Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062852

ABSTRACT

Therapeutic needs for hair loss are intended to find small interfering ribonucleic acid (siRNA) therapeutics for breakthrough. Since naked siRNA is restricted to meet a druggable target in clinic,, delivery systems are indispensable to overcome intrinsic and pathophysiological barriers, enhancing targetability and persistency to ensure safety, efficacy, and effectiveness. Diverse carriers repurposed from small molecules to siRNA can be systematically or locally employed in hair loss therapy, followed by the adoption of new compositions associated with structural and environmental modification. The siRNA delivery systems have been extensively studied via conjugation or nanoparticle formulation to improve their fate in vitro and in vivo. In this review, we introduce clinically tunable siRNA delivery systems for hair loss based on design principles, after analyzing clinical trials in hair loss and currently approved siRNA therapeutics. We further discuss a strategic research framework for optimized siRNA delivery in hair loss from the scientific perspective of clinical translation.


Subject(s)
Alopecia , RNA, Small Interfering , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Humans , Alopecia/therapy , Alopecia/genetics , Animals , Nanoparticles/chemistry , Genetic Therapy/methods , Drug Delivery Systems/methods , Gene Transfer Techniques
2.
Int J Mol Sci ; 25(3)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38338982

ABSTRACT

We had previously investigated the expression and functional role of C-X-C Motif Chemokine Ligand 12 (CXCL12) during the hair cycle progression. CXCL12 was highly expressed in stromal cells such as dermal fibroblasts (DFs) and inhibition of CXCL12 increased hair growth. Therefore, we further investigated whether a CXCL12 neutralizing antibody (αCXCL12) is effective for androgenic alopecia (AGA) and alopecia areata (AA) and studied the underlying molecular mechanism for treating these diseases. In the AGA model, CXCL12 is highly expressed in DFs. Subcutaneous (s.c.) injection of αCXCL12 significantly induced hair growth in AGA mice, and treatment with αCXCL12 attenuated the androgen-induced hair damage in hair organ culture. Androgens increased the secretion of CXCL12 from DFs through the androgen receptor (AR). Secreted CXCL12 from DFs increased the expression of the AR and C-X-C Motif Chemokine Receptor 4 (CXCR4) in dermal papilla cells (DPCs), which induced hair loss in AGA. Likewise, CXCL12 expression is increased in AA mice, while s.c. injection of αCXCL12 significantly inhibited hair loss in AA mice and reduced the number of CD8+, MHC-I+, and MHC-II+ cells in the skin. In addition, injection of αCXCL12 also prevented the onset of AA and reduced the number of CD8+ cells. Interferon-γ (IFNγ) treatment increased the secretion of CXCL12 from DFs through the signal transducer and activator of transcription 3 (STAT3) pathway, and αCXCL12 treatment protected the hair follicle from IFNγ in hair organ culture. Collectively, these results indicate that CXCL12 is involved in the progression of AGA and AA and antibody therapy for CXCL12 is promising for hair loss treatment.


Subject(s)
Alopecia Areata , Antibodies, Neutralizing , Animals , Mice , Alopecia/metabolism , Alopecia Areata/drug therapy , Alopecia Areata/metabolism , Androgens/metabolism , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/metabolism , Hair , Hair Follicle/metabolism , Skin/metabolism , Chemokine CXCL12/immunology
3.
Int J Mol Sci ; 23(16)2022 08 22.
Article in English | MEDLINE | ID: mdl-36012732

ABSTRACT

The fibroblast growth factor (FGF) family has various biological functions, including cell growth, tissue regeneration, embryonic development, metabolism, and angiogenesis. In the case of hair growth, several members of the FGF family, such as FGF1 and FGF2, are involved in hair growth, while FGF5 has the opposite effect. In this study, the regulation of the hair growth cycle by FGF12 was investigated. To observe its effect, the expression of FGF12 was downregulated in mice and outer root sheath (ORS) by siRNA transfection, while FGF12 overexpression was carried out using FGF12 adenovirus. For the results, FGF12 was primarily expressed in ORS cells with a high expression during the anagen phase of hair follicles. Knockdown of FGF12 delayed telogen-to-anagen transition in mice and decreased the hair length in vibrissae hair follicles. It also inhibited the proliferation and migration of ORS cells. On the contrary, FGF12 overexpression increased the migration of ORS cells. FGF12-overexpressed ORS cells induced the telogen-to-anagen transition in the animal model. In addition, FGF12 overexpression regulated the expression of PDGF-CC, MDK, and HB-EGF, and treatment of these factors exhibited hair growth promotion. Altogether, FGF12 promoted hair growth by inducing the anagen phase of hair follicles, suggesting the potential for hair loss therapy.


