converges on a higher load of proinflammatory cytokines in the fetal brain. Rodent models of maternal exposure to SSRIs generate ASD-like behavior in offspring, but clinical correlations with these preclinical findings are inconclusive. Hypertensive disorders of pregnancy and advanced parental age increase the risk of ASD in humans, but the mechanisms have been poorly investigated in animal models. Evidence of the mechanisms by which environmental factors are related to ASD is discussed, which may contribute to the development of preventive and therapeutic interventions for ASD.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan.,Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | - Miho Tanaka
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihisa Katoh
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
28
|
Williams OOF, Coppolino M, Perreault ML. Sex differences in neuronal systems function and behaviour: beyond a single diagnosis in autism spectrum disorders. Transl Psychiatry 2021; 11:625. [PMID: 34887388 PMCID: PMC8660826 DOI: 10.1038/s41398-021-01757-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that is associated with functional brain alterations that underlie the expression of behaviour. Males are diagnosed up to four times more than females, and sex differences have been identified in memory, cognitive flexibility, verbal fluency, and social communication. Unfortunately, there exists a lack of information on the sex-dependent mechanisms of ASD, as well as biological markers to distinguish sex-specific symptoms in ASD. This can often result in a standardized diagnosis for individuals across the spectrum, despite significant differences in the various ASD subtypes. Alterations in neuronal connectivity and oscillatory activity, such as is observed in ASD, are highly coupled to behavioural states. Yet, despite the well-identified sexual dimorphisms that exist in ASD, these functional patterns have rarely been analyzed in the context of sex differences or symptomology. This review summarizes alterations in neuronal oscillatory function in ASD, discusses the age, region, symptom and sex-specific differences that are currently observed across the spectrum, and potential targets for regulating neuronal oscillatory activity in ASD. The need to identify sex-specific biomarkers, in order to facilitate specific diagnostic criteria and allow for more targeted therapeutic approaches for ASD will also be discussed.
Collapse
Affiliation(s)
| | | | - Melissa L Perreault
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
29
|
Wu Q, Zhou D, Wu R, Shi R, Shen X, Jin N, Gu J, Gu JH, Liu F, Chu D. Excess folic acid supplementation before and during pregnancy and lactation activates β-catenin in the brain of male mouse offspring. Brain Res Bull 2021; 178:133-143. [PMID: 34808323 DOI: 10.1016/j.brainresbull.2021.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/03/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
Folic acid (FA) supplementation in early pregnancy is recommended to protect against birth defects. But excess FA has exhibited neurodevelopmental toxicity. We previously reported that the mice treated with 2.5-fold the dietary requirement of FA one week before mating and throughout pregnancy and lactation displayed abnormal behaviors in the offspring. Here we found the levels of non-phosphorylated β-catenin (active) were increased in the brains of weaning and adult FA-exposed offspring. Meanwhile, demethylation of protein phosphatase 2 A catalytic subunit (PP2Ac), which suppresses its enzyme activity in regulatory subunit dependent manner, was significantly inhibited. Among the upstream regulators of β-catenin, PI3K/Akt/GSK-3β but not Wnt signaling was stimulated in FA-exposed brains only at weaning. In mouse neuroblastoma N2a cells, knockdown of PP2Ac or leucine carboxyl methyltransferase-1 (LCMT-1), or overexpression of PP2Ac methylation-deficient mutant decreased β-catenin dephosphorylation. These results suggest that excess FA may activate β-catenin via suppressing PP2Ac demethylation, providing a novel mechanism for the influence of FA on neurodevelopment.
Collapse
Affiliation(s)
- Qian Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Dingwei Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Ruozhen Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Ruirui Shi
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong University, 226018 Nantong, China
| | - Xin Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Nana Jin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Jianlan Gu
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, 226001 Nantong, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong University, 226018 Nantong, China.
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, United States.
| | - Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China.
| |
Collapse
|
30
|
Wang X, Qu M, Li Z, Long Y, Hong K, Li H. Valproic acid promotes the in vitro differentiation of human pluripotent stem cells into spermatogonial stem cell-like cells. Stem Cell Res Ther 2021; 12:553. [PMID: 34715904 PMCID: PMC8555208 DOI: 10.1186/s13287-021-02621-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022] Open
Abstract
Background Studying human germ cell development and male infertility is heavily relied on mouse models. In vitro differentiation of human pluripotent stem cells into spermatogonial stem cell-like cells (SSCLCs) can be used as a model to study human germ cells and infertility. The current study aimed to develop the SSCLC induction protocol and assess the effects of the developed protocol on SSCLC induction. Methods We examined the effects of valproic acid (VPA), vitamin C (VC) and the combination of VPA and VC on the SSCLC induction efficiency and determined the expression of spermatogonial genes of differentiated cells. Haploid cells and cells expressed meiotic genes were also detected. RNA-seq analysis was performed to compare the transcriptome between cells at 0 and 12 days of differentiation and differently expressed genes were confirmed by RT-qPCR. We further evaluated the alteration in histone marks (H3K9ac and H3K27me3) at 12 days of differentiation. Moreover, the SSCLC induction efficiency of two hiPSC lines of non-obstructive azoospermia (NOA) patients was assessed using different induction protocols. Results The combination of low concentrations of VPA and VC in the induction medium was most effective to induce SSCLCs expressing several spermatogonial genes from human pluripotent stem cells at 12 days of differentiation. The high concentration of VPA was more effective to induce cells expressing meiotic genes and haploid cells. RNA-seq analysis revealed that the induction of SSCLC involved the upregulated genes in Wnt signaling pathway, and cells at 12 days of differentiation showed increased H3K9ac and decreased H3K27me3. Additionally, two hiPSC lines of NOA patients showed low SSCLC induction efficiency and decreased expression of genes in Wnt signaling pathway. Conclusions VPA robustly promoted the differentiation of human pluripotent stem cells into SSCLCs, which involved the upregulated genes in Wnt signaling pathway and epigenetic changes. hiPSCs from NOA patients showed decreased SSCLC induction efficiency and Wnt signaling pathway gene expression, suggesting that SSC depletion in azoospermia testes might be associated with inactivation of Wnt signaling pathway. Our developed SSCLC induction protocol provides a reliable tool and model to study human germ cell development and male infertility. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02621-1.
