Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Biochemistry ; 63(6): 767-776, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38439718

ABSTRACT

Interferon regulatory factor 4 (IRF4) is a crucial transcription factor that plays a vital role in lymphocyte development, including in the fate-determining steps in terminal differentiation. It is also implicated in the development of lymphoid tumors such as multiple myeloma and adult T-cell leukemia. IRF4 can form a homodimer and multiple heterocomplexes with other transcription factors such as purine-rich box1 and activator protein 1. Each protein complex binds to specific DNA sequences to regulate a distinct set of genes. However, the precise relationship among these complex formations remains unclear. Herein, we investigated the abilities of IRF4 proteins with functional mutations in the IRF-association domain and autoinhibitory region to form complexes using luciferase reporter assays. The assays allowed us to selectively assess the activity of each complex. Our results revealed that certain IRF-association domain mutants, previously known to have impaired heterocomplex formation, maintained or even enhanced homodimer activity. This discrepancy suggests that the mutated amino acid residues selectively influence homodimer activity. Conversely, a phosphomimetic serine mutation in the autoinhibitory region displayed strong activating effects in all complexes. Furthermore, we observed that partner proteins involved in heterocomplex formation could disrupt the activity of the homodimer, suggesting a potential competition between homocomplexes and heterocomplexes. Our findings provide new insights into the mechanistic function of IRF4.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factors , Base Sequence , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Mutation , Transcription Factor AP-1/metabolism , Humans , HEK293 Cells
2.
Pigment Cell Melanoma Res ; 36(1): 53-70, 2023 01.
Article in English | MEDLINE | ID: mdl-36318272

ABSTRACT

Conditional and inducible gene targeting using Cre/loxP-mediated recombination is a powerful reverse genetics approach used to study spatiotemporal gene functions in specified cell types. To enable temporal gene manipulation in the melanocyte lineage, we established a novel inducible Cre-driver mouse line by targeting an all-in-one tetracycline/doxycycline (Dox)-inducible Cre expression cassette into the Pmel locus (PmelP2A-TetON3G-TRE3G-iCre ), a gene locus preferentially expressed in pigment cells. By crossing these Cre-driver mice with a strong Cre-reporter mouse line, Gt(ROSA)26Sortm9(CAG-tdTomato)Hze , we show the effectiveness of the PmelP2A-TetON3G-TRE3G-iCre mouse line in facilitating Dox-inducible Cre/loxP recombination in a wide variety of pigment cell lineages including hair follicle melanocytes and their stem cells. Furthermore, to demonstrate proof of concept, we ablated Notch signaling postnatally in the PmelP2A-TetON3G-TRE3G-iCre mice. In agreement with the previously reported phenotype, induced ablation of Notch signaling in the melanocyte lineage resulted in premature hair graying, demonstrating the utility of the PmelP2A-TetON3G-TRE3G-iCre allele. Therefore, the PmelP2A-TetON3G-TRE3G-iCre mouse line is suitable for assessing gene functions in melanocytes using an in vivo inducible reverse genetics approach. Furthermore, we unexpectedly identified previously unrecognized PMEL-expressing cells in non-pigmentary organs in the mice, suggesting unanticipated functions of PMEL other than melanosome formation.


Subject(s)
Integrases , Melanocytes , Mice , Animals , Mice, Transgenic , Integrases/metabolism , Melanocytes/metabolism , Phenotype
3.
Life Sci Alliance ; 4(9)2021 09.
Article in English | MEDLINE | ID: mdl-34244422

ABSTRACT

Acetylcholine (ACh) signaling through activation of nicotinic and muscarinic ACh receptors regulates expression of specific genes that mediate and sustain proliferation, differentiation, and homeostasis in the intestinal crypts. This signaling plays a pivotal role in the regulation of intestinal stem cell function, but the details have not been clarified. Here, we performed experiments using type 3 muscarinic acetylcholine receptor (M3) knockout mice and their intestinal organoids and report that endogenous ACh affects the size of the intestinal stem niche via M3 signaling. RNA sequencing of crypts identified up-regulation of the EphB/ephrin-B signaling pathway. Furthermore, using an MEK inhibitor (U0126), we found that mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling, which is downstream of EphB/ephrin-B signaling, is activated in M3-deficient crypts. Collectively, M3, EphB/ephrin-B, and the MAPK/ERK signaling cascade work together to maintain the homeostasis of intestinal epithelial cell growth and differentiation following modifications of the cholinergic intestinal niche.


