Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
Nature ; 618(7966): 808-817, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37344645

ABSTRACT

Niche signals maintain stem cells in a prolonged quiescence or transiently activate them for proper regeneration1. Altering balanced niche signalling can lead to regenerative disorders. Melanocytic skin nevi in human often display excessive hair growth, suggesting hair stem cell hyperactivity. Here, using genetic mouse models of nevi2,3, we show that dermal clusters of senescent melanocytes drive epithelial hair stem cells to exit quiescence and change their transcriptome and composition, potently enhancing hair renewal. Nevus melanocytes activate a distinct secretome, enriched for signalling factors. Osteopontin, the leading nevus signalling factor, is both necessary and sufficient to induce hair growth. Injection of osteopontin or its genetic overexpression is sufficient to induce robust hair growth in mice, whereas germline and conditional deletions of either osteopontin or CD44, its cognate receptor on epithelial hair cells, rescue enhanced hair growth induced by dermal nevus melanocytes. Osteopontin is overexpressed in human hairy nevi, and it stimulates new growth of human hair follicles. Although broad accumulation of senescent cells, such as upon ageing or genotoxic stress, is detrimental for the regenerative capacity of tissue4, we show that signalling by senescent cell clusters can potently enhance the activity of adjacent intact stem cells and stimulate tissue renewal. This finding identifies senescent cells and their secretome as an attractive therapeutic target in regenerative disorders.


Subject(s)
Hair , Melanocytes , Signal Transduction , Animals , Mice , Hair/cytology , Hair/growth & development , Hair Follicle/cytology , Hair Follicle/physiology , Hyaluronan Receptors/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Nevus/metabolism , Nevus/pathology , Osteopontin/metabolism , Stem Cells/cytology
2.
Genes Cells ; 26(2): 45-55, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33211397

ABSTRACT

Rest (RE1-silencing transcription factor, also called Nrsf) is involved in the maintenance of the undifferentiated state of neuronal stem/progenitor cells by preventing precocious expression of neuronal genes. In order to further investigate the function of Rest in neurons, we generated and examined mice evoking genetic ablation of Rest specifically in neural tissues by generating Rest conditional knockout mice. As the Rest knockout mice are embryonically lethal, we used a Sox1-Cre allele to excise the floxed Rest gene from the early stage of nerve cell differentiation including neural crest-derived nerve cells. Using this conditional Rest knockout Sox1-Cre; Restflox/flox mice, we have revealed the role of Rest in the parasympathetic nervous system in the stomach and heart.


Subject(s)
Gene Deletion , Repressor Proteins/genetics , Vagus Nerve/physiology , Animals , Electric Stimulation , Electrophysiological Phenomena , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Neurons/metabolism , Pressure , Repressor Proteins/metabolism , Stomach/innervation , Synaptic Transmission
3.
J Periodontal Res ; 57(1): 162-172, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34826339

ABSTRACT

BACKGROUND AND OBJECTIVE: Exosomes are small vesicles secreted from many cell types. Their biological effects largely depend on their cellular origin and the physiological state of the originating cells. Exosomes secreted by mesenchymal stem cells exert therapeutic effects against multiple diseases and may serve as potential alternatives to stem cell therapies. We previously established and characterized human leukocyte antigen (HLA) haplotype homo (HHH) dental pulp cell (DPC) lines from human wisdom teeth. In this study, we aimed to investigate the effect of local administration of HHH-DPC exosomes in a mouse model of periodontitis. METHODS: Exosomes purified from HHH-DPCs were subjected to particle size analysis, and expression of exosome markers was confirmed by western blotting. We also confirmed the effect of exosomes on the migration of both HHH-DPCs and mouse osteoblastic MC3T3-E1 cells. A mouse experimental periodontitis model was used to evaluate the effect of exosomes in vivo. The morphology of alveolar bone was assessed by micro-computed tomography (µCT) and histological analysis. The effect of exosomes on osteoclastogenesis was evaluated using a co-culture system. RESULTS: The exosomes purified from HHH-DPCs were homogeneous and had a spherical membrane structure. HHH-DPC exosomes promoted the migration of both human DPCs and mouse osteoblastic cells. The MTT assay showed a positive effect on the proliferation of human DPCs, but not on mouse osteoblastic cells. Treatment with HHH-DPC exosomes did not alter the differentiation of osteoblastic cells. Imaging with µCT revealed that the exosomes suppressed alveolar bone resorption in the mouse model of periodontitis. Although no change was apparent in the dominance of TRAP-positive osteoclast-like cells in decalcified tissue sections upon exosome treatment, HHH-DPC exosomes significantly suppressed osteoclast formation in vitro. CONCLUSIONS: HHH-DPC exosomes stimulated the migration of human DPCs and mouse osteoblastic cells and effectively attenuated bone loss due to periodontitis.


