1
|
Chan CK, Lim KS, Chan CY, Kumar TS, Audrey C, Narayanan V, Fong SL, Ng CC. A review of epilepsy syndromes and epileptogenic mechanism affiliated with brain tumor related genes. Gene 2025; 962:149531. [PMID: 40294709 DOI: 10.1016/j.gene.2025.149531] [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: 11/24/2024] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
Epilepsy is one of the comorbidities often manifested by patients with brain tumors. While there are reviews commenting on the epileptogenicity of brain-tumor-related genes, the reviews are commonly restricted to BRAF, IDH and PIK3CA. According to World Health Organization (WHO), at least 50 genes have been proposed as brain-tumor-related genes. Hence, we aimed to provide a more comprehensive review of the epileptogenicity of the brain-tumor-related genes. We performed an extensive literature search on PubMed, classified the studies, and provided an overview of the associated epilepsy phenotype and epileptogenic mechanism of the brain-tumor-related genes advocated by WHO. Through our analysis, we found a minor overlap between brain-tumor-related genes and epilepsy-associated genes, as some brain-tumor-related genes have been classified as epilepsy-associated genes in earlier studies. Besides reviewing the well-studied genes like TSC1 and TSC2, we identified several under-discovered brain-tumor-related genes, including TP53, CIC, IDH1 and NOTCH1, that warrant future exploration due to the existence of clinical or in vivo evidence substantiating their pathogenic role in epileptogenesis. We also propounded some methodologies that can be applied in future research to enhance the study of the epileptogenic mechanism of brain-tumor-related genes. To date, this article covers the greatest number of brain-tumor-related genes.
Collapse
Affiliation(s)
- Chung-Kin Chan
- Microbiology and Molecular Genetics, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kheng-Seang Lim
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Chet-Ying Chan
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Thinisha Sathis Kumar
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia; Department of Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Christine Audrey
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Vairavan Narayanan
- Department of Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Si-Lei Fong
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ching-Ching Ng
- Microbiology and Molecular Genetics, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
2
|
Goldberg AR, Dovas A, Torres D, Pereira B, Viswanathan A, Das Sharma S, Mela A, Merricks EM, Megino-Luque C, McInvale JJ, Olabarria M, Shokooh LA, Zhao HT, Chen C, Kotidis C, Calvaresi P, Banu MA, Razavilar A, Sudhakar TD, Saxena A, Chokran C, Humala N, Mahajan A, Xu W, Metz JB, Bushong EA, Boassa D, Ellisman MH, Hillman EMC, Hargus G, Bravo-Cordero JJ, McKhann GM, Gill BJA, Rosenfeld SS, Schevon CA, Bruce JN, Sims PA, Peterka DS, Canoll P. Glioma-induced alterations in excitatory neurons are reversed by mTOR inhibition. Neuron 2025; 113:858-875.e10. [PMID: 39837324 PMCID: PMC11925689 DOI: 10.1016/j.neuron.2024.12.026] [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: 01/23/2024] [Revised: 09/27/2024] [Accepted: 12/24/2024] [Indexed: 01/23/2025]
Abstract
Gliomas are aggressive neoplasms that diffusely infiltrate the brain and cause neurological symptoms, including cognitive deficits and seizures. Increased mTOR signaling has been implicated in glioma-induced neuronal hyperexcitability, but the molecular and functional consequences have not been identified. Here, we show three types of changes in tumor-associated neurons: (1) downregulation of transcripts encoding excitatory and inhibitory postsynaptic proteins and dendritic spine development and upregulation of cytoskeletal transcripts via neuron-specific profiling of ribosome-bound mRNA, (2) marked decreases in dendritic spine density via light and electron microscopy, and (3) progressive functional alterations leading to neuronal hyperexcitability via in vivo calcium imaging. A single acute dose of AZD8055, a combined mTORC1/2 inhibitor, reversed these tumor-induced changes. These findings reveal mTOR-driven pathological plasticity in neurons at the infiltrative margin of glioma and suggest new strategies for treating glioma-associated neurological symptoms.
Collapse
Affiliation(s)
- Alexander R Goldberg
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Daniela Torres
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Brianna Pereira
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ashwin Viswanathan
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sohani Das Sharma
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Edward M Merricks
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cristina Megino-Luque
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10027, USA
| | - Julie J McInvale
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Markel Olabarria
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | | | - Hanzhi T Zhao
- Laboratory for Functional Optical Imaging, Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Cady Chen
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Corina Kotidis
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter Calvaresi
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Matei A Banu
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aida Razavilar
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Tejaswi D Sudhakar
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ankita Saxena
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cole Chokran
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nelson Humala
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aayushi Mahajan
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Weihao Xu
- Laboratory for Functional Optical Imaging, Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Jordan B Metz
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Eric A Bushong
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniela Boassa
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Elizabeth M C Hillman
- Laboratory for Functional Optical Imaging, Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jose Javier Bravo-Cordero
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10027, USA
| | - Guy M McKhann
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Brian J A Gill
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | | | - Catherine A Schevon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Darcy S Peterka
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
3
|
Kambli L, Raut D, Bhatt LK. Sulfamethizole Attenuates Pentylenetetrazole-Induced Seizures in Mice via mTOR Inhibition. Drug Dev Res 2025; 86:e70039. [PMID: 39723688 DOI: 10.1002/ddr.70039] [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: 06/01/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
Epilepsy affects at least 1% of the global population of all socioeconomic backgrounds. Data obtained from previous studies suggest the role of mTOR signaling in epileptogenesis. The present study aimed to investigate the hypothesis that mTOR inhibitor sulfamethizole might produce antiepileptic effects in pentylenetetrazole (PTZ)-induced kindling seizures in mice. For induction of kindling, mice were administered 40 mg/kg PTZ on alternate days for 13 days. The severity of kindling was analyzed using a seizure intensity score. Rotarod performance, actophotometer, and chimney tests were performed to check muscle coordination and locomotor functions. mTOR and IL-6 levels were measured in the brain homogenate. Histological analyses were done using hematoxylin-eosin and cresyl violet stains. Sulfamethizole was administered daily at 10 and 50 mg/kg doses. PTZ administration resulted in kindling seizures in the PTZ-veh group. In addition, mice from the PTZ-veh group showed decreased fall time in rotarod performance, reduced locomotor activity, and failed chimney tests. mTOR and IL-6 levels were also increased in the brain, along with neuronal degeneration and a decreased layer of neuronal cells in the hippocampus. Treatment with sulfamethizole at 50 mg/kg significantly ameliorated seizure intensity score, seizure latency and duration, muscle coordination, and locomotor functions compared to the PTZ-veh group. It also downregulated brain mTOR and IL-6 expression significantly. In conclusion, sulfamethizole showed antiepileptic activity against PTZ-induced kindling seizure in mice via mTOR inhibition.
Collapse
Affiliation(s)
- Lazari Kambli
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, Maharashtra, India
| | - Dezaree Raut
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, Maharashtra, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, Maharashtra, India
| |
Collapse
|
4
|
Yui K, Imataka G, Yuge K, Sasaki H, Shiohama T, Asada K, Tachiki H. The Development of Methods of BLOTCHIP ®-MS for Peptidome: Small Samples in Tuberous Sclerosis. Curr Issues Mol Biol 2025; 47:34. [PMID: 39852149 PMCID: PMC11763708 DOI: 10.3390/cimb47010034] [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: 11/17/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
Mutations in TSC1 or TSC2 in axons induce tuberous sclerosis complex. Neurological manifestations mainly include epilepsy and autism spectrum disorder (ASD). ASD is the presenting symptom (25-50% of patients). ASD was observed at significantly higher frequencies in participants with TSC2 than those with TSC1 mutations. The occurrence of TSC2 mutations is about 50% larger than TSC1. Therefore, ASD may develop due to TSC2 deficiency. TSC2 regulates microRNA biogenesis and Microprocessor activity via GSK3β. Of reference, everolimus has the best treatment target because of the higher potency of interactions with mTORC2 rather than rapamycin. Mutations in the TSC1 and TSC2 genes result in the constitutive hyperactivation of the mammalian target of the rapamycin (mTOR) pathway, contributing to the growth of benign tumors or hamartomas in various organs. TSC2 mutations were associated with a more severe phenotypic spectrum than TSC1 mutations because of the inhibition of the mTOR cascade. There are few studies on the peptide analysis of this disorder in relation to everolimus. Only one study reported that, in ten plasma samples, pre-melanosome protein (PMEL) and S-adenosylmethionine (SAM) were significantly changed as diagnostic prognostic effects. Our study on peptide analysis in Protosera Inc (Osaka, Japan) revealed that three peptides that were related to inflammation in two patients with tuberous sclerosis, who showed a 30% decrease in ASD symptoms following everolimus treatment. TSC2 mutations were associated with a more severe phenotypic spectrum due to the inhibition of the mTOR cascade. PMEL and SAM were significantly changed as diagnostic effects.
Collapse
Affiliation(s)
- Kunio Yui
- Department of Pediatrics, Chiba University, Chiba-Shi 260-8677, Chiba, Japan;
| | - George Imataka
- Department of Pediatrics, Dokkyo Medical University, Tochigi 321-0293, Tochigi, Japan;
| | - Kotaro Yuge
- Department of Pediatrics, Kurume University, Kurume-Shi 830-0011, Fukuoka, Japan;
| | - Hitomi Sasaki
- Department of Urology, Fujita University, Toyoake-Shi 470-1192, Aichi, Japan;
| | - Tadashi Shiohama
- Department of Pediatrics, Chiba University, Chiba-Shi 260-8677, Chiba, Japan;
| | - Kyoichi Asada
- Protosera Inc., Settsu-Shi 566-0002, Osaka, Japan; (K.A.); (H.T.)
| | - Hidehisa Tachiki
- Protosera Inc., Settsu-Shi 566-0002, Osaka, Japan; (K.A.); (H.T.)
| |
Collapse
|
5
|
Aungaroon G, Cooke A, Ritter D, Krueger D, Horn P, Franz DN. Cenobamate's Efficacy for Seizure Treatment in Tuberous Sclerosis Complex. Pediatr Neurol 2024; 161:201-207. [PMID: 39426344 DOI: 10.1016/j.pediatrneurol.2024.09.023] [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: 03/18/2024] [Revised: 08/16/2024] [Accepted: 09/20/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Epilepsy is prevalent, and seizure control is challenging in patients with tuberous sclerosis complex (TSC). Cenobamate (CBM) has proven efficacy in several studies; however, its benefit in the TSC population is not known. METHODS We performed a retrospective review of patients with TSC who received adjunctive CBM for seizure treatments. We assessed treatment efficacy by comparing seizure frequencies three months before CBM (baseline) and those at 3-, 6-, 12-, and 18- month follow-ups. RESULTS We identified 70 patients with TSC receiving CBM and excluded 16 with insufficient data. Fifty-four patients aged 2 to 39 years, with an average baseline seizure of 66.1 ± 88.9 per month, were analyzed. Treatment retention rates at 3, 6, 12, and 18 months were 94.4%, 79.6%, 66.7%, 44.4%, and responder rates (proportions of patients who remained on treatment and had ≥50% seizure reduction) were 38.1%, 51.7%, 53.1%, and 59.1%, respectively. Seizure-free rates at these respective follow-ups were 7.1%, 13.8%, 6.3%, and 9.1%. For patients experiencing reduced seizures, the mean percentage of change ranged from 61.5% to 74.6%. Side effects were common (64.8%), particularly sedation (42.6%), behavioral disturbance (24.1%), and gastrointestinal disturbance (22.2%). CONCLUSIONS Most patients in this study showed seizure reduction; however, the overall responder and seizure-free rates were lower than the literature, likely due to the unique underlying epileptogenesis in TSC and the challenges of tolerating CBM. The lower treatment retention rates signal areas for improvement in concurrent medication adjustment practices.
Collapse
Affiliation(s)
- Gewalin Aungaroon
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Pediatrics, University of Cincinnati, Cincinnati, Ohio.
| | - Alexander Cooke
- University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David Ritter
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Darcy Krueger
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Paul Horn
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - David N Franz
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
6
|
Nord C, Jones I, Garcia-Maestre M, Hägglund AC, Carlsson L. Reduced mTORC1-signaling in progenitor cells leads to retinal lamination deficits. Dev Dyn 2024; 253:922-939. [PMID: 38546215 DOI: 10.1002/dvdy.707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 10/02/2024] Open
Abstract
BACKGROUND Neuronal lamination is a hallmark of the mammalian central nervous system (CNS) and underlies connectivity and function. Initial formation of this tissue architecture involves the integration of various signaling pathways that regulate the differentiation and migration of neural progenitor cells. RESULTS Here, we demonstrate that mTORC1 mediates critical roles during neuronal lamination using the mouse retina as a model system. Down-regulation of mTORC1-signaling in retinal progenitor cells by conditional deletion of Rptor led to decreases in proliferation and increased apoptosis during embryogenesis. These developmental deficits preceded aberrant lamination in adult animals which was best exemplified by the fusion of the outer and inner nuclear layer and the absence of an outer plexiform layer. Moreover, ganglion cell axons originating from each Rptor-ablated retina appeared to segregate to an equal degree at the optic chiasm with both contralateral and ipsilateral projections displaying overlapping termination topographies within several retinorecipient nuclei. In combination, these visual pathway defects led to visually mediated behavioral deficits. CONCLUSIONS This study establishes a critical role for mTORC1-signaling during retinal lamination and demonstrates that this pathway regulates diverse developmental mechanisms involved in driving the stratified arrangement of neurons during CNS development.