Subject(s)
Cell Self Renewal/genetics , Intestines/cytology , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Receptors, Eph Family/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Cell Proliferation , Female , Fluorescent Antibody Technique , Gene Expression , Immunohistochemistry , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , Models, Biological , Organoids
4.
Int Immunopharmacol ; 88: 106984, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33182055

ABSTRACT

BACKGROUNDS: Recent studies have shown that various mammalian non-neuronal cells synthesize acetylcholine (ACh) in situ and operate cholinergic signaling via nicotinic and muscarinic ACh receptors (nAChRs and mAChRs). Understanding the mechanisms that control intestinal stem cell (ISC) function through activation of nAChR signaling is critical for developing therapeutic interventions for diseases such as inflammatory bowel disease (IBD). Previously, by conducting RNA sequencing (RNA-Seq) analysis using crypt-villus organoid cultures, we found that the Hippo signaling pathway, a stem cell regulating network, is upregulated in ISCs after treatment with nicotine. Here, we explored the roles of nAChR signaling through activation of the Hippo signaling pathway. METHODS: RNA-Seq data were validated by quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis. ß4-knock-in mice were generated, and experiments using the knock-in mice and their intestinal organoids were carried out. RESULTS: RNA-Seq and qRT-PCR analyses demonstrated that the expression of YAP1/TAZ and Notch1/Dll1 was upregulated after treatment with nicotine. However, a nAChR antagonist, mecamylamine, strongly inhibited the expression of these genes. Notably, we found that in ß4-knock-in mouse small intestines, expression of YAP1 and Notch1 was significantly reduced, but not that of TAZ and Dll1, suggesting that Hippo and Notch signaling pathways are putative targets for nAChR signaling. Furthermore, fluorescent signals were detected in Paneth cells that interact with ISCs at the crypt bottom, indicating an interaction between Paneth cells and ISCs via nAChR signaling through the activation of Hippo and Notch signaling pathways. CONCLUSION: Our results indicate that upregulated nAChR signaling contributes to the maintenance of ISC activity and balances differentiation through activation of Hippo and Notch signaling pathways.


Subject(s)
Intestines/cytology , Nicotine/pharmacology , Receptors, Nicotinic/physiology , Receptors, Notch/metabolism , Stem Cells/physiology , Animals , Gene Expression Regulation/drug effects , Hippo Signaling Pathway , Mice , Nicotinic Agonists/pharmacology , Protein Serine-Threonine Kinases , Receptors, Notch/genetics , Signal Transduction/drug effects , Up-Regulation
5.
J Inherit Metab Dis ; 43(5): 960-968, 2020 09.
Article in English | MEDLINE | ID: mdl-32279332

ABSTRACT

d-3-Hydroxy-n-butyrate dehydrogenase (BDH1; EC 1.1.1.30), encoded by BDH1, catalyzes the reversible reduction of acetoacetate (AcAc) to 3-hydroxybutyrate (3HB). BDH1 is the last enzyme of hepatic ketogenesis and the first enzyme of ketolysis. The hereditary deficiency of BDH1 has not yet been described in humans. To define the features of BDH1 deficiency in a mammalian model, we generated Bdh1-deficient mice (Bdh1 KO mice). Under normal housing conditions, with unrestricted access to food, Bdh1 KO mice showed normal growth, appearance, behavior, and fertility. In contrast, fasting produced marked differences from controls. Although Bdh1 KO mice survive fasting for at least 48 hours, blood 3HB levels remained very low in Bdh1 KO mice, and despite AcAc levels moderately higher than in controls, total ketone body levels in Bdh1 KO mice were significantly lower than in wild-type (WT) mice after 16, 24, and 48 hours fasting. Hepatic fat content at 24 hours of fasting was greater in Bdh1 KO than in WT mice. Systemic BDH1 deficiency was well tolerated under normal fed conditions but manifested during fasting with a marked increase in AcAc/3HB ratio and hepatic steatosis, indicating the importance of ketogenesis for lipid energy balance in the liver.


