Figure - uploaded by Mahmmood Barati
Content may be subject to copyright.
Characteristics of hair follicle stem cell (HFSC) cultures shown by inverted microscopy. The migratory cells are present on the collagen substratum. (A) HFSC one day after the primary culture, (B) migration of HFSC after the colony formation, (C) HFSC proliferation, and (D) HFSC culture after 10 days (Scale bar = 20 µm).

Characteristics of hair follicle stem cell (HFSC) cultures shown by inverted microscopy. The migratory cells are present on the collagen substratum. (A) HFSC one day after the primary culture, (B) migration of HFSC after the colony formation, (C) HFSC proliferation, and (D) HFSC culture after 10 days (Scale bar = 20 µm).

Source publication
Article
Full-text available
Background: The seladin-1 (selective Alzheimer disease indicator-1), also known as DHCR24, is a gene found to be down-regulated in brain region affected by Alzheimer disease (AD). Whereas, hair follicle stem cells (HFSC), which are affected in with neurogenic potential, it might to hypothesize that this multipotent cell compartment is the predomin...

Context in source publication

Context 1
... successfully cultured and propagated in vitro, and homogeneous population of stem cells covered the bottom of the cell cultured flask after 10 days (Fig. 1). Fluorescent cell sorting at passage 1 demonstrated that the cultured cells had neural crest characteristic, because they were positive for nestin (neuron progenitor cell marker) and CD34 (stem cell specific marker) and negative for K15 (keratinocyte cell marker) antibodies (Fig. 2). The immunostaining results of 14-day HFSC with β-III ...

Similar publications

Article
Full-text available
Significance Stem cells in the hair follicle are quiescent when the follicle is resting, but rapidly expand and differentiate upon proper cues to fuel hair regeneration, skin repair, and even cancer formation. Here we identify an important regulator of this activation process, namely Pygopus homolog 2 (Pygo2), a protein that acts in Wnt/β-catenin s...

Citations

... Numerous lines of evidence (such as gene expression, functional annotation, protein-protein interactions, and a literature survey) showed that we identified nine important candidate genes, LAMA5, OVOL1, SRF, DHCR24, NGFR, SMO, CDSN, KDF1, and HOXC13, implicated in regulating hair