1
|
Liu MH, Xu YG, Bai XN, Lin JH, Xiang ZQ, Wang T, Xu L, Chen G. Efficient Dlx2-mediated astrocyte-to-neuron conversion and inhibition of neuroinflammation by NeuroD1. Dev Neurobiol 2024; 84:274-290. [PMID: 39034481 DOI: 10.1002/dneu.22951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 06/05/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024]
Abstract
In vivo astrocyte-to-neuron (AtN) conversion induced by overexpression of neural transcriptional factors has great potential for neural regeneration and repair. Here, we demonstrate that a single neural transcriptional factor, Dlx2, converts mouse striatal astrocytes into neurons in a dose-dependent manner. Lineage-tracing studies in Aldh1l1-CreERT2 mice confirm that Dlx2 can convert striatal astrocytes into DARPP32+ and Ctip2+ medium spiny neurons (MSNs). Time-course studies reveal a gradual conversion from astrocytes to neurons in 1 month, with a distinct intermediate state in between astrocytes and neurons. Interestingly, when Dlx2-infected astrocytes start to lose astrocytic markers, the other local astrocytes proliferate to maintain astrocytic levels in the converted areas. Unexpectedly, although Dlx2 efficiently reprograms astrocytes into neurons in the gray matter striatum, it also induces partial reprogramming of astrocytes in the white matter corpus callosum. Such partial reprogramming of white matter astrocytes is associated with neuroinflammation, which can be suppressed by the addition of NeuroD1. Our results highlight the importance of investigating AtN conversion in both the gray matter and white matter to thoroughly evaluate therapeutic potentials. This study also unveils the critical role of anti-inflammation by NeuroD1 during AtN conversion.
Collapse
Affiliation(s)
- Min-Hui Liu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
- VIB Center for Brain and Disease, KU Leuven, Herestraat 49, Leuven, Belgium
| | - Yu-Ge Xu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Xiao-Ni Bai
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Jian-Hua Lin
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Zong-Qin Xiang
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
- Department of Neurosurgery, the First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Tao Wang
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Liang Xu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Gong Chen
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| |
Collapse
|
2
|
Ju Y, Li S, Kong X, Zhao Q. EBF1 is a potential biomarker for predicting progression from mild cognitive impairment to Alzheimer's disease: an in silico study. Front Aging Neurosci 2024; 16:1397696. [PMID: 39347016 PMCID: PMC11427346 DOI: 10.3389/fnagi.2024.1397696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/19/2024] [Indexed: 10/01/2024] Open
Abstract
Introduction The prediction of progression from mild cognitive impairment (MCI) to Alzheimer's disease (AD) is an important clinical challenge. This study aimed to identify the independent risk factors and develop a nomogram model that can predict progression from MCI to AD. Methods Data of 141 patients with MCI were obtained from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. We set a follow-up time of 72 months and defined patients as stable MCI (sMCI) or progressive MCI (pMCI) according to whether or not the progression of MCI to AD occurred. We identified and screened independent risk factors by utilizing weighted gene co-expression network analysis (WGCNA), where we obtained 14,893 genes after data preprocessing and selected the soft threshold β = 7 at an R 2 of 0.85 to achieve a scale-free network. A total of 14 modules were discovered, with the midnightblue module having a strong association with the prognosis of MCI. Using machine learning strategies, which included the least absolute selection and shrinkage operator and support vector machine-recursive feature elimination; and the Cox proportional-hazards model, which included univariate and multivariable analyses, we identified and screened independent risk factors. Subsequently, we developed a nomogram model for predicting the progression from MCI to AD. The performance of our nomogram was evaluated by the C-index, calibration curve, and decision curve analysis (DCA). Bioinformatics analysis and immune infiltration analysis were conducted to clarify the function of early B cell factor 1 (EBF1). Results First, the results showed that 40 differentially expressed genes (DEGs) related to the prognosis of MCI were generated by weighted gene co-expression network analysis. Second, five hub variables were obtained through the abovementioned machine learning strategies. Third, a low Montreal Cognitive Assessment (MoCA) score [hazard ratio (HR): 4.258, 95% confidence interval (CI): 1.994-9.091] and low EBF1 expression (hazard ratio: 3.454, 95% confidence interval: 1.813-6.579) were identified as the independent risk factors through the Cox proportional-hazards regression analysis. Finally, we developed a nomogram model including the MoCA score, EBF1, and potential confounders (age and gender). By evaluating our nomogram model and validating it in both internal and external validation sets, we demonstrated that our nomogram model exhibits excellent predictive performance. Through the Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes Genomes (KEGG) functional enrichment analysis, and immune infiltration analysis, we found that the role of EBF1 in MCI was closely related to B cells. Conclusion EBF1, as a B cell-specific transcription factor, may be a key target for predicting progression from MCI to AD. Our nomogram model was able to provide personalized risk factors for the progression from MCI to AD after evaluation and validation.
Collapse
Affiliation(s)
- Yanxiu Ju
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Memory and Cognitive Impairment Disease of Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Songtao Li
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Memory and Cognitive Impairment Disease of Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiangyi Kong
- Key Laboratory of Lymphatic Surgery of Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qing Zhao
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Memory and Cognitive Impairment Disease of Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Galimberti M, Nucera MR, Bocchi VD, Conforti P, Vezzoli E, Cereda M, Maffezzini C, Iennaco R, Scolz A, Falqui A, Cordiglieri C, Cremona M, Espuny-Camacho I, Faedo A, Felsenfeld DP, Vogt TF, Ranzani V, Zuccato C, Besusso D, Cattaneo E. Huntington's disease cellular phenotypes are rescued non-cell autonomously by healthy cells in mosaic telencephalic organoids. Nat Commun 2024; 15:6534. [PMID: 39095390 PMCID: PMC11297310 DOI: 10.1038/s41467-024-50877-x] [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: 05/10/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
Huntington's disease (HD) causes selective degeneration of striatal and cortical neurons, resulting in cell mosaicism of coexisting still functional and dysfunctional cells. The impact of non-cell autonomous mechanisms between these cellular states is poorly understood. Here we generated telencephalic organoids with healthy or HD cells, grown separately or as mosaics of the two genotypes. Single-cell RNA sequencing revealed neurodevelopmental abnormalities in the ventral fate acquisition of HD organoids, confirmed by cytoarchitectural and transcriptional defects leading to fewer GABAergic neurons, while dorsal populations showed milder phenotypes mainly in maturation trajectory. Healthy cells in mosaic organoids restored HD cell identity, trajectories, synaptic density, and communication pathways upon cell-cell contact, while showing no significant alterations when grown with HD cells. These findings highlight cell-type-specific alterations in HD and beneficial non-cell autonomous effects of healthy cells, emphasizing the therapeutic potential of modulating cell-cell communication in disease progression and treatment.
Collapse
Affiliation(s)
- Maura Galimberti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Maria R Nucera
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Stem Cell Biology Department; Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Vittoria D Bocchi
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Paola Conforti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Elena Vezzoli
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- ALEMBIC Advanced Light and Electron Microscopy BioImaging Center, San Raffaele Scientific Institute, DIBIT 1, Via Olgettina 58, 20132, Milan, Italy
| | - Matteo Cereda
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Camilla Maffezzini
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Raffaele Iennaco
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andrea Scolz
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andrea Falqui
- Department of Physics "Aldo Pontremoli", University of Milan, Via Celoria 16, 20133, Milan, Italy
| | - Chiara Cordiglieri
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Martina Cremona
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Swiss Stem Cell Foundation, Via Petrini 2, 6900, Lugano, Switzerland
| | - Ira Espuny-Camacho
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- GIGA-Neuroscience, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, C.H.U. Sart Tilman, 4000, Liège, Belgium
| | - Andrea Faedo
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Axxam, OpenZone, Via Meucci 3, 20091, Bresso, Milan, Italy
| | | | | | - Valeria Ranzani
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Chiara Zuccato
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Dario Besusso
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Elena Cattaneo
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy.
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy.
| |
Collapse
|
4
|
Foster M, Dwibhashyam S, Patel D, Gupta K, Matz OC, Billings BK, Bitterman K, Bertelson M, Tang CY, Mars RB, Raghanti MA, Hof PR, Sherwood CC, Manger PR, Spocter MA. Comparative anatomy of the caudate nucleus in canids and felids: Associations with brain size, curvature, cross-sectional properties, and behavioral ecology. J Comp Neurol 2024; 532:e25618. [PMID: 38686628 DOI: 10.1002/cne.25618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 03/30/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
The evolutionary history of canids and felids is marked by a deep time separation that has uniquely shaped their behavior and phenotype toward refined predatory abilities. The caudate nucleus is a subcortical brain structure associated with both motor control and cognitive, emotional, and executive functions. We used a combination of three-dimensional imaging, allometric scaling, and structural analyses to compare the size and shape characteristics of the caudate nucleus. The sample consisted of MRI scan data obtained from six canid species (Canis lupus lupus, Canis latrans, Chrysocyon brachyurus, Lycaon pictus, Vulpes vulpes, Vulpes zerda), two canid subspecies (Canis lupus familiaris, Canis lupus dingo), as well as three felids (Panthera tigris, Panthera uncia, Felis silvestris catus). Results revealed marked conservation in the scaling and shape attributes of the caudate nucleus across species, with only slight deviations. We hypothesize that observed differences in caudate nucleus size and structure for the domestic canids are reflective of enhanced cognitive and emotional pathways that possibly emerged during domestication.
Collapse
Affiliation(s)
- Michael Foster
- Department of Anatomy, Des Moines University, West Des Moines, Iowa, USA
| | - Sai Dwibhashyam
- Department of Anatomy, Des Moines University, West Des Moines, Iowa, USA
| | - Devan Patel
- Department of Anatomy, Des Moines University, West Des Moines, Iowa, USA
| | - Kanika Gupta
- Department of Anatomy, Des Moines University, West Des Moines, Iowa, USA
| | - Olivia C Matz
- Department of Anatomy, Des Moines University, West Des Moines, Iowa, USA
| | - Brendon K Billings
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Kathleen Bitterman
- Department of Anatomy, Des Moines University, West Des Moines, Iowa, USA
| | - Mads Bertelson
- Center for Zoo and Wild Animal Health, Copenhagen Zoo, Frederiksberg, Denmark
| | - Cheuk Y Tang
- Departments of Radiology and Psychiatry, BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rogier B Mars
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- New York Consortium in Evolutionary Primatology, New York, New York, USA
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
| | - Paul R Manger
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Muhammad A Spocter
- Department of Anatomy, Des Moines University, West Des Moines, Iowa, USA
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Republic of South Africa
- College of Veterinary Medicine, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
5
|
Xue X, Kim YS, Ponce-Arias AI, O'Laughlin R, Yan RZ, Kobayashi N, Tshuva RY, Tsai YH, Sun S, Zheng Y, Liu Y, Wong FCK, Surani A, Spence JR, Song H, Ming GL, Reiner O, Fu J. A patterned human neural tube model using microfluidic gradients. Nature 2024; 628:391-399. [PMID: 38408487 PMCID: PMC11006583 DOI: 10.1038/s41586-024-07204-7] [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: 01/26/2022] [Accepted: 02/16/2024] [Indexed: 02/28/2024]
Abstract
The human nervous system is a highly complex but organized organ. The foundation of its complexity and organization is laid down during regional patterning of the neural tube, the embryonic precursor to the human nervous system. Historically, studies of neural tube patterning have relied on animal models to uncover underlying principles. Recently, models of neurodevelopment based on human pluripotent stem cells, including neural organoids1-5 and bioengineered neural tube development models6-10, have emerged. However, such models fail to recapitulate neural patterning along both rostral-caudal and dorsal-ventral axes in a three-dimensional tubular geometry, a hallmark of neural tube development. Here we report a human pluripotent stem cell-based, microfluidic neural tube-like structure, the development of which recapitulates several crucial aspects of neural patterning in brain and spinal cord regions and along rostral-caudal and dorsal-ventral axes. This structure was utilized for studying neuronal lineage development, which revealed pre-patterning of axial identities of neural crest progenitors and functional roles of neuromesodermal progenitors and the caudal gene CDX2 in spinal cord and trunk neural crest development. We further developed dorsal-ventral patterned microfluidic forebrain-like structures with spatially segregated dorsal and ventral regions and layered apicobasal cellular organizations that mimic development of the human forebrain pallium and subpallium, respectively. Together, these microfluidics-based neurodevelopment models provide three-dimensional lumenal tissue architectures with in vivo-like spatiotemporal cell differentiation and organization, which will facilitate the study of human neurodevelopment and disease.