Collapse
Affiliation(s)
- Christoffer Nord
- Umeå Center for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
| | - Iwan Jones
- Umeå Center for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
| | | | | | - Leif Carlsson
- Umeå Center for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
| |
Collapse
|
7
|
Kommaddi RP, Gowaikar R, P A H, Diwakar L, Singh K, Mondal A. Akt activation ameliorates deficits in hippocampal-dependent memory and activity-dependent synaptic protein synthesis in an Alzheimer's disease mouse model. J Biol Chem 2024; 300:105619. [PMID: 38182004 PMCID: PMC10839450 DOI: 10.1016/j.jbc.2023.105619] [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: 08/24/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/07/2024] Open
Abstract
Protein kinase-B (Akt) and the mechanistic target of rapamycin (mTOR) signaling pathways are implicated in Alzheimer's disease (AD) pathology. Akt/mTOR signaling pathways, activated by external inputs, enable new protein synthesis at the synapse and synaptic plasticity. The molecular mechanisms impeding new protein synthesis at the synapse in AD pathogenesis remain elusive. Here, we aimed to understand the molecular mechanisms prior to the manifestation of histopathological hallmarks by characterizing Akt1/mTOR signaling cascades and new protein synthesis in the hippocampus of WT and amyloid precursor protein/presenilin-1 (APP/PS1) male mice. Intriguingly, compared to those in WT mice, we found significant decreases in pAkt1, pGSK3β, pmTOR, pS6 ribosomal protein, and p4E-BP1 levels in both post nuclear supernatant and synaptosomes isolated from the hippocampus of one-month-old (presymptomatic) APP/PS1 mice. In synaptoneurosomes prepared from the hippocampus of presymptomatic APP/PS1 mice, activity-dependent protein synthesis at the synapse was impaired and this deficit was sustained in young adults. In hippocampal neurons from C57BL/6 mice, downregulation of Akt1 precluded synaptic activity-dependent protein synthesis at the dendrites but not in the soma. In three-month-old APP/PS1 mice, Akt activator (SC79) administration restored deficits in memory recall and activity-dependent synaptic protein synthesis. C57BL/6 mice administered with an Akt inhibitor (MK2206) resulted in memory recall deficits compared to those treated with vehicle. We conclude that dysregulation of Akt1/mTOR and its downstream signaling molecules in the hippocampus contribute to memory recall deficits and loss of activity-dependent synaptic protein synthesis. In AD mice, however, Akt activation ameliorates deficits in memory recall and activity-dependent synaptic protein synthesis.
Collapse
Affiliation(s)
| | - Ruturaj Gowaikar
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Haseena P A
- Centre for Brain Research, Indian Institute of Science, Bangalore, India; Manipal Academy of Higher Education, Manipal, India
| | - Latha Diwakar
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| | - Kunal Singh
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Amrita Mondal
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| |
Collapse
|
8
|
Rahman M, Nguyen TM, Lee GJ, Kim B, Park MK, Lee CH. Unraveling the Role of Ras Homolog Enriched in Brain (Rheb1 and Rheb2): Bridging Neuronal Dynamics and Cancer Pathogenesis through Mechanistic Target of Rapamycin Signaling. Int J Mol Sci 2024; 25:1489. [PMID: 38338768 PMCID: PMC10855792 DOI: 10.3390/ijms25031489] [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: 12/15/2023] [Revised: 01/14/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024] Open
Abstract
Ras homolog enriched in brain (Rheb1 and Rheb2), small GTPases, play a crucial role in regulating neuronal activity and have gained attention for their implications in cancer development, particularly in breast cancer. This study delves into the intricate connection between the multifaceted functions of Rheb1 in neurons and cancer, with a specific focus on the mTOR pathway. It aims to elucidate Rheb1's involvement in pivotal cellular processes such as proliferation, apoptosis resistance, migration, invasion, metastasis, and inflammatory responses while acknowledging that Rheb2 has not been extensively studied. Despite the recognized associations, a comprehensive understanding of the intricate interplay between Rheb1 and Rheb2 and their roles in both nerve and cancer remains elusive. This review consolidates current knowledge regarding the impact of Rheb1 on cancer hallmarks and explores the potential of Rheb1 as a therapeutic target in cancer treatment. It emphasizes the necessity for a deeper comprehension of the molecular mechanisms underlying Rheb1-mediated oncogenic processes, underscoring the existing gaps in our understanding. Additionally, the review highlights the exploration of Rheb1 inhibitors as a promising avenue for cancer therapy. By shedding light on the complicated roles between Rheb1/Rheb2 and cancer, this study provides valuable insights to the scientific community. These insights are instrumental in guiding the identification of novel targets and advancing the development of effective therapeutic strategies for treating cancer.
Collapse
Affiliation(s)
- Mostafizur Rahman
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea; (M.R.); (G.J.L.)
| | - Tuan Minh Nguyen
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea; (M.R.); (G.J.L.)
| | - Gi Jeong Lee
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea; (M.R.); (G.J.L.)
| | - Boram Kim
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea; (M.R.); (G.J.L.)
| | - Mi Kyung Park
- Department of BioHealthcare, Hwasung Medi-Science University, Hwaseong-si 18274, Republic of Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea; (M.R.); (G.J.L.)
| |
Collapse
|
9
|
Goldberg AR, Dovas A, Torres D, Sharma SD, Mela A, Merricks EM, Olabarria M, Shokooh LA, Zhao HT, Kotidis C, Calvaresi P, Viswanathan A, Banu MA, Razavilar A, Sudhakar TD, Saxena A, Chokran C, Humala N, Mahajan A, Xu W, Metz JB, Chen C, Bushong EA, Boassa D, Ellisman MH, Hillman EM, McKhann GM, Gill BJA, Rosenfeld SS, Schevon CA, Bruce JN, Sims PA, Peterka DS, Canoll P. Glioma-Induced Alterations in Excitatory Neurons are Reversed by mTOR Inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575092. [PMID: 38293120 PMCID: PMC10827113 DOI: 10.1101/2024.01.10.575092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Gliomas are highly aggressive brain tumors characterized by poor prognosis and composed of diffusely infiltrating tumor cells that intermingle with non-neoplastic cells in the tumor microenvironment, including neurons. Neurons are increasingly appreciated as important reactive components of the glioma microenvironment, due to their role in causing hallmark glioma symptoms, such as cognitive deficits and seizures, as well as their potential ability to drive glioma progression. Separately, mTOR signaling has been shown to have pleiotropic effects in the brain tumor microenvironment, including regulation of neuronal hyperexcitability. However, the local cellular-level effects of mTOR inhibition on glioma-induced neuronal alterations are not well understood. Here we employed neuron-specific profiling of ribosome-bound mRNA via 'RiboTag,' morphometric analysis of dendritic spines, and in vivo calcium imaging, along with pharmacological mTOR inhibition to investigate the impact of glioma burden and mTOR inhibition on these neuronal alterations. The RiboTag analysis of tumor-associated excitatory neurons showed a downregulation of transcripts encoding excitatory and inhibitory postsynaptic proteins and dendritic spine development, and an upregulation of transcripts encoding cytoskeletal proteins involved in dendritic spine turnover. Light and electron microscopy of tumor-associated excitatory neurons demonstrated marked decreases in dendritic spine density. In vivo two-photon calcium imaging in tumor-associated excitatory neurons revealed progressive alterations in neuronal activity, both at the population and single-neuron level, throughout tumor growth. This in vivo calcium imaging also revealed altered stimulus-evoked somatic calcium events, with changes in event rate, size, and temporal alignment to stimulus, which was most pronounced in neurons with high-tumor burden. A single acute dose of AZD8055, a combined mTORC1/2 inhibitor, reversed the glioma-induced alterations on the excitatory neurons, including the alterations in ribosome-bound transcripts, dendritic spine density, and stimulus evoked responses seen by calcium imaging. These results point to mTOR-driven pathological plasticity in neurons at the infiltrative margin of glioma - manifested by alterations in ribosome-bound mRNA, dendritic spine density, and stimulus-evoked neuronal activity. Collectively, our work identifies the pathological changes that tumor-associated excitatory neurons experience as both hyperlocal and reversible under the influence of mTOR inhibition, providing a foundation for developing therapies targeting neuronal signaling in glioma.
Collapse
Affiliation(s)
- Alexander R. Goldberg
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Daniela Torres
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sohani Das Sharma
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Edward M. Merricks
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Markel Olabarria
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | | | - Hanzhi T. Zhao
- Laboratory for Functional Optical Imaging, Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Corina Kotidis
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter Calvaresi
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ashwin Viswanathan
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Matei A. Banu
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aida Razavilar
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Tejaswi D. Sudhakar
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ankita Saxena
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cole Chokran
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nelson Humala
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aayushi Mahajan
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Weihao Xu
- Laboratory for Functional Optical Imaging, Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Jordan B. Metz
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Cady Chen
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Eric A. Bushong
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniela Boassa
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mark H. Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Elizabeth M.C. Hillman
- Laboratory for Functional Optical Imaging, Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Guy M. McKhann
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Brian J. A. Gill
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | | | - Catherine A. Schevon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jeffrey N. Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter A. Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032
- Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY, 10032
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, 10032
| | - Darcy S. Peterka
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
10
|
McArdle CJ, Arnone AA, Heaney CF, Raab-Graham KF. A paradoxical switch: the implications of excitatory GABAergic signaling in neurological disorders. Front Psychiatry 2024; 14:1296527. [PMID: 38268565 PMCID: PMC10805837 DOI: 10.3389/fpsyt.2023.1296527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/04/2023] [Indexed: 01/26/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system. In the mature brain, inhibitory GABAergic signaling is critical in maintaining neuronal homeostasis and vital human behaviors such as cognition, emotion, and motivation. While classically known to inhibit neuronal function under physiological conditions, previous research indicates a paradoxical switch from inhibitory to excitatory GABAergic signaling that is implicated in several neurological disorders. Various mechanisms have been proposed to contribute to the excitatory switch such as chloride ion dyshomeostasis, alterations in inhibitory receptor expression, and modifications in GABAergic synaptic plasticity. Of note, the hypothesized mechanisms underlying excitatory GABAergic signaling are highlighted in a number of neurodevelopmental, substance use, stress, and neurodegenerative disorders. Herein, we present an updated review discussing the presence of excitatory GABAergic signaling in various neurological disorders, and their potential contributions towards disease pathology.
Collapse
Affiliation(s)
- Colin J. McArdle
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Alana A. Arnone
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of General Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Chelcie F. Heaney
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Kimberly F. Raab-Graham
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
11
|
Kumari S, Brewster AL. Exploring Dendritic and Spine Structural Profiles in Epilepsy: Insights From Human Studies and Experimental Animal Models. Epilepsy Curr 2024; 24:40-46. [PMID: 38327540 PMCID: PMC10846509 DOI: 10.1177/15357597231218603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Dendrites are tree-like structures with tiny spines specialized to receive excitatory synaptic transmission. Spino-dendritic plasticity, driven by neural activity, underlies the maintenance of neuronal connections crucial for proper circuit function. Abnormalities in dendritic morphology are frequently seen in epilepsy. However, the exact etiology or functional implications are not yet known. Therefore, to better comprehend the structure-function significance of this dendritic pathology in epilepsy, it is necessary to identify the common spino-dendritic disturbances present in both human and experimental models. Here, we describe the dendritic and spine structural profiles found across human refractory epilepsy as well as in animal models of developmental, acquired, and genetic epilepsies.
Collapse
Affiliation(s)
- Shikha Kumari
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, USA
| | - Amy L. Brewster
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, USA
| |
Collapse
|
12
|
Park SB, Lim B, Kim KY, Koh B. Long and Short-Term Effect of mTOR Regulation on Cerebral Organoid Growth and Differentiations. Tissue Eng Regen Med 2024; 21:159-169. [PMID: 38153672 PMCID: PMC10764682 DOI: 10.1007/s13770-023-00611-3] [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: 08/03/2023] [Revised: 09/20/2023] [Accepted: 10/29/2023] [Indexed: 12/29/2023] Open
Abstract
BACKGROUND The mammalian target of rapamycin (mTOR) signaling is critical for the maintenance and differentiation of neurogenesis, and conceivably for many other brain developmental processes. However, in vivo studies of mTOR functions in the brain are often hampered due to the essential role of the associated signaling in brain development. METHODS We monitored the long- and short-term effects of mTOR signaling regulation on cerebral organoids growth, differentiation and function using an mTOR inhibitor (everolimus) and an mTOR activator (MHY1485). RESULTS Short-term treatment with MHY1485 induced faster organoid growth and differentiation, while long-term treatment induced the maturation of cerebral organoids. CONCLUSION These data suggest that the optimal activity of mTOR is crucial in maintaining normal brain development, and its role is not confined to the early neurogenic phase of brain development.
