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.
FASEB J ; 31(1): 256-265, 2017 01.
Article in English | MEDLINE | ID: mdl-27825105

ABSTRACT

Corneal avascularity is critical for achieving transparency necessary for proper transmission of light to the lens and visual acuity. Although much is known about angiogenesis and angiostasis, the precise regulation of these processes in the cornea is unclear. MicroRNA (miR)-184, the most abundant corneal epithelial miRNA, has been suggested to function in corneal angiostasis by altering VEGF signaling; however, the mechanism(s) underlying this regulation have not been addressed. Using a combination of in vitro and in vivo assays to evaluate angiogenesis, we demonstrated that human limbal epithelial keratinocytes (HLEKs) engineered to overexpress miR-184 secreted lower amounts of angiogenic mitogens. Human dermal microvascular cells exposed to conditioned medium from miR-184-overexpressing HLEKs were less proliferative and failed to seal linear scratch wounds. The in vivo Matrigel plug assay showed that conditioned medium from miR-184-expressing HLEKs elicited a lesser degree of neovascularization compared with controls. We found that miR-184 directly targets and represses the proangiogenic factors, friend of Gata 2 (FOG2), platelet-derived growth factor (PDGF)-ß, and phosphatidic acid phosphatase 2b (PPAP2B). FOG2 regulates VEGF expression, whereas PDGF-ß and PPAP2B regulate Akt activity. By attenuating both VEGF and Akt signaling, miR-184 acts as a broad-spectrum negative regulator of corneal angiogenesis.-Park, J. K., Peng, H., Yang, W., Katsnelson, J., Volpert, O., Lavker, R. M. miR-184 exhibits angiostatic properties via regulation of Akt and VEGF signaling pathways.


Subject(s)
Gene Expression Regulation/physiology , Keratinocytes/metabolism , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inducing Agents , Animals , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/genetics
2.
J Cell Biol ; 215(5): 667-685, 2016 Dec 05.
Article in English | MEDLINE | ID: mdl-27872138

ABSTRACT

Macropinocytosis, by which cells ingest large amounts of fluid, and autophagy, the lysosome-based catabolic process, involve vesicular biogenesis (early stage) and turnover (end stage). Much is known about early-stage events; however, our understanding of how the end stages of these processes are governed is incomplete. Here we demonstrate that the microRNA-103/107(miR-103/107) family, which is preferentially expressed in the stem cell-enriched limbal epithelium, coordinately regulates aspects of both these activities. Loss of miR-103/107 causes dysregulation of macropinocytosis with the formation of large vacuoles, primarily through up-regulation of Src, Ras, and Ankfy1. Vacuole accumulation is not a malfunction of early-stage autophagy; rather, miR-103/107 ensure proper end-stage autophagy by regulating diacylglycerol/protein kinase C and cyclin-dependent kinase 5 signaling, which enables dynamin to function in vacuole clearance. Our findings unveil a key biological function for miR-103/107 in coordinately suppressing macropinocytosis and preserving end-stage autophagy, thereby contributing to maintenance of a stem cell-enriched epithelium.


Subject(s)
Autophagy , MicroRNAs/metabolism , Pinocytosis , Animals , Biomarkers/metabolism , Cell Proliferation , Diglycerides/metabolism , Dynamins/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/ultrastructure , Lysosomes/metabolism , Membrane Proteins , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Phosphate-Binding Proteins , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism , Vacuoles/metabolism , Vacuoles/ultrastructure , rab GTP-Binding Proteins/metabolism
3.
PLoS One ; 10(8): e0134853, 2015.
Article in English | MEDLINE | ID: mdl-26248284

ABSTRACT

MicroRNAs are critical regulators of stem cell behavior. The miR-103/107 family is preferentially expressed in the stem cell-enriched corneal limbal epithelium and plays an important role in coordinating several intrinsic characteristics of limbal epithelial stem cells. To elucidate further the mechanisms by which miRs-103/107 function in regulating limbal epithelial stem cells, we investigate the global effects of miRs-103/107 on gene expression in an unbiased manner. Using antagomirs-103/107, we knocked down endogenous miRs-103/107 in keratinocytes and conducted an mRNA profiling study. We show that miRs-103/107 target mitogen-activated protein kinase kinase kinase 7 (MAP3K7) and thereby negatively regulate the p38/AP-1 pathway, which directs epithelial cells towards a differentiated state. Pharmacological inhibition of p38 increases holoclone colony formation, a measure of proliferative capacity. This suggests that the negative regulation of p38 by miRs-103/107 contributes to enhanced proliferative capacity, which is a hallmark of stem cells. Since miRs-103/107 also promote increased holoclone colony formation by regulating JNK activation through non-canonical Wnt signaling, we believe that this microRNA family preserves "stemness" by mediating the crosstalk between the Wnt/JNK and MAP3K7/p38/AP-1 pathways.


