1
|
Liu C, Zhang C, Glatt SJ. Psychiatric Genomics 2025: State of the Art and the Path Forward. Psychiatr Clin North Am 2025; 48:217-240. [PMID: 40348414 DOI: 10.1016/j.psc.2025.01.002] [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] [Indexed: 05/14/2025]
Abstract
Psychiatric genetics has evolved from candidate-gene studies to whole-genome sequencing efforts. With hundreds of disease-associated loci now identified, functional interpretation of the associated loci becomes the critical next step toward translational applications. The article discusses achievements, challenges, and opportunities in psychiatric genomics associated with complexity and heterogeneity. Brain expression quantitative trait loci, single-cell ribonucleic acid-sequence, and functional genomics technologies are highlighted. It also covers newly developed techniques with improved spatiotemporal resolution, quality and sensitivity, coupled with advanced analytical methods and artificial intelligence. The power of collaborative research and inclusion of diverse populations will ensure a bright future for precision psychiatry.
Collapse
Affiliation(s)
- Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA.
| | - Chunling Zhang
- Department of Neuroscience & Physiology, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| | - Stephen J Glatt
- Department of Psychiatry, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| |
Collapse
|
2
|
Zhai Y, Gao F, Shi S, Zhong Q, Zhang J, Fang J, He F, Zhang Y, Li Y, Liu F, Xue B, Gu Y, Li S. Noninvasive Optogenetics Realized by iPSC-Derived Tentacled Carrier in Alzheimer's Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2419768. [PMID: 40434197 DOI: 10.1002/adma.202419768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/21/2025] [Indexed: 05/29/2025]
Abstract
Neural-activated optogenetics technique contributing to the "restart" of degenerative neurons offers hope for the treatment of several neurodegenerative diseases. However, the limitations of persistent invasiveness and inadequate repair of the pathological environment strongly hinder its further application. Here, a concept of differentiating stem cells is proposed to produce functional materials to enhance the therapeutic applicability of optogenetics. Induced pluripotent stem cells (iPSCs) are differentiated to generate the "tentacled" stem cells TenSCs. Their "tentacled" vesicles TenSCev, upon inheriting the biological functions of the parent cell, will possess both neural targeting capacity and pathological environment repair ability. Hence, TenSCev are utilized as functional carrier to deliver optogenetics elements for completely non-traumatic and controllable neuron activation, while also facilitating the comprehensive restoration of the pathological environment, thus effectively halted disease progression and significantly improved cognitive function in Alzheimer's disease or aged mice. Further, the concept of generating specialized biomaterials from differentiated stem cells as functional carriers holds the potential to broaden the applicability of various neuroregulatory techniques in the treatment of neurological disorders.
Collapse
Affiliation(s)
- Yuewen Zhai
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Fan Gao
- Department of Biomedical Engineering, School of Automation, Nanjing University of Aeronautics and Astronautics, No. 29 JiangJun street, Jiangning District, Nanjing, Jiangsu Province, 211106, China
| | - Shihao Shi
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Qifeng Zhong
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Jinnan Zhang
- Department of Neurosurgery, China-Japan union Hospital, Jilin University, No. 126 Sendai Street, Changchun, Jilin Province, 130033, China
| | - Ji Fang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Fang He
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Yanqin Zhang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Yu Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Fei Liu
- Department of Biomedical Engineering, Sichuan University, No.24, Wangjiang Road, Wuhou District, Chengdu, Sichuan Province, 610000, China
| | - Bing Xue
- Department of Biomedical Engineering, Sichuan University, No.24, Wangjiang Road, Wuhou District, Chengdu, Sichuan Province, 610000, China
| | - Yueqing Gu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Siwen Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| |
Collapse
|
3
|
Xie L, Sheehy RN, Muneer A, Xiong Y, Wrobel JA, Zhang F, Park KS, Velez J, Liu J, Luo YJ, Asrican B, Dong P, Li YD, Damian C, Quintanilla L, Li Y, Xu C, Deshmukh M, Coleman LG, Ming GL, Song H, Wen Z, Jin J, Song J, Chen X. Development of a brain-penetrant G9a methylase inhibitor to target Alzheimer's disease-associated proteopathology. Nat Commun 2025; 16:4222. [PMID: 40328756 PMCID: PMC12056044 DOI: 10.1038/s41467-025-59128-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Current Aβ-targeting therapeutics for Alzheimer's disease (AD) only slow cognitive decline due to poor understanding of AD pathogenesis. Here we describe a mechanism of AD pathogenesis in which the histone methyltransferase G9a noncanonically regulates translation of hippocampal proteins associated with AD pathology. Correspondingly, we developed a brain-penetrant inhibitor of G9a, MS1262, which restored both age-related learning & memory and noncognitive functions in multiple AD mouse models. Further, comparison of AD pathology-correlated mouse proteomes with those of AD patients found G9a regulates pathological pathways that promote Aβ and neurofibrillary tangles. This mouse-to-human overlap of G9a regulated AD-associated pathologic proteins supports at the molecular level the efficacy of targeting G9a translational mechanism for treating AD patients. Additionally, MS1262 treatment reversed the AD-characteristic expression or phosphorylation of multiple clinically validated biomarkers of AD that have the potential to be used for early-stage AD diagnosis and companion diagnosis of individualized drug effects.
Collapse
Affiliation(s)
- Ling Xie
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ryan N Sheehy
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adil Muneer
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John A Wrobel
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Kwang-Su Park
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia Velez
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yan-Jia Luo
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brent Asrican
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ping Dong
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ya-Dong Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Corina Damian
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Luis Quintanilla
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yongyi Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chongchong Xu
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Xian Chen
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Shen Y, Wong SZH, Ma T, Zhang F, Wang Q, Kawaguchi R, Geschwind DH, Wang J, He C, Ming GL, Song H. m 6A deficiency impairs hypothalamic neurogenesis of feeding-related neurons in mice and human organoids and leads to adult obesity in mice. Cell Stem Cell 2025; 32:727-743.e8. [PMID: 40112816 DOI: 10.1016/j.stem.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/07/2024] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
N6-methyladenosine (m6A), the most prevalent internal modification on mRNAs, plays important roles in the nervous system. Whether neurogenesis in the hypothalamus, a region critical for controlling appetite, is regulated by m6A signaling, especially in humans, remains unclear. Here, we showed that deletion of m6A writer Mettl14 in the mouse embryonic hypothalamus led to adult obesity, with impaired glucose-insulin homeostasis and increased energy intake. Mechanistically, deletion of Mettl14 leads to hypothalamic arcuate nucleus neurogenesis deficits with reduced generation of feeding-related neurons and dysregulation of neurogenesis-related m6A-tagged transcripts. Deletion of m6A writer Mettl3 or m6A reader Ythdc1 shared similar phenotypes. METTL14 or YTHDC1 knockdown also led to reduced generation of feeding-related neurons in human brain subregion-specific arcuate nucleus organoids. Our studies reveal a conserved role of m6A signaling in arcuate nucleus neurogenesis in mice and human organoids and shed light on the developmental basis of epitranscriptomic regulation of food intake and energy homeostasis.
Collapse
Affiliation(s)
- Yachen Shen
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel Zheng Hao Wong
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tong Ma
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qing Wang
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Chuan He
- Department of Chemistry, Howard Hughes Medical Institute, the University of Chicago, Chicago, IL, USA; Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute, the University of Chicago, Chicago, IL, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Zhao Y, Metzler AD, Li Y, Urlich P, Wang Y, Gilbert DM, Yao B, Tang H. Interferon-dependent R-loop induction by Zika virus contributes to growth attenuation. PNAS NEXUS 2025; 4:pgaf147. [PMID: 40386679 PMCID: PMC12082298 DOI: 10.1093/pnasnexus/pgaf147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 03/31/2025] [Indexed: 05/20/2025]
Abstract
Zika virus (ZIKV) infection in human neural progenitors triggers DNA damage and activates DNA damage response, leading to cell cycle arrest that can retard brain development. Here, we link the ZIKV-induced S phase arrest to replication fork stalling and R-loop induction. DRIP-seq reveals that ZIKV infection induces R loops at specific loci strongly enriched in the interferon (IFN)-stimulated genes (ISGs). Bromouridine sequencing results further indicate that nascent ISGs transcripts are prone to R-loop induction upon infection. Knockout of IFN receptor eliminated the R loops on ISGs and partially rescued S-phase arrest in infected cells. And overexpression of RNaseH1 reduced ZIKV-mediated DNA damage and cell cycle arrest. We conclude that unscheduled expression of ISGs induced by ZIKV alters R-loop homeostasis and perturbs replication fork progression, leading to fork stalling and eventually DNA damage. IFN-dependent R-loop induction represents a previously unknown, nucleic acid-based mechanism for cell cycle arrest.
Collapse
Affiliation(s)
- Yijing Zhao
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Anna D Metzler
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Yangping Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Paola Urlich
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Yilin Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - David M Gilbert
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
6
|
Carstens KE, Gronskaya E, Jäckel D, Bertoli J, Cuevas KR, Dorier J, Wang S, Lopez-Rodriguez D, Shafer TJ, Zurich MG, Pamies D. Application of a high-density microelectrode array assay using a 3D human iPSC-derived brain microphysiological system model for in vitro neurotoxicity screening of environmental compounds. Arch Toxicol 2025:10.1007/s00204-025-04043-x. [PMID: 40293475 DOI: 10.1007/s00204-025-04043-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/27/2025] [Indexed: 04/30/2025]
Abstract
Unraveling the associations between human exposure to environmental chemicals and potential neurotoxicity presents significant challenges. Evaluation of neurotoxicity potential using animal testing is resource-intensive (financial, labor, and animal use) and faces uncertainties regarding biological relevance to human health outcomes. Therefore, there is a need to develop efficient and human-relevant in vitro new approach methodologies (NAMs) to screen and evaluate chemicals for neurotoxicity potential. Recording of neural network activity using microelectrode array (MEA) technology has been identified as a reliable and reproducible method for evaluating neurotoxicity. Much of this research has been performed in 2D rodent-derived cell models. The 'BrainSpheres MEA assay' described in this study offers a promising functional human induced pluripotent stem cell (iPSC)-derived 3D brain model comprising neurons, astrocytes, and oligodendrocytes. We demonstrate consistent spontaneous neuronal firing and network bursting parameters from 7-week-old BrainSpheres using a high-density MEA technology. The performance of this model as a human-relevant NAM was evaluated by conducting a multi-concentration, 13 day exposure study with a set of ten chemicals. Neural activity metrics were assessed and compared to results from a 2D-MEA assay using rodent cells. Loperamide and domoic acid (two assay positive controls) demonstrated similar bioactivity profiles in the BrainSphere MEA assay to the 2D-MEA assay, while acetaminophen (assay negative control) was inactive in both assays. The 2D-MEA model demonstrated more potent bioactivity for 4/7 chemicals that were active in both assays. In the future, reducing replicate variability and testing a larger set of chemicals will likely improve the accuracy and reliability of the assay. These preliminary findings suggest that the BrainSphere assay could be used alongside the rat network formation assay (rNFA) as part of a tiered strategy, where hits in the rNFA are confirmed and further characterized in the BrainSphere model, helping move toward animal-free toxicological testing.
Collapse
Affiliation(s)
- Kelly E Carstens
- Center for Computational Toxicology and Exposure, USA Environmental Protection Agency, Research Triangle Park, North Carolina, NC, 27707, USA
| | | | | | - Jessica Bertoli
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | | | - Julien Dorier
- Bioinformatics Competence Center, University of Lausanne, CH- 1015, Lausanne, Switzerland
- Bioinformatics Competence Center, Ecole Polytechnique Fédérale de Lausanne, CH- 1015, Lausanne, Switzerland
| | - Shan Wang
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - David Lopez-Rodriguez
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland
- Institute of Earth Surface Dynamics, University of Lausanne, Canton de Vaud, Lausanne, Switzerland
| | - Timothy J Shafer
- Center for Computational Toxicology and Exposure, USA Environmental Protection Agency, Research Triangle Park, North Carolina, NC, 27707, USA
| | - Marie-Gabrielle Zurich
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland
| | - David Pamies
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland.
- Stem Cells and Organoid Facility, University of Lausanne, Rue du Bugnon 9, CH-1005, Lausanne, Switzerland.
| |
Collapse
|
7
|
Eun J, Lee JE, Yang SH. Cerebral organoid research for pediatric patients with neurological disorders. Clin Exp Pediatr 2025; 68:269-277. [PMID: 39608368 PMCID: PMC11969208 DOI: 10.3345/cep.2024.01235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/30/2024] Open
Abstract
Cerebral organoids derived from human induced pluripotent stem cells offer a groundbreaking foundation for the analysis of pediatric neurological diseases. Unlike organoids from other somatic systems, cerebral organoids present unique challenges, such as the high sensitivity of neuronal cells to environmental conditions and the complexity of replicating brain-specific architectures. Cerebral organoids replicate the human brain development and pathology, enabling research on conditions such as microcephaly, Rett syndrome, autism spectrum disorders, and brain tumors. This review explores the utility of cerebral organoids for modeling diseases and testing therapeutic interventions. Despite current limitations such as variability and lack of vascularization, recent technological advancements have improved the reliability and application of such interventions. Cerebral organoids provide valuable insight into the mechanisms underlying complex neural disorders and hold promise as novel treatment strategies for pediatric neurological diseases.