Collapse
Affiliation(s)
- Sung Bum Park
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Byungho Lim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Ki Young Kim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea.
| | - Byumseok Koh
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea.
| |
Collapse
|
13
|
Hu JS, Malik R, Sohal VS, Rubenstein JL, Vogt D. Tsc1 Loss in VIP-Lineage Cortical Interneurons Results in More VIP+ Interneurons and Enhanced Excitability. Cells 2023; 13:52. [PMID: 38201256 PMCID: PMC10777938 DOI: 10.3390/cells13010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway is a powerful regulator of cell proliferation, growth, synapse maintenance and cell fate. While intensely studied for its role in cancer, the role of mTOR signaling is just beginning to be uncovered in specific cell types that are implicated in neurodevelopmental disorders. Previously, loss of the Tsc1 gene, which results in hyperactive mTOR, was shown to affect the function and molecular properties of GABAergic cortical interneurons (CINs) derived from the medial ganglionic eminence. To assess if other important classes of CINs could be impacted by mTOR dysfunction, we deleted Tsc1 in a caudal ganglionic eminence-derived interneuron group, the vasoactive intestinal peptide (VIP)+ subtype, whose activity disinhibits local circuits. Tsc1 mutant VIP+ CINs reduced their pattern of apoptosis from postnatal days 15-20, resulting in increased VIP+ CINs. The mutant CINs exhibited synaptic and electrophysiological properties that could contribute to the high rate of seizure activity in humans that harbor Tsc1 mutations.
Collapse
Affiliation(s)
- Jia Sheng Hu
- Department of Psychiatry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Ruchi Malik
- Department of Psychiatry, University of California San Francisco, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
- Center for Integrative Neuroscience, University of California San Francisco, 1550 4th St., San Francisco, CA 94158, USA
- Sloan-Swartz Center for Theoretical Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA 94158, USA
| | - Vikaas S. Sohal
- Department of Psychiatry, University of California San Francisco, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
- Center for Integrative Neuroscience, University of California San Francisco, 1550 4th St., San Francisco, CA 94158, USA
- Sloan-Swartz Center for Theoretical Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA 94158, USA
| | - John L. Rubenstein
- Department of Psychiatry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Daniel Vogt
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI 49503, USA
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
14
|
Jiang J, Zhang L, Zou J, Liu J, Yang J, Jiang Q, Duan P, Jiang B. Phosphorylated S6K1 and 4E-BP1 play different roles in constitutively active Rheb-mediated retinal ganglion cell survival and axon regeneration after optic nerve injury. Neural Regen Res 2023; 18:2526-2534. [PMID: 37282486 PMCID: PMC10360084 DOI: 10.4103/1673-5374.371372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
Ras homolog enriched in brain (Rheb) is a small GTPase that activates mammalian target of rapamycin complex 1 (mTORC1). Previous studies have shown that constitutively active Rheb can enhance the regeneration of sensory axons after spinal cord injury by activating downstream effectors of mTOR. S6K1 and 4E-BP1 are important downstream effectors of mTORC1. In this study, we investigated the role of Rheb/mTOR and its downstream effectors S6K1 and 4E-BP1 in the protection of retinal ganglion cells. We transfected an optic nerve crush mouse model with adeno-associated viral 2-mediated constitutively active Rheb and observed the effects on retinal ganglion cell survival and axon regeneration. We found that overexpression of constitutively active Rheb promoted survival of retinal ganglion cells in the acute (14 days) and chronic (21 and 42 days) stages of injury. We also found that either co-expression of the dominant-negative S6K1 mutant or the constitutively active 4E-BP1 mutant together with constitutively active Rheb markedly inhibited axon regeneration of retinal ganglion cells. This suggests that mTORC1-mediated S6K1 activation and 4E-BP1 inhibition were necessary components for constitutively active Rheb-induced axon regeneration. However, only S6K1 activation, but not 4E-BP1 knockdown, induced axon regeneration when applied alone. Furthermore, S6K1 activation promoted the survival of retinal ganglion cells at 14 days post-injury, whereas 4E-BP1 knockdown unexpectedly slightly decreased the survival of retinal ganglion cells at 14 days post-injury. Overexpression of constitutively active 4E-BP1 increased the survival of retinal ganglion cells at 14 days post-injury. Likewise, co-expressing constitutively active Rheb and constitutively active 4E-BP1 markedly increased the survival of retinal ganglion cells compared with overexpression of constitutively active Rheb alone at 14 days post-injury. These findings indicate that functional 4E-BP1 and S6K1 are neuroprotective and that 4E-BP1 may exert protective effects through a pathway at least partially independent of Rheb/mTOR. Together, our results show that constitutively active Rheb promotes the survival of retinal ganglion cells and axon regeneration through modulating S6K1 and 4E-BP1 activity. Phosphorylated S6K1 and 4E-BP1 promote axon regeneration but play an antagonistic role in the survival of retinal ganglion cells.
Collapse
Affiliation(s)
- Jikuan Jiang
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Lusi Zhang
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Jingling Zou
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Jingyuan Liu
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Jia Yang
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Qian Jiang
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Peiyun Duan
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Bing Jiang
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| |
Collapse
|
15
|
Herron RS, Kunisky AK, Madden JR, Anyaeche VI, Maung MZ, Hwang HW. A twin UGUA motif directs the balance between gene isoforms through CFIm and the mTORC1 signaling pathway. eLife 2023; 12:e85036. [PMID: 37665675 PMCID: PMC10476966 DOI: 10.7554/elife.85036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 08/16/2023] [Indexed: 09/06/2023] Open
Abstract
Alternative polyadenylation (APA) generates mRNA isoforms and diversifies gene expression. Here we report the discovery that the mTORC1 signaling pathway balances the expression of two Trim9/TRIM9 isoforms through APA regulation in human and mouse. We showed that CFIm components, CPSF6 and NUDT21, promote the short Trim9/TRIM9 isoform (Trim9-S/TRIM9-S) expression. In addition, we identified an evolutionarily conserved twin UGUA motif, UGUAYUGUA, in TRIM9-S polyadenylation site (PAS) that is critical for its regulation by CPSF6. We found additional CPSF6-regulated PASs with similar twin UGUA motifs in human and experimentally validated the twin UGUA motif functionality in BMPR1B, MOB4, and BRD4-L. Importantly, we showed that inserting a twin UGUA motif into a heterologous PAS was sufficient to confer regulation by CPSF6 and mTORC1. Our study reveals an evolutionarily conserved mechanism to regulate gene isoform expression by mTORC1 and implicates possible gene isoform imbalance in cancer and neurological disorders with mTORC1 pathway dysregulation.
Collapse
Affiliation(s)
- R Samuel Herron
- Department of Pathology, University of PittsburghPittsburghUnited States
| | | | - Jessica R Madden
- Department of Pathology, University of PittsburghPittsburghUnited States
| | - Vivian I Anyaeche
- Department of Pathology, University of PittsburghPittsburghUnited States
| | - May Z Maung
- Department of Biological Sciences, University of PittsburghPittsburghUnited States
| | - Hun-Way Hwang
- Department of Pathology, University of PittsburghPittsburghUnited States
| |
Collapse
|
16
|
Winden KD, Pham TT, Teaney NA, Ruiz J, Chen R, Chen C, Sahin M. Increased degradation of FMRP contributes to neuronal hyperexcitability in tuberous sclerosis complex. Cell Rep 2023; 42:112838. [PMID: 37494191 PMCID: PMC10529098 DOI: 10.1016/j.celrep.2023.112838] [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: 12/16/2022] [Revised: 05/12/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023] Open
Abstract
Autism spectrum disorder (ASD) is a highly prevalent neurodevelopmental disorder, but new therapies have been impeded by a lack of understanding of the pathological mechanisms. Tuberous sclerosis complex (TSC) and fragile X syndrome are associated with alterations in the mechanistic target of rapamycin (mTOR) and fragile X messenger ribonucleoprotein 1 (FMRP), which have been implicated in the development of ASD. Previously, we observed that transcripts associated with FMRP were down-regulated in TSC2-deficient neurons. In this study, we find that FMRP turnover is dysregulated in TSC2-deficient rodent primary neurons and human induced pluripotent stem cell (iPSC)-derived neurons and is dependent on the E3 ubiquitin ligase anaphase-promoting complex. We also demonstrate that overexpression of FMRP can partially rescue hyperexcitability in TSC2-deficient iPSC-derived neurons. These data indicate that FMRP dysregulation represents an important pathological mechanism in the development of abnormal neuronal activity in TSC and illustrate a molecular convergence between these two neurogenetic disorders.
Collapse
Affiliation(s)
- Kellen D Winden
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Truc T Pham
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nicole A Teaney
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Juan Ruiz
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan Chen
- Human Neuron Core, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Cidi Chen
- Human Neuron Core, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Dong R, Li X, Flores AD, Lai KO. The translation initiating factor eIF4E and arginine methylation underlie G3BP1 function in dendritic spine development of neurons. J Biol Chem 2023; 299:105029. [PMID: 37442236 PMCID: PMC10432808 DOI: 10.1016/j.jbc.2023.105029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Communication between neurons relies on neurotransmission that takes place at synapses. Excitatory synapses are located primarily on dendritic spines that possess diverse morphologies, ranging from elongated filopodia to mushroom-shaped spines. Failure in the proper development of dendritic spines has detrimental consequences on neuronal connectivity, but the molecular mechanism that controls the balance of filopodia and mushroom spines is not well understood. G3BP1 is the key RNA-binding protein that assembles the stress granules in non-neuronal cells to adjust protein synthesis upon exogenous stress. Emerging evidence suggests that the biological significance of G3BP1 extends beyond its role in stress response, especially in the nervous system. However, the mechanism underlying the regulation and function of G3BP1 in neurons remains elusive. Here we found that G3BP1 suppresses protein synthesis and binds to the translation initiation factor eIF4E via its NTF2-like domain. Notably, the over-production of filopodia caused by G3BP1 depletion can be alleviated by blocking the formation of the translation initiation complex. We further found that the interaction of G3BP1 with eIF4E is regulated by arginine methylation. Knockdown of the protein arginine methyltransferase PRMT8 leads to elevated protein synthesis and filopodia production, which is reversed by the expression of methylation-mimetic G3BP1. Our study, therefore, reveals arginine methylation as a key regulatory mechanism of G3BP1 during dendritic spine morphogenesis and identifies eIF4E as a novel downstream target of G3BP1 in neuronal development independent of stress response.
Collapse
Affiliation(s)
- Rui Dong
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Xuejun Li
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China; Hong Kong Institute for Advanced Study, City University of Hong Kong, Hong Kong, China
| | - Angelo D Flores
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Kwok-On Lai
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China; Hong Kong Institute for Advanced Study, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
18
|
Gąssowska-Dobrowolska M, Czapski GA, Cieślik M, Zajdel K, Frontczak-Baniewicz M, Babiec L, Adamczyk A. Microtubule Cytoskeletal Network Alterations in a Transgenic Model of Tuberous Sclerosis Complex: Relevance to Autism Spectrum Disorders. Int J Mol Sci 2023; 24:7303. [PMID: 37108467 PMCID: PMC10138344 DOI: 10.3390/ijms24087303] [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: 02/19/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare genetic multisystem disorder caused by loss-of-function mutations in the tumour suppressors TSC1/TSC2, both of which are negative regulators of the mammalian target of rapamycin (mTOR) kinase. Importantly, mTOR hyperactivity seems to be linked with the pathobiology of autism spectrum disorders (ASD). Recent studies suggest the potential involvement of microtubule (MT) network dysfunction in the neuropathology of "mTORopathies", including ASD. Cytoskeletal reorganization could be responsible for neuroplasticity disturbances in ASD individuals. Thus, the aim of this work was to study the effect of Tsc2 haploinsufficiency on the cytoskeletal pathology and disturbances in the proteostasis of the key cytoskeletal proteins in the brain of a TSC mouse model of ASD. Western-blot analysis indicated significant brain-structure-dependent abnormalities in the microtubule-associated protein Tau (MAP-Tau), and reduced MAP1B and neurofilament light (NF-L) protein level in 2-month-old male B6;129S4-Tsc2tm1Djk/J mice. Alongside, pathological irregularities in the ultrastructure of both MT and neurofilament (NFL) networks as well as swelling of the nerve endings were demonstrated. These changes in the level of key cytoskeletal proteins in the brain of the autistic-like TSC mice suggest the possible molecular mechanisms responsible for neuroplasticity alterations in the ASD brain.
Collapse
Affiliation(s)
- Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
| | - Grzegorz A. Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
| | - Magdalena Cieślik
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
| | - Karolina Zajdel
- Electron Microscopy Research Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
| | - Małgorzata Frontczak-Baniewicz
- Electron Microscopy Research Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
| | - Lidia Babiec
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
| |
Collapse
|
19
|
Wang Y, Lin J, Li J, Yan L, Li W, He X, Ma H. Chronic Neuronal Inactivity Utilizes the mTOR-TFEB Pathway to Drive Transcription-Dependent Autophagy for Homeostatic Up-Scaling. J Neurosci 2023; 43:2631-2652. [PMID: 36868861 PMCID: PMC10089247 DOI: 10.1523/jneurosci.0146-23.2023] [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: 01/24/2023] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023] Open
Abstract
Activity-dependent changes in protein expression are critical for neuronal plasticity, a fundamental process for the processing and storage of information in the brain. Among the various forms of plasticity, homeostatic synaptic up-scaling is unique in that it is induced primarily by neuronal inactivity. However, precisely how the turnover of synaptic proteins occurs in this homeostatic process remains unclear. Here, we report that chronically inhibiting neuronal activity in primary cortical neurons prepared from embryonic day (E)18 Sprague Dawley rats (both sexes) induces autophagy, thereby regulating key synaptic proteins for up-scaling. Mechanistically, chronic neuronal inactivity causes dephosphorylation of ERK and mTOR, which induces transcription factor EB (TFEB)-mediated cytonuclear signaling and drives transcription-dependent autophagy to regulate αCaMKII and PSD95 during synaptic up-scaling. Together, these findings suggest that mTOR-dependent autophagy, which is often triggered by metabolic stressors such as starvation, is recruited and sustained during neuronal inactivity to maintain synaptic homeostasis, a process that ensures proper brain function and if impaired can cause neuropsychiatric disorders such as autism.SIGNIFICANCE STATEMENT In the mammalian brain, protein turnover is tightly controlled by neuronal activation to ensure key neuronal functions during long-lasting synaptic plasticity. However, a long-standing question is how this process occurs during synaptic up-scaling, a process that requires protein turnover but is induced by neuronal inactivation. Here, we report that mTOR-dependent signaling, which is often triggered by metabolic stressors such as starvation, is "hijacked" by chronic neuronal inactivation, which then serves as a nucleation point for transcription factor EB (TFEB) cytonuclear signaling that drives transcription-dependent autophagy for up-scaling. These results provide the first evidence of a physiological role of mTOR-dependent autophagy in enduing neuronal plasticity, thereby connecting major themes in cell biology and neuroscience via a servo loop that mediates autoregulation in the brain.