Collapse
Affiliation(s)
- Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yung Su Kim
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Alfredo-Isaac Ponce-Arias
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Richard O'Laughlin
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Robin Zhexuan Yan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Norio Kobayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Rami Yair Tshuva
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Yu-Hwai Tsai
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shiyu Sun
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yue Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Frederick C K Wong
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Azim Surani
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jason R Spence
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - 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
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
6
|
Reumann D, Krauditsch C, Novatchkova M, Sozzi E, Wong SN, Zabolocki M, Priouret M, Doleschall B, Ritzau-Reid KI, Piber M, Morassut I, Fieseler C, Fiorenzano A, Stevens MM, Zimmer M, Bardy C, Parmar M, Knoblich JA. In vitro modeling of the human dopaminergic system using spatially arranged ventral midbrain-striatum-cortex assembloids. Nat Methods 2023; 20:2034-2047. [PMID: 38052989 PMCID: PMC10703680 DOI: 10.1038/s41592-023-02080-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 10/10/2023] [Indexed: 12/07/2023]
Abstract
Ventral midbrain dopaminergic neurons project to the striatum as well as the cortex and are involved in movement control and reward-related cognition. In Parkinson's disease, nigrostriatal midbrain dopaminergic neurons degenerate and cause typical Parkinson's disease motor-related impairments, while the dysfunction of mesocorticolimbic midbrain dopaminergic neurons is implicated in addiction and neuropsychiatric disorders. Study of the development and selective neurodegeneration of the human dopaminergic system, however, has been limited due to the lack of an appropriate model and access to human material. Here, we have developed a human in vitro model that recapitulates key aspects of dopaminergic innervation of the striatum and cortex. These spatially arranged ventral midbrain-striatum-cortical organoids (MISCOs) can be used to study dopaminergic neuron maturation, innervation and function with implications for cell therapy and addiction research. We detail protocols for growing ventral midbrain, striatal and cortical organoids and describe how they fuse in a linear manner when placed in custom embedding molds. We report the formation of functional long-range dopaminergic connections to striatal and cortical tissues in MISCOs, and show that injected, ventral midbrain-patterned progenitors can mature and innervate the tissue. Using these assembloids, we examine dopaminergic circuit perturbations and show that chronic cocaine treatment causes long-lasting morphological, functional and transcriptional changes that persist upon drug withdrawal. Thus, our method opens new avenues to investigate human dopaminergic cell transplantation and circuitry reconstruction as well as the effect of drugs on the human dopaminergic system.
Collapse
Affiliation(s)
- Daniel Reumann
- Institute of Molecular Biotechnology (IMBA) of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
- Vienna BioCenter, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Christian Krauditsch
- Institute of Molecular Biotechnology (IMBA) of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - Maria Novatchkova
- Institute of Molecular Biotechnology (IMBA) of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - Edoardo Sozzi
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sakurako Nagumo Wong
- Institute of Molecular Biotechnology (IMBA) of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
- Vienna BioCenter, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Michael Zabolocki
- Laboratory for Human Neurophysiology and Genetics, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Marthe Priouret
- Institute of Molecular Biotechnology (IMBA) of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - Balint Doleschall
- Institute of Molecular Biotechnology (IMBA) of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
- Vienna BioCenter, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Kaja I Ritzau-Reid
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London, London, UK
| | - Marielle Piber
- Institute of Molecular Biotechnology (IMBA) of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
- Zebrafish Neurogenetics Unit, Institut Pasteur, Paris, France
| | - Ilaria Morassut
- Institute of Molecular Biotechnology (IMBA) of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Charles Fieseler
- Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria
| | - Alessandro Fiorenzano
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics 'Adriano Buzzati Traverso' (IGB), CNR, Naples, Italy
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London, London, UK
| | - Manuel Zimmer
- Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria
| | - Cedric Bardy
- Laboratory for Human Neurophysiology and Genetics, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Malin Parmar
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Jürgen A Knoblich
- Institute of Molecular Biotechnology (IMBA) of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria.
- Department of Neurology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Dell'Amico C, Angulo Salavarria MM, Takeo Y, Saotome I, Dell'Anno MT, Galimberti M, Pellegrino E, Cattaneo E, Louvi A, Onorati M. Microcephaly-associated protein WDR62 shuttles from the Golgi apparatus to the spindle poles in human neural progenitors. eLife 2023; 12:e81716. [PMID: 37272619 PMCID: PMC10241521 DOI: 10.7554/elife.81716] [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: 07/08/2022] [Accepted: 04/17/2023] [Indexed: 06/06/2023] Open
Abstract
WDR62 is a spindle pole-associated scaffold protein with pleiotropic functions. Recessive mutations in WDR62 cause structural brain abnormalities and account for the second most common cause of autosomal recessive primary microcephaly (MCPH), indicating WDR62 as a critical hub for human brain development. Here, we investigated WDR62 function in corticogenesis through the analysis of a C-terminal truncating mutation (D955AfsX112). Using induced Pluripotent Stem Cells (iPSCs) obtained from a patient and his unaffected parent, as well as isogenic corrected lines, we generated 2D and 3D models of human neurodevelopment, including neuroepithelial stem cells, cerebro-cortical progenitors, terminally differentiated neurons, and cerebral organoids. We report that WDR62 localizes to the Golgi apparatus during interphase in cultured cells and human fetal brain tissue, and translocates to the mitotic spindle poles in a microtubule-dependent manner. Moreover, we demonstrate that WDR62 dysfunction impairs mitotic progression and results in alterations of the neurogenic trajectories of iPSC neuroderivatives. In summary, impairment of WDR62 localization and function results in severe neurodevelopmental abnormalities, thus delineating new mechanisms in the etiology of MCPH.
Collapse
Affiliation(s)
- Claudia Dell'Amico
- Department of Biology, Unit of Cell and Developmental Biology, University of PisaPisaItaly
| | | | - Yutaka Takeo
- Departments of Neurosurgery and Neuroscience, Yale School of MedicineNew HavenUnited States
| | - Ichiko Saotome
- Departments of Neurosurgery and Neuroscience, Yale School of MedicineNew HavenUnited States
| | | | - Maura Galimberti
- Dipartimento di Bioscienze, Università degli Studi di MilanoMilanItaly
- INGM, Istituto Nazionale Genetica MolecolareMilanItaly
| | - Enrica Pellegrino
- Department of Biology, Unit of Cell and Developmental Biology, University of PisaPisaItaly
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Elena Cattaneo
- Dipartimento di Bioscienze, Università degli Studi di MilanoMilanItaly
- INGM, Istituto Nazionale Genetica MolecolareMilanItaly
| | - Angeliki Louvi
- Departments of Neurosurgery and Neuroscience, Yale School of MedicineNew HavenUnited States
| | - Marco Onorati
- Department of Biology, Unit of Cell and Developmental Biology, University of PisaPisaItaly
| |
Collapse
|
8
|
Brocchetti S, Conforti P. Differentiation of hPSCs to Study PRC2 Role in Cell-Fate Specification and Neurodevelopment. Methods Mol Biol 2023; 2655:211-220. [PMID: 37212999 DOI: 10.1007/978-1-0716-3143-0_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Several studies highlighted the importance of the polycomb repressive complex 2 (PRC2) already at the beginning of development. Although the crucial function of PRC2 in regulating lineage commitment and cell-fate specification has been well-established, the in vitro study of the exact mechanisms for which H3K27me3 is indispensable for proper differentiation is still challenging. In this chapter, we report a well-established and reproducible differentiation protocol to generate striatal medium spiny neurons as a tool to explore PRC2 role in brain development.
Collapse
Affiliation(s)
| | - Paola Conforti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan and INGM, Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy.
| |
Collapse
|
9
|
Kostović I, Džaja D, Raguž M, Kopić J, Blažević A, Krsnik Ž. Transient compartmentalization and accelerated volume growth coincide with the expected development of cortical afferents in the human neostriatum. Cereb Cortex 2022; 33:434-457. [PMID: 35244150 DOI: 10.1093/cercor/bhac076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 01/17/2023] Open
Abstract
The neostriatum plays a central role in cortico-subcortical circuitry underlying goal-directed behavior. The adult mammalian neostriatum shows chemical and cytoarchitectonic compartmentalization in line with the connectivity. However, it is poorly understood how and when fetal compartmentalization (AChE-rich islands, nonreactive matrix) switches to adult (AChE-poor striosomes, reactive matrix) and how this relates to the ingrowth of corticostriatal afferents. Here, we analyze neostriatal compartments on postmortem human brains from 9 postconceptional week (PCW) to 18 postnatal months (PM), using Nissl staining, histochemical techniques (AChE, PAS-Alcian), immunohistochemistry, stereology, and comparing data with volume-growth of in vivo and in vitro MRI. We find that compartmentalization (C) follows a two-compartment (2-C) pattern around 10PCW and is transformed into a midgestational labyrinth-like 3-C pattern (patches, AChE-nonreactive perimeters, matrix), peaking between 22 and 28PCW during accelerated volume-growth. Finally, compartmentalization resolves perinatally, by the decrease in transient "AChE-clumping," disappearance of AChE-nonreactive, ECM-rich perimeters, and an increase in matrix reactivity. The initial "mature" pattern appears around 9 PM. Therefore, transient, a 3-C pattern and accelerated neostriatal growth coincide with the expected timing of the nonhomogeneous distribution of corticostriatal afferents. The decrease in growth-related AChE activity and transfiguration of corticostriatal terminals are putative mechanisms underlying fetal compartments reorganization. Our findings serve as normative for studying neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Domagoj Džaja
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia.,Department of Anatomy and Clinical Anatomy, School of Medicine University of Zagreb, 10000 Zagreb, Croatia
| | - Marina Raguž
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia.,Department of Neurosurgery, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Janja Kopić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Andrea Blažević
- Department of Anatomy and Clinical Anatomy, School of Medicine University of Zagreb, 10000 Zagreb, Croatia
| | - Željka Krsnik
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
10
|
Conforti P, Bocchi VD, Campus I, Scaramuzza L, Galimberti M, Lischetti T, Talpo F, Pedrazzoli M, Murgia A, Ferrari I, Cordiglieri C, Fasciani A, Arenas E, Felsenfeld D, Biella G, Besusso D, Cattaneo E. In vitro-derived medium spiny neurons recapitulate human striatal development and complexity at single-cell resolution. CELL REPORTS METHODS 2022; 2:100367. [PMID: 36590694 PMCID: PMC9795363 DOI: 10.1016/j.crmeth.2022.100367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/06/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
Stem cell engineering of striatal medium spiny neurons (MSNs) is a promising strategy to understand diseases affecting the striatum and for cell-replacement therapies in different neurological diseases. Protocols to generate cells from human pluripotent stem cells (PSCs) are scarce and how well they recapitulate the endogenous fetal cells remains poorly understood. We have developed a protocol that modulates cell seeding density and exposure to specific morphogens that generates authentic and functional D1- and D2-MSNs with a high degree of reproducibility in 25 days of differentiation. Single-cell RNA sequencing (scRNA-seq) shows that our cells can mimic the cell-fate acquisition steps observed in vivo in terms of cell type composition, gene expression, and signaling pathways. Finally, by modulating the midkine pathway we show that we can increase the yield of MSNs. We expect that this protocol will help decode pathogenesis factors in striatal diseases and eventually facilitate cell-replacement therapies for Huntington's disease (HD).