Collapse
Affiliation(s)
- Jin Eun
- Department of Neurosurgery, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jung Eun Lee
- Department of Neurosurgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Ho Yang
- Department of Neurosurgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
8
|
Wu X, Xiong D, Liu R, Lai X, Tian Y, Xie Z, Chen L, Hu L, Duan J, Gao X, Zeng X, Dong W, Xu T, Fu F, Yang X, Cheng X, Plewczynski D, Kim M, Xin W, Wang T, Xiang AP, Tang Z. Evolutionary divergence in CTCF-mediated chromatin topology drives transcriptional innovation in humans. Nat Commun 2025; 16:2941. [PMID: 40140405 PMCID: PMC11947266 DOI: 10.1038/s41467-025-58275-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Chromatin topology can impact gene regulation, but how evolutionary divergence in chromatin topology has shaped gene regulatory landscapes for distinctive human traits remains poorly understood. CTCF sites determine chromatin topology by forming domains and loops. Here, we show evolutionary divergence in CTCF-mediated chromatin topology at the domain and loop scales during primate evolution, elucidating distinct mechanisms for shaping regulatory landscapes. Human-specific divergent domains lead to a broad rewiring of transcriptional landscapes. Divergent CTCF loops concord with species-specific enhancer activity, influencing enhancer connectivity to target genes in a concordant yet constrained manner. Under this concordant mechanism, we establish the role of human-specific CTCF loops in shaping transcriptional isoform diversity, with functional implications for disease susceptibility. Furthermore, we validate the function of these human-specific CTCF loops using human forebrain organoids. This study advances our understanding of genetic evolution from the perspective of genome architecture.
Collapse
Affiliation(s)
- Xia Wu
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Dan Xiong
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Rong Liu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangdong, China
| | - Xingqiang Lai
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangdong, China
| | - Yuhan Tian
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Ziying Xie
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Li Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Lanqi Hu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Jingjing Duan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Xinyu Gao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Xian Zeng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Wei Dong
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Ting Xu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Fang Fu
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Xin Yang
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Xinlai Cheng
- Buchmann Institute for Molecular Life Sciences, Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany
| | - Dariusz Plewczynski
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Minji Kim
- Department of Computational Medicine and Bioinformatics, University of Michigan, Michigan, MI, USA
| | - Wenjun Xin
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Tianyun Wang
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, China
- Autism Research Center, Peking University Health Science Center, Beijing, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangdong, China
| | - Zhonghui Tang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.
| |
Collapse
|
9
|
Chopra S, Worhunsky PD, Naganawa M, Zhang XH, Segal A, Orchard E, Cropley V, Wood S, Angarita GA, Cosgrove K, Matuskey D, Nabulsi NB, Huang Y, Carson RE, Esterlis I, Skosnik PD, D’Souza DC, Holmes AJ, Radhakrishnan R. Network-based Molecular Constraints on in vivo Synaptic Density Alterations in Schizophrenia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.22.25324465. [PMID: 40166544 PMCID: PMC11957185 DOI: 10.1101/2025.03.22.25324465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Converging neuroimaging, genetic, and post-mortem evidence show a fundamental role of synaptic deficits in schizophrenia pathogenesis. However, the underlying molecular and cellular mechanisms that drive the onset and progression of synaptic pathology remain to be established. Here, we used synaptic density positron emission tomography (PET) imaging using the [11C]UCB-J radiotracer to reveal a prominent widespread pattern (p FWE < 0.05) of lower synaptic density in individuals with schizophrenia (n=29), compared to a large sample of healthy controls (n=93). We found that the spatial pattern of lower synaptic density in schizophrenia is spatially aligned (r cca = 0.67; p < 0.001) with higher normative distributions of GABAA/BZ, 5HT1B, 5HT2A, and 5HT6, and lower levels of CB1 and 5HT1A. Competing neighborhood deformation network models revealed that regional synaptic pathology strongly correlated with estimates predicted using a model constrained by both interregional structural connectivity and molecular similarity (.42 < r < .61; p FWE < 0.05). These data suggest that synaptic pathology in schizophrenia is jointly constrained by both global axonal connectivity and local molecular vulnerability. Simulation-based network diffusion models were used to identify regions that may represent the initial sources of pathology, nominating left prefrontal areas (p FWE < 0.05) as potential foci from which synaptic pathology initiates and propagates to molecularly similar areas. Overall, our findings provide in vivo evidence for widespread deficit in synaptic density in schizophrenia that is jointly constrained by axonal connectivity and molecular similarity between regions, and that synaptic deficits spread from initial source regions to axonally connected and molecularly similar territories.
Collapse
Affiliation(s)
- Sidhant Chopra
- Department of Psychology, Yale University, New Haven, CT, USA
- Orygen, Parkville, Melbourne, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | | | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Xi-Han Zhang
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Ashlea Segal
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Edwina Orchard
- Department of Psychology, Yale University, New Haven, CT, USA
- Ann S. Bowers Women’s Brain Health Initiative, University of California Santa Barbara, CA, USA
| | - Vanessa Cropley
- Orygen, Parkville, Melbourne, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Stephen Wood
- Orygen, Parkville, Melbourne, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
- School of Psychology, University of Birmingham, UK
| | | | - Kelly Cosgrove
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - David Matuskey
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Nabeel B. Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Richard E. Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Irina Esterlis
- Department of Psychology, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | | | | | - Avram J. Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, USA
| | - Rajiv Radhakrishnan
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| |
Collapse
|
10
|
Seah C, Sidamon-Eristoff AE, Huckins LM, Brennand KJ. Implications of gene × environment interactions in post-traumatic stress disorder risk and treatment. J Clin Invest 2025; 135:e185102. [PMID: 40026250 PMCID: PMC11870735 DOI: 10.1172/jci185102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
Exposure to traumatic stress is common in the general population. Variation in the brain's molecular encoding of stress potentially contributes to the heterogeneous clinical outcomes in response to traumatic experiences. For instance, only a minority of those exposed to trauma will develop post-traumatic stress disorder (PTSD). Risk for PTSD is at least partially heritable, with a growing number of genetic factors identified through GWAS. A major limitation of genetic studies is that they capture only the genetic component of risk, whereas PTSD by definition requires an environmental traumatic exposure. Furthermore, the extent, timing, and type of trauma affects susceptibility. Here, we discuss the molecular mechanisms of PTSD risk together with gene × environment interactions, with a focus on how either might inform genetic screening for individuals at high risk for disease, reveal biological mechanisms that might one day yield novel therapeutics, and impact best clinical practices even today. To close, we discuss the interaction of trauma with sex, gender, and race, with a focus on the implications for treatment. Altogether, we suggest that predicting, preventing, and treating PTSD will require integrating both genotypic and environmental information.
Collapse
Affiliation(s)
- Carina Seah
- Department of Genetics and Genomics and
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anne Elizabeth Sidamon-Eristoff
- Department of Psychiatry, Division of Molecular Psychiatry
- Interdepartmental Neuroscience Program, Wu Tsai Institute, and
- MD-PhD Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Kristen J. Brennand
- Department of Genetics and Genomics and
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Division of Molecular Psychiatry
- Interdepartmental Neuroscience Program, Wu Tsai Institute, and
| |
Collapse
|
11
|
Rui T, Huang H, Zheng K, Fan H, Zhang J, Guo Z, Man H, Brazhe N, Semyanov A, Lu Y, Liu D, Zhu L. Tau Pathology Drives Disease-Associated Astrocyte Reactivity in Salt-Induced Neurodegeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410799. [PMID: 39853966 PMCID: PMC11923866 DOI: 10.1002/advs.202410799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/11/2024] [Indexed: 01/26/2025]
Abstract
Dietary high salt intake is increasingly recognized as a risk factor for cognitive decline and dementia, including Alzheimer's disease (AD). Recent studies have identified a population of disease-associated astrocytes (DAA)-like astrocytes closely linked to amyloid deposition and tau pathology in an AD mouse model. However, the presence and role of these astrocytes in high-salt diet (HSD) models remain unexplored. In this study, it is demonstrated that HSD significantly induces enhanced reactivity of DAA-like astrocytes in the hippocampal CA3 region of mice, with this reactivity being critically dependent on neuronal tau pathology. Neuronal tau pathology activates adenosine A1R signaling, exacerbating tau pathology by inhibiting the Cers1 pathway, which sustains astrocyte reactivity. Additionally, neurons burdened with tau pathology promote astrocyte reactivity via releasing Proteins Associated with Promoting DAA-like Astrocyte Reactivity (PAPD), with Lcn2 playing a pivotal role. Knockout of Lcn2 or its receptor 24p3R significantly mitigates HSD-induced DAA reactivity and neuroinflammation. These findings suggest a vicious cycle between tau pathology and A1R signaling, driving DAA-like astrocyte reactivity. Targeting the Tau-A1R axis may provide a novel therapeutic strategy for reducing HSD-induced neuroinflammation and cognitive deficits.
Collapse
Affiliation(s)
- Tong‐Yu Rui
- Department of PathophysiologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| | - He‐Zhou Huang
- Department of PathophysiologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Kai Zheng
- Department of GeriatricsTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Hong‐Wei Fan
- Department of PathophysiologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Juan Zhang
- Department of PathophysiologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Zi‐Yuan Guo
- Center for Stem Cell and Organoid Medicine (CuSTOM)Division of Developmental BiologyCincinnati Children's Hospital Medical CenterCincinnatiOH45229USA
| | - Heng‐Ye Man
- Department of BiologyBoston UniversityBostonMA02215USA
| | - Nadezhda Brazhe
- Faculty of BiologyM.V. Lomonosov Moscow State UniversityMoscow119234Russia
| | - Alexey Semyanov
- College of MedicineJiaxing UniversityJiaxingZhejiang Province314001China
| | - You‐Ming Lu
- Department of PathophysiologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Dan Liu
- Department of Medical GeneticsSchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Ling‐Qiang Zhu
- Department of PathophysiologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| |
Collapse
|
12
|
Werner S, Pamies D, Zurich MG, Suter-Dick L. Hepatic and extra-hepatic metabolism of propylene glycol ethers in the context of central nervous system toxicity. Toxicology 2025; 512:154081. [PMID: 39929342 DOI: 10.1016/j.tox.2025.154081] [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: 01/09/2025] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Propylene glycol ethers (PGEs) are mixtures of an α-isomer and a β-isomer (β-PGE) that is oxidized via alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH) to potentially neurotoxic alkoxy propionic acids (β-metabolites). While the liver is the primary organ for ADH- and ALDH-mediated metabolism, the contribution to the metabolism of β-PGEs by the blood-brain barrier (BBB) and the brain remains unknown. Here, we aimed to assess the neurotoxic potential of PGEs after systemic exposure by (1) comparing 3D HepaRG and human liver subcellular fraction (S9) for the in vitro determination of the kinetics of hepatic metabolism for β-PGEs, (2) evaluating the BBB-permeability of PGEs and β-metabolites, (3) determining the presence of ADH1 and ALDH2 and the extent of metabolization of β-PGEs in the BBB and brain. The results show that 3D HepaRG and S9 served as competent systems to estimate the enzymatic kinetic (clearance) for β-metabolite formation. We observed that PGEs and the β-metabolites could cross the BBB, based on their permeance across a cellular barrier consisting of the hCMEC/D3 cell line. Metabolic enzymes were not exclusive to the liver, as expression of ADH1 and ALDH2 was demonstrated using RT-qPCR, Western blot, and immunostainings in the BBB in vitro models and in BrainSpheres. Furthermore, LC-MS/MS quantification of the β-metabolites in all in vitro models revealed that 3D HepaRG had a similar metabolic capacity to primary human hepatocytes and that the amount of β-metabolite formed per protein in the BBB was approximately 10-30 % of that in the liver. We also demonstrated active metabolism in the BrainSpheres. In conclusion, the hepatic in vitro models provided data that will help to refine toxicokinetic models and predict internal exposures, thereby supporting the risk assessment of PGEs. In addition, the high permeance of the PGEs and the β-metabolites across the BBB increases the plausibility of neurotoxicity upon systemic exposure. This is further supported by the presence of active ADH1 and ALDH2 enzymes in the BBB in vitro systems and in BrainSpheres, suggesting metabolite formation in the central nervous system. Hence, we suggest that BBB-permeance and extra-hepatic metabolism of the β-PGEs may contribute to the neurotoxicity of PGEs.
