Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 19(5): 654-672, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38579710

ABSTRACT

Here, we used single-cell RNA sequencing (scRNA-seq), single-cell ATAC sequencing (scATAC-seq), and single-cell spatial transcriptomics to characterize murine cortical OPCs throughout postnatal life. During development, we identified two groups of differentially localized PDGFRα+ OPCs that are transcriptionally and epigenetically distinct. One group (active, or actOPCs) is metabolically active and enriched in white matter. The second (homeostatic, or hOPCs) is less active, enriched in gray matter, and predicted to derive from actOPCs. In adulthood, these two groups are transcriptionally but not epigenetically distinct, and relative to developing OPCs are less active metabolically and have less open chromatin. When adult oligodendrogenesis is enhanced during experimentally induced remyelination, adult OPCs do not reacquire a developmental open chromatin state, and the oligodendrogenesis trajectory is distinct from that seen neonatally. These data suggest that there are two OPC groups subserving distinct postnatal functions and that neonatal and adult OPC-mediated oligodendrogenesis are fundamentally different.


Subject(s)
Oligodendrocyte Precursor Cells , Single-Cell Analysis , Animals , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/cytology , Mice , Cell Differentiation/genetics , Oligodendroglia/metabolism , Oligodendroglia/cytology , Epigenesis, Genetic , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Transcriptome , Gene Expression Regulation, Developmental , Mice, Inbred C57BL , White Matter/metabolism , White Matter/cytology
2.
Cell Rep ; 42(3): 112242, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36924490

ABSTRACT

Here, we ask how developing precursors maintain the balance between cell genesis for tissue growth and establishment of adult stem cell pools, focusing on postnatal forebrain neural precursor cells (NPCs). We show that these NPCs are transcriptionally primed to differentiate and that the primed mRNAs are associated with the translational repressor 4E-T. 4E-T also broadly associates with other NPC mRNAs encoding transcriptional regulators, and these are preferentially depleted from ribosomes, consistent with repression. By contrast, a second translational regulator, Cpeb4, associates with diverse target mRNAs that are largely ribosome associated. The 4E-T-dependent mRNA association is functionally important because 4E-T knockdown or conditional knockout derepresses proneurogenic mRNA translation and perturbs maintenance versus differentiation of early postnatal NPCs in culture and in vivo. Thus, early postnatal NPCs are primed to differentiate, and 4E-T regulates the balance between cell genesis and stem cell expansion by sequestering and repressing mRNAs encoding transcriptional regulators.


Subject(s)
Neural Stem Cells , Cell Differentiation/physiology , Neural Stem Cells/metabolism , Neurons/metabolism , Processing Bodies , Protein Biosynthesis , Repressor Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Nucleocytoplasmic Transport Proteins/metabolism
3.
Cell Rep ; 41(12): 111853, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36543145

ABSTRACT

Here, we ask why the nail base is essential for mammalian digit tip regeneration, focusing on the inductive nail mesenchyme. We identify a transcriptional signature for these cells that includes Lmx1b and show that the Lmx1b-expressing nail mesenchyme is essential for blastema formation. We use a combination of Lmx1bCreERT2-based lineage-tracing and single-cell transcriptional analyses to show that the nail mesenchyme contributes cells for two pro-regenerative mechanisms. One group of cells maintains their identity and regenerates the new nail mesenchyme. A second group contributes specifically to the dorsal blastema, loses their nail mesenchyme phenotype, acquires a blastema transcriptional state that is highly similar to blastema cells of other origins, and ultimately contributes to regeneration of the dorsal but not ventral dermis and bone. Thus, the regenerative necessity for an intact nail base is explained, at least in part, by a requirement for the inductive nail mesenchyme.


Subject(s)
Mesenchymal Stem Cells , Animals , Bone and Bones , Cells, Cultured , Extremities , Mammals
4.
Article in English | MEDLINE | ID: mdl-35667791

ABSTRACT

Animals such as amphibians have an incredible capacity for regeneration with some being able to regrow their tail or appendages. Although some mammalian tissues like the skin and bones can repair following injury, there are only a few examples of true multilineage regeneration, including the distal portion of the digit tip. In both amphibians and mammals, however, to achieve successful repair or regeneration, it is now appreciated that intact nerve innervation is a necessity. Here, we review the current state of literature and discuss recent advances that identify axon-derived signals, Schwann cells, and nerve-derived mesenchymal cells as direct and indirect supporters of adult tissue homeostasis and repair. We posit that understanding how nerves positively influence repair and regeneration could lead to targeted regenerative medicine strategies to enhance tissue repair in humans.