Subject(s)
Fasting/metabolism , Fatty Liver/genetics , Hydroxybutyrate Dehydrogenase/genetics , Ketone Bodies/metabolism , Liver/metabolism , Animals , Disease Models, Animal , Energy Metabolism , Fatty Liver/enzymology , Fatty Liver/physiopathology , Female , Hydroxybutyrate Dehydrogenase/deficiency , Hydroxybutyrate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
J Biochem ; 165(4): 369-378, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30561706

ABSTRACT

Disrupted-in-schizophrenia 1 (DISC1) is strongly associated with schizophrenia, but it remains elusive how the modification of the intermolecular interaction of DISC1 affects the information processing in brain. We show that a DISC1 point mutation alters intermolecular cohesiveness promoting the phase separation, and disrupts sensorimotor gating monitored by the prepulse inhibition in a mouse model of schizophrenia. Although the conformation of DISC1 partial peptide with the schizophrenia-related mutation L607F in human or the corresponding L604F in mouse was essentially indistinguishable from the wild type (WT) as long as monitored by fluorescence, circular dichroism, ultracentrifugation, dynamic light scattering and nuclear magnetic resonance, the atomic force microscopy was able to detect their morphological distinctions. The WT peptides were round and well dispersed, while mutants were inhomogeneous and disrupted to form dimer to trimer that aligned along one direction without apparent aggregate formation. Homozygous L604F mutant mice created by CRISPR exhibited the significant decrease in DISC1 level in the immunohistopathology at the hippocampal region compared to the WTs. The ratio of prepulse inhibition of the homozygous mutant mice was significantly impaired compared to WTs. Altered DISC1 distribution or function caused by aberrant intermolecular interactions may contribute to information processing characteristics in schizophrenia.


Subject(s)
Nerve Tissue Proteins , Point Mutation , Protein Multimerization , Schizophrenia , Amino Acid Substitution , Animals , Disease Models, Animal , Humans , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology , Schizophrenia/physiopathology
7.
Brain Res ; 1688: 81-90, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29571668

ABSTRACT

Olfaxin, which is a BNIP2 and Cdc42GAP homology (BCH) domain-containing protein, is predominantly expressed in mitral and tufted (M/T) cells in the olfactory bulb (OB). Olfaxin and Caytaxin, which share 56.3% amino acid identity, are similar in their glutamatergic terminal localization, kidney-type glutaminase (KGA) interaction, and caspase-3 substrate. Although the deletion of Caytaxin protein causes human Cayman ataxia and ataxia in the mutant mouse, the function of Olfaxin is largely unknown. In this study, we generated Prune2 gene mutant mice (Prune2Ex16-/-; knock out [KO] mice) using the CRISPR/Cas9 system, during which the exon 16 containing start codon of Olfaxin mRNA was deleted. Exon 16 has 80 nucleotides and is contained in four of five Prune2 isoforms, including PRUNE2, BMCC1, BNIPXL, and Olfaxin/BMCC1s. The levels of Olfaxin mRNA and Olfaxin protein in the OB and piriform cortex of KO mice significantly decreased. Although Prune2 mRNA also significantly decreased in the spinal cord, the gross anatomy of the spinal cord and dorsal root ganglion (DRG) was intact. Further, disturbance of the sensory and motor system was not observed in KO mice. Therefore, in the current study, we examined the role of Olfaxin in the olfactory system where PRUNE2, BMCC1, and BNIPXL are scarcely expressed. Odor preference was impaired in KO mice using opposite-sex urinary scents as well as a non-social odor stimulus (almond). Results of the odor-aversion test demonstrated that odor-associative learning was disrupted in KO mice. Moreover, the NMDAR2A/NMDAR2B subunits switch in the piriform cortex was not observed in KO mice. These results indicated that Olfaxin may play a critical role in odor preference and olfactory memory.


