Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Dermatol Sci ; 2024 May 29.
Article in English | MEDLINE | ID: mdl-38876908

ABSTRACT

BACKGROUND: Recessive dystrophic epidermolysis bullosa (RDEB) is a severe skin fragility disorder caused by mutations in the COL7A1 gene, which encodes type VII collagen (COL7), the main constituent of anchoring fibrils for attaching the epidermis to the dermis. Persistent skin erosions frequently result in intractable ulcers in RDEB patients. Adipose-derived mesenchymal stromal cells (AD-MSCs) are easily harvested in large quantities and have low immunogenicity. Therefore, they are suitable for clinical use, including applications involving allogeneic cell transplantation. Keratinocyte-like cells transdifferentiated from AD-MSCs (KC-AD-MSCs) express more COL7 than undifferentiated AD-MSCs and facilitate skin wound healing with less contracture. Therefore, these cells can be used for skin ulcer treatment in RDEB patients. OBJECTIVE: We investigated whether KC-AD-MSCs transplantation ameliorated the RDEB phenotype severity in the grafted skin of a RDEB mouse model (col7a1-null) on the back of the immunodeficient mouse. METHODS: KC-AD-MSCs were intradermally injected into the region surrounding the skin grafts, and this procedure was repeated after 7 days. After a further 7-day interval, the skin grafts were harvested. RESULTS: Neodeposition of COL7 and generation of anchoring fibrils at the dermal-epidermal junction were observed, although experiments were based on qualitative. CONCLUSION: KC-AD-MSCs may correct the COL7 insufficiency, repair defective/reduced anchoring fibrils, and improve skin integrity in RDEB patients.

3.
Ann Dermatol ; 33(4): 324-332, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34341633

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) have been reported to promote wound healing in both animal models and human studies. Among MSCs, adipose-derived stem cells (ADSCs) can be easily harvested in large quantities. OBJECTIVE: We investigated whether skin wound healing in mice can be facilitated by keratinocyte-like cells differentiated from ADSCs (KC-ADSCs). METHODS: For the wound contraction and epithelialization model, a 20 mm×20 mm fullthickness skin wound was made on the dorsum. For the wound epithelialization model, a 6 mm×6 mm full-thickness skin wound was made on the dorsum. A nitrile rubber stent with an inner diameter of 8 mm was sutured around the wounds to minimize wound contraction. Undifferentiated ADSCs (uADSCs) or KC-ADSCs was injected around the wound base in both models. To evaluate whether the injected ADSCs could enhance wound contraction in a skin wound, the contractile activity of ADSCs was assessed by an in vitro type I collagen gel contraction assay. Alpha-smooth muscle actin (αSMA) expressions in uADSCs and KC-ADSCs were also evaluated by flow cytometry and real-time polymerase chain reaction. RESULTS: In a wound contraction and epithelialization model, KC-ADSCs further facilitated wound healing compared with uADSCs. In a wound epithelialization model, KC-ADSCs also further facilitated wound epithelialization compared with uADSCs. The contractile activity of KC-ADSCs was lower than that of uADSCs. The uADSCs expressed high levels of αSMA, which decreased after the differentiation into keratinocyte-like cells. CONCLUSION: Our results suggest that the wound healing effect of KC-ADSCs depends primarily on re-epithelialization rather than wound contraction.

4.
Biomedicines ; 9(7)2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34356904

ABSTRACT

We recently discovered a nonsynonymous variant in the coiled-coil alpha-helical rod protein 1 (CCHCR1) gene within the alopecia areata (AA) risk haplotype. We also reported that the engineered mice with this risk allele exhibited. To investigate more about the involvement of the CCHCR1 gene in AA pathogenesis, we developed an AA model using C57BL/6N cchcr1 gene knockout mice. In this study, mice (6-8 weeks) were divided into two groups: cchcr1-/- mice and wild-type (WT) littermates. Both groups were subjected to a water avoidance stress (WAS) test. Eight weeks after the WAS test, 25% of cchcr1-/- mice exhibited non-inflammatory foci of alopecia on the dorsal skin. On the other hand, none of wild-type littermates cause hair loss. The foci resembled human AA in terms of gross morphology, trichoscopic findings and histological findings. Additionally, gene expression microarray analysis of cchcr1-/- mice revealed abnormalities of hair related genes compared to the control. Our results strongly suggest that CCHCR1 is associated with AA pathogenesis and that cchcr1-/- mice are a good model for investigating AA.