Subject(s)
Alveolar Bone Loss , Exosomes , Periodontitis , Alveolar Bone Loss/diagnostic imaging , Alveolar Bone Loss/therapy , Animals , Cell Differentiation , Dental Pulp , Mice , Periodontitis/therapy , X-Ray Microtomography
4.
Biol Pharm Bull ; 43(10): 1451-1454, 2020.
Article in English | MEDLINE | ID: mdl-32999155

ABSTRACT

Hair follicular keratinocyte stem cells (HFKSC) which provide a functional niche for melanocyte stem cells (MSC) are the primary target of hair graying. However, little research has been done on anti-hair graying medicines targeting HFKSC. We focused on Eriodictyon angustifolium (Ea), which reduces human hair graying when applied topically. To investigate the protective effect of dietary Ea tea (EaT) on hair pigmentation, we used an acute mouse model of hair graying that mimics X-ray-induced DNA damage associated with age-related hair graying. Our results suggest that dietary EaT maintained the niche HFKSC function against X-ray-induced DNA damage and hair graying. These results indicate that dietary EaT may prevent age-related hair graying and serve as an anti-hair graying herbal medicine.


Subject(s)
DNA Damage/drug effects , Eriodictyon , Hair Color/drug effects , Hair Follicle/drug effects , Plant Extracts/administration & dosage , Tea , Animals , Antigens, CD34/analysis , Antigens, CD34/metabolism , DNA Damage/physiology , Hair Color/physiology , Hair Follicle/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Stem Cells/drug effects , Stem Cells/metabolism
5.
Proc Natl Acad Sci U S A ; 114(4): 758-763, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28057861

ABSTRACT

The spectrum of genetic mutations differs among cancers in different organs, implying a cellular context-dependent effect for genetic aberrations. However, the extent to which the cellular context affects the consequences of oncogenic mutations remains to be fully elucidated. We reprogrammed colon tumor cells in an ApcMin/+ (adenomatous polyposis coli) mouse model, in which the loss of the Apc gene plays a critical role in tumor development and subsequently, established reprogrammed tumor cells (RTCs) that exhibit pluripotent stem cell (PSC)-like signatures of gene expression. We show that the majority of the genes in RTCs that were affected by Apc mutations did not overlap with the genes affected in the intestine. RTCs lacked pluripotency but exhibited an increased expression of Cdx2 and a differentiation propensity that was biased toward the trophectoderm cell lineage. Genetic rescue of the mutated Apc allele conferred pluripotency on RTCs and enabled their differentiation into various cell types in vivo. The redisruption of Apc in RTC-derived differentiated cells resulted in neoplastic growth that was exclusive to the intestine, but the majority of the intestinal lesions remained as pretumoral microadenomas. These results highlight the significant influence of cellular context on gene regulation, cellular plasticity, and cellular behavior in response to the loss of the Apc function. Our results also imply that the transition from microadenomas to macroscopic tumors is reprogrammable, which underscores the importance of epigenetic regulation on tumor promotion.