Collapse
Affiliation(s)
- Sophie Werner
- University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland.
| | - David Pamies
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland; Department of Biomedical Science, University of Lausanne, Lausanne, Switzerland.
| | - Marie-Gabrielle Zurich
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland; Department of Biomedical Science, University of Lausanne, Lausanne, Switzerland.
| | - Laura Suter-Dick
- University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland.
| |
Collapse
|
13
|
Zhou Y, Su Y, Yang Q, Li J, Hong Y, Gao T, Zhong Y, Ma X, Jin M, Liu X, Yuan N, Kennedy BC, Wang L, Yan L, Viaene AN, Helbig I, Kessler SK, Kleinman JE, Hyde TM, Nauen DW, Liu C, Liu Z, Shen Z, Li C, Xu S, He J, Weinberger DR, Ming GL, Song H. Comparative molecular landscapes of immature neurons in the mammalian dentate gyrus across species reveal special features in humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.16.638557. [PMID: 40027814 PMCID: PMC11870590 DOI: 10.1101/2025.02.16.638557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Immature dentate granule cells (imGCs) arising from adult hippocampal neurogenesis contribute to plasticity, learning and memory, but their evolutionary changes across species and specialized features in humans remain poorly understood. Here we performed machine learning-augmented analysis of published single-cell RNA-sequencing datasets and identified macaque imGCs with transcriptome-wide immature neuronal characteristics. Our cross-species comparisons among humans, monkeys, pigs, and mice showed few shared (such as DPYSL5), but mostly species-specific gene expression in imGCs that converged onto common biological processes regulating neuronal development. We further identified human-specific transcriptomic features of imGCs and demonstrated functional roles of human imGC-enriched expression of a family of proton-transporting vacuolar-type ATPase subtypes in development of imGCs derived from human pluripotent stem cells. Our study reveals divergent gene expression patterns but convergent biological processes in the molecular characteristics of imGCs across species, highlighting the importance of conducting independent molecular and functional analyses for adult neurogenesis in different species.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Yijing Su
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qian Yang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiaqi Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Yan Hong
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Taosha Gao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yanqing Zhong
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xueting Ma
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Mengmeng Jin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Xinglan Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Nini Yuan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Benjamin C. Kennedy
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lizhou Wang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Longying Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Angela N. Viaene
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ingo Helbig
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sudha K. Kessler
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, The Solomon H. Snyder Department of Neuroscience, Department of Neurology, and Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, The Solomon H. Snyder Department of Neuroscience, Department of Neurology, and Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David W. Nauen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cirong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Zhen Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Zhiming Shen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Chao Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Shengjin Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Jie He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Daniel R. Weinberger
- Lieber Institute for Brain Development, The Solomon H. Snyder Department of Neuroscience, Department of Neurology, and Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Guo-li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
14
|
Charles S, Jackson-Holmes E, Sun G, Zhou Y, Siciliano B, Niu W, Han H, Nikitina A, Kemp ML, Wen Z, Lu H. Non-Invasive Quality Control of Organoid Cultures Using Mesofluidic CSTR Bioreactors and High-Content Imaging. ADVANCED MATERIALS TECHNOLOGIES 2025; 10:2400473. [PMID: 40248044 PMCID: PMC12002419 DOI: 10.1002/admt.202400473] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Indexed: 04/19/2025]
Abstract
Human brain organoids produce anatomically relevant cellular structures and recapitulate key aspects of in vivo brain function, which holds great potential to model neurological diseases and screen therapeutics. However, the long growth time of 3D systems complicates the culturing of brain organoids and results in heterogeneity across samples hampering their applications. We developed an integrated platform to enable robust and long-term culturing of 3D brain organoids. We designed a mesofluidic bioreactor device based on a reaction-diffusion scaling theory, which achieves robust media exchange for sufficient nutrient delivery in long-term culture. We integrated this device with longitudinal tracking and machine learning-based classification tools to enable non-invasive quality control of live organoids. This integrated platform allows for sample pre-selection for downstream molecular analysis. Transcriptome analyses of organoids revealed that our mesofluidic bioreactor promoted organoid development while reducing cell death. Our platform thus offers a generalizable tool to establish reproducible culture standards for 3D cellular systems for a variety of applications beyond brain organoids.
Collapse
Affiliation(s)
- Seleipiri Charles
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Emily Jackson-Holmes
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Gongchen Sun
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Ying Zhou
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Benjamin Siciliano
- Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, 615 Michael Street, Atlanta, GA, 30322, U.S.A
| | - Weibo Niu
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Haejun Han
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- School of Biological Sciences, Georgia Institute of Technology, 310 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Arina Nikitina
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Melissa L Kemp
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Hang Lu
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| |
Collapse
|
15
|
Blasco MB, Nisha Aji K, Ramos-Jiménez C, Leppert IR, Tardif CL, Cohen J, Rusjan PM, Mizrahi R. Synaptic Density in Early Stages of Psychosis and Clinical High Risk. JAMA Psychiatry 2025; 82:171-180. [PMID: 39535765 PMCID: PMC11561726 DOI: 10.1001/jamapsychiatry.2024.3608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
Importance Synaptic dysfunction is involved in schizophrenia pathophysiology. However, whether in vivo synaptic density is reduced in early stages of psychosis, including its high-risk states, remains unclear. Objective To investigate whether synaptic density (synaptic vesicle glycoprotein 2A [SV2A] binding potential) is reduced in first-episode psychosis (FEP) and in clinical high risk (CHR) and investigate the effect of cannabis use on synaptic density and examine its relationship with psychotic symptoms and gray matter microstructure across groups. Design, Setting, and Participants This cross-sectional study was performed in a tertiary care psychiatric hospital from July 2021 to October 2023. Participants were patients with antipsychotic-free or minimally exposed FEP or CHR and healthy controls with a clean urine drug screen (except cannabis). Main Outcomes and Measures Synaptic density was quantified with dynamic 90-minute [18F]SynVesT-1 positron emission tomography (PET) scans across prioritized brain regions of interest (ROIs) delineated in individual magnetic resonance images (MRIs). Cannabis use was confirmed with urine drug screens. Gray matter microstructure was assessed using diffusion-weighted MRI to estimate neurite density. Results A total of 49 participants were included, including 16 patients with FEP (mean [SD] age, 26.1 [4.6] years; 9 males and 7 females), 17 patients at CHR (mean [SD] age, 21.2 [3.5] years; 8 males and 9 females), and 16 healthy controls (mean [SD] age, 23.4 [3.6] years; 7 males and 9 females). Synaptic density was significantly different between groups (F2,273 = 4.02, P = .02, Cohen F = 0.17; ROI: F5,273 = 360.18, P < .01, Cohen F = 2.55) with a group × ROI interaction (F10,273 = 2.67, P < .01, Cohen F = 0.32). Synaptic density was lower in cannabis users (F1,272 = 5.31, P = .02, Cohen F = 0.14). Lower synaptic density across groups was associated with more negative symptoms (Positive and Negative Syndrome Scale negative scores: F1,81 = 4.31, P = .04, Cohen F = 0.23; Scale of Psychosis-Risk Symptoms negative scores: F1,90 = 4.12, P = .04, Cohen F = 0.21). SV2A binding potential was significantly associated with neurite density index (F1,138 = 6.76, P = .01, Cohen F = 0.22). Conclusions and Relevance This study found that synaptic density reductions were present during the early stages of psychosis and its risk states and associated with negative symptoms. The implications of SV2A for negative symptoms in psychosis and CHR warrant further investigation. Future studies should investigate the impact of cannabis use on synaptic density in CHR longitudinally.
Collapse
Affiliation(s)
- M. Belen Blasco
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
| | - Kankana Nisha Aji
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
| | - Christian Ramos-Jiménez
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
| | - Ilana Ruth Leppert
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Christine Lucas Tardif
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Johan Cohen
- Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Pablo M. Rusjan
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Romina Mizrahi
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Lu L, Sarkar AK, Dao L, Liu Y, Ma C, Thwin PH, Chang X, Yoshida G, Li A, Wang C, Westerkamp C, Schmitt L, Chelsey M, Stephanie M, Zhao Y, Liu Y, Wang X, Zhu LQ, Liu D, Tchieu J, Miyakoshi M, Zhu H, Gross C, Pedapati E, Salomonis N, Erickson C, Guo Z. An iPSC model of fragile X syndrome reflects clinical phenotypes and reveals m 6 A- mediated epi-transcriptomic dysregulation underlying synaptic dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.14.618205. [PMID: 39464060 PMCID: PMC11507714 DOI: 10.1101/2024.10.14.618205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Fragile X syndrome (FXS), the leading genetic cause of intellectual disability, arises from FMR1 gene silencing and loss of the FMRP protein. N6-methyladenosine (m 6 A) is a prevalent mRNA modification essential for post-transcriptional regulation. FMRP is known to bind to and regulate the stability of m 6 A-containing transcripts. However, how loss of FMRP impacts on transcriptome-wide m 6 A modifications in FXS patients remains unknown. To answer this question, we generated cortical neurons differentiated from induced pluripotent stem cells (iPSC) derived from healthy subjects and FXS patients. In electrophysiology recordings, we validated that synaptic and neuronal network defects in iPSC-derived FXS neurons corresponded to the clinical EEG data of the patients from which the corresponding iPSC line was derived. In analysis of transcriptome-wide methylation, we show that FMRP deficiency led to increased translation of m 6 A writers, resulting in hypermethylation that primarily affecting synapse-associated transcripts and increased mRNA decay. Conversely, in the presence of an m 6 A writer inhibitor, synaptic defects in FXS neurons were rescued. Taken together, our findings uncover that an FMRP-dependent epi-transcriptomic mechanism contributes to FXS pathogenesis by disrupting m 6 A modifications in FXS, suggesting a promising avenue for m 6 A- targeted therapies.
Collapse
|
17
|
Liu C, Gershon ES. Endophenotype 2.0: updated definitions and criteria for endophenotypes of psychiatric disorders, incorporating new technologies and findings. Transl Psychiatry 2024; 14:502. [PMID: 39719446 PMCID: PMC11668880 DOI: 10.1038/s41398-024-03195-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/26/2024] Open
Abstract
Recent genetic studies have linked numerous loci to psychiatric disorders. However, the biological pathways that connect these genetic associations to psychiatric disorders' specific pathophysiological processes are largely unclear. Endophenotypes, first defined over five decades ago, are heritable traits, independent of disease state that are associated with a disease, encompassing a broad range of neurophysiological, biochemical, endocrinological, neuroanatomical, cognitive, and neuropsychological characteristics. Considering the advancements in genetics and genomics over recent decades, we propose a revised definition of endophenotypes as 'genetically influenced phenotypes linked to disease or treatment characteristics and their related events.' We also updated endophenotype criteria to include (1) reliable measurement, (2) association with the disease or its related events, and (3) genetic mediation. 'Genetic mediation' is introduced to differentiate between causality and pleiotropic effects and allows non-linear relationships. Furthermore, this updated Endophenotype 2.0 framework expands to encompass genetically regulated responses to disease-related factors, including environmental risks, illness progression, treatment responses, and resilience phenotypes, which may be state-dependent. This broadened definition paves the way for developing new endophenotypes crucial for genetic analyses in psychiatric disorders. Integrating genetics, genomics, and diverse endophenotypes into multi-dimensional mechanistic models is vital for advancing our understanding of psychiatric disorders. Crucially, elucidating the biological underpinnings of endophenotypes will enhance our grasp of psychiatric genetics, thereby improving disease risk prediction and treatment approaches.
Collapse
Affiliation(s)
- Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
- School of Life Sciences, Central South University, Changsha, China.
| | - Elliot S Gershon
- Departments of Psychiatry and Human Genetics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
18
|
Johns AE, Taga A, Charalampopoulou A, Gross SK, Rust K, McCray BA, Sullivan JM, Maragakis NJ. Exploring P2X7 receptor antagonism as a therapeutic target for neuroprotection in an hiPSC motor neuron model. Stem Cells Transl Med 2024; 13:1198-1212. [PMID: 39419765 PMCID: PMC11631223 DOI: 10.1093/stcltm/szae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 09/01/2024] [Indexed: 10/19/2024] Open
Abstract
ATP is present in negligible concentrations in the interstitium of healthy tissues but accumulates to significantly higher concentrations in an inflammatory microenvironment. ATP binds to 2 categories of purine receptors on the surface of cells, the ionotropic P2X receptors and metabotropic P2Y receptors. Included in the family of ionotropic purine receptors is P2X7 (P2X7R), a non-specific cation channel with unique functional and structural properties that suggest it has distinct roles in pathological conditions marked by increased extracellular ATP. The role of P2X7R has previously been explored in microglia and astrocytes within the context of neuroinflammation, however the presence of P2X7R on human motor neurons and its potential role in neurodegenerative diseases has not been the focus of the current literature. We leveraged the use of human iPSC-derived spinal motor neurons (hiPSC-MN) as well as human and rodent tissue to demonstrate the expression of P2X7R on motor neurons. We extend this observation to demonstrate that these receptors are functionally active on hiPSC-MN and that ATP can directly induce death via P2X7R activation in a dose dependent manner. Finally, using a highly specific P2X7R blocker, we demonstrate how modulation of P2X7R activation on motor neurons is neuroprotective and could provide a unique pharmacologic target for ATP-induced MN death that is distinct from the role of ATP as a modulator of neuroinflammation.
