229 results on '"Guo-Li Ming"'
Search Results
2. Epigenetic and epitranscriptomic regulation of axon regeneration
- Author
-
Yating Cheng, Hongjun Song, Guo-li Ming, and Yi-Lan Weng
- Subjects
Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
3. Opportunities and limitations for studying neuropsychiatric disorders using patient-derived induced pluripotent stem cells
- Author
-
Yan Hong, Qian Yang, Hongjun Song, and Guo-li Ming
- Subjects
Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
4. The IAP antagonist birinapant enhances chimeric antigen receptor T cell therapy for glioblastoma by overcoming antigen heterogeneity
- Author
-
Edward Z. Song, Xin Wang, Benjamin I. Philipson, Qian Zhang, Radhika Thokala, Logan Zhang, Charles-Antoine Assenmacher, Zev A. Binder, Guo-li Ming, Donald M. O’Rourke, Hongjun Song, and Michael C. Milone
- Subjects
Cancer Research ,Oncology ,Molecular Medicine ,Pharmacology (medical) - Abstract
Antigen heterogeneity that results in tumor antigenic escape is one of the major obstacles to successful chimeric antigen receptor (CAR) T cell therapies in solid tumors including glioblastoma multiforme (GBM). To address this issue and improve the efficacy of CAR T cell therapy for GBM, we developed an approach that combines CAR T cells with inhibitor of apoptosis protein (IAP) antagonists, a new class of small molecules that mediate the degradation of IAPs, to treat GBM. Here, we demonstrated that the IAP antagonist birinapant could sensitize GBM cell lines and patient-derived primary GBM organoids to apoptosis induced by CAR T cell-derived cytokines, such as tumor necrosis factor. Therefore, birinapant could enhance CAR T cell-mediated bystander death of antigen-negative GBM cells, thus preventing tumor antigenic escape in antigen-heterogeneous tumor models
- Published
- 2022
- Full Text
- View/download PDF
5. CYFIP1 Dosages Exhibit Divergent Behavioral Impact via Diametric Regulation of NMDA Receptor Complex Translation in Mouse Models of Psychiatric Disorders
- Author
-
Valina L. Dawson, Guo Li Ming, Kimberly M. Christian, Stephanie J. Temme, Weidong Li, Stephen M. Eacker, Kuei Sen Hsu, Stefan Canzar, Ki Jun Yoon, Francisca Rojas Ringeling, Ted M. Dawson, Ha Nam Nguyen, Bo Xiao, Paul F. Worley, Yu Ting Lin, Namshik Kim, Hongjun Song, and Ying Zhou
- Subjects
0301 basic medicine ,N-Methylaspartate ,DNA Copy Number Variations ,Autism Spectrum Disorder ,RNA-binding protein ,Biology ,Receptors, N-Methyl-D-Aspartate ,Gene dosage ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Postsynaptic potential ,mental disorders ,medicine ,Animals ,RNA, Messenger ,Copy-number variation ,Biological Psychiatry ,Adaptor Proteins, Signal Transducing ,Messenger RNA ,Mental Disorders ,Translation (biology) ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Schizophrenia ,RNA ,NMDA receptor ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Background Gene dosage imbalance caused by copy number variations (CNVs) is a prominent contributor to brain disorders. In particular, 15q11.2 CNV duplications and deletions have been associated with autism spectrum disorder and schizophrenia, respectively. The mechanism underlying these diametric contributions remains unclear. Methods We established both loss-of-function and gain-of-function mouse models of Cyfip1, one of four genes within 15q11.2 CNVs. To assess the functional consequences of altered CYFIP1 levels, we performed systematic investigations on behavioral, electrophysiological, and biochemical phenotypes in both mouse models. In addition, we utilized RNA immunoprecipitation sequencing (RIP-seq) analysis to reveal molecular targets of CYFIP1 in vivo. Results Cyfip1 loss-of-function and gain-of function mouse models exhibited distinct and shared behavioral abnormalities related to autism spectrum disorder and schizophrenia. RIP-seq analysis identified messenger RNA targets of CYFIP1 in vivo, including postsynaptic NMDA receptor (NMDAR) complex components. In addition, these mouse models showed diametric changes in levels of postsynaptic NMDAR complex components at synapses because of dysregulated protein translation, resulting in bidirectional alteration of NMDAR-mediated signaling. Importantly, pharmacological balancing of NMDAR signaling in these mouse models with diametric Cyfip1 dosages rescues behavioral abnormalities. Conclusions CYFIP1 regulates protein translation of NMDAR and associated complex components at synapses to maintain normal synaptic functions and behaviors. Our integrated analyses provide insight into how gene dosage imbalance caused by CNVs may contribute to divergent neuropsychiatric disorders.
- Published
- 2022
- Full Text
- View/download PDF
6. A nomenclature consensus for nervous system organoids and assembloids
- Author
-
Sergiu P. Pașca, Paola Arlotta, Helen S. Bateup, J. Gray Camp, Silvia Cappello, Fred H. Gage, Jürgen A. Knoblich, Arnold R. Kriegstein, Madeline A. Lancaster, Guo-Li Ming, Alysson R. Muotri, In-Hyun Park, Orly Reiner, Hongjun Song, Lorenz Studer, Sally Temple, Giuseppe Testa, Barbara Treutlein, and Flora M. Vaccarino
- Subjects
Multidisciplinary - Published
- 2022
- Full Text
- View/download PDF
7. Special properties of adult neurogenesis in the human hippocampus: Implications for its clinical applications
- Author
-
Yi Zhou, Yijing Su, Guo‐li Ming, and Hongjun Song
- Subjects
Molecular Medicine ,Medicine (miscellaneous) - Published
- 2023
- Full Text
- View/download PDF
8. Single-Soma Deep Rna Sequencing of Human DRG Neurons Reveals Novel Molecular and Cellular Mechanisms Underlying Somatosensation
- Author
-
Huasheng Yu, Dmitry Usoskin, Saad S. Nagi, Yizhou Hu, Jussi Kupari, Otmane Bouchatta, Suna Li Cranfill, Yijing Su, You Lv, Hongjun Song, Guo-Li Ming, Stephen Prouty, John Seykora, Hao Wu, Minghong Ma, Håkan Olausson, Patrik Ernfors, and Wenqin Luo
- Subjects
Article - Abstract
The versatility of somatosensation arises from heterogenous human dorsal root ganglion (DRG) neurons. The critical information to decipher their functions, i.e., the soma transcriptome, is lacking due to technical difficulties. Here we developed a novel approach to isolate individual human DRG neuron somas for deep RNA sequencing (RNA-seq). On average, >9000 unique genes per neuron were detected, and 16 neuronal types were identified. Cross-species analyses revealed that touch-, cold-, and itch-sensing neuronal types were relatively conserved, while the pain-sensing neurons displayed marked divergence. Soma transcriptomes of human DRG neurons predicted novel functional features, which were confirmed using single-cellin vivoelectrophysiological recordings. These results support a close relationship the between physiological properties of human sensory afferents and molecular profiles uncovered by the single-soma RNA-seq dataset. In summary, by conducting single-soma RNA-seq of human DRG neurons, we generated an unprecedented neural atlas for human somatosensation.
- Published
- 2023
- Full Text
- View/download PDF
9. 454 Effects of Human Brain Organoid Age on Post-Transplant Growth in the Rat Motor Cortex
- Author
-
Rachel Blue, Marissa Fu, Paul Harary, Dennis Jgamadze, Imon Rahaman, Shikha Singh, Guo-Li Ming, Hongjun Song, and H. Isaac Chen
- Subjects
Surgery ,Neurology (clinical) - Published
- 2023
- Full Text
- View/download PDF
10. Global remapping in granule cells and mossy cells of the mouse dentate gyrus
- Author
-
Sang Hoon Kim, Douglas GoodSmith, Stephanie J. Temme, Fumika Moriya, Guo-li Ming, Kimberly M. Christian, Hongjun Song, and James J. Knierim
- Subjects
General Biochemistry, Genetics and Molecular Biology - Abstract
SUMMARYHippocampal place cells exhibit spatially modulated firing, or place fields, which can remap to encode changes in the environment or other variables. Unique among hippocampal subregions, the dentate gyrus (DG) has two excitatory populations of place cells, granule cells and mossy cells, which are among the least and most active spatially modulated cells in the hippocampus, respectively. Previous studies of remapping in the DG have drawn different conclusions about whether granule cells exhibit global remapping and contribute to the encoding of context specificity. By recording granule cells and mossy cells as mice foraged in different environments, we found that by most measures, both granule cells and mossy cells remapped robustly but through different mechanisms that are consistent with firing properties of each cell type. Our results resolve the ambiguity surrounding remapping in the DG and suggest that most spatially modulated granule cells contribute to orthogonal representations of distinct spatial contexts.