Subject(s)
Keratinocytes/cytology , MicroRNAs/metabolism , Stem Cells/cytology , Animals , Base Sequence , Cell Proliferation , Cells, Cultured , Female , Gene Regulatory Networks , Humans , Keratinocytes/metabolism , Limbus Corneae/cytology , MAP Kinase Kinase Kinases/chemistry , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred BALB C , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oligonucleotides, Antisense/metabolism , Sequence Alignment , Signal Transduction , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Stem Cells ; 33(5): 1642-56, 2015 May.
Article in English | MEDLINE | ID: mdl-25639731

ABSTRACT

The stem cell niche is thought to affect cell cycle quiescence, proliferative capacity, and communication between stem cells and their neighbors. How these activities are controlled is not completely understood. Here we define a microRNA family (miRs-103/107) preferentially expressed in the stem cell-enriched limbal epithelium that regulates and integrates these stem cell characteristics. miRs-103/107 target the ribosomal kinase p90RSK2, thereby arresting cells in G0/G1 and contributing to a slow-cycling phenotype. Furthermore, miRs-103/107 increase the proliferative capacity of keratinocytes by targeting Wnt3a, which enhances Sox9 and YAP1 levels and thus promotes a stem cell phenotype. This miRNA family also regulates keratinocyte cell-cell communication by targeting: (a) the scaffolding protein NEDD9, preserving E-cadherin-mediated cell adhesion; and (b) the tyrosine phosphatase PTPRM, which negatively regulates connexin 43-based gap junctions. We propose that such regulation of cell communication and adhesion molecules maintains the integrity of the stem cell niche ultimately preserving self-renewal, a hallmark of epithelial stem cells.


Subject(s)
Epithelial Cells/metabolism , MicroRNAs/metabolism , Stem Cells/metabolism , Animals , Cadherins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Gap Junctions/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Limbus Corneae/cytology , Male , Mice, Inbred BALB C , MicroRNAs/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Stem Cells/drug effects , Wnt3A Protein/pharmacology
5.
Invest Ophthalmol Vis Sci ; 54(4): 2781-6, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23548624

ABSTRACT

PURPOSE: Corneal epithelial cells have large stores of glycogen, which serve as their primary energy source. Recently, we demonstrated that factor-inhibiting hypoxia-inducible factor 1 (FIH-1) diminished glycogen stores in vitro and in vivo, working through the Akt/Glycogen Synthase Kinase (GSK)-3ß pathway. In this study we investigated the relationship between FIH-1 and c-kit as it pertains to limbal and corneal epithelial glycogen stores. METHODS: Limbal and corneal epithelia from wild-type FIH-1(-/-) and Kit(W/Wv) mice were stained with periodic acid Schiff (PAS) to detect glycogen. RNA samples prepared from laser-capture microdissected populations of limbal epithelium were subjected to real-time quantitative PCR to determine c-kit ligand expression. Submerged cultures of primary human corneal epithelial keratinocytes (HCEKs) transduced with FIH-1 were treated with c-kit ligand to establish further a FIH-1/c-kit interaction via Western analysis. Akt phosphorylation was assessed by Western blotting. RESULTS: The limbal epithelial cells of FIH-1 null mice had an increase in glycogen levels as well as increased c-kit ligand mRNA compared with wild-type controls. Consistent with a FIH-1/c-kit association, the diminished Akt signaling observed in FIH-1-overexpressing HCEKs could be restored by the addition of c-kit ligand. Interestingly, Akt signaling and glycogen content of the corneal epithelium were significantly decreased in c-kit mutant mice. CONCLUSIONS: c-Kit signaling has been shown to affect glucose metabolism via the Akt/GSK-3ß pathway. An inverse relationship between FIH-1 and c-kit signaling pathways accounts, in part, for differences in glycogen content between corneal and limbal epithelial cells.