Collapse
Affiliation(s)
- Yang Wang
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Jingran Lin
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Jiarui Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Lu Yan
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Wenwen Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Xingzhi He
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Huan Ma
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
- Research Units for Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing 100050, China
| |
Collapse
|
20
|
D’Antona L, Amato R, Brescia C, Rocca V, Colao E, Iuliano R, Blazer-Yost BL, Perrotti N. Kinase Inhibitors in Genetic Diseases. Int J Mol Sci 2023; 24:ijms24065276. [PMID: 36982349 PMCID: PMC10048847 DOI: 10.3390/ijms24065276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Over the years, several studies have shown that kinase-regulated signaling pathways are involved in the development of rare genetic diseases. The study of the mechanisms underlying the onset of these diseases has opened a possible way for the development of targeted therapies using particular kinase inhibitors. Some of these are currently used to treat other diseases, such as cancer. This review aims to describe the possibilities of using kinase inhibitors in genetic pathologies such as tuberous sclerosis, RASopathies, and ciliopathies, describing the various pathways involved and the possible targets already identified or currently under study.
Collapse
Affiliation(s)
- Lucia D’Antona
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Rosario Amato
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Carolina Brescia
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
| | - Valentina Rocca
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
- Department of Experimental and Clinical Medicine, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
| | - Emma Colao
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Rodolfo Iuliano
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Bonnie L. Blazer-Yost
- Department of Biology, Indiana University Purdue University, Indianapolis, IN 46202, USA
| | - Nicola Perrotti
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
- Correspondence:
| |
Collapse
|
21
|
Role of mTOR1 signaling in the antidepressant effects of ketamine and the potential of mTORC1 activators as novel antidepressants. Neuropharmacology 2023; 223:109325. [PMID: 36334763 DOI: 10.1016/j.neuropharm.2022.109325] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022]
Abstract
Conventional antidepressant medications act on monoaminergic systems and have important limitations, including a therapeutic delay of weeks to months and low rates of efficacy. Recently, clinical findings have indicated that ketamine, a dissociative anesthetic that blocks N-methyl-d-aspartate receptor channel activity, causes rapid and long-lasting antidepressant effects. Although the exact mechanisms underlying the antidepressant effects of ketamine are not fully known, preclinical studies have demonstrated a key role for mechanistic target of rapamycin complex 1 (mTORC1) signaling and a subsequent increase in synapse formation in the medial prefrontal cortex. In this review, we discuss the role of mTORC1 and its subsequent signaling cascade in the antidepressant effects of ketamine and other compounds with glutamatergic mechanisms of action. We also present the possibility that mTORC1 signaling itself is a drug discovery target.
Collapse
|
22
|
Huang KC, Gomes C, Shiga Y, Belforte N, VanderWall KB, Lavekar SS, Fligor CM, Harkin J, Di Polo A, Meyer JS. Autophagy disruption reduces mTORC1 activation leading to retinal ganglion cell neurodegeneration associated with glaucoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.04.522687. [PMID: 36711831 PMCID: PMC9881969 DOI: 10.1101/2023.01.04.522687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Autophagy dysfunction has been associated with several neurodegenerative diseases including glaucoma, characterized by the degeneration of retinal ganglion cells (RGCs). However, the mechanisms by which autophagy dysfunction promotes RGC damage remain unclear. Here, we hypothesized that perturbation of the autophagy pathway results in increased autophagic demand, thereby downregulating signaling through mammalian target of rapamycin complex 1 (mTORC1), a negative regulator of autophagy, contributing to the degeneration of RGCs. We identified an impairment of autophagic-lysosomal degradation and decreased mTORC1 signaling via activation of the stress sensor adenosine monophosphate-activated protein kinase (AMPK), along with subsequent neurodegeneration in RGCs differentiated from human pluripotent stem cells (hPSCs) with a glaucoma-associated variant of Optineurin (OPTN-E50K). Similarly, the microbead occlusion model of glaucoma resulting in ocular hypertension also exhibited autophagy disruption and mTORC1 downregulation. Pharmacological inhibition of mTORC1 in hPSC-derived RGCs recapitulated disease-related neurodegenerative phenotypes in otherwise healthy RGCs, while the mTOR-independent induction of autophagy reduced protein accumulation and restored neurite outgrowth in diseased OPTN-E50K RGCs. Taken together, these results highlight an important balance between autophagy and mTORC1 signaling essential for RGC homeostasis, while disruption to these pathways contributes to neurodegenerative features in glaucoma, providing a potential therapeutic target to prevent neurodegeneration.
Collapse
Affiliation(s)
- Kang-Chieh Huang
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis IN USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN USA
| | - Cátia Gomes
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis IN USA
| | - Yukihiro Shiga
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada
| | - Nicolas Belforte
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada
| | - Kirstin B. VanderWall
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis IN USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN USA
| | - Sailee S. Lavekar
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis IN USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN USA
| | - Clarisse M. Fligor
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis IN USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN USA
| | - Jade Harkin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis IN USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada
| | - Jason S. Meyer
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis IN USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis IN USA
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis IN USA
| |
Collapse
|
23
|
Granak S, Tuckova K, Kutna V, Vojtechova I, Bajkova L, Petrasek T, Ovsepian SV. Developmental effects of constitutive mTORC1 hyperactivity and environmental enrichment on structural synaptic plasticity and behaviour in a rat model of autism spectrum disorder. Eur J Neurosci 2023; 57:17-31. [PMID: 36380588 DOI: 10.1111/ejn.15864] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/18/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition causing a range of social and communication impairments. Although the role of multiple genes and environmental factors has been reported, the effects of the interplay between genes and environment on the onset and progression of the disease remains elusive. We housed wild-type (Tsc2+/+) and tuberous sclerosis 2 deficient (Tsc2+/-) Eker rats (ASD model) in individually ventilated cages or enriched conditions and conducted a series of behavioural tests followed by the histochemical analysis of dendritic spines and plasticity in three age groups (days 45, 90 and 365). The elevated plus-maze test revealed a reduction of anxiety by enrichment, whereas the mobility of young and adult Eker rats in the open field was lower compared to the wild type. In the social interaction test, an enriched environment reduced social contact in the youngest group and increased anogenital exploration in 90- and 365-day-old rats. Self-grooming was increased by environmental enrichment in young and adult rats and decreased in aged Eker rats. Dendritic spine counts revealed an increased spine density in the cingulate gyrus in adult Ekers irrespective of housing conditions, whereas spine density in hippocampal pyramidal neurons was comparable across all genotypes and groups. Morphometric analysis of dendritic spines revealed age-related changes in spine morphology and density, which were responsive to animal genotype and environment. Taken together, our findings suggest that under TSC2 haploinsufficiency and mTORC1 hyperactivity, the expression of behavioural signs and neuroplasticity in Eker rats can be differentially influenced by the developmental stage and environment.
Collapse
Affiliation(s)
- Simon Granak
- National Institute of Mental Health, Klecany, Czech Republic.,Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Klara Tuckova
- National Institute of Mental Health, Klecany, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Viera Kutna
- National Institute of Mental Health, Klecany, Czech Republic
| | - Iveta Vojtechova
- National Institute of Mental Health, Klecany, Czech Republic.,Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Laura Bajkova
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomas Petrasek
- National Institute of Mental Health, Klecany, Czech Republic.,Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Saak V Ovsepian
- Faculty of Science and Engineering, University of Greenwich London, Chatham Maritime, UK
| |
Collapse
|
24
|
Shimada T, Yamagata K. Spine morphogenesis and synapse formation in tubular sclerosis complex models. Front Mol Neurosci 2022; 15:1019343. [PMID: 36606143 PMCID: PMC9807618 DOI: 10.3389/fnmol.2022.1019343] [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: 08/15/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is caused by mutations in the Tsc1 or Tsc2 genes, whose products form a complex and inactivate the small G-protein Rheb1. The activation of Rheb1 may cause refractory epilepsy, intellectual disability, and autism, which are the major neuropsychiatric manifestations of TSC. Abnormalities in dendritic spines and altered synaptic structure are hallmarks of epilepsy, intellectual disability, and autism. In addition, spine dysmorphology and aberrant synapse formation are observed in TSC animal models. Therefore, it is important to investigate the molecular mechanism underlying the regulation of spine morphology and synapse formation in neurons to identify therapeutic targets for TSC. In this review, we focus on the representative proteins regulated by Rheb1 activity, mTORC1 and syntenin, which are pivotal downstream factors of Rheb1 in the alteration of spine formation and synapse function in TSC neurons.
Collapse
Affiliation(s)
- Tadayuki Shimada
- Child Brain Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan,*Correspondence: Tadayuki Shimada,
| | - Kanato Yamagata
- Child Brain Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan,Department of Psychiatry, Takada Nishishiro Hospital, Niigata, Japan,Kanato Yamagata,
| |
Collapse
|
25
|
Ye Z, Lin S, Zhao X, Bennett MF, Brown NJ, Wallis M, Gao X, Sun L, Wu J, Vedururu R, Witkowski T, Gardiner F, Stutterd C, Duan J, Mullen SA, McGillivray G, Bodek S, Valente G, Reagan M, Yao Y, Li L, Chen L, Boys A, Adikari TN, Cao D, Hu Z, Beshay V, Zhang VW, Berkovic SF, Scheffer IE, Liao J, Hildebrand MS. Mosaicism in tuberous sclerosis complex: Lowering the threshold for clinical reporting. Hum Mutat 2022; 43:1956-1969. [PMID: 36030538 DOI: 10.1002/humu.24454] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/20/2022] [Accepted: 08/21/2022] [Indexed: 01/25/2023]
Abstract
Tuberous sclerosis complex (TSC) is a multi-system genetic disorder. Most patients have germline mutations in TSC1 or TSC2 but, 10%-15% patients do not have TSC1/TSC2 mutations detected on routine clinical genetic testing. We investigated the contribution of low-level mosaic TSC1/TSC2 mutations in unsolved sporadic patients and families with TSC. Thirty-one sporadic TSC patients negative on routine testing and eight families with suspected parental mosaicism were sequenced using deep panel sequencing followed by droplet digital polymerase chain reaction. Pathogenic variants were found in 22/31 (71%) unsolved sporadic patients, 16 were mosaic (median variant allele fraction [VAF] 6.8% in blood) and 6 had missed germline mutations. Parental mosaicism was detected in 5/8 families (median VAF 1% in blood). Clinical testing laboratories typically only report pathogenic variants with allele fractions above 10%. Our findings highlight the critical need to change laboratory practice by implementing higher sensitivity assays to improve diagnostic yield, inform patient management and guide reproductive counseling.