5.
Stem Cell Reports ; 17(2): 259-275, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35063124

ABSTRACT

Senescent cells are responsible, in part, for tissue decline during aging. Here, we focused on CNS neural precursor cells (NPCs) to ask if this is because senescent cells in stem cell niches impair precursor-mediated tissue maintenance. We demonstrate an aging-dependent accumulation of senescent cells, largely senescent NPCs, within the hippocampal stem cell niche coincident with declining adult neurogenesis. Pharmacological ablation of senescent cells via acute systemic administration of the senolytic drug ABT-263 (Navitoclax) caused a rapid increase in NPC proliferation and neurogenesis. Genetic ablation of senescent cells similarly activated hippocampal NPCs. This acute burst of neurogenesis had long-term effects in middle-aged mice. One month post-ABT-263, adult-born hippocampal neuron numbers increased and hippocampus-dependent spatial memory was enhanced. These data support a model where senescent niche cells negatively influence neighboring non-senescent NPCs during aging, and ablation of these senescent cells partially restores neurogenesis and hippocampus-dependent cognition.


Subject(s)
Cellular Senescence/physiology , Neural Stem Cells/metabolism , Stem Cell Niche/physiology , Aging , Aniline Compounds/pharmacology , Animals , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Female , Hippocampus/cytology , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neurogenesis/drug effects , Spatial Memory/drug effects , Sulfonamides/pharmacology
6.
eNeuro ; 9(1)2022.
Article in English | MEDLINE | ID: mdl-35027446

ABSTRACT

Adult neural stem cells (NSCs) reside in two distinct niches in the mammalian brain, the ventricular-subventricular zone (V-SVZ) of the forebrain lateral ventricles and the subgranular zone (SGZ) of the hippocampal dentate gyrus. They are thought to be molecularly distinct since V-SVZ NSCs produce inhibitory olfactory bulb (OB) interneurons and SGZ NSCs excitatory dentate granule neurons. Here, we have asked whether this is so by directly comparing V-SVZ and SGZ NSCs from embryogenesis to adulthood using single-cell transcriptional data. We show that the embryonic radial glial precursor (RP) parents of these two NSC populations are very similar, but differentially express a small cohort of genes involved in glutamatergic versus GABAergic neurogenesis. These different RPs then undergo a similar gradual transition to a dormant adult NSC state over the first three postnatal weeks. This dormancy state involves transcriptional shutdown of genes that maintain an active, proliferative, prodifferentiation state and induction of genes involved in sensing and regulating their niche environment. Moreover, when reactivated to generate adult-born progeny, both populations reacquire a development-like state and re-express proneurogenic genes. Thus, V-SVZ and SGZ NSCs share a common transcriptional state throughout their lifespans and transition into and out of dormancy via similar trajectories.


Subject(s)
Neural Stem Cells , Adult , Animals , Dentate Gyrus , Embryonic Development , Humans , Lateral Ventricles , Mammals , Neurogenesis/physiology , Prosencephalon
7.
Curr Opin Genet Dev ; 70: 1-6, 2021 10.
Article in English | MEDLINE | ID: mdl-34044269

ABSTRACT

In mammals, multi-tissue regeneration is largely restricted to the distal portion of the digit tip and involves the formation of a blastema, a transient, proliferating cell mass that reforms the diverse tissues of the digit. Historically little was known about the mammalian blastema but with recent advances in single cell transcriptomic approaches and genetic lineage tracing, a more precise understanding of this critical structure has begun to emerge. In this review we summarise the cellular mechanisms underlying adult mammalian digit tip regeneration. We posit that understanding how some mammals naturally regenerate complex tissues will lead to strategies for enhancing regenerative abilities in humans.


Subject(s)
Regeneration/physiology , Toes/physiology , Animals , Cell Differentiation , Mice , Stem Cells/physiology , Wound Healing/physiology
8.
Cell Rep ; 33(2): 108257, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053360

ABSTRACT

Here, we ask how neural stem cells (NSCs) transition in the developing neocortex from a rapidly to a slowly proliferating state, a process required to maintain lifelong stem cell pools. We identify LRIG1, known to regulate receptor tyrosine kinase signaling in other cell types, as a negative regulator of cortical NSC proliferation. LRIG1 is expressed in murine cortical NSCs as they start to proliferate more slowly during embryogenesis and then peaks postnatally when they transition to give rise to a portion of adult NSCs. Constitutive or acute loss of Lrig1 in NSCs over this developmental time frame causes stem cell expansion due to increased proliferation. LRIG1 controls NSC proliferation by associating with and negatively regulating the epidermal growth factor receptor (EGFR). These data support a model in which LRIG1 dampens the stem cell response to EGFR ligands within the cortical environment to slow their proliferation as they transition to postnatal adult NSCs.