- Published
- 2022
- Full Text
- View/download PDF
11. PUS7: a targetable epitranscriptomic regulator of glioblastoma growth
- Author
-
Daniel Y. Zhang, Hongjun Song, and Guo-li Ming
- Subjects
Pharmacology ,chemistry.chemical_classification ,Regulator ,Translation (biology) ,Toxicology ,Small molecule ,Pseudouridine ,Cell biology ,chemistry.chemical_compound ,Enzyme ,RNA, Transfer ,chemistry ,Tyrosine kinase 2 ,Interferon ,Transfer RNA ,medicine ,Humans ,Glioblastoma ,Intramolecular Transferases ,medicine.drug - Abstract
Pseudouridine is the most abundant yet unexplored RNA modification in glioblastoma. Cui and coworkers find that PUS7, a pseudouridine depositing enzyme, promotes tumor growth and can be targeted by small molecule inhibitors. Mechanistically, PUS7 modifies tRNAs, reduces TYK2 translation, and downregulates a proliferation-restricting interferon-STAT1 pathway in glioblastoma.
- Published
- 2021
- Full Text
- View/download PDF
12. Author response: Label-free three-photon imaging of intact human cerebral organoids for tracking early events in brain development and deficits in Rett syndrome
- Author
-
Murat Yildirim, Chloe Delepine, Danielle Feldman, Vincent A Pham, Stephanie Chou, Jacque Ip, Alexi Nott, Li-Huei Tsai, Guo-Li Ming, Peter TC So, and Mriganka Sur
- Published
- 2022
- Full Text
- View/download PDF
13. Basal forebrain cholinergic circuits orchestrate diverse cell types in the adult dentate gyrus to support neural stem cell function and spatial memory
- Author
-
Luis Quintanilla, Yijing Su, Jeremy M. Simon, Yan-Jia Luo, Brent Asrican, Seth Tart, Ryan N. Sheehy, Ya-Dong Li, Guo-li Ming, Hongjun Song, and Juan Song
- Abstract
SummaryDentate gyrus (DG) is a critical structure involved in spatial memory and adult neurogenesis, two distinct processes dynamically regulated by local circuits comprising diverse populations of DG cells. It remains unknown how these DG cells are orchestrated to regulate these distinct hippocampal functions. Here we report activation of a cholinergic circuit from the Diagonal Band of Broca to DG promotes quiescent radial neural stem cell (rNSC) activation and spatial memory. Furthermore, single-nucleus RNA-sequencing reveals broad transcriptomic changes across DG mature and adult-born cells in response to cholinergic-circuit activation. Notably, neuronal populations exhibit cholinergic-activity-induced molecular changes related to synaptic functions crucial for spatial memory; while rNSCs exhibit changes related to structural remodeling and neurogenic proliferation crucial for hippocampal neurogenesis. Electrophysiology and NicheNet analyses reveal granule cells, endothelial cells, and astrocytes as potential intermediaries for cholinergic regulation of rNSCs. Our findings reveal cell-type-specific signaling mechanisms underlying cholinergic regulation of distinct DG functions.
- Published
- 2022
- Full Text
- View/download PDF
14. Loss-of-function mutation in human Oxidation Resistance gene 1 disrupts the spatial-temporal regulation of histone arginine methylation in early brain development
- Author
-
Xiaolin Lin, Wei Wang, Mingyi Yang, Nadirah Damseh, Mirta Mittelstedt Leal de Sousa, Fadi Jacob, Anna Lång, Elise Kristiansen, Marco Pannone, Miroslava Kissova, Runar Almaas, Anna Kuśnierczyk, Richard Siller, Maher Shahrour, Motee Al-Ashhab, Bassam Abu-Libdeh, Wannan Tang, Geir Slupphaug, Orly Elpeleg, Stig Ove Bøe, Lars Eide, Gareth J Sullivan, Johanne Egge Rinholm, Hongjun Song, Guo-li Ming, Barbara van Loon, Simon Edvardson, Jing Ye, and Magnar Bjørås
- Abstract
We report a loss-of-function mutation in the TLDc domain of human Oxidation Resistance 1 (OXR1) gene, resulting in early-onset epilepsy, developmental delay, cognitive disabilities, and cerebellar atrophy. Patient lymphoblasts show impaired cell survival, proliferation, and hypersensitivity to oxidative stress. These phenotypes are rescued by TLDc domain replacement. We generated patient derived induced pluripotent stem cells (iPSCs) revealing impaired neural differentiation along with dysregulation of genes essential for neurodevelopment. We identified that OXR1 influences histone arginine methylation by activating protein arginine methyltransferases (PRMTs), suggesting OXR1 dependent mechanisms regulating gene expression during neurodevelopment. We modeled the function of OXR1 in early human brain development using patient derived brain organoids revealing that OXR1 contributes to the spatial-temporal regulation of histone arginine methylation in specific brain regions. Our work provides new insights into pathological features and molecular underpinnings associated with OXR1 deficiency, highlighting the therapeutic potential of OXR1 in numerous neurodegenerative and neurodevelopmental disorders.
- Published
- 2022
- Full Text
- View/download PDF
15. Generation and biobanking of patient-derived glioblastoma organoids and their application in CAR T cell testing
- Author
-
Fadi Jacob, Hongjun Song, and Guo Li Ming
- Subjects
0303 health sciences ,T cell ,Tissue Processing ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Chimeric antigen receptor ,Cryopreservation ,In vitro ,03 medical and health sciences ,Tissue culture ,Chemically defined medium ,0302 clinical medicine ,medicine.anatomical_structure ,Cancer research ,medicine ,Receptor ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Glioblastoma tumors exhibit extensive inter- and intratumoral heterogeneity, which has contributed to the poor outcomes of numerous clinical trials and continues to complicate the development of effective therapeutic strategies. Most in vitro models do not preserve the cellular and mutational diversity of parent tumors and often require a lengthy generation time with variable efficiency. Here, we describe detailed procedures for generating glioblastoma organoids (GBOs) from surgically resected patient tumor tissue using a chemically defined medium without cell dissociation. By preserving cell-cell interactions and minimizing clonal selection, GBOs maintain the cellular heterogeneity of parent tumors. We include details of how to passage and cryopreserve GBOs for continued use, biobanking and long-term recovery. In addition, we describe procedures for investigating patient-specific responses to immunotherapies by co-culturing GBOs with chimeric antigen receptor (CAR) T cells. It takes ~2–4 weeks to generate GBOs and 5–7 d to perform CAR T cell co-culture using this protocol. Competence with human cell culture, tissue processing, immunohistology and microscopy is required for optimal results. The authors describe procedures for generating and biobanking glioblastoma organoids from patient tumor tissue and testing of chimeric antigen receptor T cell efficacy by co-culture. Tissue processing, immunohistology and detection of hypoxic gradients and actively proliferating cells are also described.
- Published
- 2020
- Full Text
- View/download PDF
16. Modeling traumatic brain injury with human brain organoids
- Author
-
Guo Li Ming, Victoria E. Johnson, H. Isaac Chen, Dennis Jgamadze, John A. Wolf, Douglas H. Smith, Hongjun Song, and D. Kacy Cullen
- Subjects
0303 health sciences ,business.industry ,Traumatic brain injury ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,Cognition ,02 engineering and technology ,Human brain ,Neural tissues ,021001 nanoscience & nanotechnology ,medicine.disease ,Article ,Biomaterials ,03 medical and health sciences ,medicine.anatomical_structure ,Organoid ,Medicine ,0210 nano-technology ,business ,Neuroscience ,030304 developmental biology - Abstract
Traumatic brain injury (TBI) remains a prominent public health concern despite several decades of attempts to develop therapies for the associated neurological and cognitive deficits. Effective models of this condition are imperative for better defining its pathophysiology and testing therapeutics. Human brain organoids are stem cell–derived neural tissues that recapitulate many of the steps of normal neurodevelopment, resulting in the reproduction of a substantial degree of brain architecture. Organoids are highly relevant to clinical conditions because of their human nature and three-dimensional tissue structure, yet they are easier to manipulate and interrogate experimentally than animals. Thus, they have the potential to serve as a novel platform for studying TBI. In this article, we discuss available in vitro models of TBI, active areas of inquiry on brain organoids, and how these two concepts could be merged.
- Published
- 2020
- Full Text
- View/download PDF
17. Label-free three-photon imaging of intact human cerebral organoids: tracking early events in brain development and deficits in Rett Syndrome
- Author
-
Murat Yildirim, Chloe Delepine, Danielle Feldman, Vincent Pham, Stephanie Chou, Jacque Pak Kan Ip, Alexi Nott, Li-Huei Tsai, Guo-li Ming, Peter T. C. So, and Mriganka Sur
- Abstract
Human cerebral organoids are unique in their development of progenitor-rich zones akin to ventricular zones from which neuronal progenitors differentiate and migrate radially. Analyses of cerebral organoids thus far have been performed in sectioned tissue or in superficial layers due to their high scattering properties. Here, we demonstrate label-free three-photon imaging of whole, uncleared intact organoids (∼2 mm depth) to assess early events of early human brain development. Optimizing a custom-made three-photon microscope to image intact cerebral organoids generated from Rett Syndrome patients, we show defects in the ventricular zone volumetric structure of mutant organoids compared to isogenic control organoids. Long-term imaging live organoids reveals that shorter migration distances and slower migration speeds of mutant radially migrating neurons are associated with more tortuous trajectories. Our label-free imaging system constitutes a particularly useful platform for tracking normal and abnormal development in individual organoids, as well as for screening therapeutic molecules via intact organoid imaging.
