Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
EMBO Rep ; 25(5): 2202-2219, 2024 May.
Article in English | MEDLINE | ID: mdl-38600346

ABSTRACT

Neural progenitor cells within the cerebral cortex undergo a characteristic switch between symmetric self-renewing cell divisions early in development and asymmetric neurogenic divisions later. Yet, the mechanisms controlling this transition remain unclear. Previous work has shown that early but not late neural progenitor cells (NPCs) endogenously express the autism-linked transcription factor Foxp1, and both loss and gain of Foxp1 function can alter NPC activity and fate choices. Here, we show that premature loss of Foxp1 upregulates transcriptional programs regulating angiogenesis, glycolysis, and cellular responses to hypoxia. These changes coincide with a premature destabilization of HIF-1α, an elevation in HIF-1α target genes, including Vegfa in NPCs, and precocious vascular network development. In vitro experiments demonstrate that stabilization of HIF-1α in Foxp1-deficient NPCs rescues the premature differentiation phenotype and restores NPC maintenance. Our data indicate that the endogenous decline in Foxp1 expression activates the HIF-1α transcriptional program leading to changes in the tissue environment adjacent to NPCs, which, in turn, might alter their self-renewal and neurogenic capacities.


Subject(s)
Cerebral Cortex , Forkhead Transcription Factors , Hypoxia-Inducible Factor 1, alpha Subunit , Neural Stem Cells , Repressor Proteins , Signal Transduction , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Animals , Mice , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Repressor Proteins/metabolism , Repressor Proteins/genetics , Neovascularization, Physiologic/genetics , Cell Differentiation/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Neurogenesis/genetics , Glycolysis , Angiogenesis
2.
bioRxiv ; 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-37546781

ABSTRACT

Dorsal interneurons (dIs) in the spinal cord encode the perception of touch, pain, heat, itch, and proprioception. While previous studies using genetic strategies in animal models have revealed important insights into dI development, the molecular details by which dIs arise as distinct populations of neurons remain incomplete. We have developed a resource to investigate dI fate specification by combining a single-cell RNA-Seq atlas of mouse ESC-derived dIs with pseudotime analyses. To validate this in silico resource as a useful tool, we used it to first identify novel genes that are candidates for directing the transition states that lead to distinct dI lineage trajectories, and then validated them using in situ hybridization analyses in the developing mouse spinal cord in vivo . We have also identified a novel endpoint of the dI5 lineage trajectory and found that dIs become more transcriptionally homogenous during terminal differentiation. Together, this study introduces a valuable tool for further discovery about the timing of gene expression during dI differentiation and demonstrates its utility clarifying dI lineage relationships. Summary statement: Pseudotime analyses of embryonic stem cell-derived dorsal spinal interneurons reveals both novel regulators and lineage relationships between different interneuron populations.

3.
Science ; 381(6665): 1480-1487, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37769108

ABSTRACT

After heart injury, dead heart muscle is replaced by scar tissue. Fibroblasts can electrically couple with myocytes, and changes in fibroblast membrane potential can lead to myocyte excitability, which suggests that fibroblast-myocyte coupling in scar tissue may be responsible for arrhythmogenesis. However, the physiologic relevance of electrical coupling of myocytes and fibroblasts and its impact on cardiac excitability in vivo have never been demonstrated. We genetically engineered a mouse that expresses the optogenetic cationic channel ChR2 (H134R) exclusively in cardiac fibroblasts. After myocardial infarction, optical stimulation of scar tissue elicited organ-wide cardiac excitation and induced arrhythmias in these animals. Complementing computational modeling with experimental approaches, we showed that gap junctional and ephaptic coupling, in a synergistic yet functionally redundant manner, excited myocytes coupled to fibroblasts.