Subject(s)
ErbB Receptors/metabolism , Membrane Glycoproteins/metabolism , Neocortex/cytology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Signal Transduction , Animals , Animals, Newborn , Cell Proliferation , Cell Self Renewal , Embryo, Mammalian/cytology , Embryonic Development , Mice , Mice, Knockout , Neurogenesis
9.
Open Biol ; 10(9): 200194, 2020 09.
Article in English | MEDLINE | ID: mdl-32993414

ABSTRACT

Digit tip regeneration is one of the few examples of true multi-tissue regeneration in an adult mammal. The key step in this process is the formation of the blastema, a transient proliferating cell mass that generates the different cell types of the digit to replicate the original structure. Failure to form the blastema results in a lack of regeneration and has been postulated to be the reason why mammalian limbs cannot regrow following amputation. Understanding how the blastema forms and functions will help us to determine what is required for mammalian regeneration to occur and will provide insights into potential therapies for mammalian tissue regeneration and repair. This review summarizes the cellular and molecular mechanisms that influence murine blastema formation and govern digit tip regeneration.


Subject(s)
Cell Differentiation , Fingers , Regeneration , Toes , Animals , Biomarkers , Cell Differentiation/genetics , Fingers/anatomy & histology , Fingers/physiology , Gene Expression Regulation , Humans , Mammals , Signal Transduction , Toes/anatomy & histology , Toes/physiology
10.
Cell Rep ; 32(6): 108022, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32783944

ABSTRACT

The transitions from developing to adult quiescent and activated neural stem cells (NSCs) are not well understood. Here, we use single-cell transcriptional profiling and lineage tracing to characterize these transitions in the murine forebrain. We show that the two forebrain NSC parental populations, embryonic cortex and ganglionic eminence radial precursors (RPs), are highly similar even though they make glutamatergic versus gabaergic neurons. Both RP populations progress linearly to transition from a highly active embryonic to a dormant adult stem cell state that still shares many similarities with embryonic RPs. When adult NSCs of either embryonic origin become reactivated to make gabaergic neurons, they acquire a developing ganglionic eminence RP-like identity. Thus, transitions from embryonic RPs to adult NSCs and back to neuronal progenitors do not involve fundamental changes in cell identity, but rather reflect conversions between activated and dormant NSC states that may be determined by the niche environment.


Subject(s)
Neural Stem Cells/metabolism , Neurogenesis/genetics , Prosencephalon/physiopathology , Animals , Cell Differentiation , Mice
11.
Nat Med ; 26(8): 1285-1294, 2020 08.
Article in English | MEDLINE | ID: mdl-32719487

ABSTRACT

We asked whether pharmacological stimulation of endogenous neural precursor cells (NPCs) may promote cognitive recovery and brain repair, focusing on the drug metformin, in parallel rodent and human studies of radiation injury. In the rodent cranial radiation model, we found that metformin enhanced the recovery of NPCs in the dentate gyrus, with sex-dependent effects on neurogenesis and cognition. A pilot double-blind, placebo-controlled crossover trial was conducted (ClinicalTrials.gov, NCT02040376) in survivors of pediatric brain tumors who had been treated with cranial radiation. Safety, feasibility, cognitive tests and MRI measures of white matter and the hippocampus were evaluated as endpoints. Twenty-four participants consented and were randomly assigned to complete 12-week cycles of metformin (A) and placebo (B) in either an AB or BA sequence with a 10-week washout period at crossover. Blood draws were conducted to monitor safety. Feasibility was assessed as recruitment rate, medication adherence and procedural adherence. Linear mixed modeling was used to examine cognitive and MRI outcomes as a function of cycle, sequence and treatment. We found no clinically relevant safety concerns and no serious adverse events associated with metformin. Sequence effects were observed for all cognitive outcomes in our linear mixed models. For the subset of participants with complete data in cycle 1, metformin was associated with better performance than placebo on tests of declarative and working memory. We present evidence that a clinical trial examining the effects of metformin on cognition and brain structure is feasible in long-term survivors of pediatric brain tumors and that metformin is safe to use and tolerable in this population. This pilot trial was not intended to test the efficacy of metformin for cognitive recovery and brain growth, but the preliminary results are encouraging and warrant further investigation in a large multicenter phase 3 trial.