Subject(s)
Fibroblast Growth Factors , Hair Follicle , Hair , Animals , Cell Cycle , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hair/growth & development , Hair Follicle/metabolism , Mice , Vibrissae
4.
Int J Mol Sci ; 22(8)2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33924406

ABSTRACT

Nuclear factor erythroid 2-related factor 2 (Nrf2), which is linked to autophagy regulation and melanogenesis regulation, is activated by marliolide. In this study, we investigated the effect of a marliolide derivative on melanosome degradation through the autophagy pathway. The effect of the marliolide derivative on melanosome degradation was investigated in α-melanocyte stimulating hormone (α-MSH)-treated melanocytes, melanosome-incorporated keratinocyte, and ultraviolet (UV)B-exposed HRM-2 mice (melanin-possessing hairless mice). The marliolide derivative, 5-methyl-3-tetradecylidene-dihydro-furan-2-one (DMF02), decreased melanin pigmentation by melanosome degradation in α-MSH-treated melanocytes and melanosome-incorporated keratinocytes, evidenced by premelanosome protein (PMEL) expression, but did not affect melanogenesis-associated proteins. The UVB-induced hyperpigmentation in HRM-2 mice was also reduced by a topical application of DMF02. DMF02 activated Nrf2 and induced autophagy in vivo, evidenced by decreased PMEL in microtubule-associated proteins 1A/1B light chain 3B (LC3)-II-expressed areas. DMF02 also induced melanosome degradation via autophagy in vitro, and DMF02-induced melanosome degradation was recovered by chloroquine (CQ), which is a lysosomal inhibitor. In addition, Nrf2 silencing by siRNA attenuated the DMF02-induced melanosome degradation via the suppression of p62. DMF02 induced melanosome degradation in melanocytes and keratinocytes by regulating autophagy via Nrf2-p62 activation. Therefore, Nrf2 activator could be a promising therapeutic agent for reducing hyperpigmentation.


Subject(s)
Autophagy , Lactones/pharmacology , Melanosomes/metabolism , NF-E2-Related Factor 2/metabolism , Sequestosome-1 Protein/metabolism , Animals , Autophagy/drug effects , Autophagy/radiation effects , Gene Knockdown Techniques , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/radiation effects , Lactones/chemistry , Male , Melanins/metabolism , Melanocytes/drug effects , Melanocytes/metabolism , Melanocytes/radiation effects , Melanoma, Experimental/pathology , Mice , Skin Pigmentation/drug effects , Skin Pigmentation/radiation effects , Ultraviolet Rays
5.
Arterioscler Thromb Vasc Biol ; 39(10): 2120-2131, 2019 10.
Article in English | MEDLINE | ID: mdl-31434494

ABSTRACT

OBJECTIVE: Aberrant neovascularization is a leading cause of blindness in several eye diseases, including age-related macular degeneration and proliferative diabetic retinopathy. The identification of key regulators of pathological ocular neovascularization has been a subject of extensive research and great therapeutic interest. Here, we explored the previously unrecognized role of cKIT and its ligand, SCF (stem cell factor), in the pathological ocular neovascularization process. Approach and Results: Compared with normoxia, hypoxia, a crucial driver of neovascularization, caused cKIT to be highly upregulated in endothelial cells, which significantly enhanced the angiogenic response of endothelial cells to SCF. In murine models of pathological ocular neovascularization, such as oxygen-induced retinopathy and laser-induced choroidal neovascularization models, cKIT and SCF expression was significantly increased in ocular tissues, and blockade of cKIT and SCF using cKit mutant mice and anti-SCF neutralizing IgG substantially suppressed pathological ocular neovascularization. Mechanistically, SCF/cKIT signaling induced neovascularization through phosphorylation of glycogen synthase kinase-3ß and enhancement of the nuclear translocation of ß-catenin and the transcription of ß-catenin target genes related to angiogenesis. Inhibition of ß-catenin-mediated transcription using chemical inhibitors blocked SCF-induced in vitro angiogenesis in hypoxia, and injection of a ß-catenin agonist into cKit mutant mice with oxygen-induced retinopathy significantly enhanced pathological neovascularization in the retina. Conclusions; Our data reveal that SCF and cKIT are promising novel therapeutic targets for treating vision-threatening ocular neovascular diseases.