Collapse
Affiliation(s)
- Xiaotong Wang
- Institute of Reproductive Health/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mengyuan Qu
- Institute of Reproductive Health/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zili Li
- Institute of Reproductive Health/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuting Long
- Wuhan Tongji Reproductive Hospital, Wuhan, 430013, China
| | - Kai Hong
- Department of Urology, Peking University Third Hospital, Beijing, 100191, China.
| | - Honggang Li
- Institute of Reproductive Health/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Wuhan Tongji Reproductive Hospital, Wuhan, 430013, China.
| |
Collapse
|
31
|
Sato A, Ikeda K. Genetic and Environmental Contributions to Autism Spectrum Disorder Through Mechanistic Target of Rapamycin. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2021; 2:95-105. [PMID: 36325164 PMCID: PMC9616270 DOI: 10.1016/j.bpsgos.2021.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that affects an individual’s reciprocal social interaction and communication ability. Numerous genetic and environmental conditions are associated with ASD, including tuberous sclerosis complex, phosphatase and tensin homolog hamartoma tumor syndrome, fragile X syndrome, and neurofibromatosis 1. The pathogenic molecular mechanisms of these diseases are integrated into the hyperactivation of mTORC1 (mechanistic target of rapamycin complex 1). Rodent models of these diseases have shown high mTORC1 activity in the brain and ASD-related behavioral deficits, which were reversed by the mTORC1 inhibitor rapamycin. Environmental stress can also affect this signaling pathway. In utero exposure to valproate caused ASD in offspring and enhanced mTORC1 activity in the brain, which was sensitive to mTORC1 inhibition. mTORC1 is a signaling hub for diverse cellular functions, including protein synthesis, through the phosphorylation of its targets, such as ribosomal protein S6 kinases. Metabotropic glutamate receptor 5–mediated synaptic function is also affected by the dysregulation of mTORC1 activity, such as in fragile X syndrome and tuberous sclerosis complex. Reversing these downstream changes that are associated with mTORC1 activation normalizes behavioral defects in rodents. Despite abundant preclinical evidence, few clinical studies have investigated the treatment of ASD and cognitive deficits. Therapeutics other than mTORC1 inhibitors failed to show efficacy in fragile X syndrome and neurofibromatosis 1. mTORC1 inhibitors have been tested mainly in tuberous sclerosis complex, and their effects on ASD and neuropsychological deficits are promising. mTORC1 is a promising target for the pharmacological treatment of ASD associated with mTORC1 activation.
Collapse
|
32
|
Balachandar V, Rajagopalan K, Jayaramayya K, Jeevanandam M, Iyer M. Mitochondrial dysfunction: A hidden trigger of autism? Genes Dis 2021; 8:629-639. [PMID: 34291134 PMCID: PMC8278534 DOI: 10.1016/j.gendis.2020.07.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Autism is a heterogeneous neurodevelopmental and neuropsychiatric disorder with no precise etiology. Deficits in cognitive functions uncover at early stages and are known to have an environmental and genetic basis. Since autism is multifaceted and also linked with other comorbidities associated with various organs, there is a possibility that there may be a fundamental cellular process responsible for this. These reasons place mitochondria at the point of interest as it is involved in multiple cellular processes predominantly involving metabolism. Mitochondria encoded genes were taken into consideration lately because it is inherited maternally, has its own genome and also functions the time of embryo development. Various researches have linked mitochondrial mishaps like oxidative stress, ROS production and mt-DNA copy number variations to autism. Despite dramatic advances in autism research worldwide, the studies focusing on mitochondrial dysfunction in autism is rather minimal, especially in India. India, owing to its rich diversity, may be able to contribute significantly to autism research. It is vital to urge more studies in this domain as it may help to completely understand the basics of the condition apart from a genetic standpoint. This review focuses on the worldwide and Indian scenario of autism research; mitochondrial abnormalities in autism and possible therapeutic approaches to combat it.