- Published
- 2022
- Full Text
- View/download PDF
18. Label-free three-photon imaging of intact human cerebral organoids for tracking early events in brain development and deficits in Rett syndrome
- Author
-
Murat Yildirim, Chloe Delepine, Danielle Feldman, Vincent A Pham, Stephanie Chou, Jacque Ip, Alexi Nott, Li-Huei Tsai, Guo-Li Ming, Peter TC So, and Mriganka Sur
- Subjects
Neurons ,Organoids ,General Immunology and Microbiology ,General Neuroscience ,Rett Syndrome ,Brain ,Humans ,General Medicine ,General Biochemistry, Genetics and Molecular Biology - Abstract
Human cerebral organoids are unique in their development of progenitor-rich zones akin to ventricular zones from which neuronal progenitors differentiate and migrate radially. Analyses of cerebral organoids thus far have been performed in sectioned tissue or in superficial layers due to their high scattering properties. Here, we demonstrate label-free three-photon imaging of whole, uncleared intact organoids (~2 mm depth) to assess early events of early human brain development. Optimizing a custom-made three-photon microscope to image intact cerebral organoids generated from Rett Syndrome patients, we show defects in the ventricular zone volumetric structure of mutant organoids compared to isogenic control organoids. Long-term imaging live organoids reveals that shorter migration distances and slower migration speeds of mutant radially migrating neurons are associated with more tortuous trajectories. Our label-free imaging system constitutes a particularly useful platform for tracking normal and abnormal development in individual organoids, as well as for screening therapeutic molecules via intact organoid imaging.
- Published
- 2022
19. What Makes Organoids Good Models of Human Neurogenesis?
- Author
-
Qian Yang, Yan Hong, Ting Zhao, Hongjun Song, and Guo-li Ming
- Subjects
General Neuroscience - Abstract
Human neurogenesis occurs mainly in embryonic, fetal, and neonatal stages and generates tremendously diverse neural cell types that constitute the human nervous system. Studies on human neurogenesis have been limited due to a lack of access to human embryonic and fetal tissues. Brain organoids derived from human pluripotent stem cells not only recapitulate major developmental processes during neurogenesis, but also exhibit human-specific features, thus providing an unprecedented opportunity to study human neurodevelopment. First, three-dimensional brain organoids resemble early human neurogenesis with diverse stem cell pools, including the presence of primate-enriched outer radial glia cells. Second, brain organoids recapitulate human neurogenesis at the cellular level, generating diverse neuronal cell types and forming stratified cortical layers. Third, brain organoids also capture gliogenesis with the presence of human-specific astrocytes. Fourth, combined with genome-editing technologies, brain organoids are promising models for investigating functions of human-specific genes at different stages of human neurogenesis. Finally, human organoids derived from patient iPSCs can recapitulate specific disease phenotypes, providing unique models for studying developmental brain disorders of genetic and environmental causes, and for mechanistic studies and drug screening. The aim of this review is to illustrate why brain organoids are good models to study various steps of human neurogenesis, with a focus on corticogenesis. We also discuss limitations of current brain organoid models and future improvements.
- Published
- 2022
20. Epitranscriptomic regulation of cortical neurogenesis via Mettl8-dependent mitochondrial tRNA m3C modification
- Author
-
Feng Zhang, Kijun Yoon, Daniel Y. Zhang, Nam-Shik Kim, Guo-li Ming, and Hongjun Song
- Subjects
Genetics ,Molecular Medicine ,Cell Biology - Published
- 2023
- Full Text
- View/download PDF
21. Structural and functional integration of human forebrain organoids with the injured adult rat visual system
- Author
-
Dennis Jgamadze, James T. Lim, Zhijian Zhang, Paul M. Harary, James Germi, Kobina Mensah-Brown, Christopher D. Adam, Ehsan Mirzakhalili, Shikha Singh, Jiahe Ben Gu, Rachel Blue, Mehek Dedhia, Marissa Fu, Fadi Jacob, Xuyu Qian, Kimberly Gagnon, Matthew Sergison, Oceane Fruchet, Imon Rahaman, Huadong Wang, Fuqiang Xu, Rui Xiao, Diego Contreras, John A. Wolf, Hongjun Song, Guo-li Ming, and Han-Chiao Isaac Chen
- Subjects
Genetics ,Molecular Medicine ,Cell Biology - Published
- 2023
- Full Text
- View/download PDF
22. Seq-ing out cell types across the isocortex and hippocampal formation
- Author
-
Guo Li Ming and Hongjun Song
- Subjects
0303 health sciences ,Cell type ,Neocortex ,Hippocampal formation ,Biology ,Hippocampus ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Neuroscience ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
The isocortex and hippocampal formation (HPF) in the mammalian brain play critical roles in perception, cognition, emotion and learning. We profiled ~1.3 million cells covering the entire adult mouse isocortex and HPF and derived a transcriptomic cell type taxonomy revealing a comprehensive repertoire of glutamatergic and GABAergic neuron types. Contrary to the traditional view of HPF as having a simpler cellular organization, we discover a complete set of glutamatergic types in HPF homologous to all major subclasses found in the six-layered isocortex, suggesting that HPF and isocortex share a common circuit organization. We also identify large-scale continuous and graded variation of cell types along isocortical depth, across isocortical sheet and in multiple dimensions in hippocampus and subiculum. Overall, our study establishes a molecular architecture of mammalian isocortex and hippocampal formation and begins to shed light on its underlying relationship with the development, evolution, connectivity and function of these brain structures.
- Published
- 2021
- Full Text
- View/download PDF
23. A nomenclature consensus for nervous system organoids and assembloids
- Author
-
Sergiu P, Pașca, Paola, Arlotta, Helen S, Bateup, J Gray, Camp, Silvia, Cappello, Fred H, Gage, Jürgen A, Knoblich, Arnold R, Kriegstein, Madeline A, Lancaster, Guo-Li, Ming, Alysson R, Muotri, In-Hyun, Park, Orly, Reiner, Hongjun, Song, Lorenz, Studer, Sally, Temple, Giuseppe, Testa, Barbara, Treutlein, and Flora M, Vaccarino
- Subjects
Organoids ,Pluripotent Stem Cells ,Consensus ,Terminology as Topic ,Humans ,Models, Biological ,Nervous System - Abstract
Self-organizing three-dimensional cellular models derived from human pluripotent stem cells or primary tissue have great potential to provide insights into how the human nervous system develops, what makes it unique and how disorders of the nervous system arise, progress and could be treated. Here, to facilitate progress and improve communication with the scientific community and the public, we clarify and provide a basic framework for the nomenclature of human multicellular models of nervous system development and disease, including organoids, assembloids and transplants.
- Published
- 2022
24. 3D spatial genome organization in the nervous system: From development and plasticity to disease
- Author
-
Yuki Fujita, Sarshan R. Pather, Guo-li Ming, and Hongjun Song
- Subjects
Genome ,General Neuroscience ,Nervous System ,Chromatin - Abstract
Chromatin is organized into multiscale three-dimensional structures, including chromosome territories, A/B compartments, topologically associating domains, and chromatin loops. This hierarchically organized genomic architecture regulates gene transcription, which, in turn, is essential for various biological processes during brain development and adult plasticity. Here, we review different aspects of spatial genome organization and their functions in regulating gene expression in the nervous system, as well as their dysregulation in brain disorders. We also highlight new technologies to probe and manipulate chromatin architecture and discuss how investigating spatial genome organization can lead to a better understanding of the nervous system and associated disorders.
- Published
- 2021
25. A single-cell transcriptome atlas of glial diversity in the human hippocampus across the postnatal lifespan
- Author
-
Yijing Su, Yi Zhou, Mariko L. Bennett, Shiying Li, Marc Carceles-Cordon, Lu Lu, Sooyoung Huh, Dennisse Jimenez-Cyrus, Benjamin C. Kennedy, Sudha K. Kessler, Angela N. Viaene, Ingo Helbig, Xiaosong Gu, Joel E. Kleinman, Thomas M. Hyde, Daniel R. Weinberger, David W. Nauen, Hongjun Song, and Guo-li Ming
- Subjects
Alzheimer Disease ,Astrocytes ,Longevity ,Genetics ,Humans ,Molecular Medicine ,Cell Biology ,Transcriptome ,Neuroglia ,Hippocampus ,Article - Abstract
The molecular diversity of glia in the human hippocampus and their temporal dynamics over the lifespan remain largely unknown. Here, we performed single-nucleus RNA sequencing to generate a transcriptome atlas of the human hippocampus across the postnatal lifespan. Detailed analyses of astrocytes, oligodendrocyte lineages, and microglia identified subpopulations with distinct molecular signatures and revealed their association with specific physiological functions, age-dependent changes in abundance, and disease relevance. We further characterized spatiotemporal heterogeneity of GFAP-enriched astrocyte subpopulations in the hippocampal formation using immunohistology. Leveraging glial subpopulation classifications as a reference map, we revealed the diversity of glia differentiated from human pluripotent stem cells and identified dysregulated genes and pathological processes in specific glial subpopulations in Alzheimer's disease (AD). Together, our study significantly extends our understanding of human glial diversity, population dynamics across the postnatal lifespan, and dysregulation in AD and provides a reference atlas for stem-cell-based glial differentiation.