Subject(s)
Arrhythmias, Cardiac , Channelrhodopsins , Cicatrix , Fibroblasts , Myocytes, Cardiac , Animals , Mice , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Cicatrix/pathology , Cicatrix/physiopathology , Fibroblasts/physiology , Myocytes, Cardiac/physiology , Channelrhodopsins/genetics , Channelrhodopsins/physiology , Optogenetics , Connexin 43/genetics , Connexin 43/physiology , Gene Knockout Techniques
4.
NPJ Regen Med ; 8(1): 16, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36922514

ABSTRACT

We developed an on-slide decellularization approach to generate acellular extracellular matrix (ECM) myoscaffolds that can be repopulated with various cell types to interrogate cell-ECM interactions. Using this platform, we investigated whether fibrotic ECM scarring affected human skeletal muscle progenitor cell (SMPC) functions that are essential for myoregeneration. SMPCs exhibited robust adhesion, motility, and differentiation on healthy muscle-derived myoscaffolds. All SPMC interactions with fibrotic myoscaffolds from dystrophic muscle were severely blunted including reduced motility rate and migration. Furthermore, SMPCs were unable to remodel laminin dense fibrotic scars within diseased myoscaffolds. Proteomics and structural analysis revealed that excessive collagen deposition alone is not pathological, and can be compensatory, as revealed by overexpression of sarcospan and its associated ECM receptors in dystrophic muscle. Our in vivo data also supported that ECM remodeling is important for SMPC engraftment and that fibrotic scars may represent one barrier to efficient cell therapy.

5.
bioRxiv ; 2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36824905

ABSTRACT

Embryonic neural stem cells (NSCs, i.e., radial glia) in the ventricular-subventricular zone (V-SVZ) generate the majority of neurons and glia in the forebrain. Postnatally, embryonic radial glia disappear and a subpopulation of radial glia transition into adult NSCs. As this transition occurs, widespread neurogenesis in brain regions such as the cerebral cortex ends. The mechanisms that regulate the postnatal disappearance of radial glia and the ending of embryonic neurogenesis remain poorly understood. Here, we show that PR domain-containing 16 (Prdm16) promotes the disappearance of radial glia and the ending of neurogenesis in the cerebral cortex. Genetic deletion of Prdm16 from NSCs leads to the persistence of radial glia in the adult V-SVZ and prolonged postnatal cortical neurogenesis. Mechanistically, Prdm16 induces the postnatal reduction in Vascular Cell Adhesion Molecule 1 (Vcam1). The postnatal disappearance of radial glia and the ending of cortical neurogenesis occur normally in Prdm16-Vcam1 double conditional knockout mice. These observations reveal novel molecular regulators of the postnatal disappearance of radial glia and the ending of embryonic neurogenesis, filling a key knowledge gap in NSC biology.

6.
J Neurosci ; 42(45): 8542-8555, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36198499

ABSTRACT

The oligodendrocyte (OL) lineage transcription factor Olig2 is expressed throughout oligodendroglial development and is essential for oligodendroglial progenitor specification and differentiation. It was previously reported that deletion of Olig2 enhanced the maturation and myelination of immature OLs and accelerated the remyelination process. However, by analyzing multiple Olig2 conditional KO mouse lines (male and female), we conclude that Olig2 has the opposite effect and is required for OL maturation and remyelination. We found that deletion of Olig2 in immature OLs driven by an immature OL-expressing Plp1 promoter resulted in defects in OL maturation and myelination, and did not enhance remyelination after demyelination. Similarly, Olig2 deletion during premyelinating stages in immature OLs using Mobp or Mog promoter-driven Cre lines also did not enhance OL maturation in the CNS. Further, we found that Olig2 was not required for myelin maintenance in mature OLs but was critical for remyelination after lysolecithin-induced demyelinating injury. Analysis of genomic occupancy in immature and mature OLs revealed that Olig2 targets the enhancers of key myelination-related genes for OL maturation from immature OLs. Together, by leveraging multiple immature OL-expressing Cre lines, these studies indicate that Olig2 is essential for differentiation and myelination of immature OLs and myelin repair. Our findings raise fundamental questions about the previously proposed role of Olig2 in opposing OL myelination and highlight the importance of using Cre-dependent reporter(s) for lineage tracing in studying cell state progression.SIGNIFICANCE STATEMENT Identification of the regulators that promote oligodendrocyte (OL) myelination and remyelination is important for promoting myelin repair in devastating demyelinating diseases. Olig2 is expressed throughout OL lineage development. Ablation of Olig2 was reported to induce maturation, myelination, and remyelination from immature OLs. However, lineage-mapping analysis of Olig2-ablated cells was not conducted. Here, by leveraging multiple immature OL-expressing Cre lines, we observed no evidence that Olig2 ablation promotes maturation or remyelination of immature OLs. Instead, we find that Olig2 is required for immature OL maturation, myelination, and myelin repair. These data raise fundamental questions about the proposed inhibitory role of Olig2 against OL maturation and remyelination. Our findings highlight the importance of validating genetic manipulation with cell lineage tracing in studying myelination.


