Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters








Publication year range
1.
Dev Cell ; 59(16): 2171-2188.e7, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39106860

ABSTRACT

Proneural transcription factors establish molecular cascades to orchestrate neuronal diversity. One such transcription factor, Atonal homolog 1 (Atoh1), gives rise to cerebellar excitatory neurons and over 30 distinct nuclei in the brainstem critical for hearing, breathing, and balance. Although Atoh1 lineage neurons have been qualitatively described, the transcriptional programs that drive their fate decisions and the full extent of their diversity remain unknown. Here, we analyzed single-cell RNA sequencing and ATOH1 DNA binding in Atoh1 lineage neurons of the developing mouse hindbrain. This high-resolution dataset identified markers for specific brainstem nuclei and demonstrated that transcriptionally heterogeneous progenitors require ATOH1 for proper migration. Moreover, we identified a sizable population of proliferating unipolar brush cell progenitors in the mouse Atoh1 lineage, previously described in humans as the origin of one medulloblastoma subtype. Collectively, our data provide insights into the developing mouse hindbrain and markers for functional assessment of understudied neuronal populations.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cell Lineage , Neurons , Rhombencephalon , Single-Cell Analysis , Transcriptome , Animals , Rhombencephalon/metabolism , Rhombencephalon/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Mice , Neurons/metabolism , Neurons/cytology , Cell Lineage/genetics , Single-Cell Analysis/methods , Transcriptome/genetics , Cell Differentiation , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Cell Movement
2.
Sci Adv ; 9(26): eadg1671, 2023 06 30.
Article in English | MEDLINE | ID: mdl-37390208

ABSTRACT

Pontine nuclei (PN) neurons mediate the communication between the cerebral cortex andthe cerebellum to refine skilled motor functions. Prior studies showed that PN neurons fall into two subtypes based on their anatomic location and region-specific connectivity, but the extent of their heterogeneity and its molecular drivers remain unknown. Atoh1 encodes a transcription factor that is expressed in the PN precursors. We previously showed that partial loss of Atoh1 function in mice results in delayed PN development and impaired motor learning. In this study, we performed single-cell RNA sequencing to elucidate the cell state-specific functions of Atoh1 during PN development and found that Atoh1 regulates cell cycle exit, differentiation, migration, and survival of PN neurons. Our data revealed six previously not known PN subtypes that are molecularly and spatially distinct. We found that the PN subtypes exhibit differential vulnerability to partial loss of Atoh1 function, providing insights into the prominence of PN phenotypes in patients with ATOH1 missense mutations.


Subject(s)
Cerebellum , Neurons , Animals , Mice , Cell Differentiation , Cell Cycle , Cell Division , Basic Helix-Loop-Helix Transcription Factors/genetics
3.
Front Cell Neurosci ; 15: 684792, 2021.
Article in English | MEDLINE | ID: mdl-34408627

ABSTRACT

The spinal cord contains a diverse array of sensory and motor circuits that are essential for normal function. Spinal cord injury (SCI) permanently disrupts neural circuits through initial mechanical damage, as well as a cascade of secondary injury events that further expand the spinal cord lesion, resulting in permanent paralysis. Tissue clearing and 3D imaging have recently emerged as promising techniques to improve our understanding of the complex neural circuitry of the spinal cord and the changes that result from damage due to SCI. However, the application of this technology for studying the intact and injured spinal cord remains limited. Here, we optimized the passive CLARITY technique (PACT) to obtain gentle and efficient clearing of the murine spinal cord without the need for specialized equipment. We demonstrate that PACT clearing enables 3D imaging of multiple fluorescent labels in the spinal cord to assess molecularly defined neuronal populations, acute inflammation, long-term tissue damage, and cell transplantation. Collectively, these procedures provide a framework for expanding the utility of tissue clearing to enhance the study of spinal cord neural circuits, as well as cellular- and tissue-level changes that occur following SCI.