Collapse
Affiliation(s)
- Paola Conforti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Vittoria Dickinson Bocchi
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Ilaria Campus
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Linda Scaramuzza
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Maura Galimberti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Tiziana Lischetti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Francesca Talpo
- Department of Biology and Biotechnologies, University of Pavia, Via Adolfo Ferrata, 9, 27100 Pavia, Italy
| | - Matteo Pedrazzoli
- Department of Biology and Biotechnologies, University of Pavia, Via Adolfo Ferrata, 9, 27100 Pavia, Italy
| | - Alessio Murgia
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Ivan Ferrari
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Chiara Cordiglieri
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Alessandra Fasciani
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Ernest Arenas
- Laboratory for Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, Solnavägen 9, 17177 Stockholm, Sweden
| | | | - Gerardo Biella
- Department of Biology and Biotechnologies, University of Pavia, Via Adolfo Ferrata, 9, 27100 Pavia, Italy
| | - Dario Besusso
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Elena Cattaneo
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| |
Collapse
|
11
|
Chen X, Saiyin H, Liu Y, Wang Y, Li X, Ji R, Ma L. Human striatal organoids derived from pluripotent stem cells recapitulate striatal development and compartments. PLoS Biol 2022; 20:e3001868. [PMID: 36395338 PMCID: PMC9714809 DOI: 10.1371/journal.pbio.3001868] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 12/01/2022] [Accepted: 10/05/2022] [Indexed: 11/18/2022] Open
Abstract
The striatum links neuronal circuits in the human brain, and its malfunction causes neuronal disorders such as Huntington's disease (HD). A human striatum model that recapitulates fetal striatal development is vital to decoding the pathogenesis of striatum-related neurological disorders and developing therapeutic strategies. Here, we developed a method to construct human striatal organoids (hStrOs) from human pluripotent stem cells (hPSCs), including hStrOs-derived assembloids. Our hStrOs partially replicated the fetal striatum and formed striosome and matrix-like compartments in vitro. Single-cell RNA sequencing revealed distinct striatal lineages in hStrOs, diverging from dorsal forebrain fate. Using hStrOs-derived assembloids, we replicated the striatal targeting projections from different brain parts. Furthermore, hStrOs can serve as hosts for striatal neuronal allografts to test allograft neuronal survival and functional integration. Our hStrOs are suitable for studying striatal development and related disorders, characterizing the neural circuitry between different brain regions, and testing therapeutic strategies.
Collapse
Affiliation(s)
- Xinyu Chen
- Department of Anatomy and Histology & Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, P.R. China
| | - Hexige Saiyin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Yang Liu
- Department of Anatomy and Histology & Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, P.R. China
| | - Yuqi Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Xuan Li
- The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, P.R. China
| | - Rong Ji
- Department of Neurology, Huadong Hospital, Fudan University, Shanghai, P.R. China
| | - Lixiang Ma
- Department of Anatomy and Histology & Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, P.R. China
- * E-mail:
| |
Collapse
|
12
|
Tian M, Xu F, Xia Q, Tang Y, Zhang Z, Lin X, Meng H, Feng L, Liu S. Morphological development of the human fetal striatum during the second trimester. Cereb Cortex 2022; 32:5072-5082. [PMID: 35078212 DOI: 10.1093/cercor/bhab532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/24/2021] [Accepted: 12/25/2021] [Indexed: 12/27/2022] Open
Abstract
The morphological development of the fetal striatum during the second trimester has remained poorly described. We manually segmented the striatum using 7.0-T MR images of the fetal specimens ranging from 14 to 22 gestational weeks. The global development of the striatum was evaluated by volume measurement. The absolute volume (Vabs) of the caudate nucleus (CN) increased linearly with gestational age, while the relative volume (Vrel) showed a quadratic growth. Both Vabs and Vrel of putamen increased linearly. Through shape analysis, the changes of local structure in developing striatum were specifically demonstrated. Except for the CN tail, the lateral and medial parts of the CN grew faster than the middle regions, with a clear rostral-caudal growth gradient as well as a distinct "outside-in" growth gradient. For putamen, the dorsal and ventral regions grew obviously faster than the other regions, with a dorsal-ventral bidirectional developmental pattern. The right CN was larger than the left, whereas there was no significant hemispheric asymmetry in the putamen. By establishing the developmental trajectories, spatial heterochrony, and hemispheric dimorphism of human fetal striatum, these data bring new insight into the fetal striatum development and provide detailed anatomical references for future striatal studies.
Collapse
Affiliation(s)
- Mimi Tian
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Feifei Xu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Qing Xia
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Yuchun Tang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Zhonghe Zhang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Department of Medical Imaging, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Xiangtao Lin
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Department of Medical Imaging, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Haiwei Meng
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Lei Feng
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Shuwei Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
13
|
Brain Regional Identity and Cell Type Specificity Landscape of Human Cortical Organoid Models. Int J Mol Sci 2022; 23:ijms232113159. [PMID: 36361956 PMCID: PMC9654943 DOI: 10.3390/ijms232113159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
Abstract
In vitro models of corticogenesis from pluripotent stem cells (PSCs) have greatly improved our understanding of human brain development and disease. Among these, 3D cortical organoid systems are able to recapitulate some aspects of in vivo cytoarchitecture of the developing cortex. Here, we tested three cortical organoid protocols for brain regional identity, cell type specificity and neuronal maturation. Overall, all protocols gave rise to organoids that displayed a time-dependent expression of neuronal maturation genes such as those involved in the establishment of synapses and neuronal function. Comparatively, guided differentiation methods without WNT activation generated the highest degree of cortical regional identity, whereas default conditions produced the broadest range of cell types such as neurons, astrocytes and hematopoietic-lineage-derived microglia cells. These results suggest that cortical organoid models produce diverse outcomes of brain regional identity and cell type specificity and emphasize the importance of selecting the correct model for the right application.
Collapse
|
14
|
Song S, Creus Muncunill J, Galicia Aguirre C, Tshilenge KT, Hamilton BW, Gerencser AA, Benlhabib H, Cirnaru MD, Leid M, Mooney SD, Ellerby LM, Ehrlich ME. Postnatal Conditional Deletion of Bcl11b in Striatal Projection Neurons Mimics the Transcriptional Signature of Huntington's Disease. Biomedicines 2022; 10:2377. [PMID: 36289639 PMCID: PMC9598565 DOI: 10.3390/biomedicines10102377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
The dysregulation of striatal gene expression and function is linked to multiple diseases, including Huntington's disease (HD), Parkinson's disease, X-linked dystonia-parkinsonism (XDP), addiction, autism, and schizophrenia. Striatal medium spiny neurons (MSNs) make up 90% of the neurons in the striatum and are critical to motor control. The transcription factor, Bcl11b (also known as Ctip2), is required for striatal development, but the function of Bcl11b in adult MSNs in vivo has not been investigated. We conditionally deleted Bcl11b specifically in postnatal MSNs and performed a transcriptomic and behavioral analysis on these mice. Multiple enrichment analyses showed that the D9-Cre-Bcl11btm1.1Leid transcriptional profile was similar to the HD gene expression in mouse and human data sets. A Gene Ontology enrichment analysis linked D9-Cre-Bcl11btm1.1Leid to calcium, synapse organization, specifically including the dopaminergic synapse, protein dephosphorylation, and HDAC-signaling, commonly dysregulated pathways in HD. D9-Cre-Bcl11btm1.1Leid mice had decreased DARPP-32/Ppp1r1b in MSNs and behavioral deficits, demonstrating the dysregulation of a subtype of the dopamine D2 receptor expressing MSNs. Finally, in human HD isogenic MSNs, the mislocalization of BCL11B into nuclear aggregates points to a mechanism for BCL11B loss of function in HD. Our results suggest that BCL11B is important for the function and maintenance of mature MSNs and Bcl11b loss of function drives, in part, the transcriptomic and functional changes in HD.
Collapse
Affiliation(s)
- Sicheng Song
- Department of Biomedical Informatics and Medical Education, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Jordi Creus Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carlos Galicia Aguirre
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90893, USA
| | | | - B. Wade Hamilton
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Houda Benlhabib
- Department of Biomedical Informatics and Medical Education, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Maria-Daniela Cirnaru
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark Leid
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Sean D. Mooney
- Department of Biomedical Informatics and Medical Education, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Lisa M. Ellerby
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90893, USA
| | - Michelle E. Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
15
|
Della Posta D, Branca JJV, Guarnieri G, Veltro C, Pacini A, Paternostro F. Modularity of the Human Musculoskeletal System: The Correlation between Functional Structures by Computer Tools Analysis. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081186. [PMID: 36013365 PMCID: PMC9410413 DOI: 10.3390/life12081186] [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: 06/07/2022] [Revised: 07/17/2022] [Accepted: 07/30/2022] [Indexed: 12/02/2022]
Abstract
Introduction: For many years, anatomical studies have been conducted with a shattered view of the body. Although the study of the different apparatuses provides a systemic view of the human body, the reconstruction of the complex network of anatomical structures is crucial for the understanding of structural and functional integration. Aim: We used network analysis to investigate the connection between the whole-body osteo-myofascial structures of the human musculoskeletal system. Materials and Methods: The musculoskeletal network was performed using the aNETomy® anatomical network with the implementation of the open-source software Cytoscape for data entry. Results: The initial graph was applied with a network consisting of 2298 body parts (nodes) and 7294 links, representing the musculoskeletal system. Considering the same weighted and unweighted osteo-myofascial network, a different distribution was obtained, suggesting both a topological organization and functional behavior of the network structure. Conclusions: Overall, we provide a deeply detailed anatomical network map of the whole-body musculoskeletal system that can be a useful tool for the comprehensive understanding of every single structure within the complex morphological organization, which could be of particular interest in the study of rehabilitation of movement dysfunctions.
Collapse
|
16
|
Lottini G, Baggiani M, Chesi G, D'Orsi B, Quaranta P, Lai M, Pancrazi L, Onorati M, Pistello M, Freer G, Costa M. Zika virus induces FOXG1 nuclear displacement and downregulation in human neural progenitors. Stem Cell Reports 2022; 17:1683-1698. [PMID: 35714598 PMCID: PMC9287670 DOI: 10.1016/j.stemcr.2022.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022] Open
Abstract
Congenital alterations in the levels of the transcription factor Forkhead box g1 (FOXG1) coding gene trigger "FOXG1 syndrome," a spectrum that recapitulates birth defects found in the "congenital Zika syndrome," such as microcephaly and other neurodevelopmental conditions. Here, we report that Zika virus (ZIKV) infection alters FOXG1 nuclear localization and causes its downregulation, thus impairing expression of genes involved in cell replication and apoptosis in several cell models, including human neural progenitor cells. Growth factors, such as EGF and FGF2, and Thr271 residue located in FOXG1 AKT domain, take part in the nuclear displacement and apoptosis protection, respectively. Finally, by progressive deletion of FOXG1 sequence, we identify the C-terminus and the residues 428-481 as critical domains. Collectively, our data suggest a causal mechanism by which ZIKV affects FOXG1, its target genes, cell cycle progression, and survival of human neural progenitors, thus contributing to microcephaly.
Collapse
Affiliation(s)
- Giulia Lottini
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy; Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Matteo Baggiani
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa 56127, Italy
| | - Giulia Chesi
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Beatrice D'Orsi
- Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, Pisa 56124, Italy; Centro Pisano ricerca e implementazione clinica Flash Radiotherapy (CPFR@CISUP), Presidio S. Chiara, ed.18 via Roma, 67, Pisa 56126, Italy
| | - Paola Quaranta
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Michele Lai
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Laura Pancrazi
- Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, Pisa 56124, Italy
| | - Marco Onorati
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa 56127, Italy
| | - Mauro Pistello
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Giulia Freer
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Mario Costa
- Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, Pisa 56124, Italy; Centro Pisano ricerca e implementazione clinica Flash Radiotherapy (CPFR@CISUP), Presidio S. Chiara, ed.18 via Roma, 67, Pisa 56126, Italy; Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Piazza dei Cavalieri, Pisa 56124, Italy.
| |
Collapse
|
17
|
Modeling and Targeting Neuroglial Interactions with Human Pluripotent Stem Cell Models. Int J Mol Sci 2022; 23:ijms23031684. [PMID: 35163606 PMCID: PMC8836094 DOI: 10.3390/ijms23031684] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/26/2022] [Accepted: 01/30/2022] [Indexed: 02/05/2023] Open
Abstract
Generation of relevant and robust models for neurological disorders is of main importance for both target identification and drug discovery. The non-cell autonomous effects of glial cells on neurons have been described in a broad range of neurodegenerative and neurodevelopmental disorders, pointing to neuroglial interactions as novel alternative targets for therapeutics development. Interestingly, the recent breakthrough discovery of human induced pluripotent stem cells (hiPSCs) has opened a new road for studying neurological and neurodevelopmental disorders “in a dish”. Here, we provide an overview of the generation and modeling of both neuronal and glial cells from human iPSCs and a brief synthesis of recent work investigating neuroglial interactions using hiPSCs in a pathophysiological context.