Subject(s)
Demyelinating Diseases , Remyelination , Animals , Female , Male , Mice , Cell Differentiation , Demyelinating Diseases/metabolism , Myelin Sheath/metabolism , Oligodendrocyte Transcription Factor 2/genetics , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/metabolism , Mice, Knockout
7.
Stem Cell Reports ; 17(10): 2220-2238, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36179695

ABSTRACT

Telencephalic organoids generated from human pluripotent stem cells (hPSCs) are a promising system for studying the distinct features of the developing human brain and the underlying causes of many neurological disorders. While organoid technology is steadily advancing, many challenges remain, including potential batch-to-batch and cell-line-to-cell-line variability, and structural inconsistency. Here, we demonstrate that a major contributor to cortical organoid quality is the way hPSCs are maintained prior to differentiation. Optimal results were achieved using particular fibroblast-feeder-supported hPSCs rather than feeder-independent cells, differences that were reflected in their transcriptomic states at the outset. Feeder-supported hPSCs displayed activation of diverse transforming growth factor ß (TGFß) superfamily signaling pathways and increased expression of genes connected to naive pluripotency. We further identified combinations of TGFß-related growth factors that are necessary and together sufficient to impart broad telencephalic organoid competency to feeder-free hPSCs and enhance the formation of well-structured brain tissues suitable for disease modeling.


Subject(s)
Organoids , Pluripotent Stem Cells , Cell Differentiation/physiology , Humans , Organoids/metabolism , Pluripotent Stem Cells/metabolism , Telencephalon/metabolism , Transforming Growth Factor beta/metabolism
8.
Cell Rep ; 40(3): 111119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858555

ABSTRACT

Restoring sensation after injury or disease requires a reproducible method for generating large quantities of bona fide somatosensory interneurons. Toward this goal, we assess the mechanisms by which dorsal spinal interneurons (dIs; dI1-dI6) can be derived from mouse embryonic stem cells (mESCs). Using two developmentally relevant growth factors, retinoic acid (RA) and bone morphogenetic protein (BMP) 4, we recapitulate the complete in vivo program of dI differentiation through a neuromesodermal intermediate. Transcriptional profiling reveals that mESC-derived dIs strikingly resemble endogenous dIs, with the correct molecular and functional signatures. We further demonstrate that RA specifies dI4-dI6 fates through a default multipotential state, while the addition of BMP4 induces dI1-dI3 fates and activates Wnt signaling to enhance progenitor proliferation. Constitutively activating Wnt signaling permits the dramatic expansion of neural progenitor cultures. These cultures retain the capacity to differentiate into diverse populations of dIs, thereby providing a method of increasing neuronal yield.


Subject(s)
Gene Expression Regulation, Developmental , Wnt Signaling Pathway , Animals , Cell Differentiation/physiology , Interneurons/metabolism , Mice , Spinal Cord/metabolism , Tretinoin/metabolism , Tretinoin/pharmacology
9.
Stem Cell Res Ther ; 13(1): 205, 2022 05 16.
Article in English | MEDLINE | ID: mdl-35578348