Collapse
Affiliation(s)
- Vellingiri Balachandar
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Kamarajan Rajagopalan
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Kaavya Jayaramayya
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu 641043, India
| | - Madesh Jeevanandam
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
- Department of Biochemistry, PSG College of Arts and Sciences, Coimbatore, Tamil Nadu 641014, India
| | - Mahalaxmi Iyer
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu 641043, India
| |
Collapse
|
33
|
Alterations in Tau Protein Level and Phosphorylation State in the Brain of the Autistic-Like Rats Induced by Prenatal Exposure to Valproic Acid. Int J Mol Sci 2021; 22:ijms22063209. [PMID: 33809910 PMCID: PMC8004207 DOI: 10.3390/ijms22063209] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/23/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficient social interaction and communication besides repetitive, stereotyped behaviours. A characteristic feature of ASD is altered dendritic spine density and morphology associated with synaptic plasticity disturbances. Since microtubules (MTs) regulate dendritic spine morphology and play an important role in spine development and plasticity the aim of the present study was to investigate the alterations in the content of neuronal α/β-tubulin and Tau protein level as well as phosphorylation state in the valproic acid (VPA)-induced rat model of autism. Our results indicated that maternal exposure to VPA induces: (1) decrease the level of α/β-tubulin along with Tau accumulation in the hippocampus and cerebral cortex; (2) excessive Tau phosphorylation and activation of Tau-kinases: CDK5, ERK1/2, and p70S6K in the cerebral cortex; (3) up-regulation of mTOR kinase-dependent signalling in the hippocampus and cerebral cortex of adolescent rat offspring. Moreover, immunohistochemical staining showed histopathological changes in neurons (chromatolysis) in both analysed brain structures of rats prenatally exposed to VPA. The observed changes in Tau protein together with an excessive decrease in α/β-tubulin level may suggest destabilization and thus dysfunction of the MT cytoskeleton network, which in consequence may lead to the disturbance in synaptic plasticity and the development of autistic-like behaviours.
Collapse
|
34
|
Rizk M, Saker Z, Harati H, Fares Y, Bahmad HF, Nabha S. Deciphering the roles of glycogen synthase kinase 3 (GSK3) in the treatment of autism spectrum disorder and related syndromes. Mol Biol Rep 2021; 48:2669-2686. [PMID: 33650079 DOI: 10.1007/s11033-021-06237-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
Autism spectrum disorder (ASD) is a complex and multifactorial neurodevelopmental disorder characterized by the presence of restricted interests and repetitive behaviors besides deficits in social communication. Syndromic ASD is a subset of ASD caused by underlying genetic disorders, most commonly Fragile X Syndrome (FXS) and Rett Syndrome (RTT). Various mutations and consequent malfunctions in core signaling pathways have been identified in ASD, including glycogen synthase kinase 3 (GSK3). A growing body of evidence suggests a key role of GSK3 dysregulation in the pathogenesis of ASD and its related disorders. Here, we provide a synopsis of the implication of GSK3 in ASD, FXS, and RTT as a promising therapeutic target for the treatment of ASD.
Collapse
Affiliation(s)
- Mahdi Rizk
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Zahraa Saker
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hayat Harati
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Youssef Fares
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.,Department of Neurosurgery, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hisham F Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL, 33140, USA
| | - Sanaa Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
35
|
Meisig J, Dreser N, Kapitza M, Henry M, Rotshteyn T, Rahnenführer J, Hengstler J, Sachinidis A, Waldmann T, Leist M, Blüthgen N. Kinetic modeling of stem cell transcriptome dynamics to identify regulatory modules of normal and disturbed neuroectodermal differentiation. Nucleic Acids Res 2020; 48:12577-12592. [PMID: 33245762 PMCID: PMC7736781 DOI: 10.1093/nar/gkaa1089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 12/22/2022] Open
Abstract
Thousands of transcriptome data sets are available, but approaches for their use in dynamic cell response modelling are few, especially for processes affected simultaneously by two orthogonal influencing variables. We approached this problem for neuroepithelial development of human pluripotent stem cells (differentiation variable), in the presence or absence of valproic acid (signaling variable). Using few basic assumptions (sequential differentiation states of cells; discrete on/off states for individual genes in these states), and time-resolved transcriptome data, a comprehensive model of spontaneous and perturbed gene expression dynamics was developed. The model made reliable predictions (average correlation of 0.85 between predicted and subsequently tested expression values). Even regulations predicted to be non-monotonic were successfully validated by PCR in new sets of experiments. Transient patterns of gene regulation were identified from model predictions. They pointed towards activation of Wnt signaling as a candidate pathway leading to a redirection of differentiation away from neuroepithelial cells towards neural crest. Intervention experiments, using a Wnt/beta-catenin antagonist, led to a phenotypic rescue of this disturbed differentiation. Thus, our broadly applicable model allows the analysis of transcriptome changes in complex time/perturbation matrices.