Subject(s)
Gene Expression Regulation , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Neovascularization/genetics , Stem Cell Factor/genetics , Vascular Endothelial Growth Factor A/metabolism , Analysis of Variance , Angiogenesis Inhibitors/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hypoxia/complications , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/genetics , Retinal Diseases/pathology , Retinal Diseases/physiopathology , Signal Transduction/genetics
6.
Int J Mol Sci ; 21(16)2020 Aug 07.
Article in English | MEDLINE | ID: mdl-32784729

ABSTRACT

Rab27a/b are known to play an important role in the transport of melanosomes, with their knockout causing silvery gray hair. However, the relationship between Rab27a/b and hair growth is not well known. To evaluate the role of Rab27a/b in hair cycle, we investigated the expression of Rab27a/b during hair cycling and human outer root sheath (hORS) cells. The expression of Rab27a in ORS cells was mainly detected at the anagen, whereas expression of Rab27b in ORS, and epidermal cells was strongly expressed at the telogen. Additionally, Rab27a/b were expressed in the Golgi of hORS cells. To evaluate the role of Rab27a/b in hair growth, telogen-to-anagen transition animal and vibrissae hair follicles (HFs) organ culture models were assayed using Rab27a/b siRNAs. The knockdown of Rab27a or Rab27b suppressed or promoted hair growth, respectively. These results were also confirmed in human dermal papilla cells (hDPCs) and hORS cells, showing the opposite mitogenic effects. Moreover, Rab27b knockdown increased the expression levels of various growth factors in the hDPCs and hORS cells. Overall, the opposite temporal expression patterns during hair cycling and roles for hair growth of Rab27a/b suggested that Rab27a/b might regulate the hair cycle. Therefore, our study may provide a novel solution for the development of hair loss treatment by regulating Rab27a/b levels.


Subject(s)
Hair/growth & development , rab GTP-Binding Proteins/antagonists & inhibitors , rab27 GTP-Binding Proteins/antagonists & inhibitors , Animals , Dermis/cytology , Hair/cytology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Models, Biological , Up-Regulation , Vibrissae/growth & development , rab GTP-Binding Proteins/metabolism , rab27 GTP-Binding Proteins/metabolism
7.
Int J Mol Sci ; 21(1)2019 Dec 23.
Article in English | MEDLINE | ID: mdl-31878047

ABSTRACT

Although adipose-derived stem cells (ASCs) have hair regenerative potential, their hair inductive capabilities are limited. The mitogenic and hair inductive effects of heparin binding-epidermal growth factor-like growth factor (HB-EGF) on ASCs were investigated in this study and the underlying mechanism of stimulation was examined. Cell growth, migration, and self-renewal assays, as well as quantitative polymerase chain reactions and immunostaining, were carried out. Telogen-to-anagen transition and organ culture using vibrissa follicles were also conducted. HB-EGF significantly increased ASC motility, including cell proliferation, migration, and self-renewal activity. The preconditioning of ASCs with HB-EGF induced telogen-to-anagen transition more rapidly in vivo, and injected PKH26-ASCs survived for longer periods of time. Conditioned medium obtained from HB-EGF-treated ASCs promoted hair growth in vivo, upregulating growth factors. In particular, thrombopoietin (THPO) also induced hair growth in vivo, stimulating dermal papilla cells (DPCs). Reactive oxygen species (ROS) appeared to play a key role in ASC stimulation as the inhibition of ROS generation and NOX4 knockout attenuated ASC stimulation and THPO upregulation by HB-EGF. In addition, the Hck phosphorylation pathway mediated the stimulation of ASCs by HB-EGF. In summary, HB-EGF increased the motility and paracrine effects of ASCs releasing THPO growth factor and THPO promoted hair growth-stimulating DPCs. ROS generation and Hck phosphorylation are key factors in HB-EGF-induced ASC stimulation. Therefore, combination therapy involving HB-EGF and ASCs may provide a novel solution for hair-loss treatment.


Subject(s)
Adipose Tissue/metabolism , Hair/physiology , Heparin-binding EGF-like Growth Factor/metabolism , Reactive Oxygen Species/metabolism , Regeneration , Stem Cells/metabolism , Vibrissae/physiology , Adipose Tissue/pathology , Animals , Humans , Male , Mice , Stem Cells/pathology
9.
Int J Mol Sci ; 19(3)2018 Feb 28.
Article in English | MEDLINE | ID: mdl-29495622