Subject(s)
Brain Neoplasms/complications , Cognitive Dysfunction/drug therapy , Metformin/administration & dosage , Pediatrics/trends , Adolescent , Adult , Brain/diagnostic imaging , Brain/drug effects , Brain/pathology , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cancer Survivors , Child , Child, Preschool , Cognition/drug effects , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Double-Blind Method , Female , Humans , Magnetic Resonance Imaging , Male , Metformin/adverse effects , Neurogenesis/drug effects , Pilot Projects , Treatment Outcome , Young Adult
12.
Stem Cell Reports ; 15(1): 140-155, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32559459

ABSTRACT

Cell transplantation for spinal cord injury (SCI) has largely been studied in sub-acute settings within 1-2 weeks of injury. In contrast, here we transplanted skin-derived precursors differentiated into Schwann cells (SKP-SCs) into the contused rat spinal cord 8 weeks post-injury (wpi). Twenty-one weeks later (29 wpi), SKP-SCs were found to have survived transplantation, integrated with host tissue, and mitigated the formation of a dense glial scar. Furthermore, transplanted SKP-SCs filled much of the lesion sites and greatly enhanced the presence of endogenous SCs, which myelinated thousands of sprouting/spared host axons in and around the injury site. In addition, SKP-SC transplantation improved locomotor outcomes and decreased pathological thickening of bladder wall. To date, functional improvements have very rarely been observed with cell transplantation beyond the sub-acute stage of injury. Hence, these findings indicate that skin-derived SCs are a promising candidate cell type for the treatment of chronic SCI.


Subject(s)
Locomotion , Schwann Cells/transplantation , Skin/pathology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy , Urinary Bladder/pathology , Animals , Axons/pathology , Chronic Disease , Female , Myelin Sheath/metabolism , Nerve Regeneration , Neuroglia/pathology , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord/physiopathology
13.
eNeuro ; 7(3)2020.
Article in English | MEDLINE | ID: mdl-32349983

ABSTRACT

Peripheral nerves provide a supportive growth environment for developing and regenerating axons and are essential for maintenance and repair of many non-neural tissues. This capacity has largely been ascribed to paracrine factors secreted by nerve-resident Schwann cells. Here, we used single-cell transcriptional profiling to identify ligands made by different injured rodent nerve cell types and have combined this with cell-surface mass spectrometry to computationally model potential paracrine interactions with peripheral neurons. These analyses show that peripheral nerves make many ligands predicted to act on peripheral and CNS neurons, including known and previously uncharacterized ligands. While Schwann cells are an important ligand source within injured nerves, more than half of the predicted ligands are made by nerve-resident mesenchymal cells, including the endoneurial cells most closely associated with peripheral axons. At least three of these mesenchymal ligands, ANGPT1, CCL11, and VEGFC, promote growth when locally applied on sympathetic axons. These data therefore identify an unexpected paracrine role for nerve mesenchymal cells and suggest that multiple cell types contribute to creating a highly pro-growth environment for peripheral axons.


Subject(s)
Nerve Regeneration , Single-Cell Analysis , Axons , Ligands , Peripheral Nerves , Schwann Cells
14.
Dev Cell ; 52(4): 509-524.e9, 2020 02 24.
Article in English | MEDLINE | ID: mdl-31902657

ABSTRACT

Here, we investigate the origin and nature of blastema cells that regenerate the adult murine digit tip. We show that Pdgfra-expressing mesenchymal cells in uninjured digits establish the regenerative blastema and are essential for regeneration. Single-cell profiling shows that the mesenchymal blastema cells are distinct from both uninjured digit and embryonic limb or digit Pdgfra-positive cells. This unique blastema state is environmentally determined; dermal fibroblasts transplanted into the regenerative, but not non-regenerative, digit express blastema-state genes and contribute to bone regeneration. Moreover, lineage tracing with single-cell profiling indicates that endogenous osteoblasts or osteocytes acquire a blastema mesenchymal transcriptional state and contribute to both dermis and bone regeneration. Thus, mammalian digit tip regeneration occurs via a distinct adult mechanism where the regenerative environment promotes acquisition of a blastema state that enables cells from tissues such as bone to contribute to the regeneration of other mesenchymal tissues such as the dermis.