ABSTRACT

BACKGROUND: Muscle denervation from trauma and motor neuron disease causes disabling morbidities. A limiting step in functional recovery is the regeneration of neuromuscular junctions (NMJs) for reinnervation. Stem cells have the potential to promote these regenerative processes, but current approaches have limited success, and the optimal types of stem cells remain to be determined. Neural crest stem cells (NCSCs), as the developmental precursors of the peripheral nervous system, are uniquely advantageous, but the role of NCSCs in neuromuscular regeneration is not clear. Furthermore, a cell delivery approach that can maintain NCSC survival upon transplantation is critical. METHODS: We established a streamlined protocol to derive, isolate, and characterize functional p75+ NCSCs from human iPSCs without genome integration of reprogramming factors. To enhance survival rate upon delivery in vivo, NCSCs were centrifuged in microwell plates to form spheroids of desirable size by controlling suspension cell density. Human bone marrow mesenchymal stem cells (MSCs) were also studied for comparison. NCSC or MSC spheroids were injected into the gastrocnemius muscle with denervation injury, and the effects on NMJ formation and functional recovery were investigated. The spheroids were also co-cultured with engineered neuromuscular tissue to assess effects on NMJ formation in vitro. RESULTS: NCSCs cultured in spheroids displayed enhanced secretion of soluble factors involved in neuromuscular regeneration. Intramuscular transplantation of spheroids enabled long-term survival and retention of NCSCs, in contrast to the transplantation of single-cell suspensions. Furthermore, NCSC spheroids significantly improved functional recovery after four weeks as shown by gait analysis, electrophysiology, and the rate of NMJ innervation. MSC spheroids, on the other hand, had insignificant effect. In vitro co-culture of NCSC or MSC spheroids with engineered myotubes and motor neurons further evidenced improved innervated NMJ formation with NCSC spheroids. CONCLUSIONS: We demonstrate that stem cell type is critical for neuromuscular regeneration and that NCSCs have a distinct advantage and therapeutic potential to promote reinnervation following peripheral nerve injury. Biophysical effects of spheroidal culture, in particular, enable long-term NCSC survival following in vivo delivery. Furthermore, synthetic neuromuscular tissue, or "tissues-on-a-chip," may offer a platform to evaluate stem cells for neuromuscular regeneration.


Subject(s)
Induced Pluripotent Stem Cells , Neural Stem Cells , Denervation , Humans , Neural Crest , Neurogenesis/physiology
10.
iScience ; 24(10): 103140, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34632335

ABSTRACT

Fukuyama congenital muscular dystrophy (FCMD) is a severe, intractable genetic disease that affects the skeletal muscle, eyes, and brain and is attributed to a defect in alpha dystroglycan (αDG) O-mannosyl glycosylation. We previously established disease models of FCMD; however, they did not fully recapitulate the phenotypes observed in human patients. In this study, we generated induced pluripotent stem cells (iPSCs) from a human FCMD patient and differentiated these cells into three-dimensional brain organoids and skeletal muscle. The brain organoids successfully mimicked patient phenotypes not reliably reproduced by existing models, including decreased αDG glycosylation and abnormal radial glial (RG) fiber migration. The basic polycyclic compound Mannan-007 (Mn007) restored αDG glycosylation in the brain and muscle models tested and partially rescued the abnormal RG fiber migration observed in cortical organoids. Therefore, our study underscores the importance of αDG O-mannosyl glycans for normal RG fiber architecture and proper neuronal migration in corticogenesis.

11.
Stem Cell Reports ; 16(10): 2548-2564, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34506726

ABSTRACT

The specification of inhibitory neurons has been described for the mouse and human brain, and many studies have shown that pluripotent stem cells (PSCs) can be used to create interneurons in vitro. It is unclear whether in vitro methods to produce human interneurons generate all the subtypes found in brain, and how similar in vitro and in vivo interneurons are. We applied single-nuclei and single-cell transcriptomics to model interneuron development from human cortex and interneurons derived from PSCs. We provide a direct comparison of various in vitro interneuron derivation methods to determine the homogeneity achieved. We find that PSC-derived interneurons capture stages of development prior to mid-gestation, and represent a minority of potential subtypes found in brain. Comparison with those found in fetal or adult brain highlighted decreased expression of synapse-related genes. These analyses highlight the potential to tailor the method of generation to drive formation of particular subtypes.