Collapse
Affiliation(s)
- Johannes Meisig
- Institute of Pathology, Charité-Universitätsmedizin, 10117 Berlin, Germany
- IRI Life Sciences, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Nadine Dreser
- In Vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden Chair foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Marion Kapitza
- In Vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden Chair foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Margit Henry
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Tamara Rotshteyn
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University, 44221 Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), TU Dortmund University, 44139 Dortmund, Germany
| | - Agapios Sachinidis
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Tanja Waldmann
- In Vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden Chair foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden Chair foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Nils Blüthgen
- Institute of Pathology, Charité-Universitätsmedizin, 10117 Berlin, Germany
- IRI Life Sciences, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| |
Collapse
|
36
|
Neurobiological Mechanisms of Autism Spectrum Disorder and Epilepsy, Insights from Animal Models. Neuroscience 2020; 445:69-82. [DOI: 10.1016/j.neuroscience.2020.02.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/22/2020] [Accepted: 02/21/2020] [Indexed: 02/09/2023]
|
37
|
Goh JY, O'Sullivan SE, Shortall SE, Zordan N, Piccinini AM, Potter HG, Fone KCF, King MV. Gestational poly(I:C) attenuates, not exacerbates, the behavioral, cytokine and mTOR changes caused by isolation rearing in a rat 'dual-hit' model for neurodevelopmental disorders. Brain Behav Immun 2020; 89:100-117. [PMID: 32485291 DOI: 10.1016/j.bbi.2020.05.076] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022] Open
Abstract
Many psychiatric illnesses have a multifactorial etiology involving genetic and environmental risk factors that trigger persistent neurodevelopmental impairments. Several risk factors have been individually replicated in rodents, to understand disease mechanisms and evaluate novel treatments, particularly for poorly-managed negative and cognitive symptoms. However, the complex interplay between various factors remains unclear. Rodent dual-hit neurodevelopmental models offer vital opportunities to examine this and explore new strategies for early therapeutic intervention. This study combined gestational administration of polyinosinic:polycytidylic acid (poly(I:C); PIC, to mimic viral infection during pregnancy) with post-weaning isolation of resulting offspring (to mirror adolescent social adversity). After in vitro and in vivo studies required for laboratory-specific PIC characterization and optimization, we administered 10 mg/kg i.p. PIC potassium salt to time-mated Lister hooded dams on gestational day 15. This induced transient hypothermia, sickness behavior and weight loss in the dams, and led to locomotor hyperactivity, elevated striatal cytokine levels, and increased frontal cortical JNK phosphorylation in the offspring at adulthood. Remarkably, instead of exacerbating the well-characterized isolation syndrome, gestational PIC exposure actually protected against a spectrum of isolation-induced behavioral and brain regional changes. Thus isolation reared rats exhibited locomotor hyperactivity, impaired associative memory and reversal learning, elevated hippocampal and frontal cortical cytokine levels, and increased mammalian target of rapamycin (mTOR) activation in the frontal cortex - which were not evident in isolates previously exposed to gestational PIC. Brains from adolescent littermates suggest little contribution of cytokines, mTOR or JNK to early development of the isolation syndrome, or resilience conferred by PIC. But notably hippocampal oxytocin, which can protect against stress, was higher in adolescent PIC-exposed isolates so might contribute to a more favorable outcome. These findings have implications for identifying individuals at risk for disorders like schizophrenia who may benefit from early therapeutic intervention, and justify preclinical assessment of whether adolescent oxytocin manipulations can modulate disease onset or progression.
Collapse
Affiliation(s)
- Jen-Yin Goh
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Saoirse E O'Sullivan
- School of Medicine, University of Nottingham, Royal Derby Hospital, Derby DE22 3DT, UK
| | - Sinead E Shortall
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Nicole Zordan
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Anna M Piccinini
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Harry G Potter
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Kevin C F Fone
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Madeleine V King
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| |
Collapse
|
38
|
Srinivasan V, Korhonen L, Lindholm D. The Unfolded Protein Response and Autophagy as Drug Targets in Neuropsychiatric Disorders. Front Cell Neurosci 2020; 14:554548. [PMID: 33132844 PMCID: PMC7550790 DOI: 10.3389/fncel.2020.554548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/02/2020] [Indexed: 12/15/2022] Open
Abstract
Neurons are polarized in structure with a cytoplasmic compartment extending into dendrites and a long axon that terminates at the synapse. The high level of compartmentalization imposes specific challenges for protein quality control in neurons making them vulnerable to disturbances that may lead to neurological dysfunctions including neuropsychiatric diseases. Synapse and dendrites undergo structural modulations regulated by neuronal activity involve key proteins requiring strict control of their turnover rates and degradation pathways. Recent advances in the study of the unfolded protein response (UPR) and autophagy processes have brought novel insights into the specific roles of these processes in neuronal physiology and synaptic signaling. In this review, we highlight recent data and concepts about UPR and autophagy in neuropsychiatric disorders and synaptic plasticity including a brief outline of possible therapeutic approaches to influence UPR and autophagy signaling in these diseases.