Subject(s)
Adenomatous Polyposis Coli/genetics , Gene Expression Regulation/genetics , Genes, APC/physiology , Mutation/genetics , Alleles , Animals , Cell Lineage/genetics , Cell Plasticity/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Epigenesis, Genetic/genetics , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Intestinal Mucosa/metabolism , Mice , Pluripotent Stem Cells/metabolism
6.
J Bone Miner Metab ; 37(3): 467-474, 2019 May.
Article in English | MEDLINE | ID: mdl-30187276

ABSTRACT

The central nervous system in adult mammals does not heal spontaneously after spinal cord injury (SCI). However, SCI treatment has been improved recently following the development of cell transplantation therapy. We recently reported that fibroblast growth factor (FGF) 2-pretreated human dental pulp cells (hDPCs) can improve recovery in a rat model of SCI. This study aimed to investigate mechanisms underlying the curative effect of SCI enhanced via FGF2 pretreatment; we selected three hDPC lines upon screening for the presence of mesenchymal stem cell markers and of their functionality in a rat model of SCI, as assessed using the Basso, Beattie, and Bresnahan score of locomotor functional scale, electrophysiological tests, and morphological analyses. We identified FGF2-responsive genes via gene expression analyses in these lines. FGF2 treatment upregulated GABRB1, MMP1, and DRD2, which suggested to contribute to SCI or central the nervous system. In an expanded screening of additional lines, GABRB1 displayed rather unique and interesting behavior; two lines with the lowest sensitivity of GABRB1 to FGF2 treatment displayed an extremely minor effect in the SCI model. These findings provide insights into the role of FGF2-responsive genes, especially GABRB1, in recovery from SCI, using hDPCs treated with FGF2.


Subject(s)
Dental Pulp/cytology , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Spinal Cord Injuries/genetics , Spinal Cord Injuries/therapy , Animals , Disease Models, Animal , Electrophysiological Phenomena/drug effects , Humans , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord Injuries/physiopathology
7.
Biol Pharm Bull ; 42(9): 1446-1449, 2019.
Article in English | MEDLINE | ID: mdl-31474706

ABSTRACT

During the process of skin regeneration following a skin injury, de novo hair follicle regeneration is initiated after wounding; however, these regenerated hairs are mostly unpigmented. The activation of epidermal melanocyte stem cells and their differentiation into regenerating hair follicles have been shown to be necessary for the pigmented hair regeneration after wounding. To determine the role of flavonoids in the regeneration of pigmented hairs, we applied the candidate flavonoids to the regenerating hair follicles after wounding and identified the flavonoid species that maximally induced pigmented hair regeneration. Flavonoids with two OH groups in the B-ring, such as sterubin, luteolin, and hydroxygenkwanin, showed promising effects in regenerating black pigmented hairs, while those with one OH group in the B-ring showed no significant change. Thus, flavonoids with two OH groups in their B-ring could be studied further as potential wound healing agents with the ability to regenerate pigmented hair.


Subject(s)
Flavonoids/pharmacology , Hair Color , Hair Follicle/drug effects , Regeneration/drug effects , Skin/injuries , Wound Healing/drug effects , Animals , Epidermal Cells/drug effects , Epidermal Cells/physiology , Flavonoids/chemistry , Hair Follicle/physiology , Luteolin/chemistry , Luteolin/pharmacology , Melanocytes/drug effects , Melanocytes/physiology , Mice, Inbred C57BL , Skin/drug effects , Structure-Activity Relationship
8.
J Cell Biochem ; 118(12): 4216-4221, 2017 12.
Article in English | MEDLINE | ID: mdl-28419513