ABSTRACT

Minoxidil directly promotes hair growth via the stimulation of dermal papilla (DP) and epithelial cells. Alternatively, there is little evidence for indirect promotion of hair growth via stimulation of adipose-derived stem cells (ASCs). We investigated whether minoxidil stimulates ASCs and if increased growth factor secretion by ASCs facilitates minoxidil-induced hair growth. Telogen-to-anagen induction was examined in mice. Cultured DP cells and vibrissae hair follicle organ cultures were used to further examine the underlying mechanisms. Subcutaneous injection of minoxidil-treated ASCs accelerated telogen-to-anagen transition in mice, and increased hair weight at day 14 post-injection. Minoxidil did not alter ASC proliferation, but increased migration and tube formation. Minoxidil also increased the secretion of growth factors from ASCs, including chemokine (C-X-C motif) ligand 1 (CXCL1), platelet-derived endothelial cell growth factor (PD-ECGF), and platelet-derived growth factor-C (PDGF-C). Minoxidil increased extracellular signal-regulated kinases 1/2 (ERK1/2) phosphorylation, and concomitant upregulation of PD-ECGF and PDGF-C mRNA levels were attenuated by an ERK inhibitor. Subcutaneous injection of CXCL1, PD-ECGF, or PDGF-C enhanced anagen induction in mice, and both CXCL1 and PDGF-C increased hair length in ex vivo organ culture. Treatment with CXCL1, PD-ECGF, or PDGF-C also increased the proliferation index in DP cells. Finally, topical application of CXCL1, PD-ECGF, or PDGF-C with 2% minoxidil enhanced anagen induction when compared to minoxidil alone. Minoxidil stimulates ASC motility and increases paracrine growth factor signaling. Minoxidil-stimulated secretion of growth factors by ASCs may enhance hair growth by promoting DP proliferation. Therefore, minoxidil can be used as an ASC preconditioning agent for hair regeneration.


Subject(s)
Adipose Tissue/cytology , Hair Follicle/drug effects , Intercellular Signaling Peptides and Proteins/biosynthesis , Minoxidil/pharmacology , Stem Cells/cytology , Stem Cells/metabolism , Biomarkers , Cell Proliferation/drug effects , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Dose-Response Relationship, Drug , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Hair/growth & development , Humans , Lymphokines/genetics , Lymphokines/metabolism , MAP Kinase Signaling System , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism
10.
Arterioscler Thromb Vasc Biol ; 36(9): 1928-36, 2016 09.
Article in English | MEDLINE | ID: mdl-27470512

ABSTRACT

OBJECTIVE: Vascular smooth muscle cells (VSMCs) modulate their phenotype between synthetic and contractile states in response to environmental changes; this modulation plays a crucial role in the pathogenesis of restenosis and atherosclerosis. Here, we identified fibroblast growth factor 12 (FGF12) as a novel key regulator of the VSMC phenotype switch. APPROACH AND RESULTS: Using murine models and human specimens, we found that FGF12 was highly expressed in contractile VSMCs of normal vessel walls but was downregulated in synthetic VSMCs from injured and atherosclerotic vessels. In human VSMCs, FGF12 expression was inhibited at the transcriptional level by platelet-derived growth factor-BB. Gain- and loss-of-function experiments showed that FGF12 was both necessary and sufficient for inducing and maintaining the quiescent and contractile phenotypes of VSMCs. FGF12 inhibited cell proliferation through the p53 pathway and upregulated the key factors involved in VSMC lineage differentiation, such as myocardin and serum response factor. Such FGF12-induced phenotypic change was mediated by the p38 MAPK (mitogen-activated protein kinase) pathway. Moreover, FGF12 promoted the differentiation of mouse embryonic stem cells and the transdifferentiation of human dermal fibroblasts into SMC-like cells. Furthermore, adenoviral infection of FGF12 substantially decreased neointima hyperplasia in a rat carotid artery injury model. CONCLUSIONS: In general, FGF family members induce a synthetic VSMC phenotype. Interestingly, the present study showed the unanticipated finding that FGF12 belonging to FGF family, strongly induced the quiescent and contractile VSMC phenotypes and directly promoted VSMC lineage differentiation. These novel findings suggested that FGF12 could be a new therapeutic target for treating restenosis and atherosclerosis.


Subject(s)
Carotid Artery Diseases/metabolism , Carotid Artery Injuries/metabolism , Cell Differentiation , Cell Plasticity , Fibroblast Growth Factors/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , 5' Untranslated Regions , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Becaplermin , Binding Sites , Carotid Artery Diseases/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Cell Differentiation/drug effects , Cell Lineage , Cell Plasticity/drug effects , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Embryonic Stem Cells/metabolism , Fibroblast Growth Factors/genetics , Genotype , Humans , Hyperplasia , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Neointima , Phenotype , Phosphatidylinositol 3-Kinase/metabolism , Protein Binding , Proto-Oncogene Proteins c-sis/pharmacology , RNA Interference , Rats, Sprague-Dawley , Signal Transduction , Transcription, Genetic , Transfection , Vasoconstriction , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Stem Cells ; 33(2): 542-56, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25332166