- Published
- 2022
- Full Text
- View/download PDF
26. STEM-10. TEMPORAL MULTI-MODAL SINGLE-CELL ANALYSES REVEAL DYNAMIC INTERACTIONS BETWEEN GLIOBLASTOMA AND CAR-T CELLS AND IMMUNOLOGIC MODULATORS OF CANCER STEM CELL STATE
- Author
-
Daniel Zhang, Xin Wang, Yusha Sun, Donald O'Rourke, Guo-Li Ming, and Hongjun Song
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
CAR-T cell therapy is a promising new immunotherapy for a number of difficult-to-treat cancers, however, it has yet to yield broad success in glioblastoma (GBM). In particular, tumor heterogeneity presents a major therapeutic challenge, and a detailed understanding of the complex interplay between different neoplastic, non-neoplastic, and CAR-T cells is critical for developing better treatments. Using a patient-derived GBM organoid model of CAR-T cell therapy, we performed single-cell multi-omics to examine the longitudinal dynamics of the adaptive tumor response, changes in cell states, and evolution of cell-to-cell interaction networks. We find that all tumor cell types - neoplastic and non-neoplastic - respond to CAR-T cell activity, and they generate to an initially anti-tumor, but subsequently pro-tumor and immune-inhibitory microenvironment, which is accompanied by eventual CAR-T cell dysfunction and exhaustion. Unexpectedly, CAR-T cell activity also leads to attenuation of glioma stem-like states in both antigen-positive and antigen-negative neoplastic cells and reduces their proliferation via diffusible factors, of which IFNɣ is required but not sufficient. These findings are supported by analyses in patient samples from CAR-T cell therapy clinical trials, and they are consistent across both de novo and recurrent tumors with different somatic mutational landscapes. Our study unravels how the complex heterogeneity of GBM interacts with CAR-T cell therapy, and we identify previously unappreciated possibilities to affect antigen-negative neoplastic cells in ways that may be further augmented for enhanced therapeutic efficacy.
- Published
- 2022
- Full Text
- View/download PDF
27. Molecular landscapes of human hippocampal immature neurons across lifespan
- Author
-
Yi Zhou, Yijing Su, Shiying Li, Benjamin C. Kennedy, Daniel Y. Zhang, Allison M. Bond, Yusha Sun, Fadi Jacob, Lu Lu, Peng Hu, Angela N. Viaene, Ingo Helbig, Sudha K. Kessler, Timothy Lucas, Ryan D. Salinas, Xiaosong Gu, H. Isaac Chen, Hao Wu, Joel E. Kleinman, Thomas M. Hyde, David W. Nauen, Daniel R. Weinberger, Guo-li Ming, and Hongjun Song
- Subjects
Adult ,Neurons ,Aging ,Multidisciplinary ,Transcription, Genetic ,Sequence Analysis, RNA ,Gene Expression Profiling ,Neurogenesis ,Longevity ,Reproducibility of Results ,Hippocampus ,Article ,Machine Learning ,Mice ,Neural Stem Cells ,Alzheimer Disease ,Dentate Gyrus ,Animals ,Humans ,Single-Cell Analysis ,Cell Proliferation - Abstract
Immature dentate granule cells (imGCs) arising from adult hippocampal neurogenesis contribute to plasticity and unique brain functions in rodents(1,2) and they are dysregulated in multiple human neurological disorders(3–5). Little is known about molecular characteristics of adult human hippocampal imGCs and even their existence is under debate(1,6–8). Here we performed single-nucleus RNA-sequencing (snRNA-seq) aided by a validated machine learning-based analytic approach to identify imGCs and quantify their abundance in the human hippocampus during infant, child, adolescent, adult, and aging stages. We identified common molecular hallmarks of human imGCs across the lifespan and discovered age-dependent transcriptional dynamics unique to human imGCs that suggest changes in cellular functionality, niche interactions, and disease relevance, in contrast to those in mice9. We also found a decreased number of imGCs with altered gene expression in Alzheimer’s disease (AD). Finally, we demonstrated the capacity for neurogenesis in the adult human hippocampus with the presence of rare dentate granule cell fate-specific proliferating neural progenitors and with cultured surgical specimens. Together, our findings suggest the presence of a significant number of imGCs in the adult human hippocampus via low frequency de novo generation and protracted maturation, and our study reveals their molecular properties across the lifespan and in AD.
- Published
- 2021
28. Evaluating Neurodevelopmental Consequences of Perinatal Exposure to Antiretroviral Drugs: Current Challenges and New Approaches
- Author
-
Hongjun Song, Brian Temsamrit, Kimberly M. Christian, Guo Li Ming, Daniel Zhang, and Jordan G. Schnoll
- Subjects
0301 basic medicine ,Cell Culture Techniques ,Human immunodeficiency virus (HIV) ,HIV Infections ,medicine.disease_cause ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Pregnancy ,Immunology and Allergy ,Pregnancy Complications, Infectious ,Maternal-Fetal Exchange ,media_common ,Perinatal Exposure ,Brain ,Mitochondria ,Prenatal Exposure Delayed Effects ,Models, Animal ,Practice Guidelines as Topic ,Childbearing age ,Female ,Cell Division ,Adult ,Drug ,medicine.medical_specialty ,Anti-HIV Agents ,media_common.quotation_subject ,Induced Pluripotent Stem Cells ,Immunology ,Neuroscience (miscellaneous) ,Article ,03 medical and health sciences ,Fetus ,medicine ,Animals ,Humans ,Intensive care medicine ,Pharmacology ,business.industry ,Infant, Newborn ,medicine.disease ,Antiretroviral therapy ,Infectious Disease Transmission, Vertical ,Clinical trial ,Oxidative Stress ,030104 developmental biology ,Neurodevelopmental Disorders ,Synapses ,Observational study ,business ,030217 neurology & neurosurgery ,Forecasting - Abstract
As antiretroviral therapy (ART) becomes increasingly affordable and accessible to women of childbearing age across the globe, the number of children who are exposed to Human Immunodeficiency Viruses (HIV) but remain uninfected is on the rise, almost all of whom were also exposed to ART perinatally. Although ART has successfully aided in the decline of mother-to-child-transmission of HIV, the long-term effects of in utero exposure to ART on fetal and postnatal neurodevelopment remain unclear. Evaluating the safety and efficacy of therapeutic drugs for pregnant women is a challenge due to the historic limitations on their inclusion in clinical trials and the dynamic physiological states during pregnancy that can alter the pharmacokinetics of drug metabolism and fetal drug exposure. Thus, much of our data on the potential consequences of ART drugs on the developing nervous system comes from preclinical animal models and clinical observational studies. In this review, we will discuss the current state of knowledge and existing approaches to investigate whether ART affects fetal brain development, and describe novel human stem cell-based strategies that may provide additional information to better predict the impact of specific drugs on the human central nervous system. Graphical Abstract Approaches to evaluate the impact of drugs on the developing brain. Dysregulation of the developing nervous system can lead to long-lasting changes. Integration of data from animal models, clinical observations, and cell culture studies is needed to predict the safety of therapeutic antiretroviral drugs during pregnancy. New approaches include human induced pluripotent stem cell (iPSC)-based 2D and 3D models of neuronal networks and brain regions, as well as single cell profiling in response to drug exposure.
- Published
- 2019
- Full Text
- View/download PDF
29. A Patient-Derived Glioblastoma Organoid Model Maintains Intertumoral and Intratumoral Heterogeneity for Therapeutic Testing
- Author
-
Daniel Zhang, Phuong D. Nguyen, Guo Li Ming, Donald M. O'Rourke, MacLean Nasrallah, Ryan D Salinas, Jay F. Dorsey, Stefan Prokop, Hongjun Song, Fadi Jacob, Steven Brem, and Saad Sheikh
- Subjects
business.industry ,MTOR Serine-Threonine Kinases ,Cancer research ,Organoid ,Medicine ,Surgery ,Neurology (clinical) ,business ,medicine.disease ,Glioblastoma - Published
- 2019
- Full Text
- View/download PDF
30. Structural interaction between DISC1 and ATF4 underlying transcriptional and synaptic dysregulation in an iPSC model of mental disorders
- Author
-
Ziyuan Guo, Fei Ye, Kimberly M. Christian, Guo Li Ming, Chuan Yu, Wei Zheng, Wei-Kai Huang, Guomin Zhou, Hongjun Song, Francisca Rojas Ringeling, Mingjie Zhang, Yijing Su, Zhexing Wen, and Xinyuan Wang
- Subjects
0301 basic medicine ,Induced Pluripotent Stem Cells ,Mutant ,Nerve Tissue Proteins ,Activating Transcription Factor 4 ,medicine.disease_cause ,Article ,Frameshift mutation ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,DISC1 ,0302 clinical medicine ,Gene expression ,medicine ,Humans ,Molecular Biology ,Gene ,Neurons ,Mutation ,biology ,Mental Disorders ,ATF4 ,Psychiatry and Mental health ,030104 developmental biology ,biology.protein ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Psychiatric disorders are a collection of heterogeneous mental disorders arising from a contribution of genetic and environmental insults, many of which molecularly converge on transcriptional dysregulation, resulting in altered synaptic functions. The underlying mechanisms linking the genetic lesion and functional phenotypes remain largely unknown. Patient iPSC-derived neurons with a rare frame-shift DISC1 (Disrupted-in-schizophrenia 1) mutation have previously been shown to exhibit aberrant gene expression and deficits in synaptic functions. How DISC1 regulates gene expression is largely unknown. Here we show that activating transcription factor 4 (ATF4), a DISC1 binding partner, is more abundant in the nucleus of DISC1 mutant human neurons and exhibits enhanced binding to a collection of dysregulated genes. Functionally, overexpressing ATF4 in control neurons recapitulates deficits seen in DISC1 mutant neurons, whereas transcriptional and synaptic deficits are rescued in DISC1 mutant neurons with CRISPR-mediated heterozygous ATF4 knockout. By solving the high-resolution atomic structure of the DISC1-ATF4 complex, we show that mechanistically, the mutation of DISC1 disrupts normal DISC1-ATF4 interaction, and results in excessive ATF4 binding to DNA targets and deregulated gene expression. Together, our study identifies the molecular and structural basis of an DISC1-ATF4 interaction underlying transcriptional and synaptic dysregulation in an iPSC model of mental disorders.