Subject(s)
Epithelium, Corneal/metabolism , Glycogen/metabolism , Mixed Function Oxygenases/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Cells, Cultured , Energy Metabolism , Epithelium, Corneal/drug effects , Female , Gene Transfer Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Limbus Corneae/cytology , Male , Mice , Mice, Transgenic , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/pharmacology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Stem Cell Factor/genetics
6.
Proc Natl Acad Sci U S A ; 109(35): 14030-4, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22891326

ABSTRACT

Notch plays a critical role in the transition from proliferation to differentiation in the epidermis and corneal epithelium. Furthermore, aberrant Notch signaling is a feature of diseases like psoriasis, eczema, nonmelanoma skin cancer, and melanoma where differentiation and proliferation are impaired. Whereas much is known about the downstream events following Notch signaling, factors responsible for negatively regulating Notch receptor signaling after ligand activation are incompletely understood. Notch can undergo hydroxylation by factor-inhibiting hypoxia-inducible factor 1 (FIH-1); however, the biological significance of this phenomenon is unclear. Here we show that FIH-1 expression is up-regulated in diseased epidermis and corneal epithelium. Elevating FIH-1 levels in primary human epidermal keratinocytes (HEKs) and human corneal epithelial keratinocytes (HCEKs) impairs differentiation in submerged cultures and in a "three-dimensional" organotypic raft model of human epidermis, in part, via a coordinate decrease in Notch signaling. Knockdown of FIH-1 enhances keratinocyte differentiation. Loss of FIH-1 in vivo increased Notch activity in the limbal epithelium, resulting in a more differentiated phenotype. microRNA-31 (miR-31) is an endogenous negative regulator of FIH-1 expression that results in keratinocyte differentiation, mediated by Notch activation. Ectopically expressing miR-31 in an undifferentiated corneal epithelial cell line promotes differentiation and recapitulates a corneal epithelium in a three-dimensional raft culture model. Our results define a previously unknown mechanism for keratinocyte fate decisions where Notch signaling potential is, in part, controlled through a miR-31/FIH-1 nexus.


Subject(s)
Keratinocytes/cytology , Keratinocytes/physiology , MicroRNAs/metabolism , Mixed Function Oxygenases/metabolism , Psoriasis/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Line, Transformed , Epidermal Cells , Epidermis/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Epithelium, Corneal/cytology , Epithelium, Corneal/physiology , Female , Humans , Mice , Mice, Transgenic , Mixed Function Oxygenases/genetics , Organ Culture Techniques , Phenotype , Psoriasis/genetics , Psoriasis/pathology , Receptors, Notch/metabolism , Repressor Proteins/genetics , Signal Transduction/physiology
7.
FASEB J ; 26(8): 3140-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22532441

ABSTRACT

Corneal epithelium relies on abundant glycogen stores as its primary energy source. MicroRNA-31 (miR-31), a corneal epithelial-preferred miRNA, negatively regulates factor inhibiting hypoxia-inducible factor-1 (FIH-1). Since HIF-1α is involved in anaerobic energy production, we investigated the role that miR-31 and FIH-1 play in regulating corneal epithelial glycogen. We used antagomirs (antago) to reduce the level of miR-31 in primary human corneal epithelial keratinocytes (HCEKs), and a miR-31-resistant FIH-1 to increase FIH-1 levels. Antago-31 raised FIH-1 levels and significantly reduced glycogen stores in HCEKs compared to irrelevant-antago treatment. Similarly, HCEKs retrovirally transduced with a miR-31-resistant FIH-1 had markedly reduced glycogen levels compared with empty vector controls. In addition, we observed no change in a HIF-1α reporter or known genes downstream of HIF-1α indicating that the action of FIH-1 and miR-31 on glycogen is HIF-1α-independent. An enzyme-dead FIH-1 mutation failed to restore glycogen stores, indicating that FIH-1 negatively regulates glycogen in a hydroxylase-independent manner. FIH-1 overexpression in HCEKs decreased AKT signaling, activated GSK-3ß, and inactivated glycogen synthase. Treatment of FIH-1-transduced HCEKs with either a myristolated Akt or a GSK-3ß inhibitor restored glycogen stores, confirming the direct involvement of Akt/GSK-3ß signaling. Silencing FIH-1 in HCEKs reversed the observed changes in Akt-signaling. Glycogen regulation in a HIF-1α-independent manner is a novel function for FIH-1 and provides new insight into how the corneal epithelium regulates its energy requirements.


Subject(s)
Epithelium, Corneal/metabolism , Glycogen/metabolism , Keratinocytes/metabolism , MicroRNAs/physiology , Mixed Function Oxygenases/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Animals , Cells, Cultured , Epithelium, Corneal/drug effects , Female , Glycogen Synthase/antagonists & inhibitors , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Keratinocytes/drug effects , Mice , Mixed Function Oxygenases/metabolism , Oligoribonucleotides/pharmacology , Proto-Oncogene Proteins c-akt/physiology , Repressor Proteins/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...