Collapse
Affiliation(s)
- Vignesh Srinivasan
- Medicum, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Helsinki, Finland
| | - Laura Korhonen
- Department of Biochemical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden.,Department of Child and Adolescent Psychiatry, Region Östergötland, Linköping, Sweden
| | - Dan Lindholm
- Medicum, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Helsinki, Finland
| |
Collapse
|
39
|
Chaliha D, Albrecht M, Vaccarezza M, Takechi R, Lam V, Al-Salami H, Mamo J. A Systematic Review of the Valproic-Acid-Induced Rodent Model of Autism. Dev Neurosci 2020; 42:12-48. [DOI: 10.1159/000509109] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/02/2020] [Indexed: 11/19/2022] Open
|
40
|
Gebuijs IGE, Metz JR, Zethof J, Carels CEL, Wagener FADTG, Von den Hoff JW. The anti-epileptic drug valproic acid causes malformations in the developing craniofacial skeleton of zebrafish larvae. Mech Dev 2020; 163:103632. [PMID: 32668265 DOI: 10.1016/j.mod.2020.103632] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/22/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022]
Abstract
Valproic acid (VPA) is an anti-epileptic drug known to cause congenital craniofacial abnormalities, including orofacial clefts (OFC). The exact mechanisms by which VPA leads to craniofacial skeletal malformations are poorly understood. In this study, we investigated the effects of VPA on cartilage and bone formation in the zebrafish larval head during 1-13 hpf (early) and 25-37 hpf (late) development in which cranial neural crest cells (CNCCs) arise and then proliferate and differentiate, respectively. Double-staining for cartilage and bone at 5 dpf revealed that VPA reduced cartilage and bone formation in a dose-dependent manner after both early or late exposure. Several different CNCC-derived cartilage and bone elements were affected in both groups. In the early group (100 μM VPA), the posterior head length and the ethmoid plate were reduced in length (both p < 0.01), while mineralization of 4 out of 9 bone elements was often lacking (all p < 0.01). In the late group (100 μM VPA), also the posterior head length was reduced as well as the length of the ceratohyals (both p < 0.01). Similar to early exposure, mineralization of 3 out of 9 bone elements was often lacking (all p < 0.01). These results indicate that both CNCC formation (early) and differentiation (late) are hampered by VPA treatment, of which the consequences for bone and cartilage formation are persistent at 5 dpf. Indeed, we also found that the expression of several genes related to cartilage and bone was upregulated at 5 dpf. These data indicate a compensatory reaction to the lack of cartilage and bone. Altogether, VPA seems to induce craniofacial malformations via disturbed CNCC function leading to defects in cartilage and bone formation.
Collapse
Affiliation(s)
- I G E Gebuijs
- Department of Dentistry - Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands; Department of Animal Ecology and Physiology, Radboud University, Nijmegen, the Netherlands
| | - J R Metz
- Department of Animal Ecology and Physiology, Radboud University, Nijmegen, the Netherlands
| | - J Zethof
- Department of Animal Ecology and Physiology, Radboud University, Nijmegen, the Netherlands
| | - C E L Carels
- Department of Oral Health Sciences, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - F A D T G Wagener
- Department of Dentistry - Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - J W Von den Hoff
- Department of Dentistry - Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands.
| |
Collapse
|
41
|
Lieberman OJ, Cartocci V, Pigulevskiy I, Molinari M, Carbonell J, Broseta MB, Post MR, Sulzer D, Borgkvist A, Santini E. mTOR Suppresses Macroautophagy During Striatal Postnatal Development and Is Hyperactive in Mouse Models of Autism Spectrum Disorders. Front Cell Neurosci 2020; 14:70. [PMID: 32296308 PMCID: PMC7136750 DOI: 10.3389/fncel.2020.00070] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 03/11/2020] [Indexed: 01/06/2023] Open
Abstract
Macroautophagy (hereafter referred to as autophagy) plays a critical role in neuronal function related to development and degeneration. Here, we investigated whether autophagy is developmentally regulated in the striatum, a brain region implicated in neurodevelopmental disease. We demonstrate that autophagic flux is suppressed during striatal postnatal development, reaching adult levels around postnatal day 28 (P28). We also find that mTOR signaling, a key regulator of autophagy, increases during the same developmental period. We further show that mTOR signaling is responsible for suppressing autophagy, via regulation of Beclin-1 and VPS34 activity. Finally, we discover that autophagy is downregulated during late striatal postnatal development (P28) in mice with in utero exposure to valproic acid (VPA), an established mouse model of autism spectrum disorder (ASD). VPA-exposed mice also display deficits in striatal neurotransmission and social behavior. Correction of hyperactive mTOR signaling in VPA-exposed mice restores social behavior. These results demonstrate that neurons coopt metabolic signaling cascades to developmentally regulate autophagy and provide additional evidence that mTOR-dependent signaling pathways represent pathogenic signaling cascades in ASD mouse models that are active during specific postnatal windows.