Collapse
|
18
|
Do foetal transplant studies continue to be justified in Huntington's disease? Neuronal Signal 2021; 5:NS20210019. [PMID: 34956650 PMCID: PMC8674623 DOI: 10.1042/ns20210019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/16/2021] [Accepted: 11/22/2021] [Indexed: 11/17/2022] Open
Abstract
Early CNS transplantation studies used foetal derived cell products to provide a foundation of evidence for functional recovery in preclinical studies and early clinical trials. However, it was soon recognised that the practical limitations of foetal tissue make it unsuitable for widespread clinical use. Considerable effort has since been directed towards producing target cell phenotypes from pluripotent stem cells (PSCs) instead, and there now exist several publications detailing the differentiation and characterisation of PSC-derived products relevant for transplantation in Huntington's disease (HD). In light of this progress, we ask if foetal tissue transplantation continues to be justified in HD research. We argue that (i) the extent to which accurately differentiated target cells can presently be produced from PSCs is still unclear, currently making them undesirable for studying wider CNS transplantation issues; (ii) foetal derived cells remain a valuable tool in preclinical research for advancing our understanding of which products produce functional striatal grafts and as a reference to further improve PSC-derived products; and (iii) until PSC-derived products are ready for human trials, it is important to continue using foetal cells to gather clinical evidence that transplantation is a viable option in HD and to use this opportunity to optimise practical parameters (such as trial design, clinical practices, and delivery strategies) to pave the way for future PSC-derived products.
Collapse
|
19
|
Monk R, Lee K, Jones KS, Connor B. Directly reprogrammed Huntington's disease neural precursor cells generate striatal neurons exhibiting aggregates and impaired neuronal maturation. STEM CELLS (DAYTON, OHIO) 2021; 39:1410-1422. [PMID: 34028139 DOI: 10.1002/stem.3420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/08/2021] [Indexed: 11/07/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterized by the progressive loss of striatal medium spiny neurons. Using a highly efficient protocol for direct reprogramming of adult human fibroblasts with chemically modified mRNA, we report the first generation of HD induced neural precursor cells (iNPs) expressing striatal lineage markers that differentiated into DARPP32+ neurons from individuals with adult-onset HD (41-57 CAG). While no transcriptional differences between normal and HD reprogrammed neurons were detected by NanoString nCounter analysis, a subpopulation of HD reprogrammed neurons contained ubiquitinated polyglutamine aggregates. Importantly, reprogrammed HD neurons exhibited impaired neuronal maturation, displaying altered neurite morphology and more depolarized resting membrane potentials. Reduced BDNF protein expression in reprogrammed HD neurons correlated with increased CAG repeat lengths and earlier symptom onset. This model represents a platform for investigating impaired neuronal maturation and screening for neuronal maturation modifiers to treat HD.
Collapse
Affiliation(s)
- Ruth Monk
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kevin Lee
- Department of Physiology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kathryn S Jones
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
20
|
Bocchi VD, Conforti P, Vezzoli E, Besusso D, Cappadona C, Lischetti T, Galimberti M, Ranzani V, Bonnal RJP, De Simone M, Rossetti G, He X, Kamimoto K, Espuny-Camacho I, Faedo A, Gervasoni F, Vuono R, Morris SA, Chen J, Felsenfeld D, Pavesi G, Barker RA, Pagani M, Cattaneo E. The coding and long noncoding single-cell atlas of the developing human fetal striatum. Science 2021; 372:372/6542/eabf5759. [PMID: 33958447 DOI: 10.1126/science.abf5759] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/29/2021] [Indexed: 12/12/2022]
Abstract
Deciphering how the human striatum develops is necessary for understanding the diseases that affect this region. To decode the transcriptional modules that regulate this structure during development, we compiled a catalog of 1116 long intergenic noncoding RNAs (lincRNAs) identified de novo and then profiled 96,789 single cells from the early human fetal striatum. We found that D1 and D2 medium spiny neurons (D1- and D2-MSNs) arise from a common progenitor and that lineage commitment is established during the postmitotic transition, across a pre-MSN phase that exhibits a continuous spectrum of fate determinants. We then uncovered cell type-specific gene regulatory networks that we validated through in silico perturbation. Finally, we identified human-specific lincRNAs that contribute to the phylogenetic divergence of this structure in humans. This work delineates the cellular hierarchies governing MSN lineage commitment.
Collapse
Affiliation(s)
- Vittoria Dickinson Bocchi
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.,INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy
| | - Paola Conforti
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.,INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy
| | - Elena Vezzoli
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.,INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy
| | - Dario Besusso
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.,INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy
| | - Claudio Cappadona
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.,INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy
| | - Tiziana Lischetti
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.,INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy
| | - Maura Galimberti
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.,INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy
| | | | | | | | | | - Xiaoling He
- WT-MRC Cambridge Stem Cell Institute and Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Kenji Kamimoto
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ira Espuny-Camacho
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.,INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy
| | - Andrea Faedo
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.,INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy
| | - Federica Gervasoni
- INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy.,Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Romina Vuono
- WT-MRC Cambridge Stem Cell Institute and Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Samantha A Morris
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jian Chen
- CHDI Management/CHDI Foundation, New York, NY, USA
| | | | - Giulio Pavesi
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | - Roger A Barker
- WT-MRC Cambridge Stem Cell Institute and Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Massimiliano Pagani
- INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy. .,Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Elena Cattaneo
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy. .,INGM, Istituto Nazionale Genetica Molecolare, Milan, Italy
| |
Collapse
|
21
|
Activity-dependent regulome of human GABAergic neurons reveals new patterns of gene regulation and neurological disease heritability. Nat Neurosci 2021; 24:437-448. [PMID: 33542524 PMCID: PMC7933108 DOI: 10.1038/s41593-020-00786-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 12/18/2020] [Indexed: 01/30/2023]
Abstract
Neuronal activity-dependent gene expression is essential for brain development. Although transcriptional and epigenetic effects of neuronal activity have been explored in mice, such an investigation is lacking in humans. Because alterations in GABAergic neuronal circuits are implicated in neurological disorders, we conducted a comprehensive activity-dependent transcriptional and epigenetic profiling of human induced pluripotent stem cell-derived GABAergic neurons similar to those of the early developing striatum. We identified genes whose expression is inducible after membrane depolarization, some of which have specifically evolved in primates and/or are associated with neurological diseases, including schizophrenia and autism spectrum disorder (ASD). We define the genome-wide profile of human neuronal activity-dependent enhancers, promoters and the transcription factors CREB and CRTC1. We found significant heritability enrichment for ASD in the inducible promoters. Our results suggest that sequence variation within activity-inducible promoters of developing human forebrain GABAergic neurons contributes to ASD risk.
Collapse
|
22
|
Dell' Amico C, Tata A, Pellegrino E, Onorati M, Conti L. Genome editing in stem cells for genetic neurodisorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 182:403-438. [PMID: 34175049 DOI: 10.1016/bs.pmbts.2020.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The recent advent of genome editing techniques and their rapid improvement paved the way in establishing innovative human neurological disease models and in developing new therapeutic opportunities. Human pluripotent (both induced or naive) stem cells and neural stem cells represent versatile tools to be applied to multiple research needs and, together with genomic snip and fix tools, have recently made possible the creation of unique platforms to directly investigate several human neural affections. In this chapter, we will discuss genome engineering tools, and their recent improvements, applied to the stem cell field, focusing on how these two technologies may be pivotal instruments to deeply unravel molecular mechanisms underlying development and function, as well as disorders, of the human brain. We will review how these frontier technologies may be exploited to investigate or treat severe neurodevelopmental disorders, such as microcephaly, autism spectrum disorder, schizophrenia, as well as neurodegenerative conditions, including Parkinson's disease, Huntington's disease, Alzheimer's disease, and spinal muscular atrophy.
Collapse
Affiliation(s)
- Claudia Dell' Amico
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - Alice Tata
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Enrica Pellegrino
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy; Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Marco Onorati
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy.
| | - Luciano Conti
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy.
| |
Collapse
|
23
|
Miura Y, Li MY, Birey F, Ikeda K, Revah O, Thete MV, Park JY, Puno A, Lee SH, Porteus MH, Pașca SP. Generation of human striatal organoids and cortico-striatal assembloids from human pluripotent stem cells. Nat Biotechnol 2020; 38:1421-1430. [PMID: 33273741 PMCID: PMC9042317 DOI: 10.1038/s41587-020-00763-w] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022]
Abstract
Cortico-striatal projections are critical components of forebrain circuitry that regulate motivated behaviors. To enable the study of the human cortico-striatal pathway and how its dysfunction leads to neuropsychiatric disease, we developed a method to convert human pluripotent stem cells into region-specific brain organoids that resemble the developing human striatum and include electrically active medium spiny neurons. We then assembled these organoids with cerebral cortical organoids in three-dimensional cultures to form cortico-striatal assembloids. Using viral tracing and functional assays in intact or sliced assembloids, we show that cortical neurons send axonal projections into striatal organoids and form synaptic connections. Medium spiny neurons mature electrophysiologically following assembly and display calcium activity after optogenetic stimulation of cortical neurons. Moreover, we derive cortico-striatal assembloids from patients with a neurodevelopmental disorder caused by a deletion on chromosome 22q13.3 and capture disease-associated defects in calcium activity, showing that this approach will allow investigation of the development and functional assembly of cortico-striatal connectivity using patient-derived cells.
Collapse
Affiliation(s)
- Yuki Miura
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Human Brain Organogenesis Program, Stanford University, Stanford, CA, USA
| | - Min-Yin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Human Brain Organogenesis Program, Stanford University, Stanford, CA, USA
| | - Fikri Birey
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Human Brain Organogenesis Program, Stanford University, Stanford, CA, USA
| | - Kazuya Ikeda
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Omer Revah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Human Brain Organogenesis Program, Stanford University, Stanford, CA, USA
| | - Mayuri Vijay Thete
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Human Brain Organogenesis Program, Stanford University, Stanford, CA, USA
| | - Jin-Young Park
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Human Brain Organogenesis Program, Stanford University, Stanford, CA, USA
| | - Alyssa Puno
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Samuel H Lee
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | | | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Human Brain Organogenesis Program, Stanford University, Stanford, CA, USA.
| |
Collapse
|
24
|
Conforti P, Besusso D, Brocchetti S, Campus I, Cappadona C, Galimberti M, Laporta A, Iennaco R, Rossi RL, Dickinson VB, Cattaneo E. RUES2 hESCs exhibit MGE-biased neuronal differentiation and muHTT-dependent defective specification hinting at SP1. Neurobiol Dis 2020; 146:105140. [PMID: 33065279 DOI: 10.1016/j.nbd.2020.105140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/09/2020] [Accepted: 10/11/2020] [Indexed: 10/23/2022] Open
Abstract
RUES2 cell lines represent the first collection of isogenic human embryonic stem cells (hESCs) carrying different pathological CAG lengths in the HTT gene. However, their neuronal differentiation potential has yet to be thoroughly evaluated. Here, we report that RUES2 during ventral telencephalic differentiation is biased towards medial ganglionic eminence (MGE). We also show that HD-RUES2 cells exhibit an altered MGE transcriptional signature in addition to recapitulating known HD phenotypes, with reduced expression of the neurodevelopmental regulators NEUROD1 and BDNF and increased cleavage of synaptically enriched N-cadherin. Finally, we identified the transcription factor SP1 as a common potential detrimental co-partner of muHTT by de novo motif discovery analysis on the LGE, MGE, and cortical genes differentially expressed in HD human pluripotent stem cells in our and additional datasets. Taken together, these observations suggest a broad deleterious effect of muHTT in the early phases of neuronal development that may unfold through its altered interaction with SP1.