Collapse
Affiliation(s)
- Zimeng Ye
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Sufang Lin
- Department of Neurology, Epilepsy Centre, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Xia Zhao
- Department of Neurology, Epilepsy Centre, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Mark F Bennett
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Natasha J Brown
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Royal Children's Hospital, The University of Melbourne, Parkville, Victoria, Australia
- Victorian Clinical Genetics Services, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Mathew Wallis
- Austin Health, Heidelberg, Victoria, Australia
- Tasmania Clinical Genetics Service, Royal Hobart Hospital, Tasmania, Australia
- School of Medicine, University of Tasmania, Tasmania, Australia
- Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia
| | - Xinyi Gao
- AmCare Genomics Laboratory, Guangzhou, Guangdong Province, China
| | - Li Sun
- AmCare Genomics Laboratory, Guangzhou, Guangdong Province, China
| | - Jiarui Wu
- AmCare Genomics Laboratory, Guangzhou, Guangdong Province, China
| | - Ravikiran Vedururu
- Molecular Diagnostic Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Tom Witkowski
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Fiona Gardiner
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Chloe Stutterd
- Victorian Clinical Genetics Services, Royal Children's Hospital, Parkville, Victoria, Australia
- Austin Health, Heidelberg, Victoria, Australia
| | - Jing Duan
- Department of Neurology, Epilepsy Centre, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Saul A Mullen
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia
- Austin Health, Heidelberg, Victoria, Australia
| | - George McGillivray
- Victorian Clinical Genetics Services, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Simon Bodek
- Austin Health, Heidelberg, Victoria, Australia
| | | | - Matthew Reagan
- Department of Medicine, Peninsula Health, Monash University, Frankston, Victoria, Australia
| | - Yi Yao
- Department of Neurology, Epilepsy Centre, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Lin Li
- Department of Neurology, Epilepsy Centre, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Li Chen
- Department of Neurology, Epilepsy Centre, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Amber Boys
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Thiuni N Adikari
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia
- School of Medicine, University of Tasmania, Tasmania, Australia
| | - Dezhi Cao
- Department of Neurology, Epilepsy Centre, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Zhanqi Hu
- Department of Neurology, Epilepsy Centre, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Victoria Beshay
- Molecular Diagnostic Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Victor W Zhang
- AmCare Genomics Laboratory, Guangzhou, Guangdong Province, China
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia
- Austin Health, Heidelberg, Victoria, Australia
| | - Ingrid E Scheffer
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Royal Children's Hospital, The University of Melbourne, Parkville, Victoria, Australia
- Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute, Parkville, Victoria, Australia
| | - Jianxiang Liao
- Department of Neurology, Epilepsy Centre, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Michael S Hildebrand
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
26
|
Litwa K. Shared mechanisms of neural circuit disruption in tuberous sclerosis across lifespan: Bridging neurodevelopmental and neurodegenerative pathology. Front Genet 2022; 13:997461. [PMID: 36506334 PMCID: PMC9732432 DOI: 10.3389/fgene.2022.997461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/06/2022] [Indexed: 11/27/2022] Open
Abstract
Tuberous Sclerosis (TS) is a rare genetic disorder manifesting with multiple benign tumors impacting the function of vital organs. In TS patients, dominant negative mutations in TSC1 or TSC2 increase mTORC1 activity. Increased mTORC1 activity drives tumor formation, but also severely impacts central nervous system function, resulting in infantile seizures, intractable epilepsy, and TS-associated neuropsychiatric disorders, including autism, attention deficits, intellectual disability, and mood disorders. More recently, TS has also been linked with frontotemporal dementia. In addition to TS, accumulating evidence implicates increased mTORC1 activity in the pathology of other neurodevelopmental and neurodegenerative disorders. Thus, TS provides a unique disease model to address whether developmental neural circuit abnormalities promote age-related neurodegeneration, while also providing insight into the therapeutic potential of mTORC1 inhibitors for both developing and degenerating neural circuits. In the following review, we explore the ability of both mouse and human brain organoid models to capture TS pathology, elucidate disease mechanisms, and shed light on how neurodevelopmental alterations may later contribute to age-related neurodegeneration.
Collapse
Affiliation(s)
- Karen Litwa
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
| |
Collapse
|
27
|
Bernard C, Exposito-Alonso D, Selten M, Sanalidou S, Hanusz-Godoy A, Aguilera A, Hamid F, Oozeer F, Maeso P, Allison L, Russell M, Fleck RA, Rico B, Marín O. Cortical wiring by synapse type-specific control of local protein synthesis. Science 2022; 378:eabm7466. [PMID: 36423280 DOI: 10.1126/science.abm7466] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Neurons use local protein synthesis to support their morphological complexity, which requires independent control across multiple subcellular compartments up to the level of individual synapses. We identify a signaling pathway that regulates the local synthesis of proteins required to form excitatory synapses on parvalbumin-expressing (PV+) interneurons in the mouse cerebral cortex. This process involves regulation of the TSC subunit 2 (Tsc2) by the Erb-B2 receptor tyrosine kinase 4 (ErbB4), which enables local control of messenger RNA {mRNA} translation in a cell type-specific and synapse type-specific manner. Ribosome-associated mRNA profiling reveals a molecular program of synaptic proteins downstream of ErbB4 signaling required to form excitatory inputs on PV+ interneurons. Thus, specific connections use local protein synthesis to control synapse formation in the nervous system.
Collapse
Affiliation(s)
- Clémence Bernard
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - David Exposito-Alonso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Stella Sanalidou
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Alicia Hanusz-Godoy
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Alfonso Aguilera
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Fursham Hamid
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Patricia Maeso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Leanne Allison
- Centre for Ultrastructural Imaging, King's College London, London SE1 1UL, UK
| | - Matthew Russell
- Centre for Ultrastructural Imaging, King's College London, London SE1 1UL, UK
| | - Roland A Fleck
- Centre for Ultrastructural Imaging, King's College London, London SE1 1UL, UK
| | - Beatriz Rico
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| |
Collapse
|
28
|
Nakhal MM, Aburuz S, Sadek B, Akour A. Repurposing SGLT2 Inhibitors for Neurological Disorders: A Focus on the Autism Spectrum Disorder. Molecules 2022; 27:7174. [PMID: 36364000 PMCID: PMC9653623 DOI: 10.3390/molecules27217174] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 09/29/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with a substantially increasing incidence rate. It is characterized by repetitive behavior, learning difficulties, deficits in social communication, and interactions. Numerous medications, dietary supplements, and behavioral treatments have been recommended for the management of this condition, however, there is no cure yet. Recent studies have examined the therapeutic potential of the sodium-glucose cotransporter 2 (SGLT2) inhibitors in neurodevelopmental diseases, based on their proved anti-inflammatory effects, such as downregulating the expression of several proteins, including the transforming growth factor beta (TGF-β), interleukin-6 (IL-6), C-reactive protein (CRP), nuclear factor κB (NF-κB), tumor necrosis factor alpha (TNF-α), and the monocyte chemoattractant protein (MCP-1). Furthermore, numerous previous studies revealed the potential of the SGLT2 inhibitors to provide antioxidant effects, due to their ability to reduce the generation of free radicals and upregulating the antioxidant systems, such as glutathione (GSH) and superoxide dismutase (SOD), while crossing the blood brain barrier (BBB). These properties have led to significant improvements in the neurologic outcomes of multiple experimental disease models, including cerebral oxidative stress in diabetes mellitus and ischemic stroke, Alzheimer's disease (AD), Parkinson's disease (PD), and epilepsy. Such diseases have mutual biomarkers with ASD, which potentially could be a link to fill the gap of the literature studying the potential of repurposing the SGLT2 inhibitors' use in ameliorating the symptoms of ASD. This review will look at the impact of the SGLT2 inhibitors on neurodevelopmental disorders on the various models, including humans, rats, and mice, with a focus on the SGLT2 inhibitor canagliflozin. Furthermore, this review will discuss how SGLT2 inhibitors regulate the ASD biomarkers, based on the clinical evidence supporting their functions as antioxidant and anti-inflammatory agents capable of crossing the blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Mohammed Moutaz Nakhal
- Department of Biochemistry, College of Medicine and Health Sciences, Al-Ain P.O. Box 15551, United Arab Emirates
| | - Salahdein Aburuz
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Al-Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Bassem Sadek
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Al-Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 17666, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Al-Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
29
|
De Meulemeester AS, Heylen L, Siekierska A, Mills JD, Romagnolo A, Van Der Wel NN, Aronica E, de Witte PAM. Hyperactivation of mTORC1 in a double hit mutant zebrafish model of tuberous sclerosis complex causes increased seizure susceptibility and neurodevelopmental abnormalities. Front Cell Dev Biol 2022; 10:952832. [PMID: 36238691 PMCID: PMC9552079 DOI: 10.3389/fcell.2022.952832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a multisystem genetic disorder caused by pathogenic variants in TSC1 and TSC2 genes. TSC patients present with seizures and brain abnormalities such as tubers and subependymal giant cells astrocytoma (SEGA). Despite common molecular and clinical features, the severity of the disease varies greatly, even intrafamilially. The second hit hypothesis suggests that an additional, inactivating mutation in the remaining functional allele causes a more severe phenotype and therefore explains the phenotypic variability. Recently, second hit mutations have been detected frequently in mTORopathies. To investigate the pathophysiological effects of second hit mutations, several mouse models have been developed. Here, we opted for a double mutant zebrafish model that carries a LOF mutation both in the tsc2 and the depdc5 gene. To the best of our knowledge, this is the first time a second-hit model has been studied in zebrafish. Significantly, the DEP domain-containing protein 5 (DEPDC5) gene has an important role in the regulation of mTORC1, and the combination of a germline TSC2 and somatic DEPDC5 mutation has been described in a TSC patient with intractable epilepsy. Our depdc5−/−x tsc2−/− double mutant zebrafish line displayed greatly increased levels of mammalian target of rapamycin (mTORC1) activity, augmented seizure susceptibility, and early lethality which could be rescued by rapamycin. Histological analysis of the brain revealed ventricular dilatation in the tsc2 and double homozygotes. RNA-sequencing showed a linear relation between the number of differentially expressed genes (DEGs) and the degree of mTORC1 hyperactivity. Enrichment analysis of their transcriptomes revealed that many genes associated with neurological developmental processes were downregulated and mitochondrial genes were upregulated. In particular, the transcriptome of human SEGA lesions overlapped strongly with the double homozygous zebrafish larvae. The data highlight the clinical relevance of the depdc5−/− x tsc2−/− double mutant zebrafish larvae that showed a more severe phenotype compared to the single mutants. Finally, analysis of gene-drug interactions identified interesting pharmacological targets for SEGA, underscoring the value of our small zebrafish vertebrate model for future drug discovery efforts.
Collapse
Affiliation(s)
| | - Lise Heylen
- Laboratory for Molecular Biodiscovery, KU Leuven, Leuven, Belgium
| | | | - James D. Mills
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
- Chalfont Centre for Epilepsy, Chalfont St Peter, United Kingdom
| | - Alessia Romagnolo
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Nicole N. Van Der Wel
- Department of Medical Biology, Electron Microscopy Center Amsterdam, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Stichting Epilepsie Instelling Nederland (SEIN), Heemstede, Netherlands
| | - Peter A. M. de Witte
- Laboratory for Molecular Biodiscovery, KU Leuven, Leuven, Belgium
- *Correspondence: Peter A. M. de Witte,
| |
Collapse
|
30
|
Casas BS, Arancibia-Altamirano D, Acevedo-La Rosa F, Garrido-Jara D, Maksaev V, Pérez-Monje D, Palma V. It takes two to tango: Widening our understanding of the onset of schizophrenia from a neuro-angiogenic perspective. Front Cell Dev Biol 2022; 10:946706. [PMID: 36092733 PMCID: PMC9448889 DOI: 10.3389/fcell.2022.946706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is a chronic debilitating mental disorder characterized by perturbations in thinking, perception, and behavior, along with brain connectivity deficiencies, neurotransmitter dysfunctions, and loss of gray brain matter. To date, schizophrenia has no cure and pharmacological treatments are only partially efficacious, with about 30% of patients describing little to no improvement after treatment. As in most neurological disorders, the main descriptions of schizophrenia physiopathology have been focused on neural network deficiencies. However, to sustain proper neural activity in the brain, another, no less important network is operating: the vast, complex and fascinating vascular network. Increasing research has characterized schizophrenia as a systemic disease where vascular involvement is important. Several neuro-angiogenic pathway disturbances have been related to schizophrenia. Alterations, ranging from genetic polymorphisms, mRNA, and protein alterations to microRNA and abnormal metabolite processing, have been evaluated in plasma, post-mortem brain, animal models, and patient-derived induced pluripotent stem cell (hiPSC) models. During embryonic brain development, the coordinated formation of blood vessels parallels neuro/gliogenesis and results in the structuration of the neurovascular niche, which brings together physical and molecular signals from both systems conforming to the Blood-Brain barrier. In this review, we offer an upfront perspective on distinctive angiogenic and neurogenic signaling pathways that might be involved in the biological causality of schizophrenia. We analyze the role of pivotal angiogenic-related pathways such as Vascular Endothelial Growth Factor and HIF signaling related to hypoxia and oxidative stress events; classic developmental pathways such as the NOTCH pathway, metabolic pathways such as the mTOR/AKT cascade; emerging neuroinflammation, and neurodegenerative processes such as UPR, and also discuss non-canonic angiogenic/axonal guidance factor signaling. Considering that all of the mentioned above pathways converge at the Blood-Brain barrier, reported neurovascular alterations could have deleterious repercussions on overall brain functioning in schizophrenia.
Collapse
|
31
|
Chaudry S, Vasudevan N. mTOR-Dependent Spine Dynamics in Autism. Front Mol Neurosci 2022; 15:877609. [PMID: 35782388 PMCID: PMC9241970 DOI: 10.3389/fnmol.2022.877609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Conditions (ASC) are a group of neurodevelopmental disorders characterized by deficits in social communication and interaction as well as repetitive behaviors and restricted range of interests. ASC are complex genetic disorders with moderate to high heritability, and associated with atypical patterns of neural connectivity. Many of the genes implicated in ASC are involved in dendritic spine pruning and spine development, both of which can be mediated by the mammalian target of rapamycin (mTOR) signaling pathway. Consistent with this idea, human postmortem studies have shown increased spine density in ASC compared to controls suggesting that the balance between autophagy and spinogenesis is altered in ASC. However, murine models of ASC have shown inconsistent results for spine morphology, which may underlie functional connectivity. This review seeks to establish the relevance of changes in dendritic spines in ASC using data gathered from rodent models. Using a literature survey, we identify 20 genes that are linked to dendritic spine pruning or development in rodents that are also strongly implicated in ASC in humans. Furthermore, we show that all 20 genes are linked to the mTOR pathway and propose that the mTOR pathway regulating spine dynamics is a potential mechanism underlying the ASC signaling pathway in ASC. We show here that the direction of change in spine density was mostly correlated to the upstream positive or negative regulation of the mTOR pathway and most rodent models of mutant mTOR regulators show increases in immature spines, based on morphological analyses. We further explore the idea that these mutations in these genes result in aberrant social behavior in rodent models that is due to these altered spine dynamics. This review should therefore pave the way for further research on the specific genes outlined, their effect on spine morphology or density with an emphasis on understanding the functional role of these changes in ASC.