Collapse
Affiliation(s)
- Alexandra E Johns
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Arens Taga
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Andriana Charalampopoulou
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Sarah K Gross
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Khalil Rust
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Brett A McCray
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Jeremy M Sullivan
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Nicholas J Maragakis
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| |
Collapse
|
19
|
LaNoce E, Zhang DY, Garcia-Epelboim A, Su Y, Sun Y, Alepa G, Angelucci AR, Akay-Espinoza C, Jordan-Sciutto KL, Song H, Ming GL, Christian KM. Exposure to the antiretroviral drug dolutegravir impairs structure and neurogenesis in a forebrain organoid model of human embryonic cortical development. Front Mol Neurosci 2024; 17:1459877. [PMID: 39569018 PMCID: PMC11576471 DOI: 10.3389/fnmol.2024.1459877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/13/2024] [Indexed: 11/22/2024] Open
Abstract
Introduction For many therapeutic drugs, including antiretroviral drugs used to treat people living with HIV-1 (PLWH), we have little data on the potential effects on the developing human brain due to limited access to tissue and historical constraints on the inclusion of pregnant populations in clinical trials. Human induced pluripotent stem cells (iPSCs) offer a new avenue to gain insight on how drugs may impact human cell types representative of the developing central nervous system. To prevent vertical transmission of HIV and promote the health of pregnant PLWH, antiretroviral therapy must be initiated and/or maintained throughout pregnancy. However, many antiretroviral drugs are approved for widespread use following clinical testing only in non-pregnant populations and there may be limited information on potential teratogenicity until pregnancy outcomes are evaluated. The integrase strand transfer inhibitor dolutegravir (DTG) is a frontline antiretroviral drug that is effective in viral suppression of HIV but was previously reported to be associated with a slight increase in the risk for neural tube defects in one study, although this has not been replicated in other cohorts. Methods To directly investigate the potential impact of DTG on human cortical neurogenesis, we measured the effects of daily drug exposure on the early stages of corticogenesis in a human iPSC-based forebrain organoid model. We quantified organoid size and structure and analyzed gene and protein expression to evaluate the impact of several doses of DTG on organoid development. Results We observed deficits in organoid structure and impaired neurogenesis in DTG-treated organoids compared to vehicle-treated control organoids after 20 or 40 days in culture. Our highest dose of DTG (10 μM) resulted in significantly smaller organoids with a reduced density of neural rosette structures compared to vehicle-treated controls. Mechanistically, RNA-sequencing and immunohistological analysis suggests dysregulated amino acid transport and activation of the integrated stress response in the DTG-treated organoids, and functionally, a small molecule integrated stress response inhibitor (ISRIB) could partially rescue increased expression of proteins related to cell cycle regulation. Discussion Together, these results illustrate the potential for human iPSC-based strategies to reveal biological processes during neurogenesis that may be affected by therapeutic drugs and provide complementary data in relevant human cell types to augment preclinical investigations of drug safety during pregnancy.
Collapse
Affiliation(s)
- Emma LaNoce
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Y. Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alan Garcia-Epelboim
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yijing Su
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yusha Sun
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Giana Alepa
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Angelina R. Angelucci
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Cagla Akay-Espinoza
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kelly L. Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guo-li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kimberly M. Christian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
20
|
Choudhary A, Peles D, Nayak R, Mizrahi L, Stern S. Current progress in understanding schizophrenia using genomics and pluripotent stem cells: A meta-analytical overview. Schizophr Res 2024; 273:24-38. [PMID: 36443183 DOI: 10.1016/j.schres.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/16/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022]
Abstract
Schizophrenia (SCZ) is a complex, heritable and polygenic neuropsychiatric disease, which disables the patients as well as decreases their life expectancy and quality of life. Common and rare variants studies on SCZ subjects have provided >100 genomic loci that hold importance in the context of SCZ pathophysiology. Transcriptomic studies from clinical samples have informed about the differentially expressed genes (DEGs) and non-coding RNAs in SCZ patients. Despite these advancements, no causative genes for SCZ were found and hence SCZ is difficult to recapitulate in animal models. In the last decade, induced Pluripotent Stem Cells (iPSCs)-based models have helped in understanding the neural phenotypes of SCZ by studying patient iPSC-derived 2D neuronal cultures and 3D brain organoids. Here, we have aimed to provide a simplistic overview of the current progress and advancements after synthesizing the enormous literature on SCZ genetics and SCZ iPSC-based models. Although further understanding of SCZ genetics and pathophysiological mechanisms using these technological advancements is required, the recent approaches have allowed to delineate important cellular mechanisms and biological pathways affected in SCZ.
Collapse
Affiliation(s)
- Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Liron Mizrahi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
21
|
Muhtaseb AW, Duan J. Modeling common and rare genetic risk factors of neuropsychiatric disorders in human induced pluripotent stem cells. Schizophr Res 2024; 273:39-61. [PMID: 35459617 PMCID: PMC9735430 DOI: 10.1016/j.schres.2022.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
Abstract
Recent genome-wide association studies (GWAS) and whole-exome sequencing of neuropsychiatric disorders, especially schizophrenia, have identified a plethora of common and rare disease risk variants/genes. Translating the mounting human genetic discoveries into novel disease biology and more tailored clinical treatments is tied to our ability to causally connect genetic risk variants to molecular and cellular phenotypes. When combined with the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) nuclease-mediated genome editing system, human induced pluripotent stem cell (hiPSC)-derived neural cultures (both 2D and 3D organoids) provide a promising tractable cellular model for bridging the gap between genetic findings and disease biology. In this review, we first conceptualize the advances in understanding the disease polygenicity and convergence from the past decade of iPSC modeling of different types of genetic risk factors of neuropsychiatric disorders. We then discuss the major cell types and cellular phenotypes that are most relevant to neuropsychiatric disorders in iPSC modeling. Finally, we critically review the limitations of iPSC modeling of neuropsychiatric disorders and outline the need for implementing and developing novel methods to scale up the number of iPSC lines and disease risk variants in a systematic manner. Sufficiently scaled-up iPSC modeling and a better functional interpretation of genetic risk variants, in combination with cutting-edge CRISPR/Cas9 gene editing and single-cell multi-omics methods, will enable the field to identify the specific and convergent molecular and cellular phenotypes in precision for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Abdurrahman W Muhtaseb
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Human Genetics, The University of Chicago, Chicago, IL 60637, United States of America
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, United States of America.
| |
Collapse
|
22
|
Santarriaga S, Gerlovin K, Layadi Y, Karmacharya R. Human stem cell-based models to study synaptic dysfunction and cognition in schizophrenia: A narrative review. Schizophr Res 2024; 273:78-97. [PMID: 36925354 PMCID: PMC10500041 DOI: 10.1016/j.schres.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is the strongest predictor of functional outcomes in schizophrenia and is hypothesized to result from synaptic dysfunction. However, targeting synaptic plasticity and cognitive deficits in patients remains a significant clinical challenge. A comprehensive understanding of synaptic plasticity and the molecular basis of learning and memory in a disease context can provide specific targets for the development of novel therapeutics targeting cognitive impairments in schizophrenia. Here, we describe the role of synaptic plasticity in cognition, summarize evidence for synaptic dysfunction in schizophrenia and demonstrate the use of patient derived induced-pluripotent stem cells for studying synaptic plasticity in vitro. Lastly, we discuss current advances and future technologies for bridging basic science research of synaptic dysfunction with clinical and translational research that can be used to predict treatment response and develop novel therapeutics.
Collapse
Affiliation(s)
- Stephanie Santarriaga
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kaia Gerlovin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Layadi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chimie ParisTech, Université Paris Sciences et Lettres, Paris, France
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
23
|
Guo Z, Su Y, Huang WK, Yao XS, Hong Y, Gordin A, Nguyen HN, Wen Z, Ringeling FR, Chen G, Li S, Lu L, Xia M, Zheng W, Sawa A, Chen G, Christian KM, Song H, Ming GL. GABAergic neuron dysregulation in a human neurodevelopmental model for major psychiatric disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614564. [PMID: 39372772 PMCID: PMC11451812 DOI: 10.1101/2024.09.23.614564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
"GABA dysfunction" is a major hypothesis for the biological basis of schizophrenia with indirect supporting evidence from human post-mortem brain and genetic studies. Patient-derived induced pluripotent stem cells (iPSCs) have emerged as a valuable platform for modeling psychiatric disorders, and previous modeling has revealed glutamatergic synapse deficits. Whether GABAergic synapse properties are affected in patient-derived human neurons and how this impacts neuronal network activity remain poorly understood. Here we optimized a protocol to differentiate iPSCs into highly enriched ganglionic eminence-like neural progenitors and GABAergic neurons. Using a collection of iPSCs derived from patients of psychiatric disorders carrying a Disrupted-in-Schizophrenia 1 ( DISC1 ) mutation and their unaffected family member, together with respective isogenic lines, we identified mutation-dependent deficits in GABAergic synapse formation and function, a phenotype similar to that of mutant glutamatergic neurons. However, mutant glutamatergic and GABAergic neurons contribute differentially to neuronal network excitability and synchrony deficits. Finally, we showed that GABAergic synaptic transmission is also defective in neurons derived from several idiopathic schizophrenia patient iPSCs. Transcriptome analysis further showed some shared gene expression dysregulation, which is more prominent in DISC1 mutant neurons. Together, our study supports a functional GABAergic synaptic deficit in major psychiatric disorders.
Collapse
|
24
|
Habela CW, Liu S, Taga A, Dastgheyb R, Haughey N, Bergles D, Song H, Ming GL, Maragakis NJ. Altered development and network connectivity in a human neuronal model of 15q11.2 deletion-related neurodevelopmental disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613912. [PMID: 39345567 PMCID: PMC11429947 DOI: 10.1101/2024.09.19.613912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The chromosome 15q11.2 locus is deleted in 1.5% of patients with genetic epilepsy and confers a risk for intellectual disability and schizophrenia. Individuals with this deletion demonstrate increased cortical thickness, decreased cortical surface area and white matter abnormalities. Human induced pluripotent stem cell (iPSC)-derived neural progenitor cells (NPC) from 15q11.2 deletion individuals exhibit early adhesion junction and migration abnormalities, but later neuronal development and function have not been fully assessed. Imaging studies indicating altered structure and network connectivity in the anterior brain regions and the cingulum suggest that in addition to alterations in progenitor dynamics, there may also be structural and functional changes within discrete networks of mature neurons. To explore this, we generated human forebrain cortical neurons from iPSCs derived from individuals with or without 15q11.2 deletion and used longitudinal imaging and multielectrode array analysis to evaluate neuronal development over time. 15q11.2 deleted neurons exhibited fewer connections and an increase in inhibitory neurons. Individual neurons had decreased neurite complexity and overall decreased neurite length. These structural changes were associated with a reduction in multiunit action potential generation, bursting and synchronization. The 15q11.2 deleted neurons also demonstrated specific functional deficits in glutamate and GABA mediated network activity and synchronization with a delay in the maturation of the inhibitory response to GABA. These data indicate that deletion of the 15q11.2 region is sufficient to impair the structural and functional maturation of cortical neuron networks which likely underlies the pathologic changes in humans with the 15q11.2 deletion.
Collapse
|
25
|
Sun Z, Kwon JS, Ren Y, Chen S, Walker CK, Lu X, Cates K, Karahan H, Sviben S, Fitzpatrick JAJ, Valdez C, Houlden H, Karch CM, Bateman RJ, Sato C, Mennerick SJ, Diamond MI, Kim J, Tanzi RE, Holtzman DM, Yoo AS. Modeling late-onset Alzheimer's disease neuropathology via direct neuronal reprogramming. Science 2024; 385:adl2992. [PMID: 39088624 PMCID: PMC11787906 DOI: 10.1126/science.adl2992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 05/31/2024] [Indexed: 08/03/2024]
Abstract
Late-onset Alzheimer's disease (LOAD) is the most common form of Alzheimer's disease (AD). However, modeling sporadic LOAD that endogenously captures hallmark neuronal pathologies such as amyloid-β (Aβ) deposition, tau tangles, and neuronal loss remains an unmet need. We demonstrate that neurons generated by microRNA (miRNA)-based direct reprogramming of fibroblasts from individuals affected by autosomal dominant AD (ADAD) and LOAD in a three-dimensional environment effectively recapitulate key neuropathological features of AD. Reprogrammed LOAD neurons exhibit Aβ-dependent neurodegeneration, and treatment with β- or γ-secretase inhibitors before (but not subsequent to) Aβ deposit formation mitigated neuronal death. Moreover inhibiting age-associated retrotransposable elements in LOAD neurons reduced both Aβ deposition and neurodegeneration. Our study underscores the efficacy of modeling late-onset neuropathology of LOAD through high-efficiency miRNA-based neuronal reprogramming.