Collapse
Affiliation(s)
- Paola Conforti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy; Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan 20122, Italy
| | - Dario Besusso
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy; Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan 20122, Italy
| | - Silvia Brocchetti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy; Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan 20122, Italy
| | - Ilaria Campus
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy; Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan 20122, Italy
| | - Claudio Cappadona
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy; Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan 20122, Italy
| | - Maura Galimberti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy; Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan 20122, Italy
| | - Angela Laporta
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy
| | - Raffaele Iennaco
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy; Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan 20122, Italy
| | - Riccardo L Rossi
- Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan 20122, Italy
| | - Vittoria Bocchi Dickinson
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy; Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan 20122, Italy
| | - Elena Cattaneo
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122 Milan, Italy; Istituto Nazionale Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan 20122, Italy.
| |
Collapse
|
25
|
Pfaff D, Barbas H. Mechanisms for the Approach/Avoidance Decision Applied to Autism. Trends Neurosci 2020; 42:448-457. [PMID: 31253250 DOI: 10.1016/j.tins.2019.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/01/2019] [Accepted: 05/01/2019] [Indexed: 02/07/2023]
Abstract
As a neurodevelopmental disorder with serious lifelong consequences, autism has received considerable attention from neuroscientists and geneticists. We present a hypothesis of mechanisms plausibly affected during brain development in autism, based on neural pathways that are associated with social behavior and connect the prefrontal cortex (PFC) to the basal ganglia (BG). We consider failure of social approach in autism as a special case of imbalance in the fundamental dichotomy between behavioral approach and avoidance. Differential combinations of genes mutated, differences in the timing of their impact during development, and graded degrees of hormonal influences may help explain the heterogeneity in symptomatology in autism and predominance in boys.
Collapse
Affiliation(s)
- Donald Pfaff
- Laboratory of Neurobiology and Behavior, Rockefeller University, New York, NY USA.
| | - Helen Barbas
- Neural Systems Laboratory, Boston University, Boston, MA, USA.
| |
Collapse
|
26
|
Castiglioni V, Faedo A, Onorati M, Bocchi VD, Li Z, Iennaco R, Vuono R, Bulfamante GP, Muzio L, Martino G, Sestan N, Barker RA, Cattaneo E. Dynamic and Cell-Specific DACH1 Expression in Human Neocortical and Striatal Development. Cereb Cortex 2020; 29:2115-2124. [PMID: 29688344 DOI: 10.1093/cercor/bhy092] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 04/12/2018] [Indexed: 02/06/2023] Open
Abstract
DACH1 is the human homolog of the Drosophila dachshund gene, which is involved in the development of the eye, nervous system, and limbs in the fly. Here, we systematically investigate DACH1 expression patterns during human neurodevelopment, from 5 to 21 postconceptional weeks. By immunodetection analysis, we found that DACH1 is highly expressed in the proliferating neuroprogenitors of the developing cortical ventricular and subventricular regions, while it is absent in the more differentiated cortical plate. Single-cell global transcriptional analysis revealed that DACH1 is specifically enriched in neuroepithelial and ventricular radial glia cells of the developing human neocortex. Moreover, we describe a previously unreported DACH1 expression in the human striatum, in particular in the striatal medium spiny neurons. This finding qualifies DACH1 as a new striatal projection neuron marker, together with PPP1R1B, BCL11B, and EBF1. We finally compared DACH1 expression profile in human and mouse forebrain, where we observed spatio-temporal similarities in its expression pattern thus providing a precise developmental description of DACH1 in the 2 mammalian species.
Collapse
Affiliation(s)
- Valentina Castiglioni
- Department of Biosciences, Istituto Nazionale di Genetica Molecolare, University of Milan and INGM, Milan, Italy
| | - Andrea Faedo
- Department of Biosciences, Istituto Nazionale di Genetica Molecolare, University of Milan and INGM, Milan, Italy.,Cell Biology Unit, Axxam, Bresso-Milan, Italy
| | - Marco Onorati
- Department of Biosciences, Istituto Nazionale di Genetica Molecolare, University of Milan and INGM, Milan, Italy.,Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy.,Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Vittoria Dickinson Bocchi
- Department of Biosciences, Istituto Nazionale di Genetica Molecolare, University of Milan and INGM, Milan, Italy
| | - Zhen Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Raffaele Iennaco
- Department of Biosciences, Istituto Nazionale di Genetica Molecolare, University of Milan and INGM, Milan, Italy
| | - Romina Vuono
- Department of Clinical Neuroscience, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Gaetano P Bulfamante
- Unit of Human Pathology and Developmental Pathology, Department of Health Sciences, Università degli Studi di Milano, San Paolo Hospital, Milan, Italy
| | - Luca Muzio
- Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Gianvito Martino
- Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Nenad Sestan
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA.,Department of Genetics, of Psychiatry and of Comparative Medicine, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - Roger A Barker
- Department of Clinical Neuroscience, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Elena Cattaneo
- Department of Biosciences, Istituto Nazionale di Genetica Molecolare, University of Milan and INGM, Milan, Italy
| |
Collapse
|
27
|
Song S, Amores D, Chen C, McConnell K, Oh B, Poon A, George PM. Controlling properties of human neural progenitor cells using 2D and 3D conductive polymer scaffolds. Sci Rep 2019; 9:19565. [PMID: 31863072 PMCID: PMC6925212 DOI: 10.1038/s41598-019-56021-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
Human induced pluripotent stem cell-derived neural progenitor cells (hNPCs) are a promising cell source for stem cell transplantation to treat neurological diseases such as stroke and peripheral nerve injuries. However, there have been limited studies investigating how the dimensionality of the physical and electrical microenvironment affects hNPC function. In this study, we report the fabrication of two- and three-dimensional (2D and 3D respectively) constructs composed of a conductive polymer to compare the effect of electrical stimulation of hydrogel-immobilized hNPCs. The physical dimension (2D vs 3D) of stimulating platforms alone changed the hNPCs gene expression related to cell proliferation and metabolic pathways. The addition of electrical stimulation was critical in upregulating gene expression of neurotrophic factors that are important in regulating cell survival, synaptic remodeling, and nerve regeneration. This study demonstrates that the applied electrical field controls hNPC properties depending on the physical nature of stimulating platforms and cellular metabolic states. The ability to control hNPC functions can be beneficial in understanding mechanistic changes related to electrical modulation and devising novel treatment methods for neurological diseases.
Collapse
Affiliation(s)
- Shang Song
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Danielle Amores
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Cheng Chen
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Kelly McConnell
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Byeongtaek Oh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Ada Poon
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Paul M George
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Stroke Center and Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
28
|
Self-organizing neuruloids model developmental aspects of Huntington's disease in the ectodermal compartment. Nat Biotechnol 2019; 37:1198-1208. [PMID: 31501559 DOI: 10.1038/s41587-019-0237-5] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 07/23/2019] [Indexed: 12/14/2022]
Abstract
Harnessing the potential of human embryonic stem cells to mimic normal and aberrant development with standardized models is a pressing challenge. Here we use micropattern technology to recapitulate early human neurulation in large numbers of nearly identical structures called neuruloids. Dual-SMAD inhibition followed by bone morphogenic protein 4 stimulation induced self-organization of neuruloids harboring neural progenitors, neural crest, sensory placode and epidermis. Single-cell transcriptomics unveiled the precise identities and timing of fate specification. Investigation of the molecular mechanism of neuruloid self-organization revealed a pulse of pSMAD1 at the edge that induced epidermis, whose juxtaposition to central neural fates specifies neural crest and placodes, modulated by fibroblast growth factor and Wnt. Neuruloids provide a unique opportunity to study the developmental aspects of human diseases. Using isogenic Huntington's disease human embryonic stem cells and deep neural network analysis, we show how specific phenotypic signatures arise in our model of early human development as a consequence of mutant huntingtin protein, outlining an approach for phenotypic drug screening.
Collapse
|
29
|
The expression of DARPP-32 in adult male zebra finches (Taenopygia guttata). Brain Struct Funct 2019; 224:2939-2972. [PMID: 31473781 DOI: 10.1007/s00429-019-01947-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/20/2019] [Indexed: 10/26/2022]
Abstract
Although the catecholaminergic circuitry in the zebra finch brain has been well studied, there is little information regarding the postsynaptic targets of dopamine. To answer this question, we looked at overall patterns of immunoreactivity for DARPP-32 (a dopamine and cAMP-regulated phosphoprotein, present mostly in dopaminoceptive neurons) in adult male zebra finches. Our results demonstrated that as in mammals and other avian species, DARPP-32 expression was highest in both medial and lateral striatum. Interestingly, a specific pattern of immunoreactivity was observed in the song control system, with 'core' song control regions, that is, LMANcore (lateral magnocellular nucleus of the anterior nidopallium), RA (nucleus robustus arcopallialis) and HVC being less immunoreactive for DARPP-32 than 'shell' areas such as LMANshell, RAcup, AId (intermediate arcopallium) and HVCshelf. Our results suggest that whereas dopamine may modulate the shell pathways at various levels of the AFP, dopaminergic modulation of the core pathway occurs mainly through Area X, a basal ganglia nucleus. Further, secondary sensory cortices including the perientopallial belt, Fields L1 and L3 had higher DARPP-32-immunoreactivity than primary sensory cortical areas such as the pallial basolateral nucleus, entopallium proper and Field L2, corresponding to somatosensory, visual and auditory systems, respectively. We also found DARPP-32-rich axon terminals surrounding dopaminergic neurons in the ventral tegmental area-substantia nigra complex which in turn project to the striatum, suggesting that there may be a reciprocal modulation between these regions. Overall, DARPP-32 expression appears to be higher in areas involved in integrating sensory information, which further supports the role of this protein as a molecular integrator of different signal processing pathways.
Collapse
|
30
|
FOXG1-Related Syndrome: From Clinical to Molecular Genetics and Pathogenic Mechanisms. Int J Mol Sci 2019; 20:ijms20174176. [PMID: 31454984 PMCID: PMC6747066 DOI: 10.3390/ijms20174176] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/23/2019] [Accepted: 08/25/2019] [Indexed: 12/29/2022] Open
Abstract
Individuals with mutations in forkhead box G1 (FOXG1) belong to a distinct clinical entity, termed “FOXG1-related encephalopathy”. There are two clinical phenotypes/syndromes identified in FOXG1-related encephalopathy, duplications and deletions/intragenic mutations. In children with deletions or intragenic mutations of FOXG1, the recognized clinical features include microcephaly, developmental delay, severe cognitive disabilities, early-onset dyskinesia and hyperkinetic movements, stereotypies, epilepsy, and cerebral malformation. In contrast, children with duplications of FOXG1 are typically normocephalic and have normal brain magnetic resonance imaging. They also have different clinical characteristics in terms of epilepsy, movement disorders, and neurodevelopment compared with children with deletions or intragenic mutations. FOXG1 is a transcriptional factor. It is expressed mainly in the telencephalon and plays a pleiotropic role in the development of the brain. It is a key player in development and territorial specification of the anterior brain. In addition, it maintains the expansion of the neural proliferating pool, and also regulates the pace of neocortical neuronogenic progression. It also facilitates cortical layer and corpus callosum formation. Furthermore, it promotes dendrite elongation and maintains neural plasticity, including dendritic arborization and spine densities in mature neurons. In this review, we summarize the clinical features, molecular genetics, and possible pathogenesis of FOXG1-related syndrome.
Collapse
|
31
|
Fjodorova M, Louessard M, Li Z, De La Fuente DC, Dyke E, Brooks SP, Perrier AL, Li M. CTIP2-Regulated Reduction in PKA-Dependent DARPP32 Phosphorylation in Human Medium Spiny Neurons: Implications for Huntington Disease. Stem Cell Reports 2019; 13:448-457. [PMID: 31447328 PMCID: PMC6739739 DOI: 10.1016/j.stemcr.2019.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 11/26/2022] Open
Abstract
The mechanisms underlying the selective degeneration of medium spiny neurons (MSNs) in Huntington disease (HD) remain largely unknown. CTIP2, a transcription factor expressed by all MSNs, is implicated in HD pathogenesis because of its interactions with mutant huntingtin. Here, we report a key role for CTIP2 in protein phosphorylation via governing protein kinase A (PKA) signaling in human striatal neurons. Transcriptomic analysis of CTIP2-deficient MSNs implicates CTIP2 target genes at the heart of cAMP-Ca2+ signal integration in the PKA pathway. These findings are further supported by experimental evidence of a substantial reduction in phosphorylation of DARPP32 and GLUR1, two PKA targets in CTIP2-deficient MSNs. Moreover, we show that CTIP2-dependent dysregulation of protein phosphorylation is shared by HD hPSC-derived MSNs and striatal tissues of two HD mouse models. This study therefore establishes an essential role for CTIP2 in human MSN homeostasis and provides mechanistic and potential therapeutic insight into striatal neurodegeneration.