ABSTRACT

The tumor microenvironment (TME) promotes tumor growth and metastasis. We previously established the color-coded EL4 lymphoma TME model with red fluorescent protein (RFP) expressing EL4 implanted in transgenic C57BL/6 green fluorescent protein (GFP) mice. Color-coded imaging of the lymphoma TME suggested an important role of stromal cells in lymphoma progression and metastasis. In the present study, we used color-coded imaging of RFP-lymphoma cells and GFP stromal cells to identify yellow-fluorescent genetically recombinant cells appearing only during metastasis. The EL4-RFP lymphoma cells were injected subcutaneously in C57BL/6-GFP transgenic mice and formed subcutaneous tumors 14 days after cell transplantation. The subcutaneous tumors were harvested and transplanted to the abdominal cavity of nude mice. Metastases to the liver, perigastric lymph node, ascites, bone marrow, and primary tumor were imaged. In addition to EL4-RFP cells and GFP-host cells, genetically recombinant yellow-fluorescent cells, were observed only in the ascites and bone marrow. These results indicate genetic exchange between the stromal and cancer cells. Possible mechanisms of genetic exchange are discussed as well as its ramifications for metastasis. J. Cell. Biochem. 118: 4216-4221, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Lymphoma/genetics , Neoplasm Metastasis , Recombination, Genetic , Stromal Cells , Animals , Cell Line, Tumor , Disease Models, Animal , Lymphoma/pathology , Mice , Mice, Transgenic , Tumor Microenvironment
9.
Dev Dyn ; 245(4): 460-71, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26773337

ABSTRACT

BACKGROUND: Melanoblasts (MBs), derived from neural crest cells, only differentiate into melanocytes (Ms) in vivo. We previously showed that MBs isolated from mouse skin were multipotent, generating neurons (Ns) and glial cells (Gs) together with Ms. Using Sox10-IRES-Venus mice and mouse embryonic stem cells, we investigated how MBs expressed their multipotency. RESULTS: MBs generated colonies containing Ns, Gs, and Ms in the presence of ST2 stromal cells, but they generated only M colonies when incubated with keratinocytes or ST2 culture supernatant, thus showing that MBs required contact with ST2 stromal cells for expression of their multipotency. Notch signaling was shown to be one of the important cues for the maintenance and differentiation of MBs through cell-cell contact. When Notch signaling was inhibited, MBs mainly generated colonies that contained just one type of cells, Ns, Gs, or Ms; the number of colonies containing two or three types of cells markedly decreased even on ST2 stromal cells, showing restriction of their differentiation potency. Whereas when Notch signaling was activated, the number of colonies containing two or three types of cells increased, indicating that their multipotency had been maintained. CONCLUSIONS: Our results demonstrate that Notch signaling played novel roles in MB multipotency.


Subject(s)
Melanocytes/metabolism , Multipotent Stem Cells/metabolism , Receptors, Notch/metabolism , Signal Transduction , Skin/metabolism , Animals , Melanocytes/cytology , Mice , Mice, Transgenic , Multipotent Stem Cells/cytology , Receptors, Notch/genetics , Skin/cytology
10.
Development ; 140(1): 66-75, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23222438

ABSTRACT

There is a gradient of ß-catenin expression along the colonic crypt axis with the highest levels at the crypt bottom. In addition, colorectal cancers show a heterogeneous subcellular pattern of ß-catenin accumulation. However, it remains unclear whether different levels of Wnt signalling exert distinct roles in the colonic epithelium. Here, we investigated the dose-dependent effect of canonical Wnt activation on colonic epithelial differentiation by controlling the expression levels of stabilised ß-catenin using a doxycycline-inducible transgenic system in mice. We show that elevated levels of Wnt signalling induce the amplification of Lgr5+ cells, which is accompanied by crypt fission and a reduction in cell proliferation among progenitor cells. By contrast, lower levels of ß-catenin induction enhance cell proliferation rates of epithelial progenitors without affecting crypt fission rates. Notably, slow-cycling cells produced by ß-catenin activation exhibit activation of Notch signalling. Consistent with the interpretation that the combination of Notch and Wnt signalling maintains crypt cells in a low proliferative state, the treatment of ß-catenin-expressing mice with a Notch inhibitor turned such slow-cycling cells into actively proliferating cells. Our results indicate that the activation of the canonical Wnt signalling pathway is sufficient for de novo crypt formation, and suggest that different levels of canonical Wnt activations, in cooperation with Notch signalling, establish a hierarchy of slower-cycling stem cells and faster-cycling progenitor cells characteristic for the colonic epithelium.