Collapse
|
32
|
Schrötter S, Yuskaitis CJ, MacArthur MR, Mitchell SJ, Hosios AM, Osipovich M, Torrence ME, Mitchell JR, Hoxhaj G, Sahin M, Manning BD. The non-essential TSC complex component TBC1D7 restricts tissue mTORC1 signaling and brain and neuron growth. Cell Rep 2022; 39:110824. [PMID: 35584673 PMCID: PMC9175135 DOI: 10.1016/j.celrep.2022.110824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/21/2022] [Accepted: 04/23/2022] [Indexed: 11/16/2022] Open
Abstract
The tuberous sclerosis complex (TSC) 1 and 2 proteins associate with TBC1D7 to form the TSC complex, which is an essential suppressor of mTOR complex 1 (mTORC1), a ubiquitous driver of cell and tissue growth. Loss-of-function mutations in TSC1 or TSC2, but not TBC1D7, give rise to TSC, a pleiotropic disorder with aberrant activation of mTORC1 in various tissues. Here, we characterize mice with genetic deletion of Tbc1d7, which are viable with normal growth and development. Consistent with partial loss of function of the TSC complex, Tbc1d7 knockout (KO) mice display variable increases in tissue mTORC1 signaling with increased muscle fiber size but with strength and motor defects. Their most pronounced phenotype is brain overgrowth due to thickening of the cerebral cortex, with enhanced neuron-intrinsic mTORC1 signaling and growth. Thus, TBC1D7 is required for full TSC complex function in tissues, and the brain is particularly sensitive to its growth-suppressing activities.
Collapse
Affiliation(s)
- Sandra Schrötter
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Christopher J Yuskaitis
- Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael R MacArthur
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sarah J Mitchell
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Aaron M Hosios
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Maria Osipovich
- Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Margaret E Torrence
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - James R Mitchell
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gerta Hoxhaj
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mustafa Sahin
- Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brendan D Manning
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Serra I, Stravs A, Osório C, Oyaga MR, Schonewille M, Tudorache C, Badura A. Tsc1 Haploinsufficiency Leads to Pax2 Dysregulation in the Developing Murine Cerebellum. Front Mol Neurosci 2022; 15:831687. [PMID: 35645731 PMCID: PMC9137405 DOI: 10.3389/fnmol.2022.831687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/04/2022] [Indexed: 12/03/2022] Open
Abstract
Tuberous sclerosis complex 1 (TSC1) is a tumor suppressor that promotes the inhibition of mechanistic target of rapamycin (mTOR) pathway, and mutations in TSC1 lead to a rare complex disorder of the same name. Despite phenotype heterogeneity, up to 50% of TSC patients present with autism spectrum disorder (ASD). Consequently, TSC models are often used to probe molecular and behavioral mechanisms of ASD development. Amongst the different brain areas proposed to play a role in the development of ASD, the cerebellum is commonly reported to be altered, and cerebellar-specific deletion of Tsc1 in mice is sufficient to induce ASD-like phenotypes. However, despite these functional changes, whether Tsc1 haploinsufficiency affects cerebellar development is still largely unknown. Given that the mTOR pathway is a master regulator of cell replication and migration, we hypothesized that dysregulation of this pathway would also disrupt the development of cell populations during critical periods of cerebellar development. Here, we used a mouse model of TSC to investigate gene and protein expression during embryonic and early postnatal periods of cerebellar development. We found that, at E18 and P7, mRNA levels of the cerebellar inhibitory interneuron marker paired box gene 2 (Pax2) were dysregulated. This dysregulation was accompanied by changes in the expression of mTOR pathway-related genes and downstream phosphorylation of S6. Differential gene correlation analysis revealed dynamic changes in correlated gene pairs across development, with an overall loss of correlation between mTOR- and cerebellar-related genes in Tsc1 mutants compared to controls. We corroborated the genetic findings by characterizing the mTOR pathway and cerebellar development on protein and cellular levels with Western blot and immunohistochemistry. We found that Pax2-expressing cells were largely unchanged at E18 and P1, while at P7, their number was increased and maturation into parvalbumin-expressing cells delayed. Our findings indicate that, in mice, Tsc1 haploinsufficiency leads to altered cerebellar development and that cerebellar interneuron precursors are particularly susceptible to mTOR pathway dysregulation.
Collapse
Affiliation(s)
- Ines Serra
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Ana Stravs
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Catarina Osório
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Maria Roa Oyaga
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | | | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
- *Correspondence: Aleksandra Badura,
| |
Collapse
|
34
|
Luo C, Ye WR, Shi W, Yin P, Chen C, He YB, Chen MF, Zu XB, Cai Y. Perfect match: mTOR inhibitors and tuberous sclerosis complex. Orphanet J Rare Dis 2022; 17:106. [PMID: 35246210 PMCID: PMC8895788 DOI: 10.1186/s13023-022-02266-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/20/2022] [Indexed: 12/22/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant syndrome that presents with diverse and complex clinical features and involves multiple human systems. TSC-related neurological abnormalities and organ dysfunction greatly affect the quality of life and can even result in death in patients with TSC. It is widely accepted that most TSC-related clinical manifestations are associated with hyperactivation of the mammalian target of rapamycin (mTOR) pathway caused by loss‑of‑function mutations in TSC1 or TSC2. Remarkable progress in basic and translational research has led to encouraging clinical advances. Although mTOR inhibitors (rapamycin/everolimus) demonstrate great potential in TSC management, two major concerns hamper their generalized application. One is the frequent manifestation of adverse events, such as stomatitis, infections, and menstrual disorders; and the other is the poor response in certain patients. Thus, indicators are required to effectively predict the efficacy of mTOR inhibitors. Herein, we have summarized the current utilization of mTOR inhibitors in the treatment of TSC and focused on their efficacy and safety, in an attempt to provide a reference to guide the treatment of TSC. Hyperactivation of mammalian target of rapamycin (mTOR) is essential in the
pathogenesis of tuberous sclerosis complex (TSC) and can serve as a therapeutic
target. mTOR inhibitors have shown considerable success in multiple clinical trials for the treatment of TSC, including neurological, pulmonary, cardiac, renal, and cutaneous
phenotypes. mTOR inhibitors are associated with adverse events, which should be considered
during the management of TSC. Indicators to predict mTOR inhibitor efficacy are required to select patients who
are likely to benefit from such therapy.
Collapse
Affiliation(s)
- Cong Luo
- Department of Urology, Disorders of Tuberous Sclerosis Complex (TSC) Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha City, 410008, Hunan Province, People's Republic of China
| | - Wen-Rui Ye
- Department of Neurosurgery, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha City, 410008, Hunan Province, People's Republic of China
| | - Wei Shi
- Department of Dermatology, Disorders of Tuberous Sclerosis Complex (TSC) Multidisciplinary Team, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha City, 410008, Hunan Province, People's Republic of China
| | - Ping Yin
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Disorders of Tuberous Sclerosis Complex (TSC) Multidisciplinary Team, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha City, 410008, Hunan Province, People's Republic of China
| | - Chen Chen
- Department of Pediatrics, Disorders of Tuberous Sclerosis Complex (TSC) Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha City, 410008, Hunan Province, People's Republic of China
| | - Yun-Bo He
- Department of Urology, Disorders of Tuberous Sclerosis Complex (TSC) Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha City, 410008, Hunan Province, People's Republic of China
| | - Min-Feng Chen
- Department of Urology, Disorders of Tuberous Sclerosis Complex (TSC) Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha City, 410008, Hunan Province, People's Republic of China
| | - Xiong-Bin Zu
- Department of Urology, Disorders of Tuberous Sclerosis Complex (TSC) Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha City, 410008, Hunan Province, People's Republic of China
| | - Yi Cai
- Department of Urology, Disorders of Tuberous Sclerosis Complex (TSC) Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha City, 410008, Hunan Province, People's Republic of China.
| |
Collapse
|
35
|
Xu L, Wu Q, Yan H, Shu C, Fan W, Tong X, Li Q. Long noncoding RNA KB-1460A1.5 inhibits glioma tumorigenesis via miR-130a-3p/TSC1/mTOR/YY1 feedback loop. Cancer Lett 2022; 525:33-45. [PMID: 34728310 DOI: 10.1016/j.canlet.2021.10.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/30/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been shown to be closely related to cancer progression and therapy. However, the clinical significance of lncRNAs and the mechanisms by which they function in glioma are largely unknown. In this study, using online data sets combined with collected clinical glioma tissues, we determined that the lncRNA KB-1460A1.5 is downregulated and positively correlated with prognosis in glioma. Functional experiments showed that overexpression of KB-1460A1.5 inhibits glioma cell proliferation, migration and invasion in vitro and in vivo, while downregulation of KB-1460A1.5 has the opposite effects. Mechanistically, tandem mass tag (TMT)-based quantitative proteomic analysis revealed that KB-1460A1.5 preferentially affects the Akt/TSC1/mTOR pathway. KB-1460A1.5 was found to function as a competing endogenous RNA (ceRNA) to regulate the expression of TSC1, a key regulatory component of the mTOR pathway, by sponging miR-130a-3p in glioma cells. Furthermore, our data demonstrate that the mTOR pathway regulates the expression of the transcription factor Yin Yang 1 (YY1), which in turn binds directly to the KB-1460A1.5 promoter and affects the expression of KB-1460A1.5. Untargeted metabolomics and quantitative real-time PCR (qRT-PCR) analysis further confirmed the effects of KB-1460A1.5 on amino acid metabolism. In conclusion, this study revealed that lncRNA KB-1460A1.5 inhibits glioma tumorigenesis via miR-130a-3p/TSC1/mTOR/YY1 feedback loop.
Collapse
Affiliation(s)
- Lixia Xu
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, 300350, PR China
| | - Qiaoli Wu
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, 300350, PR China
| | - Hua Yan
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, 300350, PR China; Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, PR China
| | - Chang Shu
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, 300350, PR China
| | - Weijia Fan
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, 300350, PR China
| | - Xiaoguang Tong
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, 300350, PR China; Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, PR China.
| | - Qingguo Li
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, 300350, PR China; Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, PR China.
| |
Collapse
|
36
|
Tong X, Wang X, Qin L, Zhou J, Guan Y, Teng P, Wang J, Yang Y, Li T, Luan G. Vagus nerve stimulation for drug-resistant epilepsy induced by tuberous sclerosis complex. Epilepsy Behav 2022; 126:108431. [PMID: 34883463 DOI: 10.1016/j.yebeh.2021.108431] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/22/2021] [Accepted: 11/05/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE This study investigated the dynamic and long-term efficacy of vagus nerve stimulation (VNS) in patients with drug-resistant epilepsy (DRE) induced by tuberous sclerosis complex (TSC). In addition, the impact of VNS on cognition and emotion after a one-year follow-up was evaluated. METHODS A total of 17 patients diagnosed with DRE induced by TSC were retrospectively recruited between 2008 and 2019. Dynamic changes in seizure frequency were observed in the responders (≥50% reduction of seizure frequency at last follow-up) and non-responders. Clinical characteristics and seizure outcomes were comprehensively analyzed to determine factors associated with seizure outcomes. The Wechsler intelligence scale was applied in a subgroup of six pediatric patients, whereas the Self-rating Anxiety Scale (SAS) and Self-rating Depression Scale (SDS) were assessed in a subgroup of nine patients to determine the impact of VNS therapy on cognitive performance and emotional state. RESULTS The follow-up duration for the 17 patients who underwent VNS treatment ranged from 0.5 to 10 years (mean ± SD: 4.1 ± 3.2 years). Monthly seizures decreased significantly from three months to four years post-treatment (p < 0.05). At the last follow-up, 70.6% of the patients achieved at least a 50% reduction in seizure frequency, and three patients were completely seizure free. Comparatively, non-responder patients experienced deterioration of seizure frequency after the first year. Notably, after one-year follow-up the mean standard score of full-scale intelligence quotient increased from 67.33 to 69.5 (p = 0.078) while the mean, standard score of SDS decreased from 49.22 to 45.67 (p = 0.003) compared to preoperative neuropsychological evaluation results. CONCLUSION VNS is a safe and effective treatment for patients with DRE caused by TSC. Although early outcomes were encouraging, a follow-up of at least one-year was required to predict long-term outcomes in patients receiving VNS treatment. Moreover, VNS may improve depressive mood in patients with DRE caused by TSC. Further investigations are needed to validate the present results.