Collapse
Affiliation(s)
- Ori J. Lieberman
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | | | - Irena Pigulevskiy
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Maya Molinari
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Josep Carbonell
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | - Michael R. Post
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - David Sulzer
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Anders Borgkvist
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Emanuela Santini
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
42
|
Hughes EM, Calcagno P, Clarke M, Sanchez C, Smith K, Kelly JP, Finn DP, Roche M. Prenatal exposure to valproic acid reduces social responses and alters mRNA levels of opioid receptor and pre-pro-peptide in discrete brain regions of adolescent and adult male rats. Brain Res 2020; 1732:146675. [PMID: 31978376 DOI: 10.1016/j.brainres.2020.146675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/21/2022]
Abstract
Altered social behaviours are a hallmark of several psychiatric and developmental disorders. Clinical and preclinical data have demonstrated that prenatal exposure to valproic acid (VPA), an anti-epileptic and mood stabiliser, is associated with impaired social responses, and thus provides a useful model for the evaluation of neurobiological mechanisms underlying altered social behaviours. The opioid system is widely recognised to regulate and modulate social behaviours, however few studies have examined if the endogenous opioid system is altered in animal models of social impairment. The present study examined social behavioural responses of adolescent and adult male rats prenatally exposed to VPA, and the expression of mRNA encoding opioid receptors and pre-pro-peptides in discrete brain regions. Adolescent and adult rats prenatally exposed to VPA spent less time engaging in social behaviours in the direct social interaction test and exhibited reduced sociability and social novelty preference in the 3-chamber sociability test, compared to saline-treated counterparts. The VPA-exposed adolescent rats exhibited significantly reduced kappa opioid receptor (oprk1) and pre-pro-dynorphin (pdyn) mRNA expression in the cerebral cortex, and reduced oprk1 and nociceptin/orphanin FQ (oprl1) mRNA expression in the hypothalamus. Adult rats prenatally exposed to VPA exhibited decreased mRNA expression of oprk1 and pdyn in hypothalamus, reduced pro-opiomelanocortin(pomc) in the striatum and an increase in delta opioid receptor (oprd1) mRNA in the amygdaloid cortex, when compared to saline-treated counterparts. Mu opioid receptor (oprm1) mRNA expression did not differ between saline and VPA-exposed rats in any region examined. The data demonstrate that impaired social behaviours in adolescent and adult rats prenatally exposed to VPA is accompanied by altered mRNA expression of opioid receptors and pre-pro-peptides in a region specific manner. In particular, both adolescent and adult VPA-exposed rats exhibit reduced oprk1-pdyn mRNA expression in several brain regions, which are associated with deficits in social behavioural responding in the model.
Collapse
Affiliation(s)
- Edel M Hughes
- Physiology, School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland; Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Patricia Calcagno
- Physiology, School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland; Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland
| | - Morgane Clarke
- Physiology, School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland; Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland
| | | | | | - John P Kelly
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland; Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland; Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Michelle Roche
- Physiology, School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland; Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland.
| |
Collapse
|
43
|
Rakitin A. Why Do Psychiatrists Still Prescribe Valproate to Women of Childbearing Potential? Front Psychiatry 2020; 11:739. [PMID: 32848919 PMCID: PMC7396713 DOI: 10.3389/fpsyt.2020.00739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 07/14/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Aleksei Rakitin
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia
| |
Collapse
|
44
|
Lieberman OJ, Cartocci V, Pigulevskiy I, Molinari M, Carbonell J, Broseta MB, Post MR, Sulzer D, Borgkvist A, Santini E. mTOR Suppresses Macroautophagy During Striatal Postnatal Development and Is Hyperactive in Mouse Models of Autism Spectrum Disorders. Front Cell Neurosci 2020; 14:70. [PMID: 32296308 DOI: 10.3389/fncel.2020.00070/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 03/11/2020] [Indexed: 05/20/2023] Open
Abstract
Macroautophagy (hereafter referred to as autophagy) plays a critical role in neuronal function related to development and degeneration. Here, we investigated whether autophagy is developmentally regulated in the striatum, a brain region implicated in neurodevelopmental disease. We demonstrate that autophagic flux is suppressed during striatal postnatal development, reaching adult levels around postnatal day 28 (P28). We also find that mTOR signaling, a key regulator of autophagy, increases during the same developmental period. We further show that mTOR signaling is responsible for suppressing autophagy, via regulation of Beclin-1 and VPS34 activity. Finally, we discover that autophagy is downregulated during late striatal postnatal development (P28) in mice with in utero exposure to valproic acid (VPA), an established mouse model of autism spectrum disorder (ASD). VPA-exposed mice also display deficits in striatal neurotransmission and social behavior. Correction of hyperactive mTOR signaling in VPA-exposed mice restores social behavior. These results demonstrate that neurons coopt metabolic signaling cascades to developmentally regulate autophagy and provide additional evidence that mTOR-dependent signaling pathways represent pathogenic signaling cascades in ASD mouse models that are active during specific postnatal windows.
Collapse
Affiliation(s)
- Ori J Lieberman
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | | | - Irena Pigulevskiy
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Maya Molinari
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Josep Carbonell
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | - Michael R Post
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - David Sulzer
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Anders Borgkvist
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Emanuela Santini
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
45
|
Dana H, Bayramov KK, Delibaşı N, Tahtasakal R, Bayramov R, Hamurcu Z, Sener EF. Disregulation of Autophagy in the Transgenerational Cc2d1a Mouse Model of Autism. Neuromolecular Med 2019; 22:239-249. [PMID: 31721010 DOI: 10.1007/s12017-019-08579-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 10/25/2019] [Indexed: 01/29/2023]
Abstract
Autism spectrum disorder (ASD) is a heterogeneously childhood neurodevelopmental disorder, believed to be under development of various genetic and environmental factors. Autophagy and related pathways have also been implicated in the etiology of ASD. We aimed to investigate autophagic markers by generating the transgenerational inheritance of ASD-like behaviors in the Cc2d1a animal model of ASD. Cc2d1a (+/-) mouse model of ASD was built in two different groups by following three generations. After behavior test, bilateral hippocampus was sliced. Western Blot assay and quantitative real-time polymerase chain reaction (QRT-PCR) were used for measurement of LC3 and Beclin-1 as key regulators of autophagy. All of the animal and laboratory studies were conducted in the Erciyes University Genome and Stem Cell Center (GENKOK). Significant LC3 and Beclin-1 mRNA expression levels were observed in mouse hippocampus between groups and generations. Western blot confirmed the changes of the proteins in the hippocampus. LC3 expressions were increased for females and decreased for males compared to the control group. Beclin-1 expression levels were found to be significantly decreased in males and females compared to controls. This study could help explain a new pathway of autophagy in ASD mouse models. Future animal studies need to investigate sex differences in mouse modeling autism-relevant genes like CC2D1A. We anticipate our results to be a starting point for more comprehensive autophagy studies in this mouse model of ASD.