4.
Development ; 148(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34142711

ABSTRACT

Axial elongation of the neural tube is crucial during mammalian embryogenesis for anterior-posterior body axis establishment and subsequent spinal cord development, but these processes cannot be interrogated directly in humans as they occur post-implantation. Here, we report an organoid model of neural tube extension derived from human pluripotent stem cell (hPSC) aggregates that have been caudalized with Wnt agonism, enabling them to recapitulate aspects of the morphological and temporal gene expression patterns of neural tube development. Elongating organoids consist largely of neuroepithelial compartments and contain TBXT+SOX2+ neuro-mesodermal progenitors in addition to PAX6+NES+ neural progenitors. A critical threshold of Wnt agonism stimulated singular axial extensions while maintaining multiple cell lineages, such that organoids displayed regionalized anterior-to-posterior HOX gene expression with hindbrain (HOXB1) regions spatially distinct from brachial (HOXC6) and thoracic (HOXB9) regions. CRISPR interference-mediated silencing of TBXT, a Wnt pathway target, increased neuroepithelial compartmentalization, abrogated HOX expression and disrupted uniaxial elongation. Together, these results demonstrate the potent capacity of caudalized hPSC organoids to undergo axial elongation in a manner that can be used to dissect the cellular organization and patterning decisions that dictate early human nervous system development.


Subject(s)
Body Patterning , Neural Tube/embryology , Organogenesis , Organoids , Body Patterning/drug effects , Cell Differentiation , Embryonic Development , Gene Expression Regulation, Developmental , Humans , Mesoderm/embryology , Mesoderm/metabolism , Neurogenesis/drug effects , Organogenesis/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Wnt Signaling Pathway/drug effects
5.
Nat Protoc ; 15(1): 181, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31705126

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Nat Protoc ; 14(11): 3033-3058, 2019 11.
Article in English | MEDLINE | ID: mdl-31628445

ABSTRACT

V2a interneurons are located in the hindbrain and spinal cord, where they provide rhythmic input to major motor control centers. Many of the phenotypic properties and functions of excitatory V2a interneurons have yet to be fully defined. Definition of these properties could lead to novel regenerative therapies for traumatic injuries and drug targets for chronic degenerative diseases. Here we describe how to produce V2a interneurons from mouse and human pluripotent stem cells (PSCs), as well as strategies to characterize and mature the cells for further analysis. The described protocols are based on a sequence of small-molecule treatments that induce differentiation of PSCs into V2a interneurons. We also include a detailed description of how to phenotypically characterize, mature, and freeze the cells. The mouse and human protocols are similar in regard to the sequence of small molecules used but differ slightly in the concentrations and durations necessary for induction. With the protocols described, scientists can expect to obtain V2a interneurons with purities of ~75% (mouse) in 7 d and ~50% (human) in 20 d.


Subject(s)
Interneurons/cytology , Neurogenesis , Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques/methods , Cell Line , Humans , Mice
7.
Proc Natl Acad Sci U S A ; 114(19): 4969-4974, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28438991

ABSTRACT

The spinal cord consists of multiple neuronal cell types that are critical to motor control and arise from distinct progenitor domains in the developing neural tube. Excitatory V2a interneurons in particular are an integral component of central pattern generators that control respiration and locomotion; however, the lack of a robust source of human V2a interneurons limits the ability to molecularly profile these cells and examine their therapeutic potential to treat spinal cord injury (SCI). Here, we report the directed differentiation of CHX10+ V2a interneurons from human pluripotent stem cells (hPSCs). Signaling pathways (retinoic acid, sonic hedgehog, and Notch) that pattern the neural tube were sequentially perturbed to identify an optimized combination of small molecules that yielded ∼25% CHX10+ cells in four hPSC lines. Differentiated cultures expressed much higher levels of V2a phenotypic markers (CHX10 and SOX14) than other neural lineage markers. Over time, CHX10+ cells expressed neuronal markers [neurofilament, NeuN, and vesicular glutamate transporter 2 (VGlut2)], and cultures exhibited increased action potential frequency. Single-cell RNAseq analysis confirmed CHX10+ cells within the differentiated population, which consisted primarily of neurons with some glial and neural progenitor cells. At 2 wk after transplantation into the spinal cord of mice, hPSC-derived V2a cultures survived at the site of injection, coexpressed NeuN and VGlut2, extended neurites >5 mm, and formed putative synapses with host neurons. These results provide a description of V2a interneurons differentiated from hPSCs that may be used to model central nervous system development and serve as a potential cell therapy for SCI.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Differentiation , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Neurons/cytology
8.
Exp Neurol ; 277: 305-316, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26784005