ABSTRACT

Platelet-derived growth factor-D (PDGF-D) was recently identified, and acts as potent mitogen for mesenchymal cells. PDGF-D also induces cellular transformation and promotes tumor growth. However, the functional role of PDGF-D in adipose-derived stem cells (ASCs) has not been identified. Therefore, we primarily investigated the autocrine and paracrine roles of PDGF-D in this study. Furthermore, we identified the signaling pathways and the molecular mechanisms involved in PDGF-D-induced stimulation of ASCs. It is of interest that PDGF-B is not expressed, but PDGF-D and PDGF receptor-ß are expressed in ASCs. PDGF-D showed the strongest mitogenic effect on ASCs, and PDGF-D regulates the proliferation and migration of ASCs through the PI3K/Akt pathways. PDGF-D also increases the proliferation and migration of ASCs through generation of mitochondrial reactive oxygen species (mtROS) and mitochondrial fission. mtROS generation and fission were mediated by p66Shc phosphorylation, and BCL2-related protein A1 and Serpine peptidase inhibitor, clade E, member 1 mediated the proliferation and migration of ASCs. In addition, PDGF-D upregulated the mRNA expression of diverse growth factors such as vascular endothelial growth factor A, fibroblast growth factor 1 (FGF1), FGF5, leukemia inhibitory factor, inhibin, beta A, interleukin 11, and heparin-binding EGF-like growth factor. Therefore, the preconditioning of PDGF-D enhanced the hair-regenerative potential of ASCs. PDGF-D-induced growth factor expression was attenuated by a pharmacological inhibitor of mitogen-activated protein kinase pathway. In summary, PDGF-D is highly expressed by ASCs, where it acts as a potent mitogenic factor. PDGF-D also upregulates growth factor expression in ASCs. Therefore, PDGF-D can be considered a novel ASC stimulator, and used as a preconditioning agent before ASC transplantation.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation/physiology , Lymphokines/biosynthesis , Platelet-Derived Growth Factor/biosynthesis , Stem Cells/metabolism , Adipose Tissue/cytology , Cells, Cultured , Cytokines/biosynthesis , Gene Expression Regulation/drug effects , Humans , Lymphokines/pharmacology , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Mitochondrial Dynamics/physiology , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/biosynthesis , Reactive Oxygen Species/metabolism , Stem Cells/cytology
12.
Int J Mol Sci ; 17(9)2016 Aug 24.
Article in English | MEDLINE | ID: mdl-27563882

ABSTRACT

Hypoxia has diverse stimulatory effects on human adipose-derived stem cells (ASCs). In the present study, we investigated whether hypoxic culture conditions (2% O2) suppress spontaneous mineralization and osteogenic differentiation of ASCs. We also investigated signaling pathways and molecular mechanisms involved in this process. We found that hypoxia suppressed spontaneous mineralization and osteogenic differentiation of ASCs, and up-regulated mRNA and protein expression of Insulin-like growth factor binding proteins (IGFBPs) in ASCs. Although treatment with recombinant IGFBPs did not affect osteogenic differentiation of ASCs, siRNA-mediated inhibition of IGFBP3 attenuated hypoxia-suppressed osteogenic differentiation of ASCs. In contrast, overexpression of IGFBP3 via lentiviral vectors inhibited ASC osteogenic differentiation. These results indicate that hypoxia suppresses spontaneous mineralization and osteogenic differentiation of ASCs via intracellular IGFBP3 up-regulation. We determined that reactive oxygen species (ROS) generation followed by activation of the MAPK and PI3K/Akt pathways play pivotal roles in IGFBP3 expression under hypoxia. For example, ROS scavengers and inhibitors for MAPK and PI3K/Akt pathways attenuated the hypoxia-induced IGFBP3 expression. Inhibition of Elk1 and NF-κB through siRNA transfection also led to down-regulation of IGFBP3 mRNA expression. We next addressed the proliferative potential of ASCs with overexpressed IGFBP3, but IGFBP3 overexpression reduced the proliferation of ASCs. In addition, hypoxia reduced the osteogenic differentiation of bone marrow-derived clonal mesenchymal stem cells. Collectively, our results indicate that hypoxia suppresses the osteogenic differentiation of mesenchymal stem cells via IGFBP3 up-regulation.


Subject(s)
Cell Differentiation/physiology , Cell Hypoxia/physiology , Insulin-Like Growth Factor Binding Protein 3/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Animals , Cell Differentiation/genetics , Cell Hypoxia/genetics , Cell Proliferation/genetics , Cell Proliferation/physiology , Cells, Cultured , Humans , Immunoprecipitation , Insulin-Like Growth Factor Binding Protein 3/genetics , Mice , Osteogenesis/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
13.
Cytotherapy ; 17(8): 1090-103, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26031742