Collapse
Affiliation(s)
- Halime Dana
- Department of Medical Biology, Medical Faculty, Erciyes University, 38039, Kayseri, Turkey
- Genome and Stem Cell Center (GENKOK), Erciyes University, 38039, Kayseri, Turkey
| | | | - Nesrin Delibaşı
- Department of Medical Biology, Medical Faculty, Erciyes University, 38039, Kayseri, Turkey
- Genome and Stem Cell Center (GENKOK), Erciyes University, 38039, Kayseri, Turkey
| | - Reyhan Tahtasakal
- Department of Medical Biology, Medical Faculty, Erciyes University, 38039, Kayseri, Turkey
- Genome and Stem Cell Center (GENKOK), Erciyes University, 38039, Kayseri, Turkey
| | - Ruslan Bayramov
- Department of Medical Genetics, Haseki Education Research Hospital, Istanbul, Turkey
| | - Zuhal Hamurcu
- Department of Medical Biology, Medical Faculty, Erciyes University, 38039, Kayseri, Turkey
- Genome and Stem Cell Center (GENKOK), Erciyes University, 38039, Kayseri, Turkey
| | - Elif Funda Sener
- Department of Medical Biology, Medical Faculty, Erciyes University, 38039, Kayseri, Turkey.
- Genome and Stem Cell Center (GENKOK), Erciyes University, 38039, Kayseri, Turkey.
| |
Collapse
|
46
|
Gender Related Changes in Gene Expression Induced by Valproic Acid in A Mouse Model of Autism and the Correction by S-adenosyl Methionine. Does It Explain the Gender Differences in Autistic Like Behavior? Int J Mol Sci 2019; 20:ijms20215278. [PMID: 31652960 PMCID: PMC6862653 DOI: 10.3390/ijms20215278] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/14/2022] Open
Abstract
In previous studies we produced autism like behavioral changes in mice by Valproic acid (VPA) with significant differences between genders. S-adenosine methionine (SAM) prevented the autism like behavior in both genders. The expression of 770 genes of pathways involved in neurophysiology and neuropathology was studied in the prefrontal cortex of 60 days old male and female mice using the NanoString nCounter. In females, VPA induced statistically significant changes in the expression of 146 genes; 71 genes were upregulated and 75 downregulated. In males, VPA changed the expression of only 19 genes, 16 were upregulated and 3 downregulated. Eight genes were similarly changed in both genders. When considering only the genes that were changed by at least 50%, VPA changed the expression of 15 genes in females and 3 in males. Only Nts was similarly downregulated in both genders. SAM normalized the expression of most changed genes in both genders. We presume that genes that are involved in autism like behavior in our model were similarly changed in both genders and corrected by SAM. The behavioral and other differences between genders may be related to genes that were differently affected by VPA in males and females and/or differently affected by SAM.
Collapse
|
47
|
Duan Q, Li S, Wen X, Sunnassee G, Chen J, Tan S, Guo Y. Valproic Acid Enhances Reprogramming Efficiency and Neuronal Differentiation on Small Molecules Staged-Induction Neural Stem Cells: Suggested Role of mTOR Signaling. Front Neurosci 2019; 13:867. [PMID: 31551670 PMCID: PMC6737087 DOI: 10.3389/fnins.2019.00867] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/02/2019] [Indexed: 12/21/2022] Open
Abstract
Inducing somatic cells into neural stem cells (iNSCs) in specific ways provides a new cell therapy in a variety of neurological diseases. In the past, iNSCs were generated by transcription factors which increased the risk of mutagenesis, tumor formations, and immune reactions by viral transduction vectors. Therefore, in this study, different small molecules were used to induce mouse embryonic fibroblasts (MEFs) into iNSCs in different reprogramming stages, which showed high reprogramming efficiency without altering the genome. We demonstrated that the small molecules staged-induction neural stem cells (SMSINS) have the characteristics of neural stem cells (NSCs) in morphology, gene expression, self-renewal and differentiation potential. Furthermore, valproic acid (VPA), one of small molecules, was showed to enhance neural induction with highest efficiency compared with six other small molecules, which were also investigated in the present study. Moreover, our results suggested that activating the mammalian target of rapamycin (mTOR) signaling enhanced the induction efficiency and neuronal differentiation. Collectively, our findings indicated that using this induction program allowed us to obtain safe and efficient iNSCs which were free of genetic manipulation. The VPA-mediated mTOR signaling pathway may enhance reprogramming efficiency and neuronal differentiation. So we suggested that this program could be a new method of obtaining iNSCs for the treatment of neurological diseases by cell replacement therapy in the future.