Subject(s)
Interneurons/metabolism , Neural Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Transcriptome , Cell Differentiation , Cellular Reprogramming Techniques/methods , Humans , Single-Cell Analysis , Transcription Factors/metabolism
12.
Nat Neurosci ; 24(10): 1488-1500, 2021 10.
Article in English | MEDLINE | ID: mdl-34426698

ABSTRACT

Brain organoids represent a powerful tool for studying human neurological diseases, particularly those that affect brain growth and structure. However, many diseases manifest with clear evidence of physiological and network abnormality in the absence of anatomical changes, raising the question of whether organoids possess sufficient neural network complexity to model these conditions. Here, we explore the network-level functions of brain organoids using calcium sensor imaging and extracellular recording approaches that together reveal the existence of complex network dynamics reminiscent of intact brain preparations. We demonstrate highly abnormal and epileptiform-like activity in organoids derived from induced pluripotent stem cells from individuals with Rett syndrome, accompanied by transcriptomic differences revealed by single-cell analyses. We also rescue key physiological activities with an unconventional neuroregulatory drug, pifithrin-α. Together, these findings provide an essential foundation for the utilization of brain organoids to study intact and disordered human brain network formation and illustrate their utility in therapeutic discovery.


Subject(s)
Brain/physiopathology , Epilepsy/physiopathology , Neurons , Adult , Benzothiazoles/pharmacology , Brain/growth & development , Calcium Signaling , Child, Preschool , Epilepsy/diagnostic imaging , Female , Humans , Induced Pluripotent Stem Cells , Methyl-CpG-Binding Protein 2/genetics , Nerve Net/physiopathology , Neurogenesis/genetics , Neuroimaging , Rett Syndrome/diagnostic imaging , Rett Syndrome/physiopathology , Single-Cell Analysis , Synapses , Toluene/analogs & derivatives , Toluene/pharmacology , Transcriptome
13.
STAR Protoc ; 2(1): 100319, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33659900

ABSTRACT

We describe two differentiation protocols to derive sensory spinal interneurons (INs) from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs). In protocol 1, we use retinoic acid (RA) to induce pain, itch, and heat mediating dI4/dI6 interneurons, and in protocol 2, RA with bone morphogenetic protein 4 (RA+BMP4) is used to induce proprioceptive dI1s and mechanosensory dI3s in hPSC cultures. These protocols provide an important step toward developing therapies for regaining sensation in spinal cord injury patients. For complete details on the use and execution of this protocol, please refer to Gupta et al. (2018).


Subject(s)
Flow Cytometry/methods , High-Throughput Nucleotide Sequencing/methods , Interneurons/cytology , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Pluripotent Stem Cells/drug effects , Spine/cytology , Tretinoin/pharmacology
14.
Cell Rep ; 30(6): 1964-1981.e3, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32049024

ABSTRACT

The laminar architecture of the mammalian neocortex depends on the orderly generation of distinct neuronal subtypes by apical radial glia (aRG) during embryogenesis. Here, we identify critical roles for the autism risk gene Foxp1 in maintaining aRG identity and gating the temporal competency for deep-layer neurogenesis. Early in development, aRG express high levels of Foxp1 mRNA and protein, which promote self-renewing cell divisions and deep-layer neuron production. Foxp1 levels subsequently decline during the transition to superficial-layer neurogenesis. Sustained Foxp1 expression impedes this transition, preserving a population of cells with aRG identity throughout development and extending the early neurogenic period into postnatal life. FOXP1 expression is further associated with the initial formation and expansion of basal RG (bRG) during human corticogenesis and can promote the formation of cells exhibiting characteristics of bRG when misexpressed in the mouse cortex. Together, these findings reveal broad functions for Foxp1 in cortical neurogenesis.


Subject(s)
Forkhead Transcription Factors/metabolism , Neural Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Self Renewal/physiology , Humans , Mice , Neural Stem Cells/cytology
15.
Curr Top Dev Biol ; 132: 417-450, 2019.
Article in English | MEDLINE | ID: mdl-30797516