Subject(s)
Cell Cycle/physiology , Colon/cytology , Intestinal Mucosa/cytology , Signal Transduction/physiology , Wnt Signaling Pathway/physiology , Animals , Cell Line , Cell Proliferation , Colon/pathology , Colon/physiology , Gene Knock-In Techniques , Growth Inhibitors/physiology , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Stem Cells/cytology , Stem Cells/metabolism , beta Catenin/biosynthesis , beta Catenin/physiology
11.
Genes Cells ; 20(5): 439-49, 2015 May.
Article in English | MEDLINE | ID: mdl-25818501

ABSTRACT

Neural crest cells (NCCs) emerge from the dorsal region of the neural tube of vertebrate embryos and have the pluripotency to differentiate into both neuronal and non-neuronal lineages including melanocytes. Rest, also known as NRSF (neuro-restrictive silencer factor), is a regulator of neuronal development and function and suggested to be involved in the lineage specification of NCCs. However, further investigations of Rest gene functions in vivo have been hampered by the fact that Rest null mice show early embryonic lethality. To investigate the function of Rest in NCC development, we recently established NCC-specific Rest conditional knockout (CKO) mice and observed their neonatal death. Here, we have established viable heterozygous NCC-specific Rest CKO mice to analyze the function of Rest in an NCC-derived melanocyte cell lineage and found that the white spotting phenotype was associated with the reduction in the number of melanoblasts in the embryonic skin. The Rest deletion induced after the specification to melanocytes did not reduce the number of melanoblasts; therefore, the expression of REST during the early neural crest specification stage was necessary for the normal development of melanoblasts to cover all of the skin.


Subject(s)
Cell Differentiation , Cell Lineage , Melanocytes/cytology , Melanocytes/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Phenotype , Repressor Proteins/genetics , Animals , Gene Expression , Gene Targeting , Genes, Reporter , Mice , Mice, Knockout , Skin Pigmentation/genetics
12.
Development ; 139(4): 667-77, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22241837

ABSTRACT

Rest (RE1-silencing transcription factor, also called Nrsf) is involved in the maintenance of the undifferentiated state of neuronal stem/progenitor cells in vitro by preventing precocious expression of neuronal genes. However, the function of Rest during neurogenesis in vivo remains to be elucidated because of the early embryonic lethal phenotype of conventional Rest knockout mice. In the present study, we have generated Rest conditional knockout mice, which allow the effect of genetic ablation of Rest during embryonic neurogenesis to be examined in vivo. We show that Rest plays a role in suppressing the expression of neuronal genes in cultured neuronal cells in vitro, as well as in non-neuronal cells outside of the central nervous system, but that it is dispensable for embryonic neurogenesis in vivo. Our findings highlight the significance of extrinsic signals for the proper intrinsic regulation of neuronal gene expression levels in the specification of cell fate during embryonic neurogenesis in vivo.


Subject(s)
Gene Deletion , Gene Expression Regulation, Developmental , Neurogenesis/physiology , Neurons/physiology , Repressor Proteins/metabolism , Animals , Cell Differentiation , Cells, Cultured , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Mice , Mice, Knockout , Neurons/cytology , Repressor Proteins/genetics , Stem Cells/cytology , Stem Cells/physiology
13.
Genes Cells ; 19(10): 723-42, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25135772