ABSTRACT

Challenges in parsing specific contributions to spinal microcircuit architecture have limited our ability to model and manipulate those networks for improved functional regeneration after injury or disease. While spinal interneurons (INs) have been implicated in driving coordinated locomotor behaviors, they constitute only a small percentage of the spinal cord and are difficult to isolate from primary tissue. In this study, we employed a genetic strategy to obtain large quantities of highly enriched mouse embryonic stem cell (ESC)-derived V2a INs, an excitatory glutamatergic IN population that is defined by expression of the homeodomain protein Chx10 during development. Puromycin N-acetyltransferase expression was driven by the native gene regulatory elements of Chx10 in the transgenic ESC line, resulting in positive selection of V2a INs after induction and treatment with puromycin. Directly after selection, approximately 80% of cells are Chx10(+), with 94% Lhx3(+); after several weeks, cultures remain free of proliferative cell types and mature into normal glutamatergic neurons as assessed by molecular markers and electrophysiological methods. Functional synapses were observed between selected ESC-derived V2a INs and motor neurons when co-cultured, demonstrating the potential of these cells to form neural networks. While ESC-derived neurons obtained in vitro are not identical to those that develop in the spinal cord, the transgenic ESCs here provide a unique tool to begin studying V2a INs in isolation or for use in in vitro models of spinal microcircuits.


Subject(s)
Embryonic Stem Cells/physiology , Homeodomain Proteins/metabolism , Interneurons/metabolism , Transcription Factors/metabolism , Acetyltransferases/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Animals , Cell Differentiation , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Synthesis Inhibitors/pharmacology , Puromycin/pharmacology , Transcription Factors/genetics
9.
Methods ; 101: 11-20, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26658353

ABSTRACT

Culture of human pluripotent stem cells (hPSC) as in vitro multicellular aggregates has been increasingly used as a method to model early embryonic development. Three-dimensional assemblies of hPSCs facilitate interactions between cells and their microenvironment to promote morphogenesis, analogous to the multicellular organization that accompanies embryogenesis. In this paper, we describe a method for reproducibly generating and maintaining populations of homogeneous three-dimensional hPSC aggregates using forced aggregation and rotary orbital suspension culture. We propose solutions to several challenges associated with the consistent formation and extended culture of cell spheroids generated from hPSCs and their differentiated progeny. Further, we provide examples to demonstrate how aggregation can be used as a tool to select specific subpopulations of cells to create homotypic spheroids, or as a means to introduce multiple cell types to create heterotypic tissue constructs. Finally, we demonstrate that the aggregation and rotary suspension method can be used to support culture and maintenance of hPSC-derived cell populations representing each of the three germ layers, underscoring the utility of this platform for culturing many different cell types.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Cell Aggregation , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Hepatocytes/physiology , Human Embryonic Stem Cells/physiology , Humans , Motor Neurons/physiology , Myocytes, Cardiac/physiology , Spheroids, Cellular/physiology
10.
IEEE Trans Biomed Circuits Syst ; 9(6): 801-14, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26812735

ABSTRACT

In this paper, we present a fully integrated multi-modality CMOS cellular sensor array with four sensing modalities to characterize different cell physiological responses, including extracellular voltage recording, cellular impedance mapping, optical detection with shadow imaging and bioluminescence sensing, and thermal monitoring. The sensor array consists of nine parallel pixel groups and nine corresponding signal conditioning blocks. Each pixel group comprises one temperature sensor and 16 tri-modality sensor pixels, while each tri-modality sensor pixel can be independently configured for extracellular voltage recording, cellular impedance measurement (voltage excitation/current sensing), and optical detection. This sensor array supports multi-modality cellular sensing at the pixel level, which enables holistic cell characterization and joint-modality physiological monitoring on the same cellular sample with a pixel resolution of 80 µm × 100 µm. Comprehensive biological experiments with different living cell samples demonstrate the functionality and benefit of the proposed multi-modality sensing in cell-based assay and drug screening.