Collapse
Affiliation(s)
- Xuezhi Tong
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Xiongfei Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China; Beijing Key Laboratory of Epilepsy, Beijing 100093, China; Epilepsy Institute, Beijing Institute for Brain Disorders, Beijing 100093, China
| | - Lang Qin
- McGovern Institute for Brain Research, Peking University, Beijing 100093, China; Center for MRI Research, Peking University, Beijing 100093, China
| | - Jian Zhou
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Yuguang Guan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Pengfei Teng
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Jing Wang
- Department of Neurology, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Yujiao Yang
- Department of Neurology, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Tianfu Li
- Epilepsy Institute, Beijing Institute for Brain Disorders, Beijing 100093, China; Department of Neurology, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Guoming Luan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China; Beijing Key Laboratory of Epilepsy, Beijing 100093, China; Epilepsy Institute, Beijing Institute for Brain Disorders, Beijing 100093, China.
| |
Collapse
|
37
|
Koene LM, Niggl E, Wallaard I, Proietti-Onori M, Rotaru DC, Elgersma Y. Identifying the temporal electrophysiological and molecular changes that contribute to TSC-associated epileptogenesis. JCI Insight 2021; 6:e150120. [PMID: 34877936 PMCID: PMC8675202 DOI: 10.1172/jci.insight.150120] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Tuberous sclerosis complex (TSC), caused by heterozygous mutations in TSC1 or TSC2, frequently results in intractable epilepsy. Here, we made use of an inducible Tsc1-knockout mouse model, allowing us to study electrophysiological and molecular changes of Tsc1-induced epileptogenesis over time. We recorded from pyramidal neurons in the hippocampus and somatosensory cortex (L2/L3) and combined this with an analysis of transcriptome changes during epileptogenesis. Deletion of Tsc1 resulted in hippocampus-specific changes in excitability and adaptation, which emerged before seizure onset and progressed over time. All phenotypes were rescued after early treatment with rapamycin, an mTOR inhibitor. Later in epileptogenesis, we observed a hippocampal increase of excitation-to-inhibition ratio. These cellular changes were accompanied by dramatic transcriptional changes, especially after seizure onset. Most of these changes were rescued upon rapamycin treatment. Of the genes encoding ion channels or belonging to the Gene Ontology term action potential, 27 were differentially expressed just before seizure onset, suggesting a potential driving role in epileptogenesis. Our data highlight the complex changes driving epileptogenesis in TSC, including the changed expression of multiple ion channels. Our study emphasizes inhibition of the TSC/mTOR signaling pathway as a promising therapeutic approach to target epilepsy in patients with TSC.
Collapse
|
38
|
Fang K, Liu D, Pathak SS, Yang B, Li J, Karthikeyan R, Chao OY, Yang YM, Jin VX, Cao R. Disruption of Circadian Rhythms by Ambient Light during Neurodevelopment Leads to Autistic-like Molecular and Behavioral Alterations in Adult Mice. Cells 2021; 10:3314. [PMID: 34943821 PMCID: PMC8699695 DOI: 10.3390/cells10123314] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 02/01/2023] Open
Abstract
Although circadian rhythms are thought to be essential for maintaining body health, the effects of chronic circadian disruption during neurodevelopment remain elusive. Here, using the "Short Day" (SD) mouse model, in which an 8 h/8 h light/dark (LD) cycle was applied from embryonic day 1 to postnatal day 42, we investigated the molecular and behavioral changes after circadian disruption in mice. Adult SD mice fully entrained to the 8 h/8 h LD cycle, and the circadian oscillations of the clock proteins, PERIOD1 and PERIOD2, were disrupted in the suprachiasmatic nucleus and the hippocampus of these mice. By RNA-seq widespread changes were identified in the hippocampal transcriptome, which are functionally associated with neurodevelopment, translational control, and autism. By western blotting and immunostaining hyperactivation of the mTOR and MAPK signaling pathways and enhanced global protein synthesis were found in the hippocampi of SD mice. Electrophysiological recording uncovered enhanced excitatory, but attenuated inhibitory, synaptic transmission in the hippocampal CA1 pyramidal neurons. These functional changes at synapses were corroborated by the immature morphology of the dendritic spines in these neurons. Lastly, autistic-like animal behavioral changes, including impaired social interaction and communication, increased repetitive behaviors, and impaired novel object recognition and location memory, were found in SD mice. Together, these results demonstrate molecular, cellular, and behavioral changes in SD mice, all of which resemble autistic-like phenotypes caused by circadian rhythm disruption. The findings highlight a critical role for circadian rhythms in neurodevelopment.
Collapse
Affiliation(s)
- Kun Fang
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (K.F.); (B.Y.)
| | - Dong Liu
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (D.L.); (S.S.P.); (J.L.); (R.K.); (O.Y.C.)
| | - Salil S. Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (D.L.); (S.S.P.); (J.L.); (R.K.); (O.Y.C.)
| | - Bowen Yang
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (K.F.); (B.Y.)
| | - Jin Li
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (D.L.); (S.S.P.); (J.L.); (R.K.); (O.Y.C.)
| | - Ramanujam Karthikeyan
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (D.L.); (S.S.P.); (J.L.); (R.K.); (O.Y.C.)
| | - Owen Y. Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (D.L.); (S.S.P.); (J.L.); (R.K.); (O.Y.C.)
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (D.L.); (S.S.P.); (J.L.); (R.K.); (O.Y.C.)
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Victor X. Jin
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (K.F.); (B.Y.)
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (D.L.); (S.S.P.); (J.L.); (R.K.); (O.Y.C.)
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
39
|
Pagani M, Barsotti N, Bertero A, Trakoshis S, Ulysse L, Locarno A, Miseviciute I, De Felice A, Canella C, Supekar K, Galbusera A, Menon V, Tonini R, Deco G, Lombardo MV, Pasqualetti M, Gozzi A. mTOR-related synaptic pathology causes autism spectrum disorder-associated functional hyperconnectivity. Nat Commun 2021; 12:6084. [PMID: 34667149 PMCID: PMC8526836 DOI: 10.1038/s41467-021-26131-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 09/17/2021] [Indexed: 11/24/2022] Open
Abstract
Postmortem studies have revealed increased density of excitatory synapses in the brains of individuals with autism spectrum disorder (ASD), with a putative link to aberrant mTOR-dependent synaptic pruning. ASD is also characterized by atypical macroscale functional connectivity as measured with resting-state fMRI (rsfMRI). These observations raise the question of whether excess of synapses causes aberrant functional connectivity in ASD. Using rsfMRI, electrophysiology and in silico modelling in Tsc2 haploinsufficient mice, we show that mTOR-dependent increased spine density is associated with ASD -like stereotypies and cortico-striatal hyperconnectivity. These deficits are completely rescued by pharmacological inhibition of mTOR. Notably, we further demonstrate that children with idiopathic ASD exhibit analogous cortical-striatal hyperconnectivity, and document that this connectivity fingerprint is enriched for ASD-dysregulated genes interacting with mTOR or Tsc2. Finally, we show that the identified transcriptomic signature is predominantly expressed in a subset of children with autism, thereby defining a segregable autism subtype. Our findings causally link mTOR-related synaptic pathology to large-scale network aberrations, revealing a unifying multi-scale framework that mechanistically reconciles developmental synaptopathy and functional hyperconnectivity in autism.
Collapse
Affiliation(s)
- Marco Pagani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
| | - Noemi Barsotti
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Alice Bertero
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Stavros Trakoshis
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | - Laura Ulysse
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Barcelona, Spain
| | - Andrea Locarno
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Ieva Miseviciute
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Alessia De Felice
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | - Carola Canella
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | | | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | | | - Raffaella Tonini
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Gustavo Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Barcelona, Spain
- Institució Catalana de la Recerca i Estudis Avançats (ICREA), Universitat Pompeu Fabra, Barcelona, Spain
| | - Michael V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Autism Research Centre, University of Cambridge, Cambridge, UK
| | - Massimo Pasqualetti
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy.
| |
Collapse
|
40
|
Moloney PB, Cavalleri GL, Delanty N. Epilepsy in the mTORopathies: opportunities for precision medicine. Brain Commun 2021; 3:fcab222. [PMID: 34632383 PMCID: PMC8495134 DOI: 10.1093/braincomms/fcab222] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 01/16/2023] Open
Abstract
The mechanistic target of rapamycin signalling pathway serves as a ubiquitous regulator of cell metabolism, growth, proliferation and survival. The main cellular activity of the mechanistic target of rapamycin cascade funnels through mechanistic target of rapamycin complex 1, which is inhibited by rapamycin, a macrolide compound produced by the bacterium Streptomyces hygroscopicus. Pathogenic variants in genes encoding upstream regulators of mechanistic target of rapamycin complex 1 cause epilepsies and neurodevelopmental disorders. Tuberous sclerosis complex is a multisystem disorder caused by mutations in mechanistic target of rapamycin regulators TSC1 or TSC2, with prominent neurological manifestations including epilepsy, focal cortical dysplasia and neuropsychiatric disorders. Focal cortical dysplasia type II results from somatic brain mutations in mechanistic target of rapamycin pathway activators MTOR, AKT3, PIK3CA and RHEB and is a major cause of drug-resistant epilepsy. DEPDC5, NPRL2 and NPRL3 code for subunits of the GTPase-activating protein (GAP) activity towards Rags 1 complex (GATOR1), the principal amino acid-sensing regulator of mechanistic target of rapamycin complex 1. Germline pathogenic variants in GATOR1 genes cause non-lesional focal epilepsies and epilepsies associated with malformations of cortical development. Collectively, the mTORopathies are characterized by excessive mechanistic target of rapamycin pathway activation and drug-resistant epilepsy. In the first large-scale precision medicine trial in a genetically mediated epilepsy, everolimus (a synthetic analogue of rapamycin) was effective at reducing seizure frequency in people with tuberous sclerosis complex. Rapamycin reduced seizures in rodent models of DEPDC5-related epilepsy and focal cortical dysplasia type II. This review outlines a personalized medicine approach to the management of epilepsies in the mTORopathies. We advocate for early diagnostic sequencing of mechanistic target of rapamycin pathway genes in drug-resistant epilepsy, as identification of a pathogenic variant may point to an occult dysplasia in apparently non-lesional epilepsy or may uncover important prognostic information including, an increased risk of sudden unexpected death in epilepsy in the GATORopathies or favourable epilepsy surgery outcomes in focal cortical dysplasia type II due to somatic brain mutations. Lastly, we discuss the potential therapeutic application of mechanistic target of rapamycin inhibitors for drug-resistant seizures in GATOR1-related epilepsies and focal cortical dysplasia type II.
Collapse
Affiliation(s)
- Patrick B Moloney
- FutureNeuro, the Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin, D02 VN51, Ireland
| | - Gianpiero L Cavalleri
- FutureNeuro, the Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin, D02 VN51, Ireland
| | - Norman Delanty
- FutureNeuro, the Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin, D02 VN51, Ireland
| |
Collapse
|
41
|
Synaptic Alterations in a Transgenic Model of Tuberous Sclerosis Complex: Relevance to Autism Spectrum Disorders. Int J Mol Sci 2021; 22:ijms221810058. [PMID: 34576223 PMCID: PMC8466868 DOI: 10.3390/ijms221810058] [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: 08/24/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 01/02/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare, multi-system genetic disease with serious neurological and mental symptoms, including autism. Mutations in the TSC1/TSC2 genes lead to the overactivation of mTOR signalling, which is also linked to nonsyndromic autism. Our aim was to analyse synaptic pathology in a transgenic model of TSC: two-month-old male B6;129S4-Tsc2tm1Djk/J mice with Tsc2 haploinsufficiency. Significant brain-region-dependent alterations in the expression of several synaptic proteins were identified. The most prominent changes were observed in the immunoreactivity of presynaptic VAMP1/2 (ca. 50% increase) and phospho-synapsin-1 (Ser62/67) (ca. 80% increase). Transmission electron microscopy demonstrated serious ultrastructural abnormalities in synapses such as a blurred structure of synaptic density and a significantly increased number of synaptic vesicles. The impairment of synaptic mitochondrial ultrastructure was represented by excessive elongation, swelling, and blurred crista contours. Polyribosomes in the cytoplasm and swollen Golgi apparatus suggest possible impairment of protein metabolism. Moreover, the delamination of myelin and the presence of vacuolar structures in the cell nucleus were observed. We also report that Tsc2+/- mice displayed increased brain weights and sizes. The behavioural analysis demonstrated the impairment of memory function, as established in the novel object recognition test. To summarise, our data indicate serious synaptic impairment in the brains of male Tsc2+/- mice.
Collapse
|
42
|
Scherrer B, Prohl AK, Taquet M, Kapur K, Peters JM, Tomas-Fernandez X, Davis PE, M Bebin E, Krueger DA, Northrup H, Y Wu J, Sahin M, Warfield SK. The Connectivity Fingerprint of the Fusiform Gyrus Captures the Risk of Developing Autism in Infants with Tuberous Sclerosis Complex. Cereb Cortex 2021; 30:2199-2214. [PMID: 31812987 DOI: 10.1093/cercor/bhz233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/05/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare genetic disorder characterized by benign tumors throughout the body; it is generally diagnosed early in life and has a high prevalence of autism spectrum disorder (ASD), making it uniquely valuable in studying the early development of autism, before neuropsychiatric symptoms become apparent. One well-documented deficit in ASD is an impairment in face processing. In this work, we assessed whether anatomical connectivity patterns of the fusiform gyrus, a central structure in face processing, capture the risk of developing autism early in life. We longitudinally imaged TSC patients at 1, 2, and 3 years of age with diffusion compartment imaging. We evaluated whether the anatomical connectivity fingerprint of the fusiform gyrus was associated with the risk of developing autism measured by the Autism Observation Scale for Infants (AOSI). Our findings suggest that the fusiform gyrus connectivity captures the risk of developing autism as early as 1 year of age and provides evidence that abnormal fusiform gyrus connectivity increases with age. Moreover, the identified connections that best capture the risk of developing autism involved the fusiform gyrus and limbic and paralimbic regions that were consistent with the ASD phenotype, involving an increased number of left-lateralized structures with increasing age.