ABSTRACT

RE1-silencing transcription factor (REST), also known as NRSF (neuron-restrictive silencer factor), is a well-known transcriptional repressor of neural genes. Rest null mice have embryonic lethality which prevents further investigations of the functions of the Rest gene in vivo. We studied neonatal but not embryonic lethality that was characterized by gastrointestinal tract dilation in the neural crest cell (NCC)-specific Rest conditional knockout (CKO) mice. While no histological abnormalities except the thinning of the digestive tract as a consequence of the gas accumulation were found in the digestive tract of the mutant mice, they do not have proper gastric retention after oral dye administration and the reduction of acetylcholinesterase (AChE) activity in NCC-derived myenteric plexus in the stomach was detected. High CO2 concentration in the dilated digestive tract of the Rest CKO mice indicates a failure of gut function by underdeveloped cholinergic transmission in the enteric nervous system. The observed gastrointestinal distension phenotype provides a model for understanding the genetic and molecular basis of NCC defects in humans.


Subject(s)
Gastrointestinal Diseases/pathology , Myenteric Plexus/pathology , Neural Crest/pathology , Repressor Proteins/genetics , Acetylcholinesterase/metabolism , Animals , Animals, Newborn , Carbon Dioxide/metabolism , Cell Lineage , Dilatation, Pathologic , Gastrointestinal Contents/chemistry , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/mortality , Gastrointestinal Tract/embryology , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/pathology , Mice , Mice, Knockout , Neurons/pathology , Repressor Proteins/metabolism , Stomach/embryology , Stomach/growth & development , Stomach/innervation , Wnt1 Protein/metabolism
14.
Photochem Photobiol Sci ; 14(6): 1179-89, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25966309

ABSTRACT

Melanocyte stem cells (MCSCs) in the upper portion of the hair follicle periodically supply melanocytes (MCs) that migrate downward into the hair bulb during anagen, the growth phase of the hair cycle. However MCs can also migrate upwards. We previously observed an increase in epidermal MC density in the mouse epidermis after a single ultraviolet radiation (UVR) exposure in neonatal, but not adult mice. To better understand MCSC activation by UVR we methodically studied the response of MCs to narrow band UVB (since UVA does not invoke this response) exposure in neonatal mice, and in adults at different stages of the hair cycle. We found that a single exposure of adult mice did not induce activation of MCSCs, in any stage of the hair cycle. When adult mice MCSCs were isolated in telogen, multiple UVB exposures resulted in their activation and production of daughter cells, which migrated upwards to the epidermis. Importantly, the MCSCs produced new progeny without themselves having incurred DNA damage after UVB exposure. This, together with examination of MC localisation in the skin of mice overexpressing stem cell factor in their keratinocytes, leads us to conclude that MCSC activation by UVB is driven via paracrine production of either SCF and/or other keratinocyte cytokines. We re-examined the increase in epidermal MC density in neonatal mouse skin. This effect was much more profound after only a single exposure than that of even multiple exposures to adult skin, and we show that in this setting also, the epidermal MCs mostly derive from activation of MC precursors in the upper hair follicle, and most likely via a cell extrinsic mechanism. Hence, although adaptive changes in the skin induced by repetitive UVB exposures are necessary in adult mice, in both the adult and neonatal context the division and migration upwards of follicular MCSCs is the major mode by which epidermal MC numbers increase after UVR exposure.


Subject(s)
Hair Follicle/cytology , Hair Follicle/radiation effects , Melanocytes/radiation effects , Ultraviolet Rays , Animals , Cell Proliferation/radiation effects , DNA Damage , Immunohistochemistry , Melanocytes/cytology , Mice , Skin/cytology , Skin/radiation effects , Stem Cells/cytology , Stem Cells/radiation effects
15.
Dev Dyn ; 243(3): 368-80, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24273191