Subject(s)
Biosensing Techniques/instrumentation , Cell Culture Techniques/instrumentation , Drug Evaluation, Preclinical/instrumentation , Tissue Array Analysis/instrumentation , Animals , Biosensing Techniques/methods , Cell Culture Techniques/methods , Cell Line, Tumor , Drug Evaluation, Preclinical/methods , Humans , Mice , Myocytes, Cardiac/cytology , Neurons/cytology , Semiconductors , Tissue Array Analysis/methods
11.
Biotechnol Bioeng ; 111(10): 2041-55, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24842774

ABSTRACT

A common problem with using embryonic stem (ES) cells as a source for analysis of gene expression, drug toxicity, or functional characterization studies is the heterogeneity that results from many differentiation protocols. The ability to generate large numbers of high purity differentiated cells from pluripotent stem cells could greatly enhance their utility for in vitro characterization studies and transplantation in pre-clinical injury models. Population heterogeneity is particularly troublesome for post-mitotic neurons, including motoneurons, because they do not proliferate and are quickly diluted in culture by proliferative phenotypes, such as glia. Studies of motoneuron biology and disease, in particular amyotrophic lateral sclerosis, can benefit from high purity motoneuron cultures. In this study, we engineered a transgenic-ES cell line where highly conserved enhancer elements for the motoneuron transcription factor Hb9 were used to drive puromycin N-acetyltransferase expression in ES cell-derived motoneurons. Antibiotic selection with puromycin was then used to obtain high purity motoneuron cultures following differentiation of mouse ES cells. Purity was maintained during maturation allowing the production of consistent, uniform populations of cholinergic ES cell-derived motoneurons. Appropriate functional properties of purified motoneurons were verified by acetylcholinesterase activity and electrophysiology. Antibiotic selection, therefore, can provide an inexpensive alternative to current methods for isolating ES cell-derived motoneurons at high purity that does not require specialized laboratory equipment and provides a unique platform for studies in motoneuron development and degeneration.


Subject(s)
Embryonic Stem Cells/cytology , Homeodomain Proteins/genetics , Motor Neurons/cytology , Neurogenesis , Transcription Factors/genetics , Acetyltransferases/genetics , Animals , Antimetabolites, Antineoplastic/metabolism , Cell Culture Techniques/methods , Cell Engineering , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Enzymologic , Mice , Motor Neurons/metabolism , Protein Synthesis Inhibitors/metabolism , Puromycin/metabolism , Transgenes
12.
Stem Cells Dev ; 23(15): 1765-76, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24650073

ABSTRACT

V2a interneurons of the ventral spinal cord and hindbrain play an important role in the central pattern generators (CPGs) involved in locomotion, skilled reaching, and respiration. However, sources of V2a interneurons for in vitro studies are limited. In this study, we developed a differentiation protocol for V2a interneurons from mouse embryonic stem cells (mESCs). Cells were induced in a 2(-)/4(+) induction protocol with varying concentrations of retinoic acid (RA) and the mild sonic hedgehog (Shh) agonist purmorphamine (Pur) in order to increase the expression of V2a interneuron transcription factors (eg, Chx10). Notch signaling, which influences the commitment of p2 progenitor cells to V2a or V2b interneurons, was inhibited in cell cultures to increase the percentage of V2a interneurons. At the end of the induction period, cell commitment was assessed using quantitative real-time polymerase chain reaction, immunocytochemistry, and flow cytometry to quantify expression of transcription factors specific to V2a interneurons and the adjacent ventral spinal cord regions. Low concentrations of RA and high concentrations of Pur led to greater expression of transcription factors specific for V2a interneurons. Notch inhibition favored V2a interneuron over V2b interneuron differentiation. The protocol established in this study can be used to further elucidate the pathways involved in V2a interneuron differentiation and help produce sources of V2a interneurons for developmental neurobiology, electrophysiology, and transplantation studies.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Interneurons/cytology , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Dipeptides/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/metabolism , Interneurons/drug effects , Interneurons/metabolism , Mice , Morpholines/pharmacology , Neural Tube/drug effects , Neural Tube/embryology , Neural Tube/metabolism , Purines/pharmacology , Receptors, Notch/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/metabolism , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL