Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Neurosci ; 23(3): 323-326, 2020 03.
Article in English | MEDLINE | ID: mdl-32066986

ABSTRACT

The presence of active neurogenic niches in adult humans is controversial. We focused attention to the human olfactory neuroepithelium, an extracranial site supplying input to the olfactory bulbs of the brain. Using single-cell RNA sequencing analyzing 28,726 cells, we identified neural stem cell and neural progenitor cell pools and neurons. Additionally, we detailed the expression of 140 olfactory receptors. These data from the olfactory neuroepithelium niche provide evidence that neuron production may continue for decades in humans.


Subject(s)
Neurogenesis/physiology , Olfactory Mucosa/innervation , Olfactory Mucosa/physiology , Single-Cell Analysis , Adult , Aging/physiology , Humans , Neural Stem Cells/physiology , Olfactory Receptor Neurons/physiology , Sequence Analysis, RNA , Smell
2.
Stem Cell Reports ; 12(6): 1354-1365, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31155504

ABSTRACT

Stem cell-based therapies have been proposed as a strategy to replace damaged tissues, especially in the nervous system. A primary sensory modality, olfaction, is impaired in 12% of the US population, but lacks treatment options. We report here the development of a novel mouse model of inducible hyposmia and demonstrate that purified tissue-specific stem cells delivered intranasally engraft to produce olfactory neurons, achieving recovery of function. Adult mice were rendered hyposmic by conditional deletion of the ciliopathy-related IFT88 gene in the olfactory sensory neuron lineage and following experimentally induced olfactory injury, received either vehicle or stem cell infusion intranasally. Engraftment-derived olfactory neurons were identified histologically, and functional improvements were measured via electrophysiology and behavioral assay. We further explored mechanisms in culture that promote expansion of engraftment-competent adult olfactory basal progenitor cells. These findings provide a basis for translational research on propagating adult tissue-specific sensory progenitor cells and testing their therapeutic potential.


Subject(s)
Ciliopathies , Neural Stem Cells , Olfaction Disorders , Olfactory Receptor Neurons , Smell , Stem Cell Transplantation , Animals , Benzilates , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/pathology , Ciliopathies/therapy , Mice, Transgenic , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/transplantation , Olfaction Disorders/genetics , Olfaction Disorders/metabolism , Olfaction Disorders/pathology , Olfaction Disorders/therapy , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
Laryngoscope Investig Otolaryngol ; 3(5): 337-344, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30410986

ABSTRACT

OBJECTIVES: To investigate epigenetic mechanisms contributing to regulation of cellular renewal and neurogenesis in adult olfactory epithelium (OE). STUDY DESIGN: Prospective basic science study. METHODS: Olfactory basal cell cultures were prepared from adult mice per established protocols. in vivo studies were performed using the mouse methimazole lesion-regeneration paradigm. Nasal tissue sections were prepared from adult mice 7 days following lesion, or from unlesioned controls. Polycomb proteins were assessed by Western blot from culture or nasal tissue lysates, and by gene expression studies from cultures. In addition, in vivo expression patterns of Polycomb proteins were examined using immunohistochemistry. Chromosome immunoprecipitation (ChIP) was performed to investigate epigenetic modifications and specific chromatin interactions for Polycomb proteins in olfactory basal cells. RESULTS: Subunits of Polycomb Repressive Complex 1 (PRC1) and Polycomb Repressive Complex 2 (PRC2) were identified in basal cell cultures and in vivo. In regenerating OE, basal progenitor cells identified by expression of the c-KIT receptor were found to coexpress the PRC2 protein EZH2. Because multiple variants of PRC1 subunits give rise to diverse PRC1 complexes serving different functions, expression of specific PRC1 variants was further examined. We identified PRC1 components including MEL18 (PCGF2) in immature neurons, and confirm BMI1 (PCGF4) expression in mature neurons. Moreover, we identified CBX8 as a neuron-specific PRC1 subunit. ChIP assays from OE cells demonstrated binding of PRC proteins to regulatory regions of specific transcription factors, consistent with PRC-mediated epigenetic silencing mechanisms active in adult OE. CONCLUSIONS: Multiple Polycomb proteins have cell type-specific expression patterns in the adult OE. Findings presented here, together with evidence from prior studies, suggest that PRC-mediated epigenetic silencing contributes to regulation of cellular renewal and tissue homeostasis in the OE. Efforts to define the mechanisms that regulate repair in the OE are essential for development of new therapeutic strategies for olfactory disorders. LEVEL OF EVIDENCE: N/A.

4.
PLoS One ; 12(11): e0187576, 2017.
Article in English | MEDLINE | ID: mdl-29107942

ABSTRACT

Despite a robust capacity for adult neurogenesis in the olfactory epithelium (OE), olfactory sensory losses are common. Identification of mechanisms regulating adult OE neurogenesis is, therefore, of interest. MicroRNAs (miRNAs) are broadly important in regulating vertebrate neurodevelopment, and are required in embryonic olfactory differentiation. We report here that a panel of miRNAs is differentially expressed by either progenitor or progeny cells in the regenerating mouse OE. Progenitor cells were purified from lesioned OE based on c-Kit expression, and miRNA expression was assayed in c-Kit (+) and c-Kit (-) cell populations. 28 miRNAs were significantly downregulated by at least 4 fold in the c-Kit (+) fraction, which marks the globose basal progenitor cell population. In addition, 10 miRNAs were upregulated in these basal cells. MiR-486, the most strongly downregulated miRNA identified, was further characterized to verify results. MiR-486 expression was confirmed in the c-Kit (-) OE layers using in situ hybridization. As a functional assay, over-expression of miR-486 in purified c-Kit (+) basal cell cultures resulted in a reduction in neurogenesis, consistent with a possible negative feedback regulatory model. Our data provide new insights regarding miRNA expression and function during adult OE neurogenesis, and identify candidate miRNAs warranting further study.