Subject(s)
Cell Differentiation , Extremities/physiology , Gene Expression Regulation, Developmental , Mesenchymal Stem Cells/cytology , Receptors, Platelet-Derived Growth Factor/physiology , Regeneration , Animals , Cell Lineage , Cells, Cultured , Extremities/embryology , Extremities/injuries , Female , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Single-Cell Analysis , Transcriptome
15.
Cell Rep ; 30(1): 215-228.e5, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31914388

ABSTRACT

PTPRD is a receptor protein tyrosine phosphatase that is genetically associated with neurodevelopmental disorders. Here, we asked whether Ptprd mutations cause aberrant neural development by perturbing neurogenesis in the murine cortex. We show that loss of Ptprd causes increases in neurogenic transit-amplifying intermediate progenitor cells and cortical neurons and perturbations in neuronal localization. These effects are intrinsic to neural precursor cells since acute Ptprd knockdown causes similar perturbations. PTPRD mediates these effects by dephosphorylating receptor tyrosine kinases, including TrkB and PDGFRß, and loss of Ptprd causes the hyperactivation of TrkB and PDGFRß and their downstream MEK-ERK signaling pathway in neural precursor cells. Moreover, inhibition of aberrant TrkB or MEK activation rescues the increased neurogenesis caused by knockdown or homozygous loss of Ptprd. These results suggest that PTPRD regulates receptor tyrosine kinases to ensure appropriate numbers of intermediate progenitor cells and neurons, suggesting a mechanism for its genetic association with neurodevelopmental disorders.


Subject(s)
Neurogenesis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Alleles , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cerebral Cortex/embryology , Embryo, Mammalian/cytology , Gene Knockdown Techniques , HEK293 Cells , Humans , Matrix Attachment Region Binding Proteins/metabolism , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Phosphorylation , Receptor-Like Protein Tyrosine Phosphatases, Class 2/deficiency , Signal Transduction , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism
16.
Neurobiol Aging ; 85: 22-37, 2020 01.
Article in English | MEDLINE | ID: mdl-31734438

ABSTRACT

The cause of midbrain dopaminergic (mDA) neuron loss in sporadic Parkinson's disease (PD) is multifactorial, involving cell autonomous factors, cell-cell interactions, and the effects of environmental toxins. Early loss of neurons in the locus coeruleus (LC), the main source of ascending noradrenergic (NA) projections, is an important feature of PD and other neurodegenerative disorders. We hypothesized that NA afferents provide trophic support for vulnerable mDA neurons. We demonstrate that depriving mDA neurons of NA input increases postnatal apoptosis and decreases cell survival in young adult rodents, with relative sparing of calbindin-positive subpopulations known to be resistant to degeneration in PD. As a mechanism, we propose that the neurotrophin brain-derived neurotrophic factor (BDNF) modulates anterograde survival effects of LC inputs to mDA neurons. We demonstrate that the LC is rich in BDNF mRNA in postnatal and young adult brains. Early postnatal NA denervation reduces both BDNF protein and activation of TrkB receptors in the ventral midbrain. Furthermore, overexpression of BDNF in NA afferents in transgenic mice increases mDA neuronal survival. Finally, increasing NA activity in primary cultures of mDA neurons improves survival, an effect that is additive or synergistic in the presence of different concentrations of BDNF. Taken together, our results point to a novel mechanism whereby LC afferents couple BDNF effects and NA activity to provide anterograde trophic support for vulnerable mDA neurons. Early loss of NA activity and anterograde neurotrophin support may contribute to degeneration of vulnerable neurons in PD and other neurodegenerative disorders.


Subject(s)
Cell Survival , Dopaminergic Neurons/pathology , Mesencephalon/cytology , Norepinephrine/physiology , Parkinson Disease/etiology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Mice, Transgenic , Parkinson Disease/pathology , Rats, Sprague-Dawley
17.
Cell Death Differ ; 26(12): 2495-2512, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31551564

ABSTRACT

The mammalian neocortex underlies our perception of sensory information, performance of motor activities, and higher-order cognition. During mammalian embryogenesis, radial glial precursor cells sequentially give rise to diverse populations of excitatory cortical neurons, followed by astrocytes and oligodendrocytes. A subpopulation of these embryonic neural precursors persists into adulthood as neural stem cells, which give rise to inhibitory interneurons and glia. Although the intrinsic mechanisms instructing the genesis of these distinct progeny have been well-studied, most work to date has focused on transcriptional, epigenetic, and cell-cycle control. Recent studies, however, have shown that posttranscriptional mechanisms also regulate the cell fate choices of transcriptionally primed neural precursors during cortical development. These mechanisms are mediated primarily by RNA-binding proteins and microRNAs that coordinately regulate mRNA translation, stability, splicing, and localization. Together, these findings point to an extensive network of posttranscriptional control and provide insight into both normal cortical development and disease. They also add another layer of complexity to brain development and raise important biological questions for future investigation.