Collapse
Affiliation(s)
- Zhao Sun
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Ji-Sun Kwon
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Program in Computational and Systems Biology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Yudong Ren
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Program in Developmental, Regenerative, and Stem Cell Biology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Shawei Chen
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Courtney K. Walker
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Xinguo Lu
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Kitra Cates
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Program in Molecular Genetics and Genomics, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Hande Karahan
- Stark Neurosciences Research Institute, Indiana University School of Medicine; Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, IN, 46202, USA
| | - Sanja Sviben
- Washington University Center for Cellular Imaging, Washington University School of Medicine; St. Louis, MO, 63110, USA
| | - James A J Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University School of Medicine; St. Louis, MO, 63110, USA
| | - Clarissa Valdez
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center; Dallas, TX, 75390, USA
| | - Henry Houlden
- UCL Institute of Neurology; Queen Square, London, WC1N 3BG, UK
| | - Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO 63110, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine; St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Randall J. Bateman
- Tracy Family SILQ Center for Neurodegenerative Biology; St. Louis, MO, MO 63110, USA
- Department of Neurology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Chihiro Sato
- Tracy Family SILQ Center for Neurodegenerative Biology; St. Louis, MO, MO 63110, USA
- Department of Neurology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Steven J. Mennerick
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Marc I. Diamond
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center; Dallas, TX, 75390, USA
| | - Jungsu Kim
- Stark Neurosciences Research Institute, Indiana University School of Medicine; Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, IN, 46202, USA
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Charlestown, Massachusetts, 02129, USA
| | - David M. Holtzman
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine; St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Andrew S. Yoo
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
| |
Collapse
|
26
|
Charles S, Jackson-Holmes E, Sun G, Zhou Y, Siciliano B, Niu W, Han H, Nikitina A, Kemp ML, Wen Z, Lu H. Non-Invasive Quality Control of Organoid Cultures Using Mesofluidic CSTR Bioreactors and High-Content Imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.19.604365. [PMID: 39091761 PMCID: PMC11291105 DOI: 10.1101/2024.07.19.604365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Human brain organoids produce anatomically relevant cellular structures and recapitulate key aspects of in vivo brain function, which holds great potential to model neurological diseases and screen therapeutics. However, the long growth time of 3D systems complicates the culturing of brain organoids and results in heterogeneity across samples hampering their applications. We developed an integrated platform to enable robust and long-term culturing of 3D brain organoids. We designed a mesofluidic bioreactor device based on a reaction-diffusion scaling theory, which achieves robust media exchange for sufficient nutrient delivery in long-term culture. We integrated this device with longitudinal tracking and machine learning-based classification tools to enable non-invasive quality control of live organoids. This integrated platform allows for sample pre-selection for downstream molecular analysis. Transcriptome analyses of organoids revealed that our mesofluidic bioreactor promoted organoid development while reducing cell death. Our platform thus offers a generalizable tool to establish reproducible culture standards for 3D cellular systems for a variety of applications beyond brain organoids.
Collapse
Affiliation(s)
- Seleipiri Charles
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Emily Jackson-Holmes
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Gongchen Sun
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Ying Zhou
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Benjamin Siciliano
- Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, 615 Michael Street, Atlanta, GA, 30322, U.S.A
| | - Weibo Niu
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Haejun Han
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- School of Biological Sciences, Georgia Institute of Technology, 310 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Arina Nikitina
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Melissa L Kemp
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Hang Lu
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| |
Collapse
|
27
|
Wahbeh MH, Boyd RJ, Yovo C, Rike B, McCallion AS, Avramopoulos D. A functional schizophrenia-associated genetic variant near the TSNARE1 and ADGRB1 genes. HGG ADVANCES 2024; 5:100303. [PMID: 38702885 PMCID: PMC11130735 DOI: 10.1016/j.xhgg.2024.100303] [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: 12/22/2023] [Revised: 05/01/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024] Open
Abstract
Recent collaborative genome-wide association studies (GWAS) have identified >200 independent loci contributing to risk for schizophrenia (SCZ). The genes closest to these loci have diverse functions, supporting the potential involvement of multiple relevant biological processes, yet there is no direct evidence that individual variants are functional or directly linked to specific genes. Nevertheless, overlap with certain epigenetic marks suggest that most GWAS-implicated variants are regulatory. Based on the strength of association with SCZ and the presence of regulatory epigenetic marks, we chose one such variant near TSNARE1 and ADGRB1, rs4129585, to test for functional potential and assay differences that may drive the pathogenicity of the risk allele. We observed that the variant-containing sequence drives reporter expression in relevant neuronal populations in zebrafish. Next, we introduced each allele into human induced pluripotent cells and differentiated four isogenic clones homozygous for the risk allele and five clones homozygous for the non-risk allele into neural progenitor cells. Employing RNA sequencing, we found that the two alleles yield significant transcriptional differences in the expression of 109 genes at a false discovery rate (FDR) of <0.05 and 259 genes at a FDR of <0.1. We demonstrate that these genes are highly interconnected in pathways enriched for synaptic proteins, axon guidance, and regulation of synapse assembly. Exploration of genes near rs4129585 suggests that this variant does not regulate TSNARE1 transcripts, as previously thought, but may regulate the neighboring ADGRB1, a regulator of synaptogenesis. Our results suggest that rs4129585 is a functional common variant that functions in specific pathways likely involved in SCZ risk.
Collapse
Affiliation(s)
- Marah H Wahbeh
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rachel J Boyd
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christian Yovo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bailey Rike
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew S McCallion
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dimitrios Avramopoulos
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
28
|
Salimpour Y, Anderson WS, Dastgheyb R, Liu S, Ming GL, Song H, Maragakis NJ, Habela CW. Phase-amplitude coupling detection and analysis of human 2-dimensional neural cultures in multi-well microelectrode array in vitro. J Neurosci Methods 2024; 407:110127. [PMID: 38615721 PMCID: PMC11351296 DOI: 10.1016/j.jneumeth.2024.110127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/13/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Human induced pluripotent stem cell (hiPSC)- derived neurons offer the possibility of studying human-specific neuronal behaviors in physiologic and pathologic states in vitro. It is unclear whether cultured neurons can achieve the fundamental network behaviors required to process information in the brain. Investigating neuronal oscillations and their interactions, as occurs in cross-frequency coupling (CFC), addresses this question. NEW METHODS We examined whether networks of two-dimensional (2D) cultured hiPSC-derived cortical neurons grown with hiPSC-derived astrocytes on microelectrode array plates recapitulate the CFC that is present in vivo. We employed the modulation index method for detecting phase-amplitude coupling (PAC) and used offline spike sorting to analyze the contribution of single neuron spiking to network behavior. RESULTS We found that PAC is present, the degree of PAC is specific to network structure, and it is modulated by external stimulation with bicuculline administration. Modulation of PAC is not driven by single neurons, but by network-level interactions. COMPARISON WITH EXISTING METHODS PAC has been demonstrated in multiple regions of the human cortex as well as in organoids. This is the first report of analysis demonstrating the presence of coupling in 2D cultures. CONCLUSION CFC in the form of PAC analysis explores communication and integration between groups of neurons and dynamical changes across networks. In vitro PAC analysis has the potential to elucidate the underlying mechanisms as well as capture the effects of chemical, electrical, or ultrasound stimulation; providing insight into modulation of neural networks to treat nervous system disorders in vivo.
Collapse
Affiliation(s)
- Yousef Salimpour
- Functional Neurosurgery Laboratory, Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - William S Anderson
- Functional Neurosurgery Laboratory, Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Raha Dastgheyb
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD, USA
| | - Shiyu Liu
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD, USA
| | - Guo-Li Ming
- 1Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA, USA
| | - Hongjun Song
- 1Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Christa W Habela
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD, USA.
| |
Collapse
|
29
|
Ho MF, Zhang C, Moon I, Zhu X, Coombes BJ, Biernacka J, Skime M, Oesterle TS, Karpyak VM, Schmidt K, Gliske K, Ngo Q, Skillon C, Seppala MD, Li H, Weinshilboum RM. Single cell transcriptomics reveals distinct transcriptional responses to oxycodone and buprenorphine by iPSC-derived brain organoids from patients with opioid use disorder. Mol Psychiatry 2024; 29:1636-1646. [PMID: 36302966 PMCID: PMC10588459 DOI: 10.1038/s41380-022-01837-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022]
Abstract
The opioid epidemic represents a national crisis. Oxycodone is one of the most prescribed opioid medications in the United States, whereas buprenorphine is currently the most prescribed medication for opioid use disorder (OUD) pharmacotherapy. Given the extensive use of prescription opioids and the global opioid epidemic, it is essential to understand how opioids modulate brain cell type function at the single-cell level. We performed single nucleus RNA-seq (snRNA-seq) using iPSC-derived forebrain organoids from three male OUD subjects in response to oxycodone, buprenorphine, or vehicle for seven days. We utilized the snRNA-seq data to identify differentially expressed genes following drug treatment using the Seurat integrative analysis pipeline. We utilized iPSC-derived forebrain organoids and single-cell sequencing technology as an unbiased tool to study cell-type-specific and drug-specific transcriptional responses. After quality control filtering, we analyzed 25787 cells and identified sixteen clusters using unsupervised clustering analysis. Our results reveal distinct transcriptional responses to oxycodone and buprenorphine by iPSC-derived brain organoids from patients with OUD. Specifically, buprenorphine displayed a significant influence on transcription regulation in glial cells. However, oxycodone induced type I interferon signaling in many cell types, including neural cells in brain organoids. Finally, we demonstrate that oxycodone, but not buprenorphine activated STAT1 and induced the type I interferon signaling in patients with OUD. These data suggest that elevation of STAT1 expression associated with OUD might play a role in transcriptional regulation in response to oxycodone. In summary, our results provide novel mechanistic insight into drug action at single-cell resolution.
Collapse
Affiliation(s)
- Ming-Fen Ho
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Irene Moon
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Xiujuan Zhu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Brandon J Coombes
- Division of Computational Biology, Quantitative Health Sciences; Mayo Clinic, Rochester, MN, USA
| | - Joanna Biernacka
- Division of Computational Biology, Quantitative Health Sciences; Mayo Clinic, Rochester, MN, USA
| | - Michelle Skime
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Tyler S Oesterle
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Victor M Karpyak
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | - Kate Gliske
- Hazelden Betty Ford Foundation, Center City, Minnesota, USA
| | - Quyen Ngo
- Hazelden Betty Ford Foundation, Center City, Minnesota, USA
| | - Cedric Skillon
- Hazelden Betty Ford Foundation, Center City, Minnesota, USA
| | | | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
30
|
Ma Y, Gao K, Sun X, Wang J, Yang Y, Wu J, Chai A, Yao L, Liu N, Yu H, Su Y, Lu T, Wang L, Yue W, Zhang X, Xu L, Zhang D, Li J. STON2 variations are involved in synaptic dysfunction and schizophrenia-like behaviors by regulating Syt1 trafficking. Sci Bull (Beijing) 2024; 69:1458-1471. [PMID: 38402028 DOI: 10.1016/j.scib.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/13/2023] [Accepted: 02/06/2024] [Indexed: 02/26/2024]
Abstract
Synaptic dysfunction is a core component of the pathophysiology of schizophrenia. However, the genetic risk factors and molecular mechanisms related to synaptic dysfunction are still not fully understood. The Stonin 2 (STON2) gene encodes a major adaptor for clathrin-mediated endocytosis (CME) of synaptic vesicles. In this study, we showed that the C-C (307Pro-851Ala) haplotype of STON2 increases the susceptibility to schizophrenia and examined whether STON2 variations cause schizophrenia-like behaviors through the regulation of CME. We found that schizophrenia-related STON2 variations led to protein dephosphorylation, which affected its interaction with synaptotagmin 1 (Syt1), a calcium sensor protein located in the presynaptic membrane that is critical for CME. STON2307Pro851Ala knockin mice exhibited deficits in synaptic transmission, short-term plasticity, and schizophrenia-like behaviors. Moreover, among seven antipsychotic drugs, patients with the C-C (307Pro-851Ala) haplotype responded better to haloperidol than did the T-A (307Ser-851Ser) carriers. The recovery of deficits in Syt1 sorting and synaptic transmission by acute administration of haloperidol effectively improved schizophrenia-like behaviors in STON2307Pro851Ala knockin mice. Our findings demonstrated the effect of schizophrenia-related STON2 variations on synaptic dysfunction through the regulation of CME, which might be attractive therapeutic targets for treating schizophrenia-like phenotypes.
Collapse
Affiliation(s)
- Yuanlin Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China; The First Affiliated Hospital, Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Kai Gao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China; Changping Laboratory, Beijing 102206, China
| | - Xiaoxuan Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China
| | - Jinxin Wang
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Yang Yang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China
| | - Jianying Wu
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Anping Chai
- Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Li Yao
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Nan Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Hao Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China
| | - Yi Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China
| | - Tianlan Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China
| | - Lifang Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China
| | - Weihua Yue
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Xiaohui Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Lin Xu
- Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Dai Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China; Changping Laboratory, Beijing 102206, China
| | - Jun Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Key Laboratory of Mental Health, Chinese Academy of Medical Sciences, Beijing 100191, China; Changping Laboratory, Beijing 102206, China.
| |
Collapse
|
31
|
Jana A, Nath A, Sen P, Kundu S, Alghamdi BS, Abujamel TS, Saboor M, Woon-Khiong C, Alexiou A, Papadakis M, Alam MZ, Ashraf GM. Unraveling the Endocannabinoid System: Exploring Its Therapeutic Potential in Autism Spectrum Disorder. Neuromolecular Med 2024; 26:20. [PMID: 38744725 PMCID: PMC11093854 DOI: 10.1007/s12017-024-08781-6] [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: 12/08/2023] [Accepted: 03/04/2024] [Indexed: 05/16/2024]
Abstract
The salient features of autism spectrum disorder (ASD) encompass persistent difficulties in social communication, as well as the presence of restricted and repetitive facets of behavior, hobbies, or pursuits, which are often accompanied with cognitive limitations. Over the past few decades, a sizable number of studies have been conducted to enhance our understanding of the pathophysiology of ASD. Preclinical rat models have proven to be extremely valuable in simulating and analyzing the roles of a wide range of established environmental and genetic factors. Recent research has also demonstrated the significant involvement of the endocannabinoid system (ECS) in the pathogenesis of several neuropsychiatric diseases, including ASD. In fact, the ECS has the potential to regulate a multitude of metabolic and cellular pathways associated with autism, including the immune system. Moreover, the ECS has emerged as a promising target for intervention with high predictive validity. Particularly noteworthy are resent preclinical studies in rodents, which describe the onset of ASD-like symptoms after various genetic or pharmacological interventions targeting the ECS, providing encouraging evidence for further exploration in this area.