ABSTRACT

BACKGROUND: Neural crest cells (NC cells) are highly migratory multipotent cells. Their multipotency is transient at the early stage of their generation; soon after emerging from the neural tube, these cells turn into lineage-restricted precursors. However, recent studies have disputed this conventionally believed paradigm. In this study, we analyzed the differentiation potency of NC-derived cells after their arrival at target tissues. RESULTS: Using Sox10-IRES-Venus mice, we found that the NC-derived cells in the skin, DRG, and inner ear could be divided into two populations: Sox10-positive/Kit-negative cells (Sox10+/Kit- cells) and Sox10- and Kit-positive cells (Sox10+/Kit+ cells). Only the Sox10+/Kit- cells were detected in the intestines. Unexpectedly, the Sox10+/Kit+ cells differentiated into neurons, glial cells, and melanocytes, showing that they had maintained their multipotency even after having entered the target tissues. The Sox10+/Kit+ cells in the DRG maintained their multipotency for a restricted period during the earlier embryonic stages, whereas those in the skin and inner ear were multipotent yet even in later embryonic stages. CONCLUSIONS: We showed that NC-derived Sox10+/Kit+ cells maintained their multipotency even after entry into the target tissues. This unexpected differentiation potency of these cells in tissues seems to have been strictly restricted by the tissue microenvironment.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells , Multipotent Stem Cells , Neural Crest , Animals , Antigens, Differentiation/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Mice , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neural Crest/cytology , Neural Crest/enzymology , Organ Specificity/physiology
16.
Birth Defects Res C Embryo Today ; 102(3): 251-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25219876

ABSTRACT

Neural crest cells (NCCs) are unique to vertebrates and emerge from the border of the neural plate and subsequently migrate extensively throughout the embryo after which they differentiate into many types of cells. This multipotency is the main reason why NCCs are regarded as a versatile tool for stem cell biology and have been gathering attention for their potential use in stem cell based therapy. Multiple sets of networks comprised of signaling molecules and transcription factors regulate every developmental phase of NCCs, including maintenance of their multipotency. Pluripotent stem cell lines, such as embryonic stem cells and induced pluripotent stem (iPS) cells, facilitate the induction of NCCs in combination with sophisticated culture systems used for neural stem cells, although at present, clinical experiments for NCC-based cell therapy need to be improved. Unexpectedly, the multipotency of NCCs is maintained after they reach the target tissues as tissue neural crest stem cells (NCSCs) that may contribute to the establishment of NCC-derived multipotential stem cells. In addition, under specific culture conditions, fate-restricted unipotent descendants of NCCs, such as melanoblasts, show multipotency to differentiate into melanocytes, neurons, and glia cells. These properties contribute to the additional versatility of NCCs for therapeutic application and to better understand NCC development.


Subject(s)
Embryonic Stem Cells/cytology , Neural Crest/cytology , Neural Stem Cells/cytology , Animals , Biological Evolution , Disease Models, Animal , Humans , Melanocytes/cytology , Neural Stem Cells/transplantation , Neurons/cytology , Pluripotent Stem Cells/cytology , Spinal Cord Injuries/therapy
17.
Nat Methods ; 8(5): 409-12, 2011 May.
Article in English | MEDLINE | ID: mdl-21460823

ABSTRACT

We report a simple method, using p53 suppression and nontransforming L-Myc, to generate human induced pluripotent stem cells (iPSCs) with episomal plasmid vectors. We generated human iPSCs from multiple donors, including two putative human leukocyte antigen (HLA)-homozygous donors who match ∼20% of the Japanese population at major HLA loci; most iPSCs are integrated transgene-free. This method may provide iPSCs suitable for autologous and allologous stem-cell therapy in the future.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Asian People/genetics , Electroporation , Gene Expression Profiling , Gene Frequency , Genetic Vectors , HLA Antigens/genetics , Humans , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Plasmids/genetics , Tissue Donors
18.
Hepatol Res ; 44(14): E408-19, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24636009