Collapse
Affiliation(s)
- Marija Fjodorova
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK.
| | - Morgane Louessard
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR861, I-Stem, AFM, 91100 Corbeil-Essonnes, France
| | - Zongze Li
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Daniel C De La Fuente
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Emma Dyke
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Simon P Brooks
- Division of Neuroscience, School of Bioscience, Cardiff University, Cardiff CF10 3AX, UK
| | - Anselme L Perrier
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR861, I-Stem, AFM, 91100 Corbeil-Essonnes, France
| | - Meng Li
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK; Division of Neuroscience, School of Bioscience, Cardiff University, Cardiff CF10 3AX, UK.
| |
Collapse
|
32
|
Iwashita M, Ohta H, Fujisawa T, Cho M, Ikeya M, Kidoaki S, Kosodo Y. Brain-stiffness-mimicking tilapia collagen gel promotes the induction of dorsal cortical neurons from human pluripotent stem cells. Sci Rep 2019; 9:3068. [PMID: 30816128 PMCID: PMC6395773 DOI: 10.1038/s41598-018-38395-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 12/21/2018] [Indexed: 11/08/2022] Open
Abstract
The mechanical properties of the extracellular microenvironment, including its stiffness, play a crucial role in stem cell fate determination. Although previous studies have demonstrated that the developing brain exhibits spatiotemporal diversity in stiffness, it remains unclear how stiffness regulates stem cell fate towards specific neural lineages. Here, we established a culture substrate that reproduces the stiffness of brain tissue using tilapia collagen for in vitro reconstitution assays. By adding crosslinkers, we obtained gels that are similar in stiffness to living brain tissue (150-1500 Pa). We further examined the capability of the gels serving as a substrate for stem cell culture and the effect of stiffness on neural lineage differentiation using human iPS cells. Surprisingly, exposure to gels with a stiffness of approximately 1500 Pa during the early period of neural induction promoted the production of dorsal cortical neurons. These findings suggest that brain-stiffness-mimicking gel has the potential to determine the terminal neural subtype. Taken together, the crosslinked tilapia collagen gel is expected to be useful in various reconstitution assays that can be used to explore the role of stiffness in neurogenesis and neural functions. The enhanced production of dorsal cortical neurons may also provide considerable advantages for neural regenerative applications.
Collapse
Affiliation(s)
- Misato Iwashita
- Korea Brain Research Institute, 61, Chemdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Hatsumi Ohta
- Ihara & Co, Ltd, 3-263-23, Zenibako, Otaru, Hokkaido, 947-0261, Japan
| | - Takahiro Fujisawa
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Minyoung Cho
- Korea Brain Research Institute, 61, Chemdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Makoto Ikeya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Satoru Kidoaki
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Yoichi Kosodo
- Korea Brain Research Institute, 61, Chemdan-ro, Dong-gu, Daegu, 41068, Republic of Korea.
| |
Collapse
|
33
|
Cortical and spinal conditioned media modify the inward ion currents and excitability and promote differentiation of human striatal primordium. J Chem Neuroanat 2018; 90:87-97. [DOI: 10.1016/j.jchemneu.2017.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 11/18/2022]
|
34
|
Karzbrun E, Kshirsagar A, Cohen SR, Hanna JH, Reiner O. Human Brain Organoids on a Chip Reveal the Physics of Folding. NATURE PHYSICS 2018; 14:515-522. [PMID: 29760764 PMCID: PMC5947782 DOI: 10.1038/s41567-018-0046-7] [Citation(s) in RCA: 259] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 01/08/2018] [Indexed: 05/18/2023]
Abstract
Human brain wrinkling has been implicated in neurodevelopmental disorders and yet its origins remain unknown. Polymer gel models suggest that wrinkling emerges spontaneously due to compression forces arising during differential swelling, but these ideas have not been tested in a living system. Here, we report the appearance of surface wrinkles during the in vitro development and self-organization of human brain organoids in a micro-fabricated compartment that supports in situ imaging over a timescale of weeks. We observe the emergence of convolutions at a critical cell density and maximal nuclear strain, which are indicative of a mechanical instability. We identify two opposing forces contributing to differential growth: cytoskeletal contraction at the organoid core and cell-cycle-dependent nuclear expansion at the organoid perimeter. The wrinkling wavelength exhibits linear scaling with tissue thickness, consistent with balanced bending and stretching energies. Lissencephalic (smooth brain) organoids display reduced convolutions, modified scaling and a reduced elastic modulus. Although the mechanism here does not include the neuronal migration seen in in vivo, it models the physics of the folding brain remarkably well. Our on-chip approach offers a means for studying the emergent properties of organoid development, with implications for the embryonic human brain.
Collapse
Affiliation(s)
- Eyal Karzbrun
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel, 7610001
| | - Aditya Kshirsagar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel, 7610001
| | - Sidney R Cohen
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel, 7610001
| | - Jacob H Hanna
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel, 7610001
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel, 7610001
| |
Collapse
|
35
|
Joven A, Simon A. Homeostatic and regenerative neurogenesis in salamanders. Prog Neurobiol 2018; 170:81-98. [PMID: 29654836 DOI: 10.1016/j.pneurobio.2018.04.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/20/2018] [Accepted: 04/07/2018] [Indexed: 01/02/2023]
Abstract
Large-scale regeneration in the adult central nervous system is a unique capacity of salamanders among tetrapods. Salamanders can replace neuronal populations, repair damaged nerve fibers and restore tissue architecture in retina, brain and spinal cord, leading to functional recovery. The underlying mechanisms have long been difficult to study due to the paucity of available genomic tools. Recent technological progress, such as genome sequencing, transgenesis and genome editing provide new momentum for systematic interrogation of regenerative processes in the salamander central nervous system. Understanding central nervous system regeneration also entails designing the appropriate molecular, cellular, and behavioral assays. Here we outline the organization of salamander brain structures. With special focus on ependymoglial cells, we integrate cellular and molecular processes of neurogenesis during developmental and adult homeostasis as well as in various injury models. Wherever possible, we correlate developmental and regenerative neurogenesis to the acquisition and recovery of behaviors. Throughout the review we place the findings into an evolutionary context for inter-species comparisons.
Collapse
Affiliation(s)
- Alberto Joven
- Department of Cell and Molecular Biology, Karolinska Institute, Berzelius väg 35, 17177, Stockholm, Sweden.
| | - András Simon
- Department of Cell and Molecular Biology, Karolinska Institute, Berzelius väg 35, 17177, Stockholm, Sweden.
| |
Collapse
|
36
|
Single-nucleus analysis of accessible chromatin in developing mouse forebrain reveals cell-type-specific transcriptional regulation. Nat Neurosci 2018; 21:432-439. [PMID: 29434377 DOI: 10.1038/s41593-018-0079-3] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/27/2017] [Indexed: 01/23/2023]
Abstract
Analysis of chromatin accessibility can reveal transcriptional regulatory sequences, but heterogeneity of primary tissues poses a significant challenge in mapping the precise chromatin landscape in specific cell types. Here we report single-nucleus ATAC-seq, a combinatorial barcoding-assisted single-cell assay for transposase-accessible chromatin that is optimized for use on flash-frozen primary tissue samples. We apply this technique to the mouse forebrain through eight developmental stages. Through analysis of more than 15,000 nuclei, we identify 20 distinct cell populations corresponding to major neuronal and non-neuronal cell types. We further define cell-type-specific transcriptional regulatory sequences, infer potential master transcriptional regulators and delineate developmental changes in forebrain cellular composition. Our results provide insight into the molecular and cellular dynamics that underlie forebrain development in the mouse and establish technical and analytical frameworks that are broadly applicable to other heterogeneous tissues.
Collapse
|
37
|
Faulty neuronal determination and cell polarization are reverted by modulating HD early phenotypes. Proc Natl Acad Sci U S A 2018; 115:E762-E771. [PMID: 29311338 DOI: 10.1073/pnas.1715865115] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Increasing evidence suggests that early neurodevelopmental defects in Huntington's disease (HD) patients could contribute to the later adult neurodegenerative phenotype. Here, by using HD-derived induced pluripotent stem cell lines, we report that early telencephalic induction and late neural identity are affected in cortical and striatal populations. We show that a large CAG expansion causes complete failure of the neuro-ectodermal acquisition, while cells carrying shorter CAGs repeats show gross abnormalities in neural rosette formation as well as disrupted cytoarchitecture in cortical organoids. Gene-expression analysis showed that control organoid overlapped with mature human fetal cortical areas, while HD organoids correlated with the immature ventricular zone/subventricular zone. We also report that defects in neuroectoderm and rosette formation could be rescued by molecular and pharmacological approaches leading to a recovery of striatal identity. These results show that mutant huntingtin precludes normal neuronal fate acquisition and highlights a possible connection between mutant huntingtin and abnormal neural development in HD.
Collapse
|
38
|
Dissection and Preparation of Human Primary Fetal Ganglionic Eminence Tissue for Research and Clinical Applications. Methods Mol Biol 2018; 1780:573-583. [PMID: 29856036 DOI: 10.1007/978-1-4939-7825-0_26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Here, we describe detailed dissection and enzymatic dissociation protocols for the ganglionic eminences from the developing human brain to generate viable quasi-single cell suspensions for subsequent use in transplantation or cell culture. These reliable and reproducible protocols can provide tissue for use in the study of the developing human brain, as well as for the preparation of donor cells for transplantation in Huntington's disease (HD). For use in the clinic as a therapy for HD, the translation of these protocols from the research laboratory to the GMP suite is described, including modification to reagents used and appropriate monitoring and tissue release criteria.
Collapse
|
39
|
Sousa AMM, Zhu Y, Raghanti MA, Kitchen RR, Onorati M, Tebbenkamp ATN, Stutz B, Meyer KA, Li M, Kawasawa YI, Liu F, Perez RG, Mele M, Carvalho T, Skarica M, Gulden FO, Pletikos M, Shibata A, Stephenson AR, Edler MK, Ely JJ, Elsworth JD, Horvath TL, Hof PR, Hyde TM, Kleinman JE, Weinberger DR, Reimers M, Lifton RP, Mane SM, Noonan JP, State MW, Lein ES, Knowles JA, Marques-Bonet T, Sherwood CC, Gerstein MB, Sestan N. Molecular and cellular reorganization of neural circuits in the human lineage. Science 2017; 358:1027-1032. [PMID: 29170230 PMCID: PMC5776074 DOI: 10.1126/science.aan3456] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 10/17/2017] [Indexed: 01/06/2023]
Abstract
To better understand the molecular and cellular differences in brain organization between human and nonhuman primates, we performed transcriptome sequencing of 16 regions of adult human, chimpanzee, and macaque brains. Integration with human single-cell transcriptomic data revealed global, regional, and cell-type-specific species expression differences in genes representing distinct functional categories. We validated and further characterized the human specificity of genes enriched in distinct cell types through histological and functional analyses, including rare subpallial-derived interneurons expressing dopamine biosynthesis genes enriched in the human striatum and absent in the nonhuman African ape neocortex. Our integrated analysis of the generated data revealed diverse molecular and cellular features of the phylogenetic reorganization of the human brain across multiple levels, with relevance for brain function and disease.