Collapse
Affiliation(s)
- Ankit Jana
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Arnab Nath
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Palash Sen
- School of Biosciences, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Swikriti Kundu
- Siksha Bhavana, Visva-Bharati University, Bolpur, West Bengal, 731235, India
| | - Badrah S Alghamdi
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Turki S Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad Saboor
- Department of Medical Laboratory Sciences, College of Health Sciences, and Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Chan Woon-Khiong
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, Greece
- Department of Research & Development, AFNP Med, 1030, Vienna, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohammad Zubair Alam
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, and Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
32
|
Yang X, Delsante M, Daneshpajouhnejad P, Fenaroli P, Mandell KP, Wang X, Takahashi S, Halushka MK, Kopp JB, Levi M, Rosenberg AZ. Bile Acid Receptor Agonist Reverses Transforming Growth Factor-β1-Mediated Fibrogenesis in Human Induced Pluripotent Stem Cells-Derived Kidney Organoids. J Transl Med 2024; 104:100336. [PMID: 38266922 DOI: 10.1016/j.labinv.2024.100336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/26/2024] Open
Abstract
Chronic kidney disease progresses through the replacement of functional tissue compartments with fibrosis, a maladaptive repair process. Shifting kidney repair toward a physiologically intact architecture, rather than fibrosis, is key to blocking chronic kidney disease progression. Much research into the mechanisms of fibrosis is performed in rodent models with less attention to the human genetic context. Recently, human induced pluripotent stem cell (iPSC)-derived organoids have shown promise in overcoming the limitation. In this study, we developed a fibrosis model that uses human iPSC-based 3-dimensional renal organoids, in which exogenous transforming growth factor-β1 (TGF-β1) induced the production of extracellular matrix. TGF-β1-treated organoids showed tubulocentric collagen 1α1 production by regulating downstream transcriptional regulators, Farnesoid X receptor, phosphorylated mothers against decapentaplegic homolog 3 (p-SMAD3), and transcriptional coactivator with PDZ-binding motif (TAZ). Increased nuclear TAZ expression was confirmed in the tubular epithelium in human kidney biopsies with tubular injury and early fibrosis. A dual bile acid receptor agonist (INT-767) increased Farnesoid X receptor and reduced p-SMAD3 and TAZ, attenuating TGF-β1-induced fibrosis in kidney organoids. Finally, we show that TAZ interacted with TEA-domain transcription factors and p-SMAD3 with TAZ and TEA-domain transcription factor 4 coregulating collagen 1α1 gene transcription. In summary, we establish a novel, readily manipulable fibrogenesis model and posit a role for bile acid receptor agonism early in renal parenchymal fibrosis.
Collapse
Affiliation(s)
- Xiaoping Yang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Marco Delsante
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland; Scuola di Specializione in Nefrologia, University of Parma, Parma, Italy
| | | | - Paride Fenaroli
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland; Scuola di Specializione in Nefrologia, University of Parma, Parma, Italy
| | | | - Xiaoxin Wang
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Shogo Takahashi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Marc K Halushka
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
33
|
Cerneckis J, Cai H, Shi Y. Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications. Signal Transduct Target Ther 2024; 9:112. [PMID: 38670977 PMCID: PMC11053163 DOI: 10.1038/s41392-024-01809-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/09/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
The induced pluripotent stem cell (iPSC) technology has transformed in vitro research and holds great promise to advance regenerative medicine. iPSCs have the capacity for an almost unlimited expansion, are amenable to genetic engineering, and can be differentiated into most somatic cell types. iPSCs have been widely applied to model human development and diseases, perform drug screening, and develop cell therapies. In this review, we outline key developments in the iPSC field and highlight the immense versatility of the iPSC technology for in vitro modeling and therapeutic applications. We begin by discussing the pivotal discoveries that revealed the potential of a somatic cell nucleus for reprogramming and led to successful generation of iPSCs. We consider the molecular mechanisms and dynamics of somatic cell reprogramming as well as the numerous methods available to induce pluripotency. Subsequently, we discuss various iPSC-based cellular models, from mono-cultures of a single cell type to complex three-dimensional organoids, and how these models can be applied to elucidate the mechanisms of human development and diseases. We use examples of neurological disorders, coronavirus disease 2019 (COVID-19), and cancer to highlight the diversity of disease-specific phenotypes that can be modeled using iPSC-derived cells. We also consider how iPSC-derived cellular models can be used in high-throughput drug screening and drug toxicity studies. Finally, we discuss the process of developing autologous and allogeneic iPSC-based cell therapies and their potential to alleviate human diseases.
Collapse
Affiliation(s)
- Jonas Cerneckis
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Hongxia Cai
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
34
|
Davis BA, Chen HY, Ye Z, Ostlund I, Tippani M, Das D, Sripathy SR, Wang Y, Martin JM, Shim G, Panchwagh NM, Moses RL, Farinelli F, Bohlen JF, Li M, Luikart BW, Jaffe AE, Maher BJ. TCF4 Mutations Disrupt Synaptic Function Through Dysregulation of RIMBP2 in Patient-Derived Cortical Neurons. Biol Psychiatry 2024; 95:662-675. [PMID: 37573005 PMCID: PMC10858293 DOI: 10.1016/j.biopsych.2023.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/21/2023] [Accepted: 07/14/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Genetic variation in the TCF4 (transcription factor 4) gene is associated with risk for a variety of developmental and psychiatric conditions, which includes a syndromic form of autism spectrum disorder called Pitt-Hopkins syndrome (PTHS). TCF4 encodes an activity-dependent transcription factor that is highly expressed during cortical development and in animal models has been shown to regulate various aspects of neuronal development and function. However, our understanding of how disease-causing mutations in TCF4 confer pathophysiology in a human context is lacking. METHODS To model PTHS, we differentiated human cortical neurons from human induced pluripotent stem cells that were derived from patients with PTHS and neurotypical individuals. To identify pathophysiology and disease mechanisms, we assayed cortical neurons with whole-cell electrophysiology, Ca2+ imaging, multielectrode arrays, immunocytochemistry, and RNA sequencing. RESULTS Cortical neurons derived from patients with TCF4 mutations showed deficits in spontaneous synaptic transmission, network excitability, and homeostatic plasticity. Transcriptomic analysis indicated that these phenotypes resulted in part from altered expression of genes involved in presynaptic neurotransmission and identified the presynaptic binding protein RIMBP2 as the most differentially expressed gene in PTHS neurons. Remarkably, TCF4-dependent deficits in spontaneous synaptic transmission and network excitability were rescued by increasing RIMBP2 expression in presynaptic neurons. CONCLUSIONS Taken together, these results identify TCF4 as a critical transcriptional regulator of human synaptic development and plasticity and specifically identifies dysregulation of presynaptic function as an early pathophysiology in PTHS.
Collapse
Affiliation(s)
- Brittany A Davis
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Huei-Ying Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Zengyou Ye
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Isaac Ostlund
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Debamitra Das
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Srinidhi Rao Sripathy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Yanhong Wang
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Jacqueline M Martin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Gina Shim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Neel M Panchwagh
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Rebecca L Moses
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Federica Farinelli
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Joseph F Bohlen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Meijie Li
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland; McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Brady J Maher
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
35
|
Ho MF, Zhang C, Moon I, Tuncturk M, Coombes BJ, Biernacka J, Skime M, Oesterle TS, Karpyak VM, Li H, Weinshilboum R. Molecular mechanisms involved in alcohol craving, IRF3, and endoplasmic reticulum stress: a multi-omics study. Transl Psychiatry 2024; 14:165. [PMID: 38531832 PMCID: PMC10965952 DOI: 10.1038/s41398-024-02880-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
Alcohol use disorder (AUD) is the most prevalent substance use disorder worldwide. Acamprosate and naltrexone are anti-craving drugs used in AUD pharmacotherapy. However, molecular mechanisms underlying their anti-craving effect remain unclear. This study utilized a patient-derived induced pluripotent stem cell (iPSC)-based model system and anti-craving drugs that are used to treat AUD as "molecular probes" to identify possible mechanisms associated with alcohol craving. We examined the pathophysiology of craving and anti-craving drugs by performing functional genomics studies using iPSC-derived astrocytes and next-generation sequencing. Specifically, RNA sequencing performed using peripheral blood mononuclear cells from AUD patients with extreme values for alcohol craving intensity prior to treatment showed that inflammation-related pathways were highly associated with alcohol cravings. We then performed a genome-wide assessment of chromatin accessibility and gene expression profiles of induced iPSC-derived astrocytes in response to ethanol or anti-craving drugs. Those experiments identified drug-dependent epigenomic signatures, with IRF3 as the most significantly enriched motif in chromatin accessible regions. Furthermore, the activation of IRF3 was associated with ethanol-induced endoplasmic reticulum (ER) stress which could be attenuated by anti-craving drugs, suggesting that ER stress attenuation might be a target for anti-craving agents. In conclusion, we found that craving intensity was associated with alcohol consumption and treatment outcomes. Our functional genomic studies suggest possible relationships among craving, ER stress, IRF3 and the actions of anti-craving drugs.
Collapse
Affiliation(s)
- Ming-Fen Ho
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Irene Moon
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Mustafa Tuncturk
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Brandon J Coombes
- Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Joanna Biernacka
- Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Michelle Skime
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Tyler S Oesterle
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Victor M Karpyak
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
36
|
Heider J, Stahl A, Sperlich D, Hartmann SM, Vogel S, Breitmeyer R, Templin M, Volkmer H. Defined co-cultures of glutamatergic and GABAergic neurons with a mutation in DISC1 reveal aberrant phenotypes in GABAergic neurons. BMC Neurosci 2024; 25:12. [PMID: 38438989 PMCID: PMC10910844 DOI: 10.1186/s12868-024-00858-z] [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: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Mutations in the gene DISC1 are associated with increased risk for schizophrenia, bipolar disorder and major depression. The study of mutated DISC1 represents a well-known and comprehensively characterized approach to understand neuropsychiatric disease mechanisms. However, previous studies have mainly used animal models or rather heterogeneous populations of iPSC-derived neurons, generated by undirected differentiation, to study the effects of DISC1 disruption. Since major hypotheses to explain neurodevelopmental, psychiatric disorders rely on altered neuronal connectivity observed in patients, an ideal iPSC-based model requires accurate representation of the structure and complexity of neuronal circuitries. In this study, we made use of an isogenic cell line with a mutation in DISC1 to study neuronal synaptic phenotypes in a culture system comprising a defined ratio of NGN2 and ASCL1/DLX2 (AD2)-transduced neurons, enriched for glutamatergic and GABAergic neurons, respectively, to mimic properties of the cortical microcircuitry. RESULTS In heterozygous DISC1 mutant neurons, we replicated the expected phenotypes including altered neural progenitor proliferation as well as neurite outgrowth, deregulated DISC1-associated signaling pathways, and reduced synaptic densities in cultures composed of glutamatergic neurons. Cultures comprising a defined ratio of NGN2 and AD2 neurons then revealed considerably increased GABAergic synapse densities, which have not been observed in any iPSC-derived model so far. Increased inhibitory synapse densities could be associated with an increased efficiency of GABAergic differentiation, which we observed in AD2-transduced cultures of mutant neurons. Additionally, we found increased neuronal activity in GABAergic neurons through calcium imaging while the activity pattern of glutamatergic neurons remained unchanged. CONCLUSIONS In conclusion, our results demonstrate phenotypic differences in a co-culture comprising a defined ratio of DISC1 mutant NGN2 and AD2 neurons, as compared to culture models comprising only one neuronal cell type. Altered synapse numbers and neuronal activity imply that DISC1 impacts the excitatory/inhibitory balance in NGN2/AD2 co-cultures, mainly through increased GABAergic input.
Collapse
Affiliation(s)
- Johanna Heider
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Aaron Stahl
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Denise Sperlich
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Sophia-Marie Hartmann
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Sabrina Vogel
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Ricarda Breitmeyer
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Markus Templin
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Hansjürgen Volkmer
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany.
| |
Collapse
|
37
|
Liu J, Chen S, Huang G, Wen P, Zhou X, Wu Y. Trisomy 21-driven metabolite alterations are linked to cellular injuries in Down syndrome. Cell Mol Life Sci 2024; 81:112. [PMID: 38433139 PMCID: PMC10909777 DOI: 10.1007/s00018-024-05127-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/10/2023] [Accepted: 01/14/2024] [Indexed: 03/05/2024]
Abstract
Down syndrome (DS) arises from a genetic anomaly characterized by an extra copy of chromosome 21 (exCh21). Despite high incidence of congenital diseases among DS patients, direct impacts of exCh21 remain elusive. Here, we established a robust DS model harnessing human-induced pluripotent stem cells (hiPSCs) from mosaic DS patient. These hiPSC lines encompassed both those with standard karyotype and those carrying an extra copy of exCh21, allowing to generate isogenic cell lines with a consistent genetic background. We unraveled that exCh21 inflicted disruption upon the cellular transcriptome, ushering in alterations in metabolic processes and triggering DNA damage. The impact of exCh21 was also manifested in profound modifications in chromatin accessibility patterns. Moreover, we identified two signature metabolites, 5-oxo-ETE and Calcitriol, whose biosynthesis is affected by exCh21. Notably, supplementation with 5-oxo-ETE promoted DNA damage, in stark contrast to the protective effect elicited by Calcitriol against such damage. We also found that exCh21 disrupted cardiogenesis, and that this impairment could be mitigated through supplementation with Calcitriol. Specifically, the deleterious effects of 5-oxo-ETE unfolded in the form of DNA damage induction and the repression of cardiogenesis. On the other hand, Calcitriol emerged as a potent activator of its nuclear receptor VDR, fostering amplified binding to chromatin and subsequent facilitation of gene transcription. Our findings provide a comprehensive understanding of exCh21's metabolic implications within the context of Down syndrome, offering potential avenues for therapeutic interventions for Down syndrome treatment.