ABSTRACT

BACKGROUND AIMS: Adipose-derived mesenchymal stromal cells (AD-MSCs) have high proliferative capacity and ability to secrete trophic factors. Although intra-arterial (IA) transplantation of stem cells induces efficient engraftment to the host brain, it is unclear whether engrafted cells exert their long-term therapeutic effects through a bystander mechanism or a cell replacement mechanism. METHODS: After induction of ischemia in rats by middle cerebral artery occlusion, we transplanted human AD-MSCs into their carotid arteries with the use of a micro-needle, and we then investigated the therapeutic effects during the early and late phases of ischemia by means of in vivo magnetic resonance imaging, functional and histological analyses. RESULTS: During the early phase of cerebral ischemia, IA transplantation of AD-MSCs attenuated inflammation and enhanced endogenous neurogenesis. Transplanted animals showed a marked improvement in functional tests during the early phase of cerebral ischemia that was less prominent but still significant during the late phase of cerebral ischemia. Although the transplanted cells effectively migrated to the infarct area, only a small number of engrafted cells survived at 8 weeks after transplantation and differentiated into neuronal, glial and endothelial cells. CONCLUSIONS: IA transplantation of human AD-MSCs provides an effective therapeutic modality in a rodent model of stroke, of which the main effects are mediated by a bystander mechanism at the early phase of ischemia.


Subject(s)
Brain Ischemia/surgery , Infarction, Middle Cerebral Artery/therapy , Mesenchymal Stem Cell Transplantation/methods , Neuroprotection/physiology , Stroke/surgery , Adipose Tissue/cytology , Adult , Animals , Bystander Effect , Cell Differentiation , Disease Models, Animal , Female , Humans , Magnetic Resonance Imaging , Male , Mesenchymal Stem Cells/cytology , Neurogenesis/physiology , Rats , Rats, Sprague-Dawley , Young Adult
14.
J Surg Res ; 194(2): 688-695, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25466519

ABSTRACT

BACKGROUND: Some malignant cancers show high levels of local invasiveness by the secretion of soluble factors that can degrade adjacent tissues and suppress surrounding cell growth. We investigated the possibility of treating fibroproliferative scars based on these properties of malignant melanoma. MATERIAL AND METHODS: B16 melanoma-conditioned medium (B16 M-CM) was added to keloid fibroblasts (KFs), and proliferation, migration, and type I collagen production were measured. The cell cycle and signaling pathways were also analyzed. Proteins associated with cell proliferation were measured with Western blot analysis. Animal experiments using a rabbit ear model was performed to confirm the effect of B16 M-CM in vivo. RESULTS: B16 M-CM reduced proliferation, migration, and type I collagen production of KFs. This treatment also increased the number of cells in the subG1 phase and decreased phosphorylation levels of AKT, extracellular signal-regulated kinase1/2, cyclin D1, and c-Myc of KFs. Additionally, B16 M-CM reduced the thickness of rabbit ear scars in the rabbit ear model in vivo. CONCLUSIONS: B16 M-CM can suppress proliferation, migration, and type I collagen production of KFs. In addition, concentrated B16 M-CM reduced scar thickness in the rabbit ear model. The specific proteins involved should be identified in a future study.


Subject(s)
Fibroblasts/drug effects , Keloid/prevention & control , Melanoma, Experimental/chemistry , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/biosynthesis , Culture Media, Conditioned/pharmacology , Drug Evaluation, Preclinical , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Mice , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rabbits
15.
Int J Mol Sci ; 16(7): 16655-68, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26204837

ABSTRACT

Fluoxetine was originally developed as an antidepressant, but it has also been used to treat obesity. Although the anti-appetite effect of fluoxetine is well-documented, its potential effects on human adipose-derived stem cells (ASCs) or mature adipocytes have not been investigated. Therefore, we investigated the mechanisms underlying the inhibitory effects of fluoxetine on the proliferation of ASCs. We also investigated its inhibitory effect on adipogenic differentiation. Fluoxetine significantly decreased ASC proliferation, and signal transduction PCR array analysis showed that it increased expression of autophagy-related genes. In addition, fluoxetine up-regulated SQSTM1 and LC3B protein expression as detected by western blotting and immunofluorescence. The autophagy inhibitor, 3-methyladenine (3-MA), significantly attenuated fluoxetine-mediated effects on ASC proliferation and SQSTM1/LC3B expression. In addition, 3-MA decreased the mRNA expression of two autophagy-related genes, beclin-1 and Atg7, in ASCs. Fluoxetine also significantly inhibited lipid accumulation and down-regulated the levels of PPAR-γ and C/EBP-α in ASCs. Collectively, these results indicate that fluoxetine decreases ASC proliferation and adipogenic differentiation. This is the first in vitro evidence that fluoxetine can reduce fat accumulation by inhibiting ASC proliferation and differentiation.