Collapse
Affiliation(s)
- André M M Sousa
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Ying Zhu
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Robert R Kitchen
- Program in Computational Biology and Bioinformatics, Departments of Molecular Biophysics and Biochemistry and Computer Science, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Marco Onorati
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Andrew T N Tebbenkamp
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Bernardo Stutz
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, New Haven, CT, USA
| | - Kyle A Meyer
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Mingfeng Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Yuka Imamura Kawasawa
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Departments of Pharmacology and Biochemistry and Molecular Biology, Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Fuchen Liu
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Raquel Garcia Perez
- Institut de Biologia Evolutiva, Consejo Superior de Investigaciones Científicas, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, Barcelona, Catalonia, Spain
| | - Marta Mele
- Institut de Biologia Evolutiva, Consejo Superior de Investigaciones Científicas, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, Barcelona, Catalonia, Spain
| | - Tiago Carvalho
- Institut de Biologia Evolutiva, Consejo Superior de Investigaciones Científicas, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, Barcelona, Catalonia, Spain
| | - Mario Skarica
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Forrest O Gulden
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Mihovil Pletikos
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Akemi Shibata
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Alexa R Stephenson
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Melissa K Edler
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - John J Ely
- Alamogordo Primate Facility, Holloman Air Force Base, NM, USA
| | - John D Elsworth
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Tamas L Horvath
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, New Haven, CT, USA
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Mark Reimers
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Richard P Lifton
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University, New Haven, CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Shrikant M Mane
- Yale Center for Genomic Analysis, Yale School of Medicine, New Haven, CT, USA
| | - James P Noonan
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Matthew W State
- Department of Psychiatry and Langley Porter Psychiatric Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA
| | - James A Knowles
- Department of Psychiatry and Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Tomas Marques-Bonet
- Institut de Biologia Evolutiva, Consejo Superior de Investigaciones Científicas, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Catalonia, Spain
- Centro Nacional de Analisis Genomico, Barcelona, Catalonia, Spain
| | - Chet C Sherwood
- Department of Anthropology, The George Washington University, Washington, DC, USA
| | - Mark B Gerstein
- Program in Computational Biology and Bioinformatics, Departments of Molecular Biophysics and Biochemistry and Computer Science, Yale University, New Haven, CT, USA
| | - Nenad Sestan
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA.
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair and Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
40
|
Tousley A, Kegel-Gleason KB. Induced Pluripotent Stem Cells in Huntington's Disease Research: Progress and Opportunity. J Huntingtons Dis 2017; 5:99-131. [PMID: 27372054 PMCID: PMC4942721 DOI: 10.3233/jhd-160199] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induced pluripotent stem cells (iPSCs) derived from controls and patients can act as a starting point for in vitro differentiation into human brain cells for discovery of novel targets and treatments for human disease without the same ethical limitations posed by embryonic stem cells. Numerous groups have successfully produced and characterized Huntington’s disease (HD) iPSCs with different CAG repeat lengths, including cells from patients with one or two HD alleles. HD iPSCs and the neural cell types derived from them recapitulate some disease phenotypes found in both human patients and animal models. Although these discoveries are encouraging, the use of iPSCs for cutting edge and reproducible research has been limited due to some of the inherent problems with cell lines and the technological differences in the way laboratories use them. The goal of this review is to summarize the current state of the HD iPSC field, and to highlight some of the issues that need to be addressed to maximize their potential as research tools.
Collapse
Affiliation(s)
| | - Kimberly B. Kegel-Gleason
- Correspondence to: Kimberly Kegel-Gleason, Assistant Professor in Neurology, Massachusetts General Hospital and Harvard Medical School, 114 16th Street, Room 2001, Charlestown, MA 02129, USA. Tel.: +1 617 724 8754; E-mail:
| |
Collapse
|
41
|
Chandrasekaran A, Avci HX, Ochalek A, Rösingh LN, Molnár K, László L, Bellák T, Téglási A, Pesti K, Mike A, Phanthong P, Bíró O, Hall V, Kitiyanant N, Krause KH, Kobolák J, Dinnyés A. Comparison of 2D and 3D neural induction methods for the generation of neural progenitor cells from human induced pluripotent stem cells. Stem Cell Res 2017; 25:139-151. [PMID: 29128818 DOI: 10.1016/j.scr.2017.10.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 02/06/2023] Open
Abstract
Neural progenitor cells (NPCs) from human induced pluripotent stem cells (hiPSCs) are frequently induced using 3D culture methodologies however, it is unknown whether spheroid-based (3D) neural induction is actually superior to monolayer (2D) neural induction. Our aim was to compare the efficiency of 2D induction with 3D induction method in their ability to generate NPCs, and subsequently neurons and astrocytes. Neural differentiation was analysed at the protein level qualitatively by immunocytochemistry and quantitatively by flow cytometry for NPC (SOX1, PAX6, NESTIN), neuronal (MAP2, TUBB3), cortical layer (TBR1, CUX1) and glial markers (SOX9, GFAP, AQP4). Electron microscopy demonstrated that both methods resulted in morphologically similar neural rosettes. However, quantification of NPCs derived from 3D neural induction exhibited an increase in the number of PAX6/NESTIN double positive cells and the derived neurons exhibited longer neurites. In contrast, 2D neural induction resulted in more SOX1 positive cells. While 2D monolayer induction resulted in slightly less mature neurons, at an early stage of differentiation, the patch clamp analysis failed to reveal any significant differences between the electrophysiological properties between the two induction methods. In conclusion, 3D neural induction increases the yield of PAX6+/NESTIN+ cells and gives rise to neurons with longer neurites, which might be an advantage for the production of forebrain cortical neurons, highlighting the potential of 3D neural induction, independent of iPSCs' genetic background.
Collapse
Affiliation(s)
- Abinaya Chandrasekaran
- BioTalentum Ltd, Gödöllő, Hungary; Molecular Animal Biotechnology Lab, Szent István University, Gödöllő, Hungary
| | - Hasan X Avci
- BioTalentum Ltd, Gödöllő, Hungary; Department of Anatomy, Embryology and Histology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Anna Ochalek
- BioTalentum Ltd, Gödöllő, Hungary; Molecular Animal Biotechnology Lab, Szent István University, Gödöllő, Hungary
| | - Lone N Rösingh
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Kinga Molnár
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Lajos László
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Tamás Bellák
- BioTalentum Ltd, Gödöllő, Hungary; Department of Anatomy, Embryology and Histology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | | | - Krisztina Pesti
- Opto-Neuropharmacology Group, MTA-ELTE NAP B, Budapest, Hungary; János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Arpad Mike
- Opto-Neuropharmacology Group, MTA-ELTE NAP B, Budapest, Hungary
| | - Phetcharat Phanthong
- BioTalentum Ltd, Gödöllő, Hungary; Stem Cell Research Group, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom Bangkok, Thailand
| | - Orsolya Bíró
- First Department of Obstetrics and Gynaecology, Semmelweis University, Budapest, Hungary
| | - Vanessa Hall
- Department of Veterinary and Animal Science, University of Copenhagen, Denmark
| | - Narisorn Kitiyanant
- Stem Cell Research Group, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom Bangkok, Thailand
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | | | - András Dinnyés
- BioTalentum Ltd, Gödöllő, Hungary; Molecular Animal Biotechnology Lab, Szent István University, Gödöllő, Hungary.
| |
Collapse
|
42
|
Kumamoto T, Hanashima C. Evolutionary conservation and conversion of Foxg1 function in brain development. Dev Growth Differ 2017; 59:258-269. [PMID: 28581027 DOI: 10.1111/dgd.12367] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 05/01/2017] [Accepted: 05/01/2017] [Indexed: 12/31/2022]
Abstract
Among the forkhead box protein family, Foxg1 is a unique transcription factor that plays pleiotropic and non-redundant roles in vertebrate brain development. The emergence of the telencephalon at the rostral end of the neural tube and its subsequent expansion that is mediated by Foxg1 was a key reason for the vertebrate brain to acquire higher order information processing, where Foxg1 is repetitively used in the sequential events of telencephalic development to control multi-steps of brain circuit formation ranging from cell cycle control to neuronal differentiation in a clade- and species-specific manner. The objective of this review is to discuss how the evolutionary changes in cis- and trans-regulatory network that is mediated by a single transcription factor has contributed to determining the fundamental vertebrate brain structure and its divergent roles in instructing species-specific neuronal circuitry and functional specialization.
Collapse
Affiliation(s)
- Takuma Kumamoto
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, 75012, Paris, France
| | - Carina Hanashima
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan.,Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan.,Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe, 650-0047, Japan
| |
Collapse
|
43
|
Characterising the developmental profile of human embryonic stem cell-derived medium spiny neuron progenitors and assessing mature neuron function using a CRISPR-generated human DARPP-32 WT/eGFP-AMP reporter line. Neurochem Int 2017; 106:3-13. [DOI: 10.1016/j.neuint.2017.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 12/21/2016] [Accepted: 01/04/2017] [Indexed: 02/06/2023]
|
44
|
Developmental alterations in Huntington's disease neural cells and pharmacological rescue in cells and mice. Nat Neurosci 2017; 20:648-660. [PMID: 28319609 PMCID: PMC5610046 DOI: 10.1038/nn.4532] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/23/2017] [Indexed: 12/11/2022]
Abstract
Neural cultures derived from Huntington's disease (HD) patient-derived induced pluripotent stem cells were used for 'omics' analyses to identify mechanisms underlying neurodegeneration. RNA-seq analysis identified genes in glutamate and GABA signaling, axonal guidance and calcium influx whose expression was decreased in HD cultures. One-third of gene changes were in pathways regulating neuronal development and maturation. When mapped to stages of mouse striatal development, the profiles aligned with earlier embryonic stages of neuronal differentiation. We observed a strong correlation between HD-related histone marks, gene expression and unique peak profiles associated with dysregulated genes, suggesting a coordinated epigenetic program. Treatment with isoxazole-9, which targets key dysregulated pathways, led to amelioration of expanded polyglutamine repeat-associated phenotypes in neural cells and of cognitive impairment and synaptic pathology in HD model R6/2 mice. These data suggest that mutant huntingtin impairs neurodevelopmental pathways that could disrupt synaptic homeostasis and increase vulnerability to the pathologic consequence of expanded polyglutamine repeats over time.
Collapse
|
45
|
Xu X, Tay Y, Sim B, Yoon SI, Huang Y, Ooi J, Utami KH, Ziaei A, Ng B, Radulescu C, Low D, Ng AYJ, Loh M, Venkatesh B, Ginhoux F, Augustine GJ, Pouladi MA. Reversal of Phenotypic Abnormalities by CRISPR/Cas9-Mediated Gene Correction in Huntington Disease Patient-Derived Induced Pluripotent Stem Cells. Stem Cell Reports 2017; 8:619-633. [PMID: 28238795 PMCID: PMC5355646 DOI: 10.1016/j.stemcr.2017.01.022] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 01/19/2017] [Accepted: 01/21/2017] [Indexed: 12/31/2022] Open
Abstract
Huntington disease (HD) is a dominant neurodegenerative disorder caused by a CAG repeat expansion in HTT. Here we report correction of HD human induced pluripotent stem cells (hiPSCs) using a CRISPR-Cas9 and piggyBac transposon-based approach. We show that both HD and corrected isogenic hiPSCs can be differentiated into excitable, synaptically active forebrain neurons. We further demonstrate that phenotypic abnormalities in HD hiPSC-derived neural cells, including impaired neural rosette formation, increased susceptibility to growth factor withdrawal, and deficits in mitochondrial respiration, are rescued in isogenic controls. Importantly, using genome-wide expression analysis, we show that a number of apparent gene expression differences detected between HD and non-related healthy control lines are absent between HD and corrected lines, suggesting that these differences are likely related to genetic background rather than HD-specific effects. Our study demonstrates correction of HD hiPSCs and associated phenotypic abnormalities, and the importance of isogenic controls for disease modeling using hiPSCs.