Collapse
Affiliation(s)
- Juli Liu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| | - Shaoxian Chen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Guiping Huang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Pengju Wen
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Xianwu Zhou
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - Yueheng Wu
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
38
|
Bellon A. Comparing stem cells, transdifferentiation and brain organoids as tools for psychiatric research. Transl Psychiatry 2024; 14:127. [PMID: 38418498 PMCID: PMC10901833 DOI: 10.1038/s41398-024-02780-8] [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: 04/17/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 03/01/2024] Open
Abstract
The inaccessibility of neurons coming directly from patients has hindered our understanding of mental illnesses at the cellular level. To overcome this obstacle, six different cellular approaches that carry the genetic vulnerability to psychiatric disorders are currently available: Olfactory Neuroepithelial Cells, Mesenchymal Stem Cells, Pluripotent Monocytes, Induced Pluripotent Stem Cells, Induced Neuronal cells and more recently Brain Organoids. Here we contrast advantages and disadvantages of each of these six cell-based methodologies. Neuronal-like cells derived from pluripotent monocytes are presented in more detail as this technique was recently used in psychiatry for the first time. Among the parameters used for comparison are; accessibility, need for reprograming, time to deliver differentiated cells, differentiation efficiency, reproducibility of results and cost. We provide a timeline on the discovery of these cell-based methodologies, but, our main goal is to assist researchers selecting which cellular approach is best suited for any given project. This manuscript also aims to help readers better interpret results from the published literature. With this goal in mind, we end our work with a discussion about the differences and similarities between cell-based techniques and postmortem research, the only currently available tools that allow the study of mental illness in neurons or neuronal-like cells coming directly from patients.
Collapse
Affiliation(s)
- Alfredo Bellon
- Penn State Hershey Medical Center, Department of Psychiatry and Behavioral Health, Hershey, PA, USA.
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, USA.
| |
Collapse
|
39
|
Huang Y, Guo X, Lu S, Chen Q, Wang Z, Lai L, Liu Q, Zhu X, Luo L, Li J, Huang Y, Gao H, Zhang Z, Bu Q, Cen X. Long-term exposure to cadmium disrupts neurodevelopment in mature cerebral organoids. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:168923. [PMID: 38065485 DOI: 10.1016/j.scitotenv.2023.168923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/25/2023] [Accepted: 11/25/2023] [Indexed: 01/18/2024]
Abstract
Cadmium (Cd) is a pervasive environmental pollutant. Increasing evidence suggests that Cd exposure during pregnancy can induce adverse neurodevelopmental outcomes. However, due to the limitations of neural cell and animal models, it is challenging to study the developmental neurotoxicity and underlying toxicity mechanism of long-term exposure to environmental pollutants during human brain development. In this study, chronic Cd exposure was performed in human mature cerebral organoids for 49 or 77 days. Our study found that prolonged exposure to Cd resulted in the inhibition of cerebral organoid growth and the disruption of neural differentiation and cortical layer organization. These potential consequences of chronic Cd exposure may include impaired GFAP expression, a reduction in SOX2+ neuronal progenitor cells, an increase in TUJ1+ immature neurons, as well as an initial increase and a subsequent decrease in both TBR2+ intermediate progenitors and CTIP2+ deep layer cortical neurons. Transcriptomic analyses revealed that long-term exposure to Cd disrupted zinc and copper ion homeostasis through excessive synthesis of metallothionein and disturbed synaptogenesis, as evidenced by inhibited postsynaptic protein. Our study employed mature cerebral organoids to evaluate the developmental neurotoxicity induced by long-term Cd exposure.
Collapse
Affiliation(s)
- Yan Huang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xinhua Guo
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Shiya Lu
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Qiqi Chen
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiqiu Wang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Li Lai
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Qian Liu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Xizhi Zhu
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Li Luo
- Department of Gynaecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yina Huang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Gao
- Department of Food Science and Technology, College of Biomass and Engineering, Sichuan University, Chengdu 610065, China
| | - Zunzhen Zhang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Qian Bu
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
40
|
Abstract
Brain development in humans is achieved through precise spatiotemporal genetic control, the mechanisms of which remain largely elusive. Recently, integration of technological advances in human stem cell-based modelling with genome editing has emerged as a powerful platform to establish causative links between genotypes and phenotypes directly in the human system. Here, we review our current knowledge of complex genetic regulation of each key step of human brain development through the lens of evolutionary specialization and neurodevelopmental disorders and highlight the use of human stem cell-derived 2D cultures and 3D brain organoids to investigate human-enriched features and disease mechanisms. We also discuss opportunities and challenges of integrating new technologies to reveal the genetic architecture of human brain development and disorders.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
41
|
Perrottelli A, Marzocchi FF, Caporusso E, Giordano GM, Giuliani L, Melillo A, Pezzella P, Bucci P, Mucci A, Galderisi S. Advances in the understanding of the pathophysiology of schizophrenia and bipolar disorder through induced pluripotent stem cell models. J Psychiatry Neurosci 2024; 49:E109-E125. [PMID: 38490647 PMCID: PMC10950363 DOI: 10.1503/jpn.230112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/04/2023] [Accepted: 01/08/2024] [Indexed: 03/17/2024] Open
Abstract
The pathophysiology of schizophrenia and bipolar disorder involves a complex interaction between genetic and environmental factors that begins in the early stages of neurodevelopment. Recent advancements in the field of induced pluripotent stem cells (iPSCs) offer a promising tool for understanding the neurobiological alterations involved in these disorders and, potentially, for developing new treatment options. In this review, we summarize the results of iPSC-based research on schizophrenia and bipolar disorder, showing disturbances in neurodevelopmental processes, imbalance in glutamatergic-GABAergic transmission and neuromorphological alterations. The limitations of the reviewed literature are also highlighted, particularly the methodological heterogeneity of the studies, the limited number of studies developing iPSC models of both diseases simultaneously, and the lack of in-depth clinical characterization of the included samples. Further studies are needed to advance knowledge on the common and disease-specific pathophysiological features of schizophrenia and bipolar disorder and to promote the development of new treatment options.
Collapse
Affiliation(s)
| | | | | | | | - Luigi Giuliani
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonio Melillo
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Paola Bucci
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Armida Mucci
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | | |
Collapse
|
42
|
Wahbeh MH, Boyd RJ, Yovo C, Rike B, McCallion AS, Avramopoulos D. A Functional Schizophrenia-associated genetic variant near the TSNARE1 and ADGRB1 genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.570831. [PMID: 38187620 PMCID: PMC10769312 DOI: 10.1101/2023.12.18.570831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Recent collaborative genome wide association studies (GWAS) have identified >200 independent loci contributing to risk for schizophrenia (SCZ). The genes closest to these loci have diverse functions, supporting the potential involvement of multiple relevant biological processes; yet there is no direct evidence that individual variants are functional or directly linked to specific genes. Nevertheless, overlap with certain epigenetic marks suggest that most GWAS-implicated variants are regulatory. Based on the strength of association with SCZ and the presence of regulatory epigenetic marks, we chose one such variant near TSNARE1 and ADGRB1, rs4129585, to test for functional potential and assay differences that may drive the pathogenicity of the risk allele. We observed that the variant-containing sequence drives reporter expression in relevant neuronal populations in zebrafish. Next, we introduced each allele into human induced pluripotent cells and differentiated 4 isogenic clones homozygous for the risk allele and 5 clones homozygous for the non-risk allele into neural precursor cells. Employing RNA-seq, we found that the two alleles yield significant transcriptional differences in the expression of 109 genes at FDR <0.05 and 259 genes at FDR <0.1. We demonstrate that these genes are highly interconnected in pathways enriched for synaptic proteins, axon guidance, and regulation of synapse assembly. Exploration of genes near rs4129585 suggests that this variant does not regulate TSNARE1 transcripts, as previously thought, but may regulate the neighboring ADGRB1, a regulator of synaptogenesis. Our results suggest that rs4129585 is a functional common variant that functions in specific pathways likely involved in SCZ risk.
Collapse
Affiliation(s)
- Marah H Wahbeh
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rachel J Boyd
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christian Yovo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bailey Rike
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew S McCallion
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dimitrios Avramopoulos
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
43
|
Chen X, Xie L, Sheehy R, Xiong Y, Muneer A, Wrobel J, Park KS, Liu J, Velez J, Luo Y, Li YD, Quintanilla L, Li Y, Xu C, Wen Z, Song J, Jin J, Deshmukh M. Novel brain-penetrant inhibitor of G9a methylase blocks Alzheimer's disease proteopathology for precision medication. RESEARCH SQUARE 2023:rs.3.rs-2743792. [PMID: 38045363 PMCID: PMC10690335 DOI: 10.21203/rs.3.rs-2743792/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Current amyloid beta-targeting approaches for Alzheimer's disease (AD) therapeutics only slow cognitive decline for small numbers of patients. This limited efficacy exists because AD is a multifactorial disease whose pathological mechanism(s) and diagnostic biomarkers are largely unknown. Here we report a new mechanism of AD pathogenesis in which the histone methyltransferase G9a noncanonically regulates translation of a hippocampal proteome that defines the proteopathic nature of AD. Accordingly, we developed a novel brain-penetrant inhibitor of G9a, MS1262, across the blood-brain barrier to block this G9a-regulated, proteopathologic mechanism. Intermittent MS1262 treatment of multiple AD mouse models consistently restored both cognitive and noncognitive functions to healthy levels. Comparison of proteomic/phosphoproteomic analyses of MS1262-treated AD mice with human AD patient data identified multiple pathological brain pathways that elaborate amyloid beta and neurofibrillary tangles as well as blood coagulation, from which biomarkers of early stage of AD including SMOC1 were found to be affected by MS1262 treatment. Notably, these results indicated that MS1262 treatment may reduce or avoid the risk of blood clot burst for brain bleeding or a stroke. This mouse-to-human conservation of G9a-translated AD proteopathology suggests that the global, multifaceted effects of MS1262 in mice could extend to relieve all symptoms of AD patients with minimum side effect. In addition, our mechanistically derived biomarkers can be used for stage-specific AD diagnosis and companion diagnosis of individualized drug effects.
Collapse
Affiliation(s)
- Xian Chen
- University of North Carolina at Chapel Hill
| | - Ling Xie
- University of North Carolina at Chapel Hill
| | | | - Yan Xiong
- Icahn School of Medicine at Mount Sinai
| | | | | | | | - Jing Liu
- Icahn School of Medicine at Mount Sinai
| | | | - Yanjia Luo
- University of North Carolina at Chapel Hill
| | - Ya-Dong Li
- University of North Carolina at Chapel Hill
| | | | - Yongyi Li
- University of North Carolina at Chapel Hill
| | | | | | - Juan Song
- University of North Carolina at Chapel Hill
| | - Jian Jin
- Icahn School of Medicine at Mount Sinai
| | | |
Collapse
|
44
|
Lv S, He E, Luo J, Liu Y, Liang W, Xu S, Zhang K, Yang Y, Wang M, Song Y, Wu Y, Cai X. Using Human-Induced Pluripotent Stem Cell Derived Neurons on Microelectrode Arrays to Model Neurological Disease: A Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301828. [PMID: 37863819 PMCID: PMC10667858 DOI: 10.1002/advs.202301828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/04/2023] [Indexed: 10/22/2023]
Abstract
In situ physiological signals of in vitro neural disease models are essential for studying pathogenesis and drug screening. Currently, an increasing number of in vitro neural disease models are established using human-induced pluripotent stem cell (hiPSC) derived neurons (hiPSC-DNs) to overcome interspecific gene expression differences. Microelectrode arrays (MEAs) can be readily interfaced with two-dimensional (2D), and more recently, three-dimensional (3D) neural stem cell-derived in vitro models of the human brain to monitor their physiological activity in real time. Therefore, MEAs are emerging and useful tools to model neurological disorders and disease in vitro using human iPSCs. This is enabling a real-time window into neuronal signaling at the network scale from patient derived. This paper provides a comprehensive review of MEA's role in analyzing neural disease models established by hiPSC-DNs. It covers the significance of MEA fabrication, surface structure and modification schemes for hiPSC-DNs culturing and signal detection. Additionally, this review discusses advances in the development and use of MEA technology to study in vitro neural disease models, including epilepsy, autism spectrum developmental disorder (ASD), and others established using hiPSC-DNs. The paper also highlights the application of MEAs combined with hiPSC-DNs in detecting in vitro neurotoxic substances. Finally, the future development and outlook of multifunctional and integrated devices for in vitro medical diagnostics and treatment are discussed.