Subject(s)
Adipogenesis , Cell Proliferation , Fluoxetine/pharmacology , Mesenchymal Stem Cells/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adipose Tissue/cytology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy , Autophagy-Related Protein 7 , Beclin-1 , Cells, Cultured , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Sequestosome-1 Protein , Signal Transduction , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
16.
Int J Mol Sci ; 16(8): 17088-100, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26225962

ABSTRACT

S-Methylmethionine sulfonium (SMMS) was reported to have wound-healing effects; we therefore have investigated the photoprotective effect of SMMS in the present study. SMMS increased the viability of keratinocyte progenitor cells (KPCs) and human dermal fibroblasts (hDFs) following ultraviolet B (UVB) irradiation, and reduced the UVB-induced apoptosis in these cells. SMMS increased the phosphorylation of extracellular signal-regulated kinases (ERK), and the inhibitor of the mitogen-activated protein kinase pathway significantly decreased the SMMS-induced viability of KPCs and hDFs. In addition, SMMS attenuated the UVB-induced reactive oxygen species (ROS) generation in KPCs and hDFs. SMMS induced the collagen synthesis and reduced the matrix metalloproteinase-1 expression in UVB-irradiated hDFs. In animal studies, application of 5% and 10% SMMS before and after UVB-irradiation significantly decreased the UVB-induced erythema index and depletion of Langerhans cells. In summary, SMMS protects KPCs and hDFs from UVB irradiation, and reduces UVB-induced skin erythema and immune suppression. Therefore, SMMS can be used as a cosmetic raw material, and protect skin from UVB.


Subject(s)
Erythema/drug therapy , Skin/drug effects , Sunscreening Agents/pharmacology , Vitamin U/pharmacology , Vitamins/pharmacology , Animals , Cell Line , Collagen/genetics , Collagen/metabolism , Erythema/etiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/radiation effects , Matrix Metalloproteinase 1/metabolism , Rats , Reactive Oxygen Species/metabolism , Skin/radiation effects , Sunscreening Agents/therapeutic use , Ultraviolet Rays/adverse effects , Vitamin U/therapeutic use , Vitamins/therapeutic use
17.
Int J Mol Sci ; 16(3): 4379-91, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25706512

ABSTRACT

Keratinocyte stem/progenitor cells (KSCs) reside in the bulge region of the hair follicles and may be involved in hair growth. Hair follicle dermal papilla cells (HFDPCs) and outer root sheath (ORS) cells were treated with conditioned medium (CM) of KSCs. Moreover, the effects of KSC-CM on hair growth were examined ex vivo and in vivo. A human growth factor chip array and RT-PCR were employed to identify enriched proteins in KSC-CM as compared with CM from keratinocytes. KSC-CM significantly increased the proliferation of HFDPCs and ORS cells, and increased the S-phase of the cell cycle in HFDPCs. KSC-CM led to the phosphorylation of ATK and ERK1/2 in both cell types. After subcutaneous injection of KSC-CM in C3H/HeN mice, a significant increase in hair growth and increased proliferation of hair matrix keratinocytes ex vivo was observed. We identified six proteins enriched in KSC-CM (amphiregulin, insulin-like growth factor binding protein-2, insulin-like growth factor binding protein-5, granulocyte macrophage-colony stimulating factor, Platelet-derived growth factor-AA, and vascular endothelial growth factor). A growth-factor cocktail that contains these six recombinant growth factors significantly increased the proliferation of HFDPCs and ORS cells and enhanced the hair growth of mouse models. These results collectively indicate that KSC-CM has the potential to increase hair growth via the proliferative capacity of HFDPCs and ORS cells.


Subject(s)
Culture Media, Conditioned/pharmacology , Hair/drug effects , Keratinocytes/metabolism , Stem Cells/metabolism , Amphiregulin/genetics , Amphiregulin/metabolism , Animals , Antigens, CD/metabolism , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/metabolism , Female , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hair/growth & development , Hair Follicle/drug effects , Hair Follicle/growth & development , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/metabolism , Integrin alpha6/metabolism , Mice, Inbred C3H , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Receptors, Transferrin/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
J Biol Chem ; 288(46): 32886-96, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24072712

ABSTRACT

The peroxisome proliferator-activated receptor γ (PPARγ) is a central regulator of adipogenesis and modulates glucose and lipid metabolism. In this study, herpesvirus-associated ubiquitin-specific protease (HAUSP) was isolated as a binding partner of PPARγ. Both endogenous and exogenous PPARγ associated with HAUSP in co-immunoprecipitation analysis. HAUSP, but not the catalytically inactive HAUSP C223S mutant, increased the stability of both endogenous and exogenous PPARγ through its deubiquitinating activity. Site-directed mutagenesis experiments showed that the Lys(462) residue of PPARγ is critical for ubiquitination. HBX 41,108, a specific inhibitor of HAUSP, abolished the increase in PPARγ stability induced by HAUSP. In addition, knockdown of endogenous HAUSP using siRNA decreased PPARγ protein levels. HAUSP enhanced the transcriptional activity of both exogenous and endogenous PPARγ in luciferase activity assays. Quantitative RT-PCR analysis showed that HAUSP increased the transcript levels of PPARγ target genes in HepG2 cells, resulting in the enhanced uptake of glucose and fatty acids, and vice versa, upon siRNA knockdown of HAUSP. In vivo analysis using adenoviruses confirmed that HAUSP, but not the HAUSP C223S mutant, decreased blood glucose and triglyceride levels, which are associated with the increased expression of endogenous PPARγ and lipid accumulation in the liver. Our results demonstrate that the stability and activity of PPARγ are modulated by the deubiquitinating activity of HAUSP, which may be a target for the development of anti-diabetic drugs.


Subject(s)
PPAR gamma/metabolism , Transcription, Genetic/physiology , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination/physiology , Adenoviridae , Amino Acid Substitution , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Blood Glucose/genetics , Blood Glucose/metabolism , COS Cells , Chlorocebus aethiops , Fatty Acids/blood , Fatty Acids/genetics , Gene Knockdown Techniques , HeLa Cells , Hep G2 Cells , Humans , Indenes/pharmacology , Male , Mice , Mutagenesis, Site-Directed , Mutation, Missense , PPAR gamma/genetics , Protein Stability , Pyrazines/pharmacology , Transcription, Genetic/drug effects , Transduction, Genetic , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitin-Specific Proteases/genetics , Ubiquitination/drug effects
19.
Cell Biol Int ; 38(5): 553-62, 2014 May.
Article in English | MEDLINE | ID: mdl-24446066

ABSTRACT

Cultivation under hypoxia has beneficial effects on adipose-derived stem cells (ASCs). Despite a history of extensive research on the responses of ASCs to hypoxia, investigations have focused on functional alterations of ASCs. Therefore, we provide novel insight in this review into the cellular and molecular changes that occur in ASCs under hypoxic conditions. Hypoxia increases the proliferation and migration of ASCs by the generation of reactive oxygen species (ROS) and downstream phosphorylation of platelet-derived growth factor receptor-beta, ERK1/2, and Akt. Chronically, activation of these signaling pathways upregulates miR-210 via phosphorylation of NF-κB and Elk1. Protein tyrosine phosphatase, non-receptor type 2 (PTPN2) is a direct miR-210 target, and downregulation of PTPN2 mediates the proliferation and migration of ASCs during hypoxia. In addition, the paracrine effect of ASCs is enhanced under hypoxic conditions, irrespective of whether ROS are generated. Hypoxic preconditioning stabilizes hypoxia inducible factor-1α under hypoxic conditions and increases secretion of vascular endothelial growth factor, thereby improving the regenerative potential of ASCs. Therefore, understanding the cellular and molecular changes that occur during hypoxia is highly relevant for the development of novel ASC therapies.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Hypoxia/physiology , Cells, Cultured , Humans , Reactive Oxygen Species/metabolism
20.
Cell Biol Int ; 38(1): 32-40, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23956071

ABSTRACT

Generation of reactive oxygen species (ROS) by NADPH oxidase 4 (Nox4) induces the proliferation and migration of adipose-derived stem cells (ASCs). However, the functional role of mitochondrial ROS (mtROS) generation in ASCs is unknown. Therefore, we have investigated whether hypoxia induces the differentiation of ASCs via ROS generation. We also have tried to identify the cellular mechanisms of ROS generation underlying adipocyte differentiation. Hypoxia (2%) and ROS generators, such as antimycin and rotenone, induced adipocyte differentiation, which was attenuated by an ROS scavenger. Although Nox4 generates ROS and regulates proliferation of ASCs, Nox4 inhibition or Nox4 silencing did not inhibit adipocyte differentiation; indeed fluorescence intensity of mito-SOX increased in hypoxia, and treatment with mito-CP, a mtROS scavenger, significantly reduced hypoxia-induced adipocyte differentiation. Phosphorylation of Akt and mTOR was induced by hypoxia, while inhibition of these molecules prevented adipocyte differentiation. Thus hypoxia induces adipocyte differentiation by mtROS generation, and the PI3K/Akt/mTOR pathway is involved.


Subject(s)
Cell Differentiation , Cell Hypoxia , Reactive Oxygen Species/metabolism , Stem Cells/cytology , Adipose Tissue/cytology , Antimycin A/analogs & derivatives , Antimycin A/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/pharmacology , Free Radical Scavengers/pharmacology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Rotenone/pharmacology , Signal Transduction , Stem Cells/metabolism , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL