Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 18(11): 2283-2296, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37832538

ABSTRACT

Adult neurogenesis occurs in the mammalian olfactory epithelium to maintain populations of neurons that are vulnerable to injury yet essential for olfaction. Multipotent olfactory basal stem cells are activated by damage, although mechanisms regulating lineage decisions are not understood. Using mouse lesion models, we focused on defining the role of Polycomb repressive complexes (PRCs) in olfactory neurogenesis. PRC2 has a well-established role in developing tissues, orchestrating transcriptional programs via chromatin modification. PRC2 proteins are expressed in olfactory globose basal cells (GBCs) and nascent neurons. Conditional PRC2 loss perturbs lesion-induced neuron production, accompanied by altered histone modifications and misexpression of lineage-specific transcription factors in GBCs. De-repression of Sox9 in PRC2-mutant GBCs is accompanied by increased Bowman's gland production, defining an unrecognized role for PRC2 in regulating gland versus neuron cell fate. Our findings support a model for PRC2-dependent mechanisms promoting sensory neuronal differentiation in an adult neurogenic niche.


Subject(s)
Polycomb Repressive Complex 2 , Smell , Mice , Animals , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Neurogenesis/physiology , Cell Differentiation/physiology , Olfactory Mucosa , Polycomb Repressive Complex 1 , Mammals/metabolism
2.
Methods Mol Biol ; 2710: 121-129, 2023.
Article in English | MEDLINE | ID: mdl-37688729

ABSTRACT

The olfactory mucosa, lining a portion of the nasal cavity, houses the primary olfactory sensory neurons responsible for odor transduction, along with supporting cell populations. Tremendous advances have come from studying the peripheral olfactory system in animal models, especially the mouse. However, acquired human olfactory disorders lack effective therapies, and many of these conditions involve pathology in the olfactory mucosa. Thus, the ability to obtain human olfactory biopsy samples from subjects with olfactory dysfunction, or controls, may be of value. Here, we describe established techniques for collecting olfactory tissue from human subjects and preparing samples for downstream assays such as immunohistochemistry, flow cytometry, single-cell RNA-sequencing, or chromatin studies.


Subject(s)
Biological Assay , Smell , Humans , Animals , Mice , Biopsy , Chromatin , Flow Cytometry
3.
Oper Tech Otolayngol Head Neck Surg ; 33(2): 141-146, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35505955

ABSTRACT

Here, we provide an overview of olfactory dysfunction associated with COVID-19. We provide background regarding the organization and function of the peripheral olfactory system. A review of the relevant literature on anosmia and parosmia due to infection with SARS-CoV-2, the virus causing COVID-19, is provided. Specific attention is focused on possible mechanisms by which the virus may interact with and damage the cell populations of peripheral olfactory system. Evidence from human studies as well as animal models is considered. Finally, we discuss current recommendations for evaluation and management of patients with persistent post-COVID olfactory dysfunction, as well as possible future research directions.

4.
Laryngoscope Investig Otolaryngol ; 5(6): 975-982, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33364385

ABSTRACT

OBJECTIVES: Mesenchymal stem cells (MSCs), classically expanded in culture from bone marrow, are of broad interest to the regenerative medicine community. Human nasal turbinate mesenchymal-like stem cell cultures have also been described, defined by an in vitro phenotype similar to bone marrow MSCs. Nonetheless, the identity in vivo of the cells that give rise to nasal MSC-like cultures remains unclear, and these cells are often suggested to be related to olfactory lineages. Here, we sought to define the in vivo phenotype of human nasal MSC-like cells. METHODS: Human turbinate tissue samples were used for RNA and immunohistochemical analysis. We also analyzed a recently published single cell RNA-sequencing dataset from adult human olfactory and respiratory mucosa samples from our lab, to focus on cell populations expressing MSC markers. Immunochemistry was performed to stain turbinate sections and nasal MSC cultures for selected markers. RESULTS: While there is no single MSC-specific gene, we identified a human nasal mucosal cell population in vivo that uniquely expressed transcripts characteristic of typical MSC cultures, including ENG (CD105), NES, and CD34, and lacked expression of other transcripts associated with surface epithelia. The expression of transcription factors such as SOX17, EBF1, and FOXP1 suggests cells in the MSC-like cluster maintain an ability to direct cell fate, consistent with the behavior of nasal MSC-like cells in vitro. SOX17 was found to be uniformly expressed by nasal MSC cultures, consistent with the in vivo data. Immunohistochemistry of human nasal tissue samples indicated that ENG, CD34, and SOX17 expression localized selectively to cells surrounding blood vessels in the lamina propria. CONCLUSION: Our findings provide evidence that the in vivo origin of nasal MSC-like cultures is likely a vascular or pericyte population, rather than cells related to the olfactory neuronal lineage. LEVEL OF EVIDENCE: NA.

5.
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
6.
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
7.
Int Forum Allergy Rhinol ; 9(9): 993-999, 2019 09.
Article in English | MEDLINE | ID: mdl-31251849

ABSTRACT

BACKGROUND: Damage to olfactory sensory neurons (OSNs), situated within the neuroepithelium of the olfactory cleft, may be associated with anosmia. Although their direct contact with the nasal airspace make OSNs vulnerable to injury and death, multiple mechanisms maintain epithelium integrity and olfactory function. We hypothesized that BMI1, a polycomb protein found to be enriched in OSNs, may function in neuroprotection. Here, we explored BMI1 function in a mouse model. METHODS: Utilizing a mouse genetic approach to delete Bmi1 selectively in mature OSNs, we investigated changes in OE homeostasis by performing immunohistochemical, biochemical, and functional assays. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR), immunostaining, and electro-olfactograms were used to compare gene expression, cell composition, and olfactory function in OSN-specific BMI1 knockout mice (n = 3 to 5) and controls. Chromatin studies were also performed to identify protein-DNA interactions between BMI1 and its target genes (n = 3). RESULTS: OSN-specific BMI1 knockout led to increased neuron death and basal cell activation. Chromatin studies suggested a mechanism of increased neurodegeneration due to de-repression of a pro-apoptosis gene, p19ARF. Despite the increased turnover, we found that olfactory neuroepithelium thickness and olfactory function remained intact. Our studies also revealed the presence of additional polycomb group proteins that may compensate for the loss of BMI1 in mature OSNs. CONCLUSION: The olfactory neuroepithelium employs multiple mechanisms to maintain epithelial homeostasis. Our findings provide evidence that in a mouse model of BMI1 deletion, the overall integrity and function of the olfactory neuroepithelium are not compromised, despite increased neuronal turnover, reflecting a remarkable reparative capacity to sustain a critical sensory system.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Epidermis/pathology , Olfaction Disorders/pathology , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/physiology , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Death/genetics , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Epigenetic Repression , Humans , Mice , Mice, Knockout , Smell/genetics
8.
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.

9.
Laryngoscope Investig Otolaryngol ; 3(1): 35-42, 2018 02.
Article in English | MEDLINE | ID: mdl-29492466

ABSTRACT

Disorders causing a loss of the sense of smell remain a therapeutic challenge. Basic research has, however, greatly expanded our knowledge of the organization and function of the olfactory system. This review describes advances in our understanding of the cellular components of the peripheral olfactory system, specifically the olfactory epithelium in the nose. The article discusses recent findings regarding the mechanisms involved in regeneration and cellular renewal from basal stem cells in the adult olfactory epithelium, considering the strategies involved in embryonic olfactory development and insights from research on other stem cell niches. In the context of clinical conditions causing anosmia, the current view of adult olfactory neurogenesis, tissue homeostasis, and failures in these processes is considered, along with current and future treatment strategies. Level of Evidence: NA.

10.
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
11.
PLoS One ; 11(7): e0159666, 2016.
Article in English | MEDLINE | ID: mdl-27428057

ABSTRACT

Age-related increases in oxidant stress (OS) play a role in regulation of estrogen receptor (ER) expression in the kidneys. In this study, we establish that in vivo 17ß-estradiol (E2) replacement can no longer upregulate glomerular ER expression by 21 months of age in female mice (anestrous). We hypothesized that advanced glycation end product (AGE) accumulation, an important source of oxidant stress, contributes to these glomerular ER expression alterations. We treated 19-month old ovariectomized female mice with pyridoxamine (Pyr), a potent AGE inhibitor, in the presence or absence of E2 replacement. Glomerular ERα mRNA expression was upregulated in mice treated with both Pyr and E2 replacement and TGFß mRNA expression decreased compared to controls. Histological sections of kidneys demonstrated decreased type IV collagen deposition in mice receiving Pyr and E2 compared to placebo control mice. In addition, anti-AGE defenses Sirtuin1 (SIRT1) and advanced glycation receptor 1 (AGER1) were also upregulated in glomeruli following treatment with Pyr and E2. Mesangial cells isolated from all groups of mice demonstrated similar ERα, SIRT1, and AGER1 expression changes to those of whole glomeruli. To demonstrate that AGE accumulation contributes to the observed age-related changes in the glomeruli of aged female mice, we treated mesangial cells from young female mice with AGE-BSA and found similar downregulation of ERα, SIRT1, and AGER1 expression. These results suggest that inhibition of intracellular AGE accumulation with pyridoxamine may protect glomeruli against age-related oxidant stress by preventing an increase of TGFß production and by regulation of the estrogen receptor.


Subject(s)
Aging/metabolism , Antioxidants/pharmacology , Estrogen Receptor alpha/genetics , Glycation End Products, Advanced/antagonists & inhibitors , Kidney Glomerulus/drug effects , Pyridoxamine/pharmacology , Aging/genetics , Animals , Collagen Type IV/genetics , Collagen Type IV/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation , Glycation End Products, Advanced/genetics , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/pharmacology , Hormone Replacement Therapy , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , Ovariectomy , Oxidative Stress , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Serum Albumin, Bovine/antagonists & inhibitors , Serum Albumin, Bovine/pharmacology , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
Transl Res ; 166(6): 554-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26432923

ABSTRACT

The observation that pulmonary inflammatory lesions and bleomycin (BLM)-induced pulmonary fibrosis spontaneously resolve in young mice, whereas remaining irreversible in aged mice suggests that impairment of pulmonary regeneration and repair is associated with aging. Because mesenchymal stem cells (MSCs) may promote repair after injury, we postulated that differences in MSCs from aged mice may underlie postinjury fibrosis in aging. The potential for young-donor MSCs to inhibit BLM-induced pulmonary fibrosis in aged male mice (>22 months) has not been studied. Adipose-derived MSCs (ASCs) from young (4 months) and old (22 months) male mice were infused 1 day after intratracheal BLM administration. At 21-day sacrifice, aged BLM mice demonstrated lung fibrosis by Ashcroft score, collagen content, and α(v)-integrin messenger RNA (mRNA) expression. Lung tissue from aged BLM mice receiving young ASCs exhibited decreased fibrosis, matrix metalloproteinase (MMP)-2 activity, oxidative stress, and markers of apoptosis vs BLM controls. Lung mRNA expression of tumor necrosis factor-alpha was also decreased in aged BLM mice receiving young-donor ASCs vs BLM controls. In contrast, old-donor ASC treatment in aged BLM mice did not reduce fibrosis and related markers. On examination of the cells, young-donor ASCs had decreased mRNA expression of MMP-2, insulin-like growth factor (IGF) receptor, and protein kinase B (AKT) activation compared with old-donor ASCs. These results show that the BLM-induced pulmonary fibrosis in aged mice could be blocked by young-donor ASCs and that the mechanisms involve changes in collagen turnover and markers of inflammation.


Subject(s)
Adipose Tissue/cytology , Age Factors , Bleomycin/toxicity , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Pulmonary Fibrosis/therapy , Animals , Biomarkers/metabolism , Disease Models, Animal , Enzyme Activation , In Situ Nick-End Labeling , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/chemically induced
13.
Endocrinology ; 155(2): 441-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24274985

ABSTRACT

The role that estrogens play in the aging lung is poorly understood. Remodeling of the aging lung with thickening of the alveolar walls and reduction in the number of peripheral airways is well recognized. The present study was designed to address whether estrogen deficiency would affect age-associated changes in the lungs of female C57BL/6J mice. Lungs isolated from old mice (24 months old, estrogen-deficient) demonstrated decreased lung volume and decreased alveolar surface area. There was no difference in alveolar number in the lungs of old and young mice (6 months old, estrogen-replete). Estrogen replacement restored lung volume, alveolar surface area, and alveolar wall thickness to that of a young mouse. Estrogen receptor-α (ERα) protein expression increased without a change in ERß protein expression in the lung tissue isolated from old mice. In the lungs of old mice, the number of apoptotic cells was increased as well as the activation of matrix metalloproteinase-2 and ERK. Young mice had the highest serum 17ß-estradiol levels that decreased with age. Our data suggest that in the aging female mouse lung, estrogen deficiency and an increase of ERα expression lead to the development of an emphysematous phenotype. Estrogen replacement partially prevents these age-associated changes in the lung architecture by restoration of interalveolar septa. Understanding the role of estrogens in the remodeling of the lung during aging may facilitate interventions and therapies for aging-related lung disease in women.


Subject(s)
Aging/physiology , Estradiol/therapeutic use , Estrogen Replacement Therapy , Lung Diseases/drug therapy , Lung/drug effects , Aging/metabolism , Animals , Apoptosis/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Lung/metabolism , Lung/pathology , Lung Diseases/metabolism , Lung Diseases/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Ovariectomy , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...