Collapse
Affiliation(s)
- Xiaohong Xu
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Yilin Tay
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Bernice Sim
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Su-In Yoon
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553, Singapore
| | - Yihui Huang
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Jolene Ooi
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Kagistia Hana Utami
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Amin Ziaei
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Bryan Ng
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Carola Radulescu
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Donovan Low
- Singapore Immunology Network (SIgN), A(∗)STAR, Singapore 138648, Singapore
| | - Alvin Yu Jin Ng
- Comparative Genomics Laboratory, Institute of Molecular and Cell Biology, A(∗)STAR, Biopolis, Singapore 138673, Singapore
| | - Marie Loh
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Byrappa Venkatesh
- Comparative Genomics Laboratory, Institute of Molecular and Cell Biology, A(∗)STAR, Biopolis, Singapore 138673, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A(∗)STAR, Singapore 138648, Singapore
| | - George J Augustine
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553, Singapore; Institute of Molecular and Cell Biology (IMCB), Singapore 138673, Singapore
| | - Mahmoud A Pouladi
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
46
|
Martín-Ibáñez R, Guardia I, Pardo M, Herranz C, Zietlow R, Vinh NN, Rosser A, Canals JM. Insights in spatio-temporal characterization of human fetal neural stem cells. Exp Neurol 2017; 291:20-35. [PMID: 28131724 DOI: 10.1016/j.expneurol.2017.01.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 01/12/2017] [Accepted: 01/24/2017] [Indexed: 11/25/2022]
Abstract
Primary human fetal cells have been used in clinical trials of cell replacement therapy for the treatment of neurodegenerative disorders such as Huntington's disease (HD). However, human fetal primary cells are scarce and difficult to work with and so a renewable source of cells is sought. Human fetal neural stem cells (hfNSCs) can be generated from human fetal tissue, but little is known about the differences between hfNSCs obtained from different developmental stages and brain areas. In the present work we characterized hfNSCs, grown as neurospheres, obtained from three developmental stages: 4-5, 6-7 and 8-9weeks post conception (wpc) and four brain areas: forebrain, cortex, whole ganglionic eminence (WGE) and cerebellum. We observed that, as fetal brain development proceeds, the number of neural precursors is diminished and post-mitotic cells are increased. In turn, primary cells obtained from older embryos are more sensitive to the dissociation process, their viability is diminished and they present lower proliferation ratios compared to younger embryos. However, independently of the developmental stage of derivation proliferation ratios were very low in all cases. Improvements in the expansion rates were achieved by mechanical, instead of enzymatic, dissociation of neurospheres but not by changes in the seeding densities. Regardless of the developmental stage, neurosphere cultures presented large variability in the viability and proliferation rates during the initial 3-4 passages, but stabilized achieving significant expansion rates at passage 5 to 6. This was true also for all brain regions except cerebellar derived cultures that did not expand. Interestingly, the brain region of hfNSC derivation influences the expansion potential, being forebrain, cortex and WGE derived cells the most expandable compared to cerebellar. Short term expansion partially compromised the regional identity of cortical but not WGE cultures. Nevertheless, both expanded cultures were multipotent and kept the ability to differentiate to region specific mature neuronal phenotypes.
Collapse
Affiliation(s)
- Raquel Martín-Ibáñez
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University of Barcelona, Casanova 143, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain; Networked Biomedical Research Centre for NeuroDegenerative Disorders (CIBERNED), Spain; Research and Development Unit, Cell Therapy Program, Faculty of Medicine and Health Sciences, University of Barcelona, Casanova 143, 08036 Barcelona, Spain.
| | - Inés Guardia
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University of Barcelona, Casanova 143, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain; Networked Biomedical Research Centre for NeuroDegenerative Disorders (CIBERNED), Spain.
| | - Mónica Pardo
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University of Barcelona, Casanova 143, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain; Networked Biomedical Research Centre for NeuroDegenerative Disorders (CIBERNED), Spain.
| | - Cristina Herranz
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University of Barcelona, Casanova 143, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain; Networked Biomedical Research Centre for NeuroDegenerative Disorders (CIBERNED), Spain; Research and Development Unit, Cell Therapy Program, Faculty of Medicine and Health Sciences, University of Barcelona, Casanova 143, 08036 Barcelona, Spain.
| | - Rike Zietlow
- Cardiff University Brain Repair Group, Schools of Biosciences and Medicine, University of Cardiff, UK.
| | - Ngoc-Nga Vinh
- Cardiff University Brain Repair Group, Schools of Biosciences and Medicine, University of Cardiff, UK.
| | - Anne Rosser
- Cardiff University Brain Repair Group, Schools of Biosciences and Medicine, University of Cardiff, UK.
| | - Josep M Canals
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University of Barcelona, Casanova 143, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain; Networked Biomedical Research Centre for NeuroDegenerative Disorders (CIBERNED), Spain; Research and Development Unit, Cell Therapy Program, Faculty of Medicine and Health Sciences, University of Barcelona, Casanova 143, 08036 Barcelona, Spain.
| |
Collapse
|
47
|
Ring KL, An MC, Zhang N, O'Brien RN, Ramos EM, Gao F, Atwood R, Bailus BJ, Melov S, Mooney SD, Coppola G, Ellerby LM. Genomic Analysis Reveals Disruption of Striatal Neuronal Development and Therapeutic Targets in Human Huntington's Disease Neural Stem Cells. Stem Cell Reports 2016; 5:1023-1038. [PMID: 26651603 PMCID: PMC4682390 DOI: 10.1016/j.stemcr.2015.11.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 11/02/2015] [Accepted: 11/12/2015] [Indexed: 12/29/2022] Open
Abstract
We utilized induced pluripotent stem cells (iPSCs) derived from Huntington's disease (HD) patients as a human model of HD and determined that the disease phenotypes only manifest in the differentiated neural stem cell (NSC) stage, not in iPSCs. To understand the molecular basis for the CAG repeat expansion-dependent disease phenotypes in NSCs, we performed transcriptomic analysis of HD iPSCs and HD NSCs compared to isogenic controls. Differential gene expression and pathway analysis pointed to transforming growth factor β (TGF-β) and netrin-1 as the top dysregulated pathways. Using data-driven gene coexpression network analysis, we identified seven distinct coexpression modules and focused on two that were correlated with changes in gene expression due to the CAG expansion. Our HD NSC model revealed the dysregulation of genes involved in neuronal development and the formation of the dorsal striatum. The striatal and neuronal networks disrupted could be modulated to correct HD phenotypes and provide therapeutic targets.
Collapse
Affiliation(s)
- Karen L Ring
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Mahru C An
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Ningzhe Zhang
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Eliana Marisa Ramos
- Departments of Neurology and Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Fuying Gao
- Departments of Neurology and Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Robert Atwood
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Sean D Mooney
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Giovanni Coppola
- Departments of Neurology and Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Lisa M Ellerby
- Buck Institute for Research on Aging, Novato, CA 94945, USA.
| |
Collapse
|
48
|
Onorati M, Li Z, Liu F, Sousa AMM, Nakagawa N, Li M, Dell'Anno MT, Gulden FO, Pochareddy S, Tebbenkamp ATN, Han W, Pletikos M, Gao T, Zhu Y, Bichsel C, Varela L, Szigeti-Buck K, Lisgo S, Zhang Y, Testen A, Gao XB, Mlakar J, Popovic M, Flamand M, Strittmatter SM, Kaczmarek LK, Anton ES, Horvath TL, Lindenbach BD, Sestan N. Zika Virus Disrupts Phospho-TBK1 Localization and Mitosis in Human Neuroepithelial Stem Cells and Radial Glia. Cell Rep 2016; 16:2576-2592. [PMID: 27568284 PMCID: PMC5135012 DOI: 10.1016/j.celrep.2016.08.038] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/27/2016] [Accepted: 08/12/2016] [Indexed: 02/08/2023] Open
Abstract
The mechanisms underlying Zika virus (ZIKV)-related microcephaly and other neurodevelopment defects remain poorly understood. Here, we describe the derivation and characterization, including single-cell RNA-seq, of neocortical and spinal cord neuroepithelial stem (NES) cells to model early human neurodevelopment and ZIKV-related neuropathogenesis. By analyzing human NES cells, organotypic fetal brain slices, and a ZIKV-infected micrencephalic brain, we show that ZIKV infects both neocortical and spinal NES cells as well as their fetal homolog, radial glial cells (RGCs), causing disrupted mitoses, supernumerary centrosomes, structural disorganization, and cell death. ZIKV infection of NES cells and RGCs causes centrosomal depletion and mitochondrial sequestration of phospho-TBK1 during mitosis. We also found that nucleoside analogs inhibit ZIKV replication in NES cells, protecting them from ZIKV-induced pTBK1 relocalization and cell death. We established a model system of human neural stem cells to reveal cellular and molecular mechanisms underlying neurodevelopmental defects associated with ZIKV infection and its potential treatment.
Collapse
Affiliation(s)
- Marco Onorati
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Zhen Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Fuchen Liu
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - André M M Sousa
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Naoki Nakagawa
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Mingfeng Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Maria Teresa Dell'Anno
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Forrest O Gulden
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sirisha Pochareddy
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Andrew T N Tebbenkamp
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Wenqi Han
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mihovil Pletikos
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Tianliuyun Gao
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ying Zhu
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Candace Bichsel
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Luis Varela
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Klara Szigeti-Buck
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Steven Lisgo
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE13BZ, UK
| | - Yalan Zhang
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Anze Testen
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Xiao-Bing Gao
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jernej Mlakar
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Mara Popovic
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Marie Flamand
- Department of Virology, Institut Pasteur, 75724 Paris Cedex 15, France
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Tamas L Horvath
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; Yale Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Brett D Lindenbach
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Nenad Sestan
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Genetics and Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
49
|
Human t-DARPP is induced during striatal development. Neuroscience 2016; 333:320-30. [PMID: 27475250 DOI: 10.1016/j.neuroscience.2016.07.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 07/06/2016] [Accepted: 07/08/2016] [Indexed: 11/21/2022]
Abstract
Human Dopamine- and cAMP-regulated phosphoprotein of molecular weight 32kDa (DARPP-32, also known as PPP1R1B) gene codes for different transcripts that are mainly translated into two DARPP-32 protein isoforms, full length (fl)-DARPP-32 and truncated (t)-DARPP. The t-DARPP lacks the first 36 residues at the N-terminal, which alters its function. In the central nervous system, fl-DARPP-32 is highly expressed in GABAergic striatal medium spiny neurons (MSNs), where it integrates dopaminergic and glutamatergic input signaling. However, no information about human DARPP-32 isoform expression during MSNs maturation is available. In this study, our aim is to determine the expression of the two DARPP-32 isoforms in human fetal and adult striatal samples. We show that DARPP-32 isoform expression is differentially regulated during human striatal development, with the t-DARPP isoform being virtually absent from whole ganglionic eminence (WGE) and highly induced in the adult striatum (in both caudate and putamen). We next compared the four most common anti-DARPP-32 antibodies used in human specimens, to study their recognition of the two isoforms in fetal and adult human striatal samples by western blot and immunohistochemistry. The four antibodies specifically identify the fl-DARPP-32 in both fetal and adult samples, while t-DARPP form was only detected in adult striatal samples. In addition, the lack of t-DARPP recognition in human adult striatum by the antibody generated against the full-length domain produces in turn different efficacy by immunohistochemical analysis. In conclusion, our results show that expression of human DARPP-32 protein isoforms depends on the striatal neurodevelopmental stage with t-DARPP being specific for the human adult striatum.
Collapse
|
50
|
Clark AR, Carter AB, Hager LE, Price EM. In Vivo Neural Tissue Engineering: Cylindrical Biocompatible Hydrogels That Create New Neural Tracts in the Adult Mammalian Brain. Stem Cells Dev 2016; 25:1109-18. [PMID: 27295980 DOI: 10.1089/scd.2016.0069] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Individuals with neurodegenerative disorders or brain injury have few treatment options and it has been proposed that endogenous adult neural stem cells can be harnessed to repopulate dysfunctional nonneurogenic regions of the brain. We have accomplished this through the development of rationally designed hydrogel implants that recruit endogenous cells from the adult subventricular zone to create new relatively long tracts of neuroblasts. These implants are biocompatible and biodegradable cylindrical hydrogels consisting of fibrin and immobilized neurotrophic factors. When implanted into rat brain such that the cylinder intersected the migratory path of endogenous neural progenitors (the rostral migratory stream) and led into the nonneurogenic striatum, we observed a robust neurogenic response in the form of migrating neuroblasts with long (>100 μm) complex neurites. The location of these new neural cells in the striatum was directly coincident with the original track of the fibrin implant, which itself had completely degraded, and covered a significant area and distance (>2.5 mm). We also observed a significant number of neuroblasts in the striatal region between the implant track and the lateral ventricle. When these fibrin cylinders were implanted into hemiparkinson rats, correction of parkinsonian behavior was observed. There were no obvious behavioral, inflammatory or tumorigenic sequelae as a consequence of the implants. In conclusion, we have successfully engineered neural tissue in vivo, using neurogenic biomaterials cast into a unique cylindrical architecture. These results represent a novel approach to efficiently induce neurogenesis in a controlled and targeted manner, which may lead toward a new therapeutic modality for neurological disorders.
Collapse
Affiliation(s)
- Amanda R Clark
- Department of Biology, Marshall University , Huntington, West Virginia
| | - Arrin B Carter
- Department of Biology, Marshall University , Huntington, West Virginia
| | - Lydia E Hager
- Department of Biology, Marshall University , Huntington, West Virginia
| | - Elmer M Price
- Department of Biology, Marshall University , Huntington, West Virginia
| |
Collapse
|