Subject(s)
MicroRNAs/genetics , Olfactory Mucosa/metabolism , Animals , Cells, Cultured , Down-Regulation , Mice , Mice, Inbred C57BL , Neurogenesis , Olfactory Mucosa/cytology , Proto-Oncogene Proteins c-kit/metabolism
5.
Development ; 143(23): 4394-4404, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27789621

ABSTRACT

Olfactory epithelium (OE) has a lifelong capacity for neurogenesis due to the presence of basal stem cells. Despite the ability to generate short-term cultures, the successful in vitro expansion of purified stem cells from adult OE has not been reported. We sought to establish expansion-competent OE stem cell cultures to facilitate further study of the mechanisms and cell populations important in OE renewal. Successful cultures were prepared using adult mouse basal cells selected for expression of c-KIT. We show that c-KIT signaling regulates self-renewal capacity and prevents neurodifferentiation in culture. Inhibition of TGFß family signaling, a known negative regulator of embryonic basal cells, is also necessary for maintenance of the proliferative, undifferentiated state in vitro Characterizing successful cultures, we identified expression of BMI1 and other Polycomb proteins not previously identified in olfactory basal cells but known to be essential for self-renewal in other stem cell populations. Inducible fate mapping demonstrates that BMI1 is expressed in vivo by multipotent OE progenitors, validating our culture model. These findings provide mechanistic insights into the renewal and potency of olfactory stem cells.


Subject(s)
Cell Self Renewal/physiology , Neurogenesis/physiology , Olfactory Mucosa/cytology , Proto-Oncogene Proteins c-kit/metabolism , Stem Cells/cytology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cell Differentiation/physiology , Cell Lineage , Cell Proliferation/physiology , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction
6.
Dev Neurobiol ; 76(3): 241-51, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26016700

ABSTRACT

Olfactory tissue undergoes lifelong renewal, due to the presence of basal neural stem cells. Multiple categories of globose basal stem cells have been identified, expressing markers such as Lgr5, Ascl1, GBC-2, and c-Kit. The differentiation potential of individual globose cells has remained unclear. Here, we utilized Cre/loxP lineage tracing with a multicolor reporter system to define c-Kit+ cell contributions at clonal resolution. We determined that reporter expression permitted identification of c-Kit derived progeny with fine cellular detail, and that clones were found to be comprised by neurons only, microvillar cells only, microvillar cells and neurons, or gland/duct cells. Quantification of reporter-labeled cells indicated that c-Kit+ cells behave as transit amplifying or immediate precursors, although we also found evidence for longer-term c-Kit+ cell contributions. Our results from the application of multicolor fate mapping delineate the clonal contributions of c-Kit+ cells to olfactory epithelial renewal, and provide novel insight into tissue maintenance of an adult neuroepithelium.


Subject(s)
Cell Differentiation/physiology , Cell Lineage , Neural Stem Cells/cytology , Olfactory Mucosa/cytology , Olfactory Receptor Neurons/cytology , Animals , Image Processing, Computer-Assisted , Immunohistochemistry , Mice , Mice, Mutant Strains , Proto-Oncogene Proteins c-kit/biosynthesis
7.
J Comp Neurol ; 523(1): 15-31, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25044230

ABSTRACT

The olfactory epithelium houses chemosensory neurons, which transmit odor information from the nose to the brain. In adult mammals, the olfactory epithelium is a uniquely robust neuroproliferative zone, with the ability to replenish its neuronal and non-neuronal populations due to the presence of germinal basal cells. The stem and progenitor cells of these germinal layers, and their regulatory mechanisms, remain incompletely defined. Here we show that progenitor cells expressing c-Kit, a receptor tyrosine kinase marking stem cells in a variety of embryonic tissues, are required for maintenance of the adult neuroepithelium. Mouse genetic fate-mapping analyses show that embryonically, a c-Kit(+) population contributes to olfactory neurogenesis. In adults under conditions of normal turnover, there is relatively sparse c-Kit(+) progenitor cell (ckPC) activity. However, after experimentally induced neuroepithelial injury, ckPCs are activated such that they reconstitute the neuronal population. There are also occasional non-neuronal cells found to arise from ckPCs. Moreover, the selective depletion of the ckPC population, utilizing temporally controlled targeted diphtheria toxin A expression, results in failure of neurogenesis after experimental injury. Analysis of this model indicates that most ckPCs reside among the globose basal cell populations and act downstream of horizontal basal cells, which can serve as stem cells. Identification of the requirement for olfactory c-Kit-expressing progenitors in olfactory maintenance provides new insight into the mechanisms involved in adult olfactory neurogenesis. Additionally, we define an important and previously unrecognized site of adult c-Kit activity.


Subject(s)
Nerve Regeneration/physiology , Neural Stem Cells/physiology , Olfactory Mucosa/physiology , Olfactory Receptor Neurons/physiology , Proto-Oncogene Proteins c-kit/metabolism , Animals , Blotting, Western , Cell Count , Dermoscopy , Immunohistochemistry , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Neurogenesis/physiology , Olfactory Mucosa/growth & development , Olfactory Mucosa/injuries , Proto-Oncogene Proteins c-kit/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...