Subject(s)
Brain/physiopathology , Neural Stem Cells/metabolism , Neurons/metabolism , Animals , Humans , Mammals
18.
Cell Stem Cell ; 24(2): 240-256.e9, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30503141

ABSTRACT

Peripheral innervation plays an important role in regulating tissue repair and regeneration. Here we provide evidence that injured peripheral nerves provide a reservoir of mesenchymal precursor cells that can directly contribute to murine digit tip regeneration and skin repair. In particular, using single-cell RNA sequencing and lineage tracing, we identify transcriptionally distinct mesenchymal cell populations within the control and injured adult nerve, including neural crest-derived cells in the endoneurium with characteristics of mesenchymal precursor cells. Culture and transplantation studies show that these nerve-derived mesenchymal cells have the potential to differentiate into non-nerve lineages. Moreover, following digit tip amputation, neural crest-derived nerve mesenchymal cells contribute to the regenerative blastema and, ultimately, to the regenerated bone. Similarly, neural crest-derived nerve mesenchymal cells contribute to the dermis during skin wound healing. These findings support a model where peripheral nerves directly contribute mesenchymal precursor cells to promote repair and regeneration of injured mammalian tissues.


Subject(s)
Mesenchymal Stem Cells/cytology , Nerve Regeneration/physiology , Nerve Tissue/pathology , Wound Healing , Animals , Bone Regeneration , Cell Differentiation , Cell Lineage , Mice , Neural Crest/cytology , Osteogenesis , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Schwann Cells/pathology , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Transcription, Genetic , Transcriptome/genetics
19.
Stem Cell Reports ; 10(5): 1464-1480, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29628394

ABSTRACT

Circulating systemic factors can regulate adult neural stem cell (NSC) biology, but the identity of these circulating cues is still being defined. Here, we have focused on the cytokine interleukin-6 (IL-6), since increased circulating levels of IL-6 are associated with neural pathologies such as autism and bipolar disorder. We show that IL-6 promotes proliferation of post-natal murine forebrain NSCs and that, when the IL-6 receptor is inducibly knocked out in post-natal or adult neural precursors, this causes a long-term decrease in forebrain NSCs. Moreover, a transient circulating surge of IL-6 in perinatal or adult mice causes an acute increase in neural precursor proliferation followed by long-term depletion of adult NSC pools. Thus, IL-6 signaling is both necessary and sufficient for adult NSC self-renewal, and acute perturbations in circulating IL-6, as observed in many pathological situations, have long-lasting effects on the size of adult NSC pools.


Subject(s)
Adult Stem Cells/cytology , Growth and Development , Interleukin-6/pharmacology , Neural Stem Cells/cytology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Animals , Animals, Newborn , Cell Count , Cell Proliferation , Interleukin-6/blood , Interleukin-6/genetics , Interleukin-6/metabolism , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Time Factors
20.
Neuron ; 97(3): 520-537.e6, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29395907

ABSTRACT

The mechanisms instructing genesis of neuronal subtypes from mammalian neural precursors are not well understood. To address this issue, we have characterized the transcriptional landscape of radial glial precursors (RPs) in the embryonic murine cortex. We show that individual RPs express mRNA, but not protein, for transcriptional specifiers of both deep and superficial layer cortical neurons. Some of these mRNAs, including the superficial versus deep layer neuron transcriptional regulators Brn1 and Tle4, are translationally repressed by their association with the RNA-binding protein Pumilio2 (Pum2) and the 4E-T protein. Disruption of these repressive complexes in RPs mid-neurogenesis by knocking down 4E-T or Pum2 causes aberrant co-expression of deep layer neuron specification proteins in newborn superficial layer neurons. Thus, cortical RPs are transcriptionally primed to generate diverse types of neurons, and a Pum2/4E-T complex represses translation of some of these neuronal identity mRNAs to ensure appropriate temporal specification of daughter neurons.


Subject(s)
Cerebral Cortex/embryology , Ependymoglial Cells/metabolism , Gene Expression Regulation, Developmental , Neural Stem Cells/metabolism , Neurogenesis , Animals , Cerebral Cortex/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Female , Male , Mice , Nerve Tissue Proteins/metabolism , POU Domain Factors/metabolism , Primary Cell Culture , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL
...