5.
PLoS One ; 16(8): e0256622, 2021.
Article in English | MEDLINE | ID: mdl-34432824

ABSTRACT

Based on the assumption that some progenitor cells in an organ might reside in neighboring adipose tissue, we investigated whether melanocyte progenitor cells reside in human subcutaneous adipose tissue. First, we examined the expression of human melanoma black 45 (HMB45) and microphthalmia-associated transcription factor (MITF) in undifferentiated adipose-derived stem cells (ADSCs) by immunostaining, RT-PCR, and western blotting. These two markers were detected in undifferentiated ADSCs, and their expression levels were increased in differentiated ADSCs in melanocyte-specific culture medium. Other melanocytic markers (Melan A, MATP, Mel2, Mel EM, tyrosinase, KIT, and PAX3) were also detected at variable levels in undifferentiated ADSCs, and the expression of some markers was increased during differentiation into the melanocyte lineage. We further showed that ADSCs differentiated in melanocyte-specific culture medium localized in the basal layer and expressed tyrosinase and HMB45 in a 3D epidermal culture system. Melanin deposits were also induced by ultraviolet-light-B (UVB) irradiation. These results demonstrate that melanocyte progenitor cells reside in human subcutaneous adipose tissue and that these cells might have the potential to differentiate into mature melanocytes. Melanocyte and keratinocyte progenitors residing in human subcutaneous tissue can be used for the treatment of skin diseases and skin rejuvenation in the future.


Subject(s)
Melanocytes/cytology , Stem Cells/cytology , Subcutaneous Tissue/anatomy & histology , Biomarkers/metabolism , Cell Differentiation , Cell Line, Tumor , Dihydroxyphenylalanine/metabolism , Down-Regulation , Epidermis/metabolism , Gene Expression Regulation , Humans , Keratinocytes/metabolism , Melanins/metabolism , Melanocytes/ultrastructure , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/metabolism , Models, Biological , Pigmentation , RNA, Small Interfering/metabolism , Stem Cells/ultrastructure
6.
Arch Dermatol Res ; 309(10): 843-849, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28940088

ABSTRACT

Type VII collagen (Col7) is a major component of the anchoring fibrils at the dermoepidermal junction. Adipose-derived stem cells (ADSCs) are a cell population highly useful in regenerative medicine because of their ease of isolation and their potential for multilineage differentiation. Based on the observations that K14 was expressed in undifferentiated ADSCs and the expression was downregulated after differentiation into adipocytes, we speculated that ADSCs are keratinocyte stem/progenitor cells. ADSCs were co-cultured with fibroblasts on type IV collagen in a medium containing all-trans retinoic acid and bone morphogenetic protein 4. At day 14 of culture in keratinocyte serum-free medium, the cells were harvested and subjected to immunofluorescence, flow cytometry, real-time PCR, and western blotting. Approximately, 45% of ADSCs were immunostained positively for anti-human cytokeratin 10, and approximately 80% were stained positively for Col7. Flow cytometry, real-time PCR, and western blotting also confirmed that differentiated ADSCs expressed higher levels of Col7. These findings support the therapeutic potential of ADSCs, not only for wound healing, but also for the correction of Col7 deficiencies.


Subject(s)
Adipocytes/physiology , Adipose Tissue/cytology , Cell Differentiation/physiology , Collagen Type VII/metabolism , Keratinocytes/physiology , Stem Cells/physiology , Cells, Cultured , Coculture Techniques , Down-Regulation , Epidermal Cells , Epidermis/metabolism , Fibroblasts , Flow Cytometry , Humans , Keratin-10/metabolism , Keratin-14/metabolism , Wound Healing/physiology
8.
PLoS One ; 10(2): e0118402, 2015.
Article in English | MEDLINE | ID: mdl-25714344

ABSTRACT

The differentiation of adipose-derived stem cells (ASCs) towards epithelial lineages has yet to be demonstrated using a standardized method. This study investigated whether keratinocyte progenitor cells are present in the ASC population. ASCs isolated from subcutaneous adipose tissue were cultured and examined for the expression of the keratinocyte progenitor markers p63 and desmoglein 3 (DSG3) by immunofluorescence microscopy and flow cytometry. In addition, p63 and DSG3 expression levels were assessed before and after differentiation of ASCs into adipocytes by real-time PCR and western blot analysis, as well as in subcutaneous adipose tissue by real-time reverse transcriptase polymerase chain reaction. Both markers were expressed in ASCs, but were downregulated after the differentiation of ASCs into adipocytes; p63-positive cells were also detected in subcutaneous adipose tissue. ASCs co-cultured with human fibroblasts and incubated with all-trans retinoic acid and bone morphologic protein 4 showed an upregulation in DSG3 level, which was also increased in the presence of type IV collagen. They also showed an upregulation in cytokeratin-5 level only in the presence of type IV collagen. These results provide the demonstration that keratinocyte progenitor cells reside in subcutaneous adipose tissue.


Subject(s)
Keratinocytes/cytology , Stem Cells/cytology , Subcutaneous Fat/cytology , Adipocytes/cytology , Adipocytes/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Separation , Cell Survival , Cell Transdifferentiation , Cells, Cultured , Gene Expression , Gene Expression Profiling , Humans , Immunophenotyping , Keratinocytes/metabolism , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...