ABSTRACT

Distinct classes of neurons arise at different positions along the dorsal-ventral axis of the spinal cord leading to spinal neurons being segregated along this axis according to their physiological properties and functions. Thus, the neurons associated with motor control are generally located in, or adjacent to, the ventral horn whereas the interneurons (INs) that mediate sensory activities are present within the dorsal horn. Here, we review classic and recent studies examining the developmental mechanisms that establish the dorsal-ventral axis in the embryonic spinal cord. Intriguingly, while the cellular organization of the dorsal and ventral halves of the spinal cord looks superficially similar during early development, the underlying molecular mechanisms that establish dorsal vs ventral patterning are markedly distinct. For example, the ventral spinal cord is patterned by the actions of a single growth factor, sonic hedgehog (Shh) acting as a morphogen, i.e., concentration-dependent signal. Recent studies have shed light on the mechanisms by which the spatial and temporal gradient of Shh is transduced by cells to elicit the generation of different classes of ventral INs, and motor neurons (MNs). In contrast, the dorsal spinal cord is patterned by the action of multiple factors, most notably by members of the bone morphogenetic protein (BMP) and Wnt families. While less is known about dorsal patterning, recent studies have suggested that the BMPs do not act as morphogens to specify dorsal IN identities as previously proposed, rather each BMP has signal-specific activities. Finally, we consider the promise that elucidation of these mechanisms holds for neural repair.


Subject(s)
Body Patterning/genetics , Gene Expression Regulation, Developmental , Neurons/metabolism , Spinal Cord/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Interneurons/cytology , Interneurons/metabolism , Motor Neurons/cytology , Motor Neurons/metabolism , Neurons/cytology , Signal Transduction/genetics , Spinal Cord/cytology , Spinal Cord/embryology , Wnt Proteins/genetics , Wnt Proteins/metabolism
16.
Dev Biol ; 436(1): 5-13, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29391164

ABSTRACT

Orofacial muscles are critical for life-sustaining behaviors, such as feeding and breathing. Centuries of work by neuroanatomists and surgeons resulted in the mapping of bulbar motor neurons in the brainstem and the course of the cranial nerves that carry their axons. Despite the sophisticated understanding of the anatomy of the region, the molecular mechanisms that dictate the development and maturation of facial motor neurons remain poorly understood. This fundamental problem has been recently revisited by physiologists with novel techniques of studying the rhythmic contraction of orofacial muscles in relationship to breathing. The molecular understanding of facial motor neuron development will not only lead to the comprehension of the neural basis of facial expression but may also unlock new avenues to generate stem cell-derived replacements. This review summarizes the current understanding of molecular programs involved in facial motor neuron generation, migration, and maturation, including neural circuit assembly.


Subject(s)
Facial Nerve/metabolism , Motor Neurons/metabolism , Neurogenesis/genetics , Animals , Body Patterning/genetics , Cell Movement/genetics , Facial Nerve/embryology , Vertebrates/embryology , Vertebrates/metabolism
17.
PLoS Biol ; 16(2): e2003127, 2018 02.
Article in English | MEDLINE | ID: mdl-29389974

ABSTRACT

During tissue development, multipotent progenitors differentiate into specific cell types in characteristic spatial and temporal patterns. We addressed the mechanism linking progenitor identity and differentiation rate in the neural tube, where motor neuron (MN) progenitors differentiate more rapidly than other progenitors. Using single cell transcriptomics, we defined the transcriptional changes associated with the transition of neural progenitors into MNs. Reconstruction of gene expression dynamics from these data indicate a pivotal role for the MN determinant Olig2 just prior to MN differentiation. Olig2 represses expression of the Notch signaling pathway effectors Hes1 and Hes5. Olig2 repression of Hes5 appears to be direct, via a conserved regulatory element within the Hes5 locus that restricts expression from MN progenitors. These findings reveal a tight coupling between the regulatory networks that control patterning and neuronal differentiation and demonstrate how Olig2 acts as the developmental pacemaker coordinating the spatial and temporal pattern of MN generation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Cycle/genetics , Motor Neurons/cytology , Neurogenesis/genetics , Oligodendrocyte Transcription Factor 2/physiology , Repressor Proteins/physiology , Single-Cell Analysis , Transcription Factor HES-1/physiology , Transcriptome , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Fluorescent Dyes/metabolism , Gene Expression Regulation/physiology , Genes, Reporter , Interneurons/cytology , Mice, Transgenic , Oligodendrocyte Transcription Factor 2/genetics , Receptors, Notch/metabolism , Regulatory Sequences, Nucleic Acid , Repressor Proteins/genetics , Signal Transduction , Transcription Factor HES-1/genetics
18.
Cell Rep ; 21(2): 517-532, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29020636