Collapse
Affiliation(s)
- Benoit Scherrer
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115 USA
| | - Anna K Prohl
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115 USA
| | - Maxime Taquet
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115 USA
| | - Kush Kapur
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115 USA
| | - Jurriaan M Peters
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115 USA
| | - Xavier Tomas-Fernandez
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115 USA
| | - Peter E Davis
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115 USA
| | - Elizabeth M Bebin
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35233 USA
| | - Darcy A Krueger
- Department of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229 USA
| | - Hope Northrup
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030 USA
| | - Joyce Y Wu
- Department of Pediatrics, UCLA Mattel Children's Hospital, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095 USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115 USA
| | - Simon K Warfield
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115 USA
| |
Collapse
|
43
|
Benthall KN, Cording KR, Agopyan-Miu AHCW, Wong CD, Chen EY, Bateup HS. Loss of Tsc1 from striatal direct pathway neurons impairs endocannabinoid-LTD and enhances motor routine learning. Cell Rep 2021; 36:109511. [PMID: 34380034 PMCID: PMC8404511 DOI: 10.1016/j.celrep.2021.109511] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 05/28/2021] [Accepted: 07/21/2021] [Indexed: 02/08/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a neurodevelopmental disorder that often presents with psychiatric conditions, including autism spectrum disorder (ASD). ASD is characterized by restricted, repetitive, and inflexible behaviors, which may result from abnormal activity in striatal circuits that mediate motor learning and action selection. To test whether altered striatal activity contributes to aberrant motor behaviors in the context of TSC, we conditionally deleted Tsc1 from direct or indirect pathway striatal projection neurons (dSPNs or iSPNs, respectively). We find that dSPN-specific loss of Tsc1 impairs endocannabinoid-mediated long-term depression (eCB-LTD) at cortico-dSPN synapses and strongly enhances corticostriatal synaptic drive, which is not observed in iSPNs. dSPN-Tsc1 KO, but not iSPN-Tsc1 KO, mice show enhanced motor learning, a phenotype observed in several mouse models of ASD. These findings demonstrate that dSPNs are particularly sensitive to Tsc1 loss and suggest that enhanced corticostriatal activation may contribute to altered motor behaviors in TSC.
Collapse
Affiliation(s)
- Katelyn N Benthall
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Katherine R Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Corinna D Wong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emily Y Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
44
|
Connecting the Neurobiology of Developmental Brain Injury: Neuronal Arborisation as a Regulator of Dysfunction and Potential Therapeutic Target. Int J Mol Sci 2021; 22:ijms22158220. [PMID: 34360985 PMCID: PMC8348801 DOI: 10.3390/ijms22158220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Neurodevelopmental disorders can derive from a complex combination of genetic variation and environmental pressures on key developmental processes. Despite this complex aetiology, and the equally complex array of syndromes and conditions diagnosed under the heading of neurodevelopmental disorder, there are parallels in the neuropathology of these conditions that suggest overlapping mechanisms of cellular injury and dysfunction. Neuronal arborisation is a process of dendrite and axon extension that is essential for the connectivity between neurons that underlies normal brain function. Disrupted arborisation and synapse formation are commonly reported in neurodevelopmental disorders. Here, we summarise the evidence for disrupted neuronal arborisation in these conditions, focusing primarily on the cortex and hippocampus. In addition, we explore the developmentally specific mechanisms by which neuronal arborisation is regulated. Finally, we discuss key regulators of neuronal arborisation that could link to neurodevelopmental disease and the potential for pharmacological modification of arborisation and the formation of synaptic connections that may provide therapeutic benefit in the future.
Collapse
|
45
|
Bassetti D, Luhmann HJ, Kirischuk S. Presynaptic GABA B receptor-mediated network excitation in the medial prefrontal cortex of Tsc2 +/- mice. Pflugers Arch 2021; 473:1261-1271. [PMID: 34279736 PMCID: PMC8302497 DOI: 10.1007/s00424-021-02576-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/21/2021] [Accepted: 05/05/2021] [Indexed: 11/02/2022]
Abstract
The TSC1 and TSC2 tumor suppressor genes control the activity of mechanistic target of rapamycin (mTOR) pathway. Elevated activity of this pathway in Tsc2+/- mouse model leads to reduction of postsynaptic GABAB receptor-mediated inhibition and hyperexcitability in the medial prefrontal cortex (mPFC). In this study, we asked whether presynaptic GABAB receptors (GABABRs) can compensate this shift of hyperexcitability. Experiments were performed in brain slices from adolescent wild-type (WT) and Tsc2+/- mice. Miniature and spontaneous postsynaptic currents (m/sPSCs) were recorded from layer 2/3 pyramidal neurons in mPFC using patch-clamp technique using a Cs+-based intrapipette solution. Presynaptic GABABRs were activated by baclofen (10 µM) or blocked by CGP55845 (1 µM). Independent on genotype, GABABR modulators bidirectionally change miniature excitatory postsynaptic current (mEPSC) frequency by about 10%, indicating presynaptic GABABR-mediated effects on glutamatergic transmission are comparable in both genotypes. In contrast, frequencies of both mIPSCs and sIPCSs were suppressed by baclofen stronger in Tsc2+/- neurons than in WT ones, whereas CGP55845 significantly increased (m/s)IPSC frequencies only in WT cells. Effects of baclofen and CGP55845 on the amplitudes of evoked (e)IPSCs confirmed these observations. These data indicate (1) that GABAergic synapses are inhibited by ambient GABA in WT but not in Tsc2+/- slices, and (2) that baclofen shifts the E/I ratio, determined as the ratio of (m/s)EPSC frequency to (m/s)IPSC frequency, towards excitation only in Tsc2+/- cells. This excitatory presynaptic GABABR-mediated action has to be taken into account for a possible medication of mental disorders using baclofen.
Collapse
Affiliation(s)
- Davide Bassetti
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128, Mainz, Germany.
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128, Mainz, Germany
| | - Sergei Kirischuk
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128, Mainz, Germany
| |
Collapse
|
46
|
Bassetti D, Luhmann HJ, Kirischuk S. Effects of Mutations in TSC Genes on Neurodevelopment and Synaptic Transmission. Int J Mol Sci 2021; 22:7273. [PMID: 34298906 PMCID: PMC8305053 DOI: 10.3390/ijms22147273] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 12/11/2022] Open
Abstract
Mutations in TSC1 or TSC2 genes are linked to alterations in neuronal function which ultimately lead to the development of a complex neurological phenotype. Here we review current research on the effects that reduction in TSC1 or TSC2 can produce on the developing neural network. A crucial feature of the disease pathophysiology appears to be an early deviation from typical neurodevelopment, in the form of structural abnormalities. Epileptic seizures are one of the primary early manifestation of the disease in the CNS, followed by intellectual deficits and autism spectrum disorders (ASD). Research using mouse models suggests that morphological brain alterations might arise from the interaction of different cellular types, and hyperexcitability in the early postnatal period might be transient. Moreover, the increased excitation-to-inhibition ratio might represent a transient compensatory adjustment to stabilize the developing network rather than a primary factor for the development of ASD symptoms. The inhomogeneous results suggest region-specificity as well as an evolving picture of functional alterations along development. Furthermore, ASD symptoms and epilepsy might originate from different but potentially overlapping mechanisms, which can explain recent observations obtained in patients. Potential treatment is determined not only by the type of medicament, but also by the time point of treatment.
Collapse
Affiliation(s)
- Davide Bassetti
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, Germany; (H.J.L.); (S.K.)
| | | | | |
Collapse
|
47
|
Sensitive period for rescuing parvalbumin interneurons connectivity and social behavior deficits caused by TSC1 loss. Nat Commun 2021; 12:3653. [PMID: 34135323 PMCID: PMC8209106 DOI: 10.1038/s41467-021-23939-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/20/2021] [Indexed: 11/08/2022] Open
Abstract
The Mechanistic Target Of Rapamycin Complex 1 (mTORC1) pathway controls several aspects of neuronal development. Mutations in regulators of mTORC1, such as Tsc1 and Tsc2, lead to neurodevelopmental disorders associated with autism, intellectual disabilities and epilepsy. The correct development of inhibitory interneurons is crucial for functional circuits. In particular, the axonal arborisation and synapse density of parvalbumin (PV)-positive GABAergic interneurons change in the postnatal brain. How and whether mTORC1 signaling affects PV cell development is unknown. Here, we show that Tsc1 haploinsufficiency causes a premature increase in terminal axonal branching and bouton density formed by mutant PV cells, followed by a loss of perisomatic innervation in adult mice. PV cell-restricted Tsc1 haploinsufficient and knockout mice show deficits in social behavior. Finally, we identify a sensitive period during the third postnatal week during which treatment with the mTOR inhibitor Rapamycin rescues deficits in both PV cell innervation and social behavior in adult conditional haploinsufficient mice. Our findings reveal a role of mTORC1 signaling in the regulation of the developmental time course and maintenance of cortical PV cell connectivity and support a mechanistic basis for the targeted rescue of autism-related behaviors in disorders associated with deregulated mTORC1 signaling.
Collapse
|
48
|
Sharma A, Mehan S. Targeting PI3K-AKT/mTOR signaling in the prevention of autism. Neurochem Int 2021; 147:105067. [PMID: 33992742 DOI: 10.1016/j.neuint.2021.105067] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 12/16/2022]
Abstract
PI3K-AKT/mTOR signaling pathway represents an essential signaling mechanism for mammalian enzyme-related receptors in transducing signals or biological processes such as cell development, differentiation, cell survival, protein synthesis, and metabolism. Upregulation of the PI3K-AKT/mTOR signaling pathway involves many human brain abnormalities, including autism and other neurological dysfunctions. Autism is a neurodevelopmental disorder associated with behavior and psychiatric illness. This research-based review discusses the functional relationship between the neuropathogenic factors associated with PI3K-AKT/mTOR signaling pathway. Ultimately causes autism-like conditions associated with genetic alterations, neuronal apoptosis, mitochondrial dysfunction, and neuroinflammation. Therefore, inhibition of the PI3K-AKT/mTOR signaling pathway may have an effective therapeutic value for autism treatment. The current review also summarizes the involvement of PI3K-AKT/mTOR signaling pathway inhibitors in the treatment of autism and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Aarti Sharma
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
49
|
Jeckel P, Kriebel M, Volkmer H. Autism Spectrum Disorder Risk Factor Met Regulates the Organization of Inhibitory Synapses. Front Mol Neurosci 2021; 14:659856. [PMID: 34054427 PMCID: PMC8155383 DOI: 10.3389/fnmol.2021.659856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/09/2021] [Indexed: 12/27/2022] Open
Abstract
A common hypothesis explains autism spectrum disorder (ASD) as a neurodevelopmental disorder linked to excitatory/inhibitory (E/I) imbalance in neuronal network connectivity. Mutation of genes including Met and downstream signaling components, e.g., PTEN, Tsc2 and, Rheb are involved in the control of synapse formation and stabilization and were all considered as risk genes for ASD. While the impact of Met on glutamatergic synapses was widely appreciated, its contribution to the stability of inhibitory, GABAergic synapses is poorly understood. The stabilization of GABAergic synapses depends on clustering of the postsynaptic scaffolding protein gephyrin. Here, we show in vivo and in vitro that Met is necessary and sufficient for the stabilization of GABAergic synapses via induction of gephyrin clustering. Likewise, we provide evidence for Met-dependent gephyrin clustering via activation of mTOR. Our results support the notion that deficient GABAergic signaling represents a pathomechanism for ASD.
Collapse
Affiliation(s)
- Pauline Jeckel
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Martin Kriebel
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Hansjürgen Volkmer
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| |
Collapse
|
50
|
Karalis V, Bateup HS. Current Approaches and Future Directions for the Treatment of mTORopathies. Dev Neurosci 2021; 43:143-158. [PMID: 33910214 PMCID: PMC8440338 DOI: 10.1159/000515672] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/13/2021] [Indexed: 11/19/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is a kinase at the center of an evolutionarily conserved signaling pathway that orchestrates cell growth and metabolism. mTOR responds to an array of intra- and extracellular stimuli and in turn controls multiple cellular anabolic and catabolic processes. Aberrant mTOR activity is associated with numerous diseases, with particularly profound impact on the nervous system. mTOR is found in two protein complexes, mTOR complex 1 (mTORC1) and 2 (mTORC2), which are governed by different upstream regulators and have distinct cellular actions. Mutations in genes encoding for mTOR regulators result in a collection of neurodevelopmental disorders known as mTORopathies. While these disorders can affect multiple organs, neuropsychiatric conditions such as epilepsy, intellectual disability, and autism spectrum disorder have a major impact on quality of life. The neuropsychiatric aspects of mTORopathies have been particularly challenging to treat in a clinical setting. Current therapeutic approaches center on rapamycin and its analogs, drugs that are administered systemically to inhibit mTOR activity. While these drugs show some clinical efficacy, adverse side effects, incomplete suppression of mTOR targets, and lack of specificity for mTORC1 or mTORC2 may limit their utility. An increased understanding of the neurobiology of mTOR and the underlying molecular, cellular, and circuit mechanisms of mTOR-related disorders will facilitate the development of improved therapeutics. Animal models of mTORopathies have helped unravel the consequences of mTOR pathway mutations in specific brain cell types and developmental stages, revealing an array of disease-related phenotypes. In this review, we discuss current progress and potential future directions for the therapeutic treatment of mTORopathies with a focus on findings from genetic mouse models.
Collapse
Affiliation(s)
- Vasiliki Karalis
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| |
Collapse
|