- Published
- 2019
- Full Text
- View/download PDF
31. Mapping cis-regulatory chromatin contacts in neural cells links neuropsychiatric disorder risk variants to target genes
- Author
-
Michela Traglia, Si-Yao Lu, Li Gan, Kenneth Wu, Bingkun Li, Lenka Maliskova, Guo Li Ming, Bruce R. Conklin, Lauren A. Weiss, Fadi Jacob, Xiaoyu Yang, Kirsty Jamieson, Yin Shen, Michael Song, Tse Wai Tam, Chao Wang, Jesse R. Dixon, Jun Yao, Xingjie Ren, Luke M. Judge, Ian R. Jones, Yun Li, and Hongjun Song
- Subjects
Male ,Medical and Health Sciences ,0302 clinical medicine ,2.1 Biological and endogenous factors ,CRISPR ,Clustered Regularly Interspaced Short Palindromic Repeats ,Aetiology ,Promoter Regions, Genetic ,Induced pluripotent stem cell ,Epigenomics ,Neurons ,Gene Editing ,Regulation of gene expression ,0303 health sciences ,Genome ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Mental Disorders ,Chromosome Mapping ,Single Nucleotide ,Biological Sciences ,Chromatin ,Enhancer Elements, Genetic ,Neurological ,Functional genomics ,Human ,Biotechnology ,Genetic Markers ,Enhancer Elements ,1.1 Normal biological development and functioning ,Induced Pluripotent Stem Cells ,Computational biology ,Biology ,Polymorphism, Single Nucleotide ,Article ,Promoter Regions ,03 medical and health sciences ,Genetic ,Underpinning research ,Genetics ,Humans ,Cell Lineage ,Polymorphism ,Enhancer ,Gene ,030304 developmental biology ,Stem Cell Research - Induced Pluripotent Stem Cell ,Genome, Human ,Human Genome ,Neurosciences ,Infant ,Stem Cell Research ,Brain Disorders ,Gene Expression Regulation ,030217 neurology & neurosurgery ,Genome-Wide Association Study ,Developmental Biology - Abstract
Mutations in gene regulatory elements have been associated with a wide range of complex neuropsychiatric disorders. However, due to their cell-type specificity and difficulties in characterizing their regulatory targets, the ability to identify causal genetic variants has remained limited. To address these constraints, we perform an integrative analysis of chromatin interactions, open chromatin regions and transcriptomes using promoter capture Hi-C, assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and RNA sequencing, respectively, in four functionally distinct neural cell types: induced pluripotent stem cell (iPSC)-induced excitatory neurons and lower motor neurons, iPSC-derived hippocampal dentate gyrus-like neurons and primary astrocytes. We identify hundreds of thousands of long-range cis-interactions between promoters and distal promoter-interacting regions, enabling us to link regulatory elements to their target genes and reveal putative processes that are dysregulated in disease. Finally, we validate several promoter-interacting regions by using clustered regularly interspaced short palindromic repeats (CRISPR) techniques in human excitatory neurons, demonstrating that CDK5RAP3, STRAP and DRD2 are transcriptionally regulated by physically linked enhancers.
- Published
- 2019
- Full Text
- View/download PDF
32. Pathophysiology and Mechanisms of Zika Virus Infection in the Nervous System
- Author
-
Hongjun Song, Guo Li Ming, and Kimberly M. Christian
- Subjects
0301 basic medicine ,Nervous system ,Microcephaly ,viruses ,Communicable Diseases, Emerging ,Disease Outbreaks ,Zika virus ,Mice ,0302 clinical medicine ,Pregnancy ,Pregnancy Complications, Infectious ,Cells, Cultured ,Zika Virus Infection ,Microbiota ,General Neuroscience ,Brain ,Gene Expression Regulation, Developmental ,Flavivirus ,medicine.anatomical_structure ,Models, Animal ,Receptors, Virus ,Twin Studies as Topic ,Female ,Microglia ,Adult ,Gene Expression Regulation, Viral ,Neurogenesis ,Genetic Vectors ,Biology ,Article ,Virus ,03 medical and health sciences ,medicine ,Animals ,Humans ,Tropism ,Host Microbial Interactions ,Infant, Newborn ,Outbreak ,Viral Vaccines ,Zika Virus ,medicine.disease ,biology.organism_classification ,Pathogenicity ,Macaca mulatta ,Virology ,030104 developmental biology ,Nervous System Diseases ,Neuroscience ,030217 neurology & neurosurgery - Abstract
In 2015, public awareness of Zika virus (ZIKV) rose in response to alarming statistics of infants with microcephaly being born to women who were infected with the virus during pregnancy, triggering global concern over these potentially devastating consequences. Although we have discovered a great deal about the genome and pathogenesis of this reemergent flavivirus since this recent outbreak, we still have much more to learn, including the nature of the virus-host interactions and mechanisms that determine its tropism and pathogenicity in the nervous system, which are in turn shaped by the continual evolution of the virus. Inevitably, we will find out more about the potential long-term effects of ZIKV exposure on the nervous system from ongoing longitudinal studies. Integrating clinical and epidemiological data with a wider range of animal and human cell culture models will be critical to understanding the pathogenetic mechanisms and developing more specific antiviral compounds and vaccines.
- Published
- 2019
- Full Text
- View/download PDF
33. Flexible encoding of objects and space in single cells of the dentate gyrus
- Author
-
Seha Kim, Guo Li Ming, James J. Knierim, Puliyadi, Douglas GoodSmith, Hongjun Song, and Kimberly M. Christian
- Subjects
Neurons ,Cell type ,Dentate gyrus ,Hippocampus ,Context (language use) ,Biology ,Space (mathematics) ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Encoding (memory) ,Dentate Gyrus ,Animals ,Spatial representation ,Spatial maps ,General Agricultural and Biological Sciences ,Neuroscience - Abstract
The hippocampus is involved in the formation of memories that require associations among stimuli to construct representations of space and the items and events within that space. Neurons in the dentate gyrus (DG), an initial input region of the hippocampus, have robust spatial tuning, but it is unclear how nonspatial information may be integrated with spatial activity in this region. We recorded from the DG of 21 adult mice as they foraged for food in an environment that contained discrete objects. We found DG cells with multiple firing fields at a fixed distance and direction from objects (landmark vector cells) as well as cells that exhibited localized changes in spatial firing when objects in the environment were manipulated. By classifying recorded DG cells into putative dentate granule cells and mossy cells, we examined how the addition or displacement of objects affected the spatial firing of these DG cell types. Object-related activity was detected in a significant proportion of mossy cells. Although few granule cells with responses to object manipulations were recorded, likely due to the sparse nature of granule cell firing, there was generally no significant difference in the proportion of granule cells and mossy cells with object responses. When mice explored a second environment with the same objects, DG spatial maps completely reorganized and a different subset of cells responded to object manipulations. Together, these data reveal the capacity of DG cells to detect small changes in the environment, while preserving a stable spatial representation of the overall context.
- Published
- 2021
- Full Text
- View/download PDF
34. Microglia modulate neurodevelopment in human neuroimmune organoids
- Author
-
Mariko L. Bennett, Hongjun Song, and Guo-li Ming
- Subjects
Microglia ,Cell ,Brain ,Cell Biology ,Human brain ,Biology ,Disease pathogenesis ,Article ,Organoids ,Transcriptome ,medicine.anatomical_structure ,nervous system ,Genetics ,medicine ,Organoid ,Humans ,Molecular Medicine ,Local environment ,Stem cell ,Neuroscience - Abstract
Microglia are resident macrophages in the brain that emerge in early development and respond to local environment by altering their molecular and phenotypic states. Fundamental questions about microglia diversity and function during development remain unanswered as we lack experimental strategies to interrogate their interactions with other cell types and responses to perturbations ex vivo. We compared human microglia states across culture models, including cultured primary and pluripotent stem cell-derived microglia. We developed a ‘report card’ of gene expression signatures across these distinct models to facilitate characterization of their responses across experimental models, perturbations, and disease conditions. Xenotransplantation of human microglia into cerebral organoids allowed us to characterize key transcriptional programs of developing microglia in vitro and reveal that microglia induce transcriptional changes in neural stem cells and decreases interferon signaling response genes. Microglia additionally accelerate the emergence of synchronized oscillatory network activity in brain organoids by modulating synaptic density.