ABSTRACT

AIM: Human induced pluripotent stem (hiPS) cells are an alternative cell source of regenerative medicine for liver disease. Because variations in hepatic differentiation efficacy among hiPS cells exist, it is important to select a hiPS cell line with hepatic differentiation propensity. In addition, nuclear receptors (NR) regulate essential biological processes including differentiation and development. In this study, we identified the hiPS cell line with hepatic differentiation propensity and examined expression levels of 48 NR during this process. METHODS: We screened 28 hiPS cell lines, which are established from various tissues of healthy persons with various reprogramming methods, using a three-step differentiation method, and examined expression levels of 48 NR by quantitative real-time polymerase chain reaction during the differentiation process in the selected cells. RESULTS: hiPS-RIKEN-2B and hiPS-RIKEN-2F cells have hepatic differentiation propensity. Differentiation propensity towards endoderm was affected by donor origin but not by reprogramming methods or cell type of origins. Expression levels of NR were closely associated with those of hepatic differentiation markers. Furthermore, expression patterns of NR were categorized as five patterns. In particular, seven NR such as chicken ovalbumin upstream promoter transcription factor 1, retinoic acid receptor α, peroxisome proliferator-activated receptor-γ, progesterone receptor, photoreceptor cell-specific nuclear receptor, tailless homolog orphan receptor and glucocorticoid receptor were identified as the genes of which expression gradually goes up with differentiation. CONCLUSION: These findings will be useful for not only elucidating mechanisms of hepatic differentiation of hiPS cells but also cell-based therapy for liver diseases.

19.
Vet Dermatol ; 25(3): 199-e50, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24840326

ABSTRACT

BACKGROUND: Previous studies reported the involvement of CC chemokine receptor 4 (CCR4)-positive CD4(+) cells in the pathogenesis of canine atopic dermatitis. In humans, CCR4 is selectively expressed on type 2 helper T (Th2) cells; however, a subset of canine CCR4(+) helper T cells has not been determined. HYPOTHESIS/OBJECTIVES: To characterize the transcription profile of CCR4(+) CD4(+) lymphocytes isolated from the peripheral blood of healthy dogs. ANIMALS: Three healthy dogs were used. METHODS: The transcription levels of type 1 helper T (Th1) and Th2 cytokines in CCR4(+) CD4(+) and CCR4(-) CD4(+) lymphocytes isolated from healthy dogs were quantified by real-time RT-PCR. RESULTS: The CCR4(+) CD4(+) lymphocytes preferentially transcribed Th2 cytokines, such as interleukin-4 and interleukin-13, but not Th1 cytokines, such as interferon-γ. CONCLUSIONS AND CLINICAL IMPORTANCE: CCR4 can be used as a specific marker of Th2 cells for elucidation of the pathogenesis or the establishment of novel therapeutics in canine Th2-associated diseases, such as canine atopic dermatitis.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Gene Expression Regulation/physiology , Receptors, CCR4 , Animals , Dogs , Female , Male , Transcriptome
20.
J Invest Dermatol ; 144(1): 96-105.e2, 2024 01.
Article in English | MEDLINE | ID: mdl-37482288

ABSTRACT

KITL signaling is important for melanocyte development in mammals; however, its function in the melanocyte stem cells in adult skin is not well-understood. In this study, we have generated genetically modified mice that express a Kitl transgene under the control of a doxycycline-inducible promoter to investigate the impact of its overexpression in embryo, young postnatal, and adult skin with intact hair follicles. We report that overexpression of KITL influences the proliferation and differentiation of melanocytes as well as the self-renewal capacity of resident melanocyte stem cells within the follicular niche. Notably, activation of Kit-KITL signaling induced the migration of melanocytes from hair follicles to the epidermis. In addition, we demonstrate that a single pulse of Kitl transgene expression in postnatal mice results in long-lasting effects on melanocyte stem cells and their differentiated progeny as pigmented skin cells that persist through adulthood. Our findings indicate that regulation of KITL signaling in melanocyte lineage is crucial for melanocyte stem cell homeostasis and melanocyte cell differentiation in postnatal and adult mice.


Subject(s)
Epidermis , Hair Follicle , Mice , Animals , Epidermis/metabolism , Hair Follicle/metabolism , Melanocytes/metabolism , Pigmentation , Epidermal Cells , Cell Differentiation , Mammals
SELECTION OF CITATIONS
SEARCH DETAIL