Subject(s)
Brain/metabolism , Neoplasm Proteins/physiology , Olfactory Perception/physiology , Smell , Animals , Association Learning/physiology , Cerebellum/metabolism , Exons , Female , Male , Mice, Knockout , Neoplasm Proteins/genetics , Odorants , Olfactory Bulb/metabolism , Piriform Cortex/metabolism , Protein Isoforms/metabolism , RNA, Messenger , Receptors, N-Methyl-D-Aspartate/metabolism
8.
Sci Rep ; 7(1): 13500, 2017 10 18.
Article in English | MEDLINE | ID: mdl-29044129

ABSTRACT

Human dental pulp cells (DPCs), adherent cells derived from dental pulp tissues, are potential tools for cell transplantation therapy. However, little work has been done to optimize such transplantation. In this study, DPCs were treated with fibroblast growth factor-2 (FGF2) for 5-6 consecutive serial passages and were transplanted into the injury site immediately after complete transection of the rat spinal cord. FGF2 priming facilitated the DPCs to promote axonal regeneration and to improve locomotor function in the rat with spinal cord injury (SCI). Additional analyses revealed that FGF2 priming protected cultured DPCs from hydrogen-peroxide-induced cell death and increased the number of DPCs in the SCI rat spinal cord even 7 weeks after transplantation. The production of major neurotrophic factors was equivalent in FGF2-treated and untreated DPCs. These observations suggest that FGF2 priming might protect DPCs from the post-trauma microenvironment in which DPCs infiltrate and resident immune cells generate cytotoxic reactive oxygen species. Surviving DPCs could increase the availability of neurotrophic factors in the lesion site, thereby promoting axonal regeneration and locomotor function recovery.


Subject(s)
Dental Pulp/cytology , Fibroblast Growth Factor 2/pharmacology , Mesenchymal Stem Cell Transplantation/methods , Nerve Regeneration , Spinal Cord Injuries/therapy , Animals , Axon Guidance , Cells, Cultured , Female , Humans , Locomotion , Mesenchymal Stem Cells/drug effects , Rats , Rats, Wistar
9.
Cell Stem Cell ; 19(4): 530-543, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27524439

ABSTRACT

Physiological stem cell function is regulated by secreted factors produced by niche cells. In this study, we describe an unbiased approach based on the differential single-cell gene expression analysis of mesenchymal osteolineage cells close to, and further removed from, hematopoietic stem/progenitor cells (HSPCs) to identify candidate niche factors. Mesenchymal cells displayed distinct molecular profiles based on their relative location. We functionally examined, among the genes that were preferentially expressed in proximal cells, three secreted or cell-surface molecules not previously connected to HSPC biology-the secreted RNase angiogenin, the cytokine IL18, and the adhesion molecule Embigin-and discovered that all of these factors are HSPC quiescence regulators. Therefore, our proximity-based differential single-cell approach reveals molecular heterogeneity within niche cells and can be used to identify novel extrinsic stem/progenitor cell regulators. Similar approaches could also be applied to other stem cell/niche pairs to advance the understanding of microenvironmental regulation of stem cell function.