Collapse
Affiliation(s)
- Shiya Lv
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Enhui He
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- The State Key Lab of Brain‐Machine IntelligenceZhejiang UniversityHangzhou321100China
| | - Jinping Luo
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yaoyao Liu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wei Liang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Shihong Xu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Kui Zhang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yan Yang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mixia Wang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yilin Song
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yirong Wu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xinxia Cai
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
45
|
Ho MF, Zhang C, Moon I, Biernacka J, Coombes B, Ngo Q, Skillon C, Skime M, Oesterle T, Croarkin PE, Karpyak VM, Li H, Weinshilboum RM. Epigenetic regulation of GABA catabolism in iPSC-derived neurons: The molecular links between FGF21 and histone methylation. Mol Metab 2023; 77:101798. [PMID: 37689244 PMCID: PMC10514449 DOI: 10.1016/j.molmet.2023.101798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/27/2023] [Accepted: 09/03/2023] [Indexed: 09/11/2023] Open
Abstract
OBJECTIVE Fibroblast growth factor 21 (FGF21) analogs have been tested as potential therapeutics for substance use disorders. Prior research suggests that FGF21 administration might affect alcohol consumption and reward behaviors. Our recent report showed that plasma FGF21 levels were positively correlated with alcohol use in patients with alcohol use disorder (AUD). FGF21 has a short half-life (0.5-2 h) and crosses the blood-brain barrier. Therefore, we set out to identify molecular mechanisms for both the naïve form of FGF21 and a long-acting FGF21 molecule (PF-05231023) in induced pluripotent stem cell (iPSC)-derived forebrain neurons. METHODS We performed RNA-seq in iPSC-derived forebrain neurons treated with naïve FGF21 or PF-05231023 at physiologically relevant concentrations. We obtained plasma levels of FGF21 and GABA from our previous AUD clinical trial (n = 442). We performed ELISA for FGF21 in both iPSC-derived forebrain neurons and forebrain organoids. We determined protein interactions using co-immunoprecipitation. Finally, we applied ChIP assays to confirm the occupancy of REST, EZH2 and H3K27me3 by FGF21 using iPSC-derived forebrain neurons with and without drug exposure. RESULTS We identified 4701 and 1956 differentially expressed genes in response to naïve FGF21 or PF-05231023, respectively (FDR < 0.05). Notably, 974 differentially expressed genes overlapped between treatment with naïve FGF21 and PF-05231023. REST was the most important upstream regulator of differentially expressed genes. The GABAergic synapse pathway was the most significant pathway identified using the overlapping genes. We also observed a significant positive correlation between plasma FGF21 and GABA concentrations in AUD patients. In parallel, FGF21 and PF-05231023 significantly induced GABA levels in iPSC-derived neurons. Finally, functional genomics studies showed a drug-dependent occupancy of REST, EZH2, and H3K27me3 in the promoter regions of genes involved in GABA catabolism which resulted in transcriptional repression. CONCLUSIONS Our results highlight a significant role in the epigenetic regulation of genes involved in GABA catabolism related to FGF21 action. (The ClinicalTrials.gov Identifier: NCT00662571).
Collapse
Affiliation(s)
- Ming-Fen Ho
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| | - Cheng Zhang
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Irene Moon
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Joanna Biernacka
- Division of Computational Biology, Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Brandon Coombes
- Division of Computational Biology, Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Quyen Ngo
- Hazelden Betty Ford Foundation, Center City, MN, USA
| | | | - Michelle Skime
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Tyler Oesterle
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Paul E Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Victor M Karpyak
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
46
|
Xie L, Sheehy RN, Xiong Y, Muneer A, Wrobel JA, Park KS, Velez J, Liu J, Luo YJ, Li YD, Quintanilla L, Li Y, Xu C, Deshmukh M, Wen Z, Jin J, Song J, Chen X. Novel brain-penetrant inhibitor of G9a methylase blocks Alzheimer's disease proteopathology for precision medication. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.25.23297491. [PMID: 37961307 PMCID: PMC10635198 DOI: 10.1101/2023.10.25.23297491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Current amyloid beta-targeting approaches for Alzheimer's disease (AD) therapeutics only slow cognitive decline for small numbers of patients. This limited efficacy exists because AD is a multifactorial disease whose pathological mechanism(s) and diagnostic biomarkers are largely unknown. Here we report a new mechanism of AD pathogenesis in which the histone methyltransferase G9a noncanonically regulates translation of a hippocampal proteome that defines the proteopathic nature of AD. Accordingly, we developed a novel brain-penetrant inhibitor of G9a, MS1262, across the blood-brain barrier to block this G9a-regulated, proteopathologic mechanism. Intermittent MS1262 treatment of multiple AD mouse models consistently restored both cognitive and noncognitive functions to healthy levels. Comparison of proteomic/phosphoproteomic analyses of MS1262-treated AD mice with human AD patient data identified multiple pathological brain pathways that elaborate amyloid beta and neurofibrillary tangles as well as blood coagulation, from which biomarkers of early stage of AD including SMOC1 were found to be affected by MS1262 treatment. Notably, these results indicated that MS1262 treatment may reduce or avoid the risk of blood clot burst for brain bleeding or a stroke. This mouse-to-human conservation of G9a-translated AD proteopathology suggests that the global, multifaceted effects of MS1262 in mice could extend to relieve all symptoms of AD patients with minimum side effect. In addition, our mechanistically derived biomarkers can be used for stage-specific AD diagnosis and companion diagnosis of individualized drug effects. One-Sentence Summary A brain-penetrant inhibitor of G9a methylase blocks G9a translational mechanism to reverse Alzheimer's disease related proteome for effective therapy.
Collapse
|
47
|
Mastwal S, Li X, Stowell R, Manion M, Zhang W, Kim NS, Yoon KJ, Song H, Ming GL, Wang KH. Adolescent neurostimulation of dopamine circuit reverses genetic deficits in frontal cortex function. eLife 2023; 12:RP87414. [PMID: 37830916 PMCID: PMC10575630 DOI: 10.7554/elife.87414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023] Open
Abstract
Dopamine system dysfunction is implicated in adolescent-onset neuropsychiatric disorders. Although psychosis symptoms can be alleviated by antipsychotics, cognitive symptoms remain unresponsive and novel paradigms investigating the circuit substrates underlying cognitive deficits are critically needed. The frontal cortex and its dopaminergic input from the midbrain are implicated in cognitive functions and undergo maturational changes during adolescence. Here, we used mice carrying mutations in Arc or Disc1 to model mesofrontal dopamine circuit deficiencies and test circuit-based neurostimulation strategies to restore cognitive functions. We found that in a memory-guided spatial navigation task, frontal cortical neurons were activated coordinately at the decision-making point in wild-type but not Arc-/- mice. Chemogenetic stimulation of midbrain dopamine neurons or optogenetic stimulation of frontal cortical dopamine axons in a limited adolescent period consistently reversed genetic defects in mesofrontal innervation, task-coordinated neuronal activity, and memory-guided decision-making at adulthood. Furthermore, adolescent stimulation of dopamine neurons also reversed the same cognitive deficits in Disc1+/- mice. Our findings reveal common mesofrontal circuit alterations underlying the cognitive deficits caused by two different genes and demonstrate the feasibility of adolescent neurostimulation to reverse these circuit and behavioral deficits. These results may suggest developmental windows and circuit targets for treating cognitive deficits in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Surjeet Mastwal
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
| | - Xinjian Li
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
| | - Rianne Stowell
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Matthew Manion
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
| | - Wenyu Zhang
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Nam-Shik Kim
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Ki-Jun Yoon
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Hongjun Song
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Guo-Li Ming
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Kuan Hong Wang
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical CenterRochesterUnited States
| |
Collapse
|
48
|
Kumar A, Karuppagounder SS, Chen Y, Corona C, Kawaguchi R, Cheng Y, Balkaya M, Sagdullaev BT, Wen Z, Stuart C, Cho S, Ming GL, Tuvikene J, Timmusk T, Geschwind DH, Ratan RR. 2-Deoxyglucose drives plasticity via an adaptive ER stress-ATF4 pathway and elicits stroke recovery and Alzheimer's resilience. Neuron 2023; 111:2831-2846.e10. [PMID: 37453419 PMCID: PMC10528360 DOI: 10.1016/j.neuron.2023.06.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/10/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023]
Abstract
Intermittent fasting (IF) is a diet with salutary effects on cognitive aging, Alzheimer's disease (AD), and stroke. IF restricts a number of nutrient components, including glucose. 2-deoxyglucose (2-DG), a glucose analog, can be used to mimic glucose restriction. 2-DG induced transcription of the pro-plasticity factor, Bdnf, in the brain without ketosis. Accordingly, 2-DG enhanced memory in an AD model (5xFAD) and functional recovery in an ischemic stroke model. 2-DG increased Bdnf transcription via reduced N-linked glycosylation, consequent ER stress, and activity of ATF4 at an enhancer of the Bdnf gene, as well as other regulatory regions of plasticity/regeneration (e.g., Creb5, Cdc42bpa, Ppp3cc, and Atf3) genes. These findings demonstrate an unrecognized role for N-linked glycosylation as an adaptive sensor to reduced glucose availability. They further demonstrate that ER stress induced by 2-DG can, in the absence of ketosis, lead to the transcription of genes involved in plasticity and cognitive resilience as well as proteostasis.
Collapse
Affiliation(s)
- Amit Kumar
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Saravanan S Karuppagounder
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Yingxin Chen
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Carlo Corona
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Riki Kawaguchi
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yuyan Cheng
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mustafa Balkaya
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Botir T Sagdullaev
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA; Regeneron Pharmaceuticals, Tarrytown, New York, NY, USA
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Charles Stuart
- East Tennessee State University Quillen College of Medicine, Johnson City, TN, USA
| | - Sunghee Cho
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Guo-Li Ming
- Department of Neuroscience, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jürgen Tuvikene
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Tõnis Timmusk
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Daniel H Geschwind
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rajiv R Ratan
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA.
| |
Collapse
|
49
|
Jgamadze D, Harary PM, Castellanos M, Blue R, Song H, Ming GL, Chen HI. Protocol for human brain organoid transplantation into a rat visual cortex to model neural repair. STAR Protoc 2023; 4:102470. [PMID: 37585295 PMCID: PMC10436235 DOI: 10.1016/j.xpro.2023.102470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/09/2023] [Accepted: 06/29/2023] [Indexed: 08/18/2023] Open
Abstract
Human stem-cell-derived organoids represent a promising substrate for transplantation-based neural repair. Here, we describe a protocol for transplanting forebrain organoids into an injured adult rat visual cortex. This protocol includes surgical details for craniectomy, aspiration injury, organoid transplantation, and cranioplasty. This platform represents a valuable tool for investigating the efficacy of organoids as structured grafts for neural repair. For complete details on the use and execution of this protocol, please refer to Jgamadze et al.1.
Collapse
Affiliation(s)
- Dennis Jgamadze
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Paul M Harary
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mackenzie Castellanos
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rachel Blue
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - H Isaac Chen
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA.
| |
Collapse
|
50
|
Zhang S, Zhang H, Forrest MP, Zhou Y, Sun X, Bagchi VA, Kozlova A, Santos MD, Piguel NH, Dionisio LE, Sanders AR, Pang ZP, He X, Penzes P, Duan J. Multiple genes in a single GWAS risk locus synergistically mediate aberrant synaptic development and function in human neurons. CELL GENOMICS 2023; 3:100399. [PMID: 37719141 PMCID: PMC10504676 DOI: 10.1016/j.xgen.2023.100399] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/22/2023] [Accepted: 08/07/2023] [Indexed: 09/19/2023]
Abstract
The mechanistic tie between genome-wide association study (GWAS)-implicated risk variants and disease-relevant cellular phenotypes remains largely unknown. Here, using human induced pluripotent stem cell (hiPSC)-derived neurons as a neurodevelopmental model, we identify multiple schizophrenia (SZ) risk variants that display allele-specific open chromatin (ASoC) and are likely to be functional. Editing the strongest ASoC SNP, rs2027349, near vacuolar protein sorting 45 homolog (VPS45) alters the expression of VPS45, lncRNA AC244033.2, and a distal gene, C1orf54. Notably, the transcriptomic changes in neurons are associated with SZ and other neuropsychiatric disorders. Neurons carrying the risk allele exhibit increased dendritic complexity and hyperactivity. Interestingly, individual/combinatorial gene knockdown shows that these genes alter cellular phenotypes in a non-additive synergistic manner. Our study reveals that multiple genes at a single GWAS risk locus mediate a compound effect on neural function, providing a mechanistic link between a non-coding risk variant and disease-related cellular phenotypes.
Collapse
Affiliation(s)
- Siwei Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Hanwen Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Marc P. Forrest
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA
| | - Yifan Zhou
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Xiaotong Sun
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Vikram A. Bagchi
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA
| | - Alena Kozlova
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Marc Dos Santos
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA
| | - Nicolas H. Piguel
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA
| | - Leonardo E. Dionisio
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA
| | - Alan R. Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Zhiping P. Pang
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Xin He
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Peter Penzes
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|