- Published
- 2021
- Full Text
- View/download PDF
35. Building the brain from scratch: Engineering region-specific brain organoids from human stem cells to study neural development and disease
- Author
-
Guo Li Ming, Fadi Jacob, Jordan G. Schnoll, and Hongjun Song
- Subjects
0303 health sciences ,Cell type ,ved/biology ,Stem Cells ,ved/biology.organism_classification_rank.species ,Brain ,Disease ,Human brain ,Biology ,Article ,Organoids ,03 medical and health sciences ,medicine.anatomical_structure ,medicine ,Organoid ,Humans ,Stem cell ,Model organism ,Induced pluripotent stem cell ,Neural development ,Neuroscience ,030304 developmental biology - Abstract
Human brain development is an intricate process that involves precisely timed coordination of cell proliferation, fate specification, neuronal differentiation, migration, and integration of diverse cell types. Understanding of these fundamental processes, however, has been largely constrained by limited access to fetal brain tissue and the inability to prospectively study neurodevelopment in humans at the molecular, cellular and system levels. Although non-human model organisms have provided important insights into mechanisms underlying brain development, these systems do not fully recapitulate many human-specific features that often relate to disease. To address these challenges, human brain organoids, self-assembled three-dimensional neural aggregates, have been engineered from human pluripotent stem cells to model the architecture and cellular diversity of the developing human brain. Recent advancements in neural induction and regional patterning using small molecules and growth factors have yielded protocols for generating brain organoids that recapitulate the structure and neuronal composition of distinct brain regions. Here, we first provide an overview of early mammalian brain development with an emphasis on molecular cues that guide region specification. We then focus on recent efforts in generating human brain organoids that model the development of specific brain regions and highlight endeavors to enhance the cellular complexity to better mimic the in vivo developing human brain. We also provide examples of how organoid models have enhanced our understanding of human neurological diseases and conclude by discussing limitations of brain organoids with our perspectives on future advancements to maximize their potential.
- Published
- 2021
36. Ontogeny of adult neural stem cells in the mammalian brain
- Author
-
Guo Li Ming, Allison M. Bond, and Hongjun Song
- Subjects
Mammals ,0303 health sciences ,Dentate gyrus ,Neurogenesis ,Hippocampus ,Subventricular zone ,Brain ,Biology ,Embryonic stem cell ,Neural stem cell ,Article ,Subgranular zone ,nervous system diseases ,03 medical and health sciences ,medicine.anatomical_structure ,nervous system ,Neural Stem Cells ,Neuroplasticity ,medicine ,Animals ,biological phenomena, cell phenomena, and immunity ,Neuroscience ,reproductive and urinary physiology ,030304 developmental biology - Abstract
Neural stem cells (NSCs) persist into adulthood in the subgranular zone (SGZ) of the dentate gyrus in the hippocampus and in the ventricular-subventricular zone (V-SVZ) of the lateral ventricles, where they generate new neurons and glia cells that contribute to neural plasticity. A better understanding of the developmental process that enables NSCs to persist beyond development will provide insight into factors that determine the size and properties of the adult NSC pool and thus the capacity for life-long neurogenesis in the adult mammalian brain. We review current knowledge regarding the developmental origins of adult NSCs and the developmental process by which embryonic NSCs transition into their adult form. We also discuss potential mechanisms that might regulate proper establishment of the adult NSC pool, and propose future directions of research that will be key to unraveling how NSCs transform to establish the adult NSC pool in the mammalian brain.
- Published
- 2021
37. High Affinity Chimeric Antigen Receptor with Cross-Reactive scFv to Clinically Relevant EGFR Oncogenic Isoforms
- Author
-
Radhika Thokala, Zev A. Binder, Yibo Yin, Logan Zhang, Jiasi Vicky Zhang, Daniel Y. Zhang, Michael C. Milone, Guo-li Ming, Hongjun Song, and Donald M. O’Rourke
- Subjects
biology ,Chemistry ,medicine.drug_class ,T cell ,Monoclonal antibody ,In vitro ,Chimeric antigen receptor ,medicine.anatomical_structure ,Tumor Escape ,Antigen ,Cell culture ,medicine ,Cancer research ,biology.protein ,Epidermal growth factor receptor ,human activities - Abstract
Tumor heterogeneity is a key reason for therapeutic failure and tumor recurrence in glioblastoma (GBM). Our chimeric antigen receptor (CAR) T cell (2173 CAR T cells) clinical trial (NCT02209376) against Epidermal growth factor receptor (EGFR) variant III (EGFRvIII) demonstrated successful trafficking of T cells across the blood brain barrier into GBM active tumor sites. However, CAR T cell infiltration was associated only with a selective loss of EGFRvIII+ tumor, demonstrating little to no effect on EGFRvIII-tumor cells. Post-CAR T treated tumor specimens showed continued presence of EGFR amplification and oncogenic EGFR extracellular domain (ECD) missense mutations, despite loss of EGFRvIII. To address tumor escape, we generated an EGFR-specific CAR by fusing monoclonal antibody (mAb) 806 to a 4-1BB co-stimulatory domain. The resulting construct was compared to 2173 CAR T cells in GBM, using in vitro and in vivo models. 806 CAR T cells specifically lysed tumor cells and secreted cytokines in response to amplified EGFR, EGFRvIII, and EGFR-ECD mutations in U87MG cells, GBM neurosphere-derived cell lines, and patient-derived GBM organoids. 806 CAR T cells did not lyse fetal brain astrocytes or primary keratinocytes to a significant degree. They also exhibited superior antitumor activity in vivo when compared to 2173 CAR T cells. The broad specificity of 806 CAR T cells to EGFR alterations gives us the potential to target multiple clones within a tumor and reduce opportunities for tumor escape via antigen loss.
- Published
- 2021
- Full Text
- View/download PDF
38. MODL-28. Patient-derived, three-dimensional organoid platform for pediatric brain tumor modeling
- Author
-
Valerie Baubet, David Beale, Santi Mariarita, Angela Viaene, Peter Madsen, Fadi Jacob, Guo-li Ming, Song Hongjun, Storm Phillip, Mateusz Koptyra, and Resnick Adam
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Brain tumors have become the leading cause of cancer-related death in children. An important hurdle to scientific and clinical progress in the field has been the limited availability of preclinical tumor models. Historically, few pediatric brain tumor cell lines have been established and these often poorly recapitulate the phenotypes of the original tumors. In recent years, the Children’s Brain Tumor Network (CBTN) has accelerated the development of patient-derived cell lines and xenografts, offering these resources to the community through open-source access. While these models are extremely valuable, their development process can be lengthy and result in clonally selected lines which presents a challenge for studying complex tumor biology. To address the need for three-dimensional tissue culture, our group in conjunction with CBTN, utilized organoid culture from fresh tissue specimens obtained directly from surgical resection of various pediatric brain tumor histologies. This resulted in the development and banking of over 30 organoid models, which included ependymoma, high-grade glioma, medulloblastoma, atypical teratoid-rhabdoid tumor, diffuse midline glioma, and low-grade glioma diagnoses. Tissue was processed within an hour post extraction and cultured with universal media composition for each diagnosis. Organoid growth was observed within 2-3 weeks of initiation and continued for up to three months before banking. Banked organoids established growth upon return to culture. Phenotypic analysis revealed organoid cell composition that represented clinical histology. Importantly, organoids returned to culture post-banking demonstrated similar cell composition to those in the original culture, indicating their utility for subsequent preclinical testing. Here we provide a simple and efficient workflow for the generation and characterization of three-dimensional tumor organoids generated from fresh surgical pediatric brain tumor tissue. The platform has the potential to accelerate investigations into tumor biology and empower a diverse array of translational studies for the pediatric brain tumor field.
- Published
- 2022
- Full Text
- View/download PDF
39. Intrinsic antiviral immunity of barrier cells revealed by an iPSC-derived blood-brain barrier cellular model
- Author
-
Yichen Cheng, Angelica Medina, Zhenlan Yao, Mausumi Basu, Janhavi P. Natekar, Jianshe Lang, Egan Sanchez, Mezindia B. Nkembo, Chongchong Xu, Xuyu Qian, Phuong T.T. Nguyen, Zhexing Wen, Hongjun Song, Guo-Li Ming, Mukesh Kumar, Margo A. Brinton, Melody M.H. Li, and Hengli Tang
- Subjects
Male ,Blood-Brain Barrier ,Induced Pluripotent Stem Cells ,Brain ,Endothelial Cells ,Humans ,Antiviral Agents ,General Biochemistry, Genetics and Molecular Biology - Abstract
Physiological blood-tissue barriers play a critical role in separating the circulation from immune-privileged sites and denying access to blood-borne viruses. The mechanism of virus restriction by these barriers is poorly understood. We utilize induced pluripotent stem cell (iPSC)-derived human brain microvascular endothelial cells (iBMECs) to study virus-blood-brain barrier (BBB) interactions. These iPSC-derived cells faithfully recapitulate a striking difference in in vivo neuroinvasion by two alphavirus isolates and are selectively permissive to neurotropic flaviviruses. A model of cocultured iBMECs and astrocytes exhibits high transendothelial electrical resistance and blocks non-neurotropic flaviviruses from getting across the barrier. We find that iBMECs constitutively express an interferon-induced gene, IFITM1, which preferentially restricts the replication of non-neurotropic flaviviruses. Barrier cells from blood-testis and blood-retinal barriers also constitutively express IFITMs that contribute to the viral resistance. Our application of a renewable human iPSC-based model for studying virus-BBB interactions reveals that intrinsic immunity at the barriers contributes to virus exclusion.