ABSTRACT

The human cerebral cortex possesses distinct structural and functional features that are not found in the lower species traditionally used to model brain development and disease. Accordingly, considerable attention has been placed on the development of methods to direct pluripotent stem cells to form human brain-like structures termed organoids. However, many organoid differentiation protocols are inefficient and display marked variability in their ability to recapitulate the three-dimensional architecture and course of neurogenesis in the developing human brain. Here, we describe optimized organoid culture methods that efficiently and reliably produce cortical and basal ganglia structures similar to those in the human fetal brain in vivo. Neurons within the organoids are functional and exhibit network-like activities. We further demonstrate the utility of this organoid system for modeling the teratogenic effects of Zika virus on the developing brain and identifying more susceptibility receptors and therapeutic compounds that can mitigate its destructive actions.


Subject(s)
Anti-Retroviral Agents/pharmacology , Cerebral Cortex/cytology , Drug Evaluation, Preclinical/methods , Organoids/virology , Primary Cell Culture/methods , Zika Virus/drug effects , Cell Line , Cerebral Cortex/virology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/virology , Humans , Neurons/cytology , Neurons/metabolism , Neurons/virology , Organoids/cytology , Organoids/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/metabolism , c-Mer Tyrosine Kinase/metabolism
19.
Proc Natl Acad Sci U S A ; 114(33): 8770-8775, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28760994

ABSTRACT

Fibrils and oligomers are the aggregated protein agents of neuronal dysfunction in ALS diseases. Whereas we now know much about fibril architecture, atomic structures of disease-related oligomers have eluded determination. Here, we determine the corkscrew-like structure of a cytotoxic segment of superoxide dismutase 1 (SOD1) in its oligomeric state. Mutations that prevent formation of this structure eliminate cytotoxicity of the segment in isolation as well as cytotoxicity of the ALS-linked mutants of SOD1 in primary motor neurons and in a Danio rerio (zebrafish) model of ALS. Cytotoxicity assays suggest that toxicity is a property of soluble oligomers, and not large insoluble aggregates. Our work adds to evidence that the toxic oligomeric entities in protein aggregation diseases contain antiparallel, out-of-register ß-sheet structures and identifies a target for structure-based therapeutics in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Superoxide Dismutase-1/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Crystallography, X-Ray/methods , Mice , Motor Neurons/metabolism , Mutation/genetics , Protein Conformation, beta-Strand , Superoxide Dismutase-1/genetics
20.
Neuron ; 94(4): 790-799.e3, 2017 May 17.
Article in English | MEDLINE | ID: mdl-28434801

ABSTRACT

Netrin1 has been proposed to act from the floor plate (FP) as a long-range diffusible chemoattractant for commissural axons in the embryonic spinal cord. However, netrin1 mRNA and protein are also present in neural progenitors within the ventricular zone (VZ), raising the question of which source of netrin1 promotes ventrally directed axon growth. Here, we use genetic approaches in mice to selectively remove netrin from different regions of the spinal cord. Our analyses show that the FP is not the source of netrin1 directing axons to the ventral midline, while local VZ-supplied netrin1 is required for this step. Furthermore, rather than being present in a gradient, netrin1 protein accumulates on the pial surface adjacent to the path of commissural axon extension. Thus, netrin1 does not act as a long-range secreted chemoattractant for commissural spinal axons but instead promotes ventrally directed axon outgrowth by haptotaxis, i.e., directed growth along an adhesive surface.


Subject(s)
Axon Guidance/genetics , Axons/metabolism , Nerve Growth Factors/genetics , Neural Stem Cells/metabolism , Spinal Cord/embryology , Tumor Suppressor Proteins/genetics , Animals , Axons/ultrastructure , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Imaging, Three-Dimensional , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Microscopy, Confocal , Nerve Growth Factors/metabolism , Netrin-1 , Neurogenesis/genetics , RNA, Messenger/metabolism , Spinal Cord/ultrastructure , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...