Subject(s)
Hematopoietic Stem Cells/cytology , Single-Cell Analysis/methods , Stem Cell Niche , Animals , Bone Marrow Cells/cytology , Bone and Bones/cytology , Cell Lineage/genetics , Cell Self Renewal/genetics , Cell Separation , Gene Deletion , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Interleukin-18/metabolism , Membrane Glycoproteins/metabolism , Ribonuclease, Pancreatic/metabolism , Time Factors , Transcription, Genetic , Vascular Cell Adhesion Molecule-1/metabolism
10.
J Invest Dermatol ; 134(6): 1627-1635, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24402046

ABSTRACT

Transcriptome analysis of the epidermis of Hes1(-/-) mouse revealed the direct relationship between Hes1 (hairy and enhancer of split-1) and BNIP3 (BCL2 and adenovirus E1B 19-kDa-interacting protein 3), a potent inducer of autophagy. Keratinocyte differentiation is going along with activation of lysosomal enzymes and organelle clearance, expecting the contribution of autophagy in this process. We found that BNIP3 was expressed in the suprabasal layer of the epidermis, where autophagosome formation is normally observed. Forced expression of BNIP3 in human primary epidermal keratinocytes (HPEKs) resulted in autophagy induction and keratinocyte differentiation, whereas knockdown of BNIP3 had the opposite effect. Intriguingly, addition of an autophagy inhibitor significantly suppressed the BNIP3-stimulated differentiation of keratinocytes, suggesting that BNIP3 plays a crucial role in keratinocyte differentiation by inducing autophagy. Furthermore, the number of dead cells increased in the human epidermal equivalent of BNIP3 knockdown keratinocytes, which suggests that BNIP3 is important for maintenance of skin epidermis. Interestingly, although UVB irradiation stimulated BNIP3 expression and cleavage of caspase3, suppression of UVB-induced BNIP3 expression led to further increase in cleaved caspase3 levels. This suggests that BNIP3 has a protective effect against UVB-induced apoptosis in keratinocytes. Overall, our data provide valuable insights into the role of BNIP3 in the differentiation and maintenance of epidermal keratinocytes.


Subject(s)
Caspase 3/metabolism , Epidermis/metabolism , Gene Expression Regulation , Keratinocytes/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis , Autophagy , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Line , Gene Expression Regulation, Enzymologic , Homeodomain Proteins/genetics , Homeostasis , Humans , Keratinocytes/cytology , Lysosomes/metabolism , Mice , Mice, Transgenic , MicroRNAs/metabolism , Transcription Factor HES-1
11.
J Invest Dermatol ; 131(12): 2358-67, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21753783

ABSTRACT

In mice, coat pigmentation requires a stem cell (SC) system in which the survival, proliferation, and differentiation of melanocytes (MCs) are regulated by microenvironments in hair follicles (HFs). In vitro systems are required to analyze the behavior of single melanocyte stem cells (MCSCs) and their potential to form SC systems in vivo. We describe here an experimental system for the isolation, self-renewal, and differentiation of MCSCs, as well as an in vivo reconstitution assay for assessing their potential. Using Dct(tm1(Cre)Bee)/CAG-CAT-GFP mice, we show that, in the presence of stem cell factor and basic fibroblast growth factor and the XB2 feeder cell line, purified MCSCs can undergo clonogenic proliferation, resulting in c-Kit(low) side scatter(low) cells. In culture, these cells maintain their capacity to differentiate and reconstitute an MCSC system in HFs. As these cells are present in the upper part of the HF near the bulge region, express only low levels of housekeeping genes, and are resistant to neonatal treatment with ACK2, it is likely that only MCSCs that are quiescent in vivo have clonogenic activity in vitro. We also found that MCSCs can be purified from wild-type mice by fluorescent cell sorting and can be characterized in vitro.