- Published
- 2022
- Full Text
- View/download PDF
40. Neurons | Biochemistry of Neurogenesis
- Author
-
Kimberly M. Christian, Hongjun Song, Guo-Li Ming, and Sarah C. Stainbrook
- Published
- 2021
- Full Text
- View/download PDF
41. Pluripotent stem cell–derived brain-region-specific organoids
- Author
-
Guo Li Ming, Phuong T.T. Nguyen, and Hongjun Song
- Subjects
Retina ,Cerebellum ,medicine.anatomical_structure ,nervous system ,Forebrain ,Functional specialization ,Thalamus ,medicine ,Organoid ,Hippocampus ,Biology ,Induced pluripotent stem cell ,Neuroscience - Abstract
Brain organoids derived from human pluripotent stem cells have emerged as invaluable tools to study human-specific features in normal brain development and diseases. Given that brain regions differ in cellular composition, molecular signature, and functional specialization, establishing brain-region-specific organoids is crucial for more precise in vitro modeling of human brains. Here, we review selected protocols for generating brain organoids with various regional identities from pluripotent stem cells, including dorsal forebrain, ventral forebrain, hippocampus, thalamus, hypothalamus, retina, midbrain, and cerebellum. These organoids possess many characteristics that resemble the fetal brain tissue of the corresponding brain regions. In addition, we highlight the utilities of using these protocols in elucidating neurodevelopmental processes, studying the impacts of genetic and environmental perturbations, and facilitating strategic development of therapies.
- Published
- 2021
- Full Text
- View/download PDF
42. Contributors
- Author
-
Rohan Bhattacharya, Dandan Cao, Wai-Yee Chan, Julia Deinsberger, Andreas M. Grabrucker, Suyu Hao, Miriel S.H. Ho, Mirabelle S.H. Ho, K.A. Kilian, Clifford L. Librach, Savitri Maddileti, Shin-Ichi Mae, Siobhan Malany, Sigita Malijauskaite, Indumathi Mariappan, Kieran McGourty, Guo-li Ming, Kenji Mishima, Kai-Kei Miu, John J.E. Mulvihill, Samira Musah, Phuong T.T. Nguyen, Arinze Emmanuel Okafor, Kenji Osafune, Maddalena Parafati, Vinay Kumar Pulimamidi, Samantha Robertson, S. Romanazzo, I. Roohani, Makoto Ryosaka, Yasuo Saijo, Hongjun Song, Masatoshi Suzuki, Junichi Tanaka, Sin-Ruow Tey, Zhangting Wang, Benedikt Weber, and Qiliang Zhou
- Published
- 2021
- Full Text
- View/download PDF
43. Partitioning RNAs by length improves transcriptome reconstruction from short-read RNA-seq data
- Author
-
Francisca Rojas Ringeling, Shounak Chakraborty, Caroline Vissers, Derek Reiman, Akshay M. Patel, Ki-Heon Lee, Ari Hong, Chan-Woo Park, Tim Reska, Julien Gagneur, Hyeshik Chang, Maria L. Spletter, Ki-Jun Yoon, Guo-li Ming, Hongjun Song, and Stefan Canzar
- Subjects
Sequence Analysis, RNA ,Gene Expression Profiling ,Biomedical Engineering ,Molecular Medicine ,High-Throughput Nucleotide Sequencing ,Protein Isoforms ,RNA ,Bioengineering ,RNA-Seq ,Transcriptome ,Applied Microbiology and Biotechnology ,Biotechnology - Abstract
The accuracy of methods for assembling transcripts from short-read RNA sequencing data is limited by the lack of long-range information. Here we introduce Ladder-seq, an approach that separates transcripts according to their lengths before sequencing and uses the additional information to improve the quantification and assembly of transcripts. Using simulated data, we show that a kallisto algorithm extended to process Ladder-seq data quantifies transcripts of complex genes with substantially higher accuracy than conventional kallisto. For reference-based assembly, a tailored scheme based on the StringTie2 algorithm reconstructs a single transcript with 30.8% higher precision than its conventional counterpart and is more than 30% more sensitive for complex genes. For de novo assembly, a similar scheme based on the Trinity algorithm correctly assembles 78% more transcripts than conventional Trinity while improving precision by 78%. In experimental data, Ladder-seq reveals 40% more genes harboring isoform switches compared to conventional RNA sequencing and unveils widespread changes in isoform usage upon m
- Published
- 2020
44. Human Pluripotent Stem Cell-Derived Neural Cells and Brain Organoids Reveal SARS-CoV-2 Neurotropism
- Author
-
Catherine Z. Chen, Juan Carlos de la Torre, Sarshan R. Pather, Samuel Zheng Hao Wong, Fadi Jacob, Wei Zheng, Manisha Pradhan, Guo Li Ming, Wei-Kai Huang, Miao Xu, Hongjun Song, Anne G. Bang, Beatrice Cubitt, Daniel Y. Zhang, Feng Zhang, and Haowen Zhou
- Subjects
Programmed cell death ,medicine.anatomical_structure ,Neurotropism ,medicine ,Organoid ,Choroid plexus ,Human brain ,Biology ,Induced pluripotent stem cell ,Pathogen ,Function (biology) ,Cell biology - Abstract
SUMMARYNeurological complications are common in patients with COVID-19. While SARS-CoV-2, the causal pathogen of COVID-19, has been detected in some patient brains, its ability to infect brain cells and impact their function are not well understood, and experimental models using human brain cells are urgently needed. Here we investigated the susceptibility of human induced pluripotent stem cell (hiPSC)-derived monolayer brain cells and region-specific brain organoids to SARS-CoV-2 infection. We found modest numbers of infected neurons and astrocytes, but greater infection of choroid plexus epithelial cells. We optimized a protocol to generate choroid plexus organoids from hiPSCs, which revealed productive SARS-CoV-2 infection that leads to increased cell death and transcriptional dysregulation indicative of an inflammatory response and cellular function deficits. Together, our results provide evidence for SARS-CoV-2 neurotropism and support use of hiPSC-derived brain organoids as a platform to investigate the cellular susceptibility, disease mechanisms, and treatment strategies for SARS-CoV-2 infection.
- Published
- 2020
- Full Text
- View/download PDF
45. Using Two- and Three-Dimensional Human iPSC Culture Systems to Model Psychiatric Disorders
- Author
-
Kimberly M, Christian, Hongjun, Song, and Guo-Li, Ming
- Subjects
Mental Disorders ,Induced Pluripotent Stem Cells ,Animals ,Humans - Abstract
Psychiatric disorders are among the most challenging human diseases to understand at a mechanistic level due to the heterogeneity of symptoms within established diagnostic categories, the general absence of focal pathology, and the genetic complexity inherent in these mostly polygenic disorders. Each of these features presents unique challenges to disease modeling for biological discovery, drug development, or improved diagnostics. In addition, live human neural tissue has been largely inaccessible to experimentation, leaving gaps in our knowledge derived from animal models that cannot fully recapitulate the features of the disease, indirect measures of brain function in human patients, and from analyses of postmortem tissue that can be confounded by comorbid conditions and medication history.
- Published
- 2020
46. Decoding neuronal composition and ontogeny of individual hypothalamic nuclei
- Author
-
Guo Li Ming, Tong Ma, Bora Lee, Samuel Zheng Hao Wong, and Hongjun Song
- Subjects
0301 basic medicine ,Hypothalamus ,Glutamic Acid ,Nerve Tissue Proteins ,Biology ,Article ,Animals, Genetically Modified ,03 medical and health sciences ,Glutamatergic ,Mice ,0302 clinical medicine ,Arcuate nucleus ,medicine ,Animals ,Cell Lineage ,gamma-Aminobutyric Acid ,Homeodomain Proteins ,Neurons ,Arc (protein) ,General Neuroscience ,Stem Cells ,Arcuate Nucleus of Hypothalamus ,Embryonic stem cell ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Ventromedial Hypothalamic Nucleus ,GABAergic ,Homeobox ,T-Box Domain Proteins ,Neuroscience ,Nucleus ,Neuroglia ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
The hypothalamus plays crucial roles in regulating endocrine, autonomic, and behavioral functions via its diverse nuclei and neuronal subtypes. The developmental mechanisms underlying ontogenetic establishment of different hypothalamic nuclei and generation of neuronal diversity remain largely unknown. Here, we show that combinatorial T-box 3 (TBX3), orthopedia homeobox (OTP), and distal-less homeobox (DLX) expression delineates all arcuate nucleus (Arc) neurons and defines four distinct subpopulations, whereas combinatorial NKX2.1/SF1 and OTP/DLX expression identifies ventromedial hypothalamus (VMH) and tuberal nucleus (TuN) neuronal subpopulations, respectively. Developmental analysis indicates that all four Arc subpopulations are mosaically and simultaneously generated from embryonic Arc progenitors, whereas glutamatergic VMH neurons and GABAergic TuN neurons are sequentially generated from common embryonic VMH progenitors. Moreover, clonal lineage-tracing analysis reveals that diverse lineages from multipotent radial glia progenitors orchestrate Arc and VMH-TuN establishment. Together, our study reveals cellular mechanisms underlying generation and organization of diverse neuronal subtypes and ontogenetic establishment of individual nuclei in the mammalian hypothalamus.
- Published
- 2020
47. Generation and biobanking of patient-derived glioblastoma organoids and their application in CAR T cell testing
- Author
-
Fadi, Jacob, Guo-Li, Ming, and Hongjun, Song
- Subjects
Cryopreservation ,Organoids ,Receptors, Chimeric Antigen ,T-Lymphocytes ,Humans ,Glioblastoma ,Coculture Techniques ,Biological Specimen Banks - Abstract
Glioblastoma tumors exhibit extensive inter- and intratumoral heterogeneity, which has contributed to the poor outcomes of numerous clinical trials and continues to complicate the development of effective therapeutic strategies. Most in vitro models do not preserve the cellular and mutational diversity of parent tumors and often require a lengthy generation time with variable efficiency. Here, we describe detailed procedures for generating glioblastoma organoids (GBOs) from surgically resected patient tumor tissue using a chemically defined medium without cell dissociation. By preserving cell-cell interactions and minimizing clonal selection, GBOs maintain the cellular heterogeneity of parent tumors. We include details of how to passage and cryopreserve GBOs for continued use, biobanking and long-term recovery. In addition, we describe procedures for investigating patient-specific responses to immunotherapies by co-culturing GBOs with chimeric antigen receptor (CAR) T cells. It takes ~2-4 weeks to generate GBOs and 5-7 d to perform CAR T cell co-culture using this protocol. Competence with human cell culture, tissue processing, immunohistology and microscopy is required for optimal results.
- Published
- 2020
48. An Integrated Systems Biology Approach Identifies the Proteasome as a Critical Host Machinery for ZIKV and DENV Replication
- Author
-
Jiang Qian, Hongjun Song, Yichen Cheng, Shu Yang, Wenwei Huang, Ruili Huang, Madhu Lal-Nag, Guang Song, Heng Zhu, Miao Xu, Ki Jun Yoon, Menghang Xia, Kimberly M. Christian, Cedric Moore, Hee Sool Rho, Guo Li Ming, Emily M. Lee, Jennifer Kouznetsova, Wei Zheng, Anton Simeonov, Samuel G. Michael, Jianbo Pan, Carleen Klumpp-Thomas, Nadia Whitt, and Hengli Tang
- Subjects
Proteasome Endopeptidase Complex ,viruses ,Systems biology ,Integrated systems ,Druggability ,Computational biology ,Dengue virus ,Biology ,medicine.disease_cause ,Virus Replication ,Biochemistry ,Protein–protein interaction ,Zika virus ,Dengue fever ,Dengue ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Molecular Biology ,Gene ,Original Research ,030304 developmental biology ,Chemical genetics screening ,Multi-omics ,0303 health sciences ,Zika Virus Infection ,Host (biology) ,Systems Biology ,Zika Virus ,Dengue Virus ,biology.organism_classification ,medicine.disease ,3. Good health ,Computational Mathematics ,Proteasome ,Viral replication ,030220 oncology & carcinogenesis ,RNAi screening ,030217 neurology & neurosurgery - Abstract
The zika virus (ZIKV) and dengue virus (DENV) flaviviruses exhibit similar replicative processes but have distinct clinical outcomes. A systematic understanding of virus-host protein-protein interaction networks can reveal cellular pathways critical to viral replication and disease pathogenesis. Here we employed three independent systems biology approaches toward this goal. First, protein array analysis of direct interactions between individual ZIKV/DENV viral proteins and 20,240 human proteins revealed multiple conserved cellular pathways and protein complexes, including proteasome complexes. Second, an RNAi screen of 10,415 druggable genes identified the host proteins required for ZIKV infection and uncovered that proteasome proteins were crucial in this process. Third, high-throughput screening of 6016 bioactive compounds for ZIKV inhibition yielded 134 effective compounds, including six proteasome inhibitors that suppress both ZIKV and DENV replication. Integrative analyses of these orthogonal datasets pinpoint proteasomes as critical host machinery for ZIKV/DENV replication. Our study provides multi-omics datasets for further studies of flavivirus-host interactions, disease pathogenesis, and new drug targets.
- Published
- 2020
- Full Text
- View/download PDF
49. m6A facilitates hippocampus-dependent learning and memory through YTHDF1
- Author
-
Yu Zhang, Hongjun Song, Piliang Hao, Hailing Shi, Yi Lan Weng, You Wu, Zongyang Lu, Guo Li Ming, Meera J. Patel, Xuliang Zhang, Zhike Lu, Tao Zhou, Bin Shen, Xiaoxi Zhuang, Yijing Su, Xiaohua Cao, Feng Zhang, Chuan He, Xingxu Huang, Yajing Liu, Jianan Li, and Jary Y. Delgado
- Subjects
0301 basic medicine ,Gene knockdown ,Multidisciplinary ,Methyltransferase complex ,Binding protein ,Hippocampus ,Long-term potentiation ,Translation (biology) ,Hippocampal formation ,Biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Protein biosynthesis ,030217 neurology & neurosurgery - Abstract
N6-methyladenosine (m6A), the most prevalent internal RNA modification on mammalian messenger RNAs, regulates the fates and functions of modified transcripts through m6A-specific binding proteins1–5. In the nervous system, m6A is abundant and modulates various neural functions6–11. Whereas m6A marks groups of mRNAs for coordinated degradation in various physiological processes12–15, the relevance of m6A for mRNA translation in vivo remains largely unknown. Here we show that, through its binding protein YTHDF1, m6A promotes protein translation of target transcripts in response to neuronal stimuli in the adult mouse hippocampus, thereby facilitating learning and memory. Mice with genetic deletion of Ythdf1 show learning and memory defects as well as impaired hippocampal synaptic transmission and long-term potentiation. Re-expression of YTHDF1 in the hippocampus of adult Ythdf1-knockout mice rescues the behavioural and synaptic defects, whereas hippocampus-specific acute knockdown of Ythdf1 or Mettl3, which encodes the catalytic component of the m6A methyltransferase complex, recapitulates the hippocampal deficiency. Transcriptome-wide mapping of YTHDF1-binding sites and m6A sites on hippocampal mRNAs identified key neuronal genes. Nascent protein labelling and tether reporter assays in hippocampal neurons showed that YTHDF1 enhances protein synthesis in a neuronal-stimulus-dependent manner. In summary, YTHDF1 facilitates translation of m6A-methylated neuronal mRNAs in response to neuronal stimulation, and this process contributes to learning and memory. Neuronal stimulation induces protein translation of m6A-methylated neuronal mRNAs facilitated by YTHDF1, and this process contributes to learning and memory.
- Published
- 2018
- Full Text
- View/download PDF
50. Applications of Human Brain Organoids to Clinical Problems
- Author
-
H. Isaac Chen, Guo Li Ming, and Hongjun Song
- Subjects
0301 basic medicine ,business.industry ,Human brain ,Biology ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Organoid ,book.journal ,Personalized medicine ,business ,Induced pluripotent stem cell ,Developmental neurobiology ,book ,Neuroscience ,030217 neurology & neurosurgery ,Developmental Biology ,Glioblastoma ,Progenitor ,Cerebral organoid - Abstract
Brain organoids are an exciting new technology with the potential to significantly change how diseases of the brain are understood and treated. These three-dimensional neural tissues are derived from the self-organization of pluripotent stem cells, and they recapitulate the developmental process of the human brain, including progenitor zones and rudimentary cortical layers. Brain organoids have been valuable in investigating different aspects of developmental neurobiology and comparative biology. Several characteristics of organoids also make them attractive as models of brain disorders. Data generated from human organoids are more generalizable to patients because of the match in species background. Personalized organoids also can be generated from patient-derived induced pluripotent stem cells. Furthermore, the three-dimensionality of brain organoids supports cellular, mechanical, and topographical cues that are lacking in planar systems. In this review, we discuss the translational potential of brain organoids, using the examples of Zika virus, autism-spectrum disorder, and glioblastoma multiforme to consider how they could contribute to disease modeling, personalized medicine, and testing of therapeutics. We then discuss areas of improvement in organoid technology that will enhance the translational potential of brain organoids, as well as the possibility of their use as substrates for repairing cerebral circuitry after injury. Developmental Dynamics 248:53-64, 2019. © 2018 Wiley Periodicals, Inc.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.