Collapse
Affiliation(s)
- Qingrui Duan
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Siyi Li
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinrui Wen
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gavin Sunnassee
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Chen
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Sheng Tan
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Guo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
48
|
Crespi BJ. Comparative psychopharmacology of autism and psychotic-affective disorders suggests new targets for treatment. Evol Med Public Health 2019; 2019:149-168. [PMID: 31548888 PMCID: PMC6748779 DOI: 10.1093/emph/eoz022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/07/2019] [Indexed: 12/13/2022] Open
Abstract
The first treatments showing effectiveness for some psychiatric disorders, such as lithium for bipolar disorder and chlorpromazine for schizophrenia, were discovered by accident. Currently, psychiatric drug design is seen as a scientific enterprise, limited though it remains by the complexity of brain development and function. Relatively few novel and effective drugs have, however, been developed for many years. The purpose of this article is to demonstrate how evolutionary biology can provide a useful framework for psychiatric drug development. The framework is based on a diametrical nature of autism, compared with psychotic-affective disorders (mainly schizophrenia, bipolar disorder and depression). This paradigm follows from two inferences: (i) risks and phenotypes of human psychiatric disorders derive from phenotypes that have evolved along the human lineage and (ii) biological variation is bidirectional (e.g. higher vs lower, faster vs slower, etc.), such that dysregulation of psychological traits varies in two opposite ways. In this context, the author review the evidence salient to the hypothesis that autism and psychotic-affective disorders represent diametrical disorders in terms of current, proposed and potential psychopharmacological treatments. Studies of brain-derived neurotrophic factor, the PI3K pathway, the NMDA receptor, kynurenic acid metabolism, agmatine metabolism, levels of the endocannabinoid anandamide, antidepressants, anticonvulsants, antipsychotics, and other treatments, demonstrate evidence of diametric effects in autism spectrum disorders and phenotypes compared with psychotic-affective disorders and phenotypes. These findings yield insights into treatment mechanisms and the development of new pharmacological therapies, as well as providing an explanation for the longstanding puzzle of antagonism between epilepsy and psychosis. Lay Summary: Consideration of autism and schizophrenia as caused by opposite alterations to brain development and function leads to novel suggestions for pharmacological treatments.
Collapse
Affiliation(s)
- Bernard J Crespi
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
49
|
[Research advances in the role of mTOR signaling pathway in autism spectrum disorder]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21. [PMID: 31315775 PMCID: PMC7389111 DOI: 10.7499/j.issn.1008-8830.2019.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Mammalian target of rapamycin (mTOR) is an intracellular signaling pathway molecule which regulates various fundamental physiological processes. The mTOR signaling pathway plays an important role in synaptic plasticity, information transmission and processing, and neuroregulation. Dysregulation of the mTOR signaling pathway is generally considered to be related to the pathogenesis of autism spectrum disorder (ASD); meanwhile, the mTOR inhibitor can ameliorate the symptoms of ASD. The role of mTOR in the pathogenesis of ASD is summarized in this article to provide a theoretical basis for targeted therapy of ASD.
Collapse
|
50
|
Vallée A, Vallée JN, Lecarpentier Y. PPARγ agonists: potential treatment for autism spectrum disorder by inhibiting the canonical WNT/β-catenin pathway. Mol Psychiatry 2019; 24:643-652. [PMID: 30104725 DOI: 10.1038/s41380-018-0131-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/24/2018] [Accepted: 06/08/2018] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that is characterized by a deficit in social interactions and communication with repetitive and restrictive behavior. No curative treatments are available for ASD. Pharmacological treatments do not address the core ASD behaviors, but target comorbid symptoms. Dysregulation of the core neurodevelopmental pathways is associated with the clinical presentation of ASD, and the canonical WNT/β-catenin pathway is one of the major pathways involved. The canonical WNT/β-catenin pathway participates in the development of the central nervous system, and its dysregulation involves developmental cognitive disorders. In numerous tissues, the canonical WNT/β-catenin pathway and peroxisome proliferator-activated receptor gamma (PPARγ) act in an opposed manner. In ASD, the canonical WNT/β-catenin pathway is increased while PPARγ seems to be decreased. PPARγ agonists present a beneficial effect in treatment for ASD children through their anti-inflammatory role. Moreover, they induce the inhibition of the canonical WNT/β-catenin pathway in several pathophysiological states. We focus this review on the hypothesis of an opposed interplay between PPARγ and the canonical WNT/β-catenin pathway in ASD and the potential role of PPARγ agonists as treatment for ASD.
Collapse
Affiliation(s)
- Alexandre Vallée
- Paris-Descartes University; Diagnosis and Therapeutic Center, Hôtel-Dieu Hospital; AP-HP, Paris, France. .,Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, Poitiers, France.
| | - Jean-Noël Vallée
- Centre Hospitalier Universitaire (CHU) Amiens Picardie, Université Picardie Jules Verne (UPJV), 80054, Amiens, France.,Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, Poitiers, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), 6-8 rue Saint-fiacre, 77100, Meaux, France
| |
Collapse
|