Subject(s)
Hair Follicle/metabolism , Melanocytes/metabolism , Stem Cells/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Female , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Hair Follicle/drug effects , Melanocytes/drug effects , Mice , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/pharmacology , Stem Cells/drug effects
12.
Pigment Cell Melanoma Res ; 24(1): 125-35, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21054816

ABSTRACT

Hair follicle reconstitution analysis was used to test the contribution of melanocytes or their precursors to regenerated hair follicles. In this study, we first confirmed the process of chimeric hair follicle regeneration by both hair keratinocytes and follicular melanocytes. Then, as first suggested from the differential growth requirements of epidermal skin melanocytes and non-cutaneous or dermal melanocytes, we confirmed the inability of the latter to be involved as follicular melanocytes to regenerate hair follicles during the hair reconstitution assay. This clear functional discrimination between non-cutaneous or dermal melanocytes and epidermal melanocytes suggests the presence of two different melanocyte cell lineages, a finding that might be important in the pathogenesis of melanocyte-related diseases and melanomas.


Subject(s)
Hair Follicle/cytology , Hair Follicle/growth & development , Melanocytes/cytology , Animals , Animals, Newborn , Biological Assay , Chimera , Dermis/cytology , Endothelin-3/metabolism , Humans , Keratin-14/genetics , Mice , Mice, Transgenic , Skin Pigmentation/physiology
13.
Stem Cells ; 28(9): 1571-80, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20641035

ABSTRACT

Adult stem cells, which are characterized by their capacity for self-renewal and differentiation, participate in tissue homeostasis and response to injury. They are thought to enter a state of relative quiescence, known as reversible cell cycle arrest, but the underlying molecular mechanisms remain poorly characterized. Previous data from our laboratory has shown that housekeeping gene expression is downregulated in melanocyte stem cells (MelSCs), suggesting a global suppression of mRNA transcription. We now show, using antibodies against specific phosphorylated forms of RNA polymerase II (RNApII), that adult MelSCs do not undergo productive mRNA transcription elongation, while RNApII is activated and initialized, ready to synthesize mRNA upon stimulation, and that the RNApII kinase CDK9 is absent in adult MelSCs. Interestingly, other adult stem cells also, including keratinocyte, muscle, spermatogonia, and hematopoietic stem cells, showed a similar absence of RNApII phosphorylation. Although it is difficult to show the functional significance of this observation in vivo, CDK9 inhibition resulted in enhanced survival of cells that are deprived from survival factors. We conclude that the absence of productive mRNA transcription is an early, specific, and conserved characteristic of adult stem cells. Downregulation of mRNA transcription may lead to decreased rates of metabolism, and protection from cellular and genetic damage. Screening heterogeneous tissues, including tumors, for transcriptionally quiescent cells may result in the identification of cells with stem cell-like phenotypes.


Subject(s)
Adult Stem Cells/enzymology , Cellular Senescence , Melanocytes/enzymology , Protein Processing, Post-Translational , RNA Polymerase II/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cellular Senescence/genetics , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Down-Regulation , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Mutation , NIH 3T3 Cells , Phenotype , Phosphorylation , Promoter Regions, Genetic , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , RNA, Untranslated , Serine , Signal Transduction , Stem Cell Factor/metabolism , Time Factors , Transcription, Genetic , Transfection
14.
Blood ; 114(20): 4383-92, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19770359

ABSTRACT

Hematopoietic stem cell (HSC) proliferation is tightly regulated by a poorly understood complex of positive and negative cell-cycle regulatory mechanisms. Necdin (Ndn) is an evolutionally conserved multifunctional protein that has been implicated in cell-cycle regulation of neuronal cells. Here, we provide evidence that necdin plays an important role in restricting excessive HSC proliferation during hematopoietic regeneration. We identify Ndn as being preferentially expressed in the HSC population on the basis of gene expression profiling and demonstrate that mice deficient in Ndn show accelerated recovery of the hematopoietic system after myelosuppressive injury, whereas no overt abnormality is seen in steady-state hematopoiesis. In parallel, after myelosuppression, Ndn-deficient mice exhibit an enhanced number of proliferating HSCs. Based on these findings, we propose that necdin functions in a negative feedback loop that prevents excessive proliferation of HSCs during hematopoietic regeneration. These data suggest that the inhibition of necdin after clinical myelosuppressive treatment (eg, chemotherapy, HSC transplantation) may provide therapeutic benefits by accelerating hematologic recovery.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Regeneration/genetics , Animals , Apoptosis , Cell Proliferation , Gene Expression , Gene Expression Profiling , Immunohistochemistry , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
15.
J Invest Dermatol ; 129(10): 2386-95, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19571816

ABSTRACT

The formation of functional skin entails multiple key signals that are implicated repeatedly in distinct processes during embryogenesis. Although Eph receptors and their membrane-bound ephrin ligands play a role in a wide variety of embryonic processes, their function in skin development has not been addressed. Here, we show that ephrin B2 is transiently expressed in hair buds during embryogenesis and in dermal mesenchymal cells during the perinatal period. Keratinocyte-specific ephrin B2-targeted mutant mice exhibit no skin phenotype, whereas postnatal systemic ephrin B2 ablation results in the enhancement of keratinocyte proliferation. Although the same treatment results in a defect of vascular remodeling, our analyses showed that the keratinocyte phenotype is not caused by hypoxia due to vascular defects. Interestingly, we found an enhanced expression of IL-1 family molecules, which have been implicated in the regulation of keratinocyte proliferation. On the basis of these observations, we propose that the transient expression of ephrin B2 in perinatal dermal mesenchymal cells plays a role in adjusting the activity of the mesenchymal microenvironment that regulates proliferation of keratinocytes.


Subject(s)
Cell Proliferation , Ephrin-B2/metabolism , Homeostasis/physiology , Keratinocytes/metabolism , Skin/metabolism , Animals , Animals, Newborn , Ephrin-B2/genetics , Interleukin-1/metabolism , Keratinocytes/cytology , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Morphogenesis/physiology , Skin/embryology , Skin/growth & development
16.
J Invest Dermatol ; 128(11): 2571-2574, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18927539

ABSTRACT

Notch signaling is an evolutionally conserved pathway that serves as a critical regulator of cell fate. From a series of studies, including a report in this issue, researchers have begun to elucidate the critical functions of Notch signaling in the regulation of melanocyte lineage development. With evidence of a recently identified role for Notch signaling in melanomagenesis, characterization of downstream molecular events may offer potential avenues for the development of novel therapeutic strategies.


Subject(s)
Melanocytes/cytology , Melanocytes/physiology , Receptors, Notch/physiology , Signal Transduction/physiology , Animals , Cell Communication/physiology , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Movement/physiology , Epidermal Cells , Epidermis/physiology , Humans , Mice , Stem Cells/cytology , Stem Cells/physiology
17.
Cancer Res ; 68(14): 5760-8, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18632629

ABSTRACT

Phosphate and tensin homologue deleted on chromosome 10 (PTEN) is a tumor suppressor gene inactivated in numerous sporadic cancers, including melanomas. To analyze Pten functions in melanocytes, we used the Cre-loxP system to delete Pten specifically in murine pigment-producing cells and generated DctCrePten(flox/flox) mice. Half of DctCrePten(flox/flox) mice died shortly after birth with enlargements of the cerebral cortex and hippocampus. Melanocytes were increased in the dermis of perinatal DctCrePten(flox/flox) mice. When the mutants were subjected to repeated depilations, melanocyte stem cells in the bulge of the hair follicle resisted exhaustion and the mice were protected against hair graying. Although spontaneous melanomas did not form in DctCrePten(flox/flox) mice, large nevi and melanomas developed after carcinogen exposure. DctCrePten(flox/flox) melanocytes were increased in size and exhibited heightened activation of Akt and extracellular signal-regulated kinases, increased expression of Bcl-2, and decreased expression of p27(Kip1). Our results show that Pten is important for the maintenance of melanocyte stem cells and the suppression of melanomagenesis.


Subject(s)
Carcinogens/pharmacology , Genetic Predisposition to Disease , Hair Color/genetics , Melanocytes/cytology , Melanoma/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/physiology , Animals , Cerebral Cortex/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hippocampus/metabolism , Immunohistochemistry , Melanocytes/metabolism , Melanoma/etiology , Melanoma/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/cytology
18.
Dev Cell ; 14(4): 594-604, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18410734

ABSTRACT

Recent studies have shown that Notch signaling plays an important role in epidermal development, but the underlying molecular mechanisms remain unclear. Here, by integrating loss- and gain-of-function studies of Notch receptors and Hes1, we describe molecular information about the role of Notch signaling in epidermal development. We show that Notch signaling determines spinous cell fate and induces terminal differentiation by a mechanism independent of Hes1, but Hes1 is required for maintenance of the immature state of spinous cells. Notch signaling induces Ascl2 expression to promote terminal differentiation, while simultaneously repressing Ascl2 through Hes1 to inhibit premature terminal differentiation. Despite the critical role of Hes1 in epidermal development, Hes1 null epidermis transplanted to adult mice showed no obvious defects, suggesting that this role of Hes1 may be restricted to developmental stages. Overall, we conclude that Notch signaling orchestrates the balance between differentiation and immature programs in suprabasal cells during epidermal development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Epidermal Cells , Epidermis/physiology , Homeodomain Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Epidermis/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Keratinocytes/cytology , Keratinocytes/physiology , Mice , Mice, Knockout , Mice, Transgenic , Phenotype , Receptors, Notch/genetics , Transcription Factor HES-1 , Transcription, Genetic
19.
J Invest Dermatol ; 128(2): 408-20, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17657242

ABSTRACT

Elucidation of the molecular mechanisms underlying stem cell regulation is of great importance both for basic biology and for clinical applications. Melanocyte stem cells (MSCs) are an excellent model in which to study the molecular basis of stem cell regulation, as the genetic alterations involved in the maintenance of the stem cells are readily identifiable by a premature hair graying phenotype. Research on MSCs has been hampered by the lack of a reliable system to assay their function. Here, by co-culturing highly purified melanoblasts (MBs) with XB2 keratinocytes, we describe an efficient culture method that allows the expansion of immature MBs in vitro. These MBs are also capable of undergoing terminal differentiation into mature melanocytes (MCs) when differentiation is induced. Furthermore, by performing a hair-follicle reconstitution assay in which expanded MBs in a mixture of epidermal and dermal cells were grafted to reconstitute a hair follicle, we demonstrate that the expanded MBs retain their capacity to become incorporated into newly developed hair follicles and repopulate the MC stem cell population there. Thus, by integrating genetic manipulations in cultured MBs in vitro, this method provides a powerful tool with which to study the molecular basis of stem cell regulation.


Subject(s)
Cell Culture Techniques/methods , Hair Follicle/cytology , Melanocytes/cytology , Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Line , Coculture Techniques , Dermis/cytology , Epidermal Cells , Female , Fetus/cytology , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Green Fluorescent Proteins/genetics , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Keratinocytes/cytology , Male , Melanocytes/metabolism , Mice , Mice, Transgenic , Pregnancy , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/metabolism
20.
Pigment Cell Res ; 20(4): 263-70, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17630959

ABSTRACT

As the ability to detect and define stem cells (SCs) has increased, attention is turning towards the definition of the niche and the identification of the signals that induce SC quiescence or proliferation. The melanocyte stem-niche system, in which the SCs and their progeny occupy geographically distinct domains within the hair follicle, provides one of the best models for studying the complex interplay between environmental cues and transcription factors that underpin cell fate. This review discusses what is known of the origin and molecular characteristics of melanocyte SCs and proposes a series of temporal events that are likely to contribute to the establishment of melanocyte SCs in the hair follicle. We also highlight the possibility of in vitro systems capable of directing cultured melanocytes/melanoblasts to a SC fate in response to specific extrinsiccues.


Subject(s)
Melanocytes/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Humans , Melanocytes/metabolism , Models, Biological , Receptors, Notch/metabolism , Stem Cells/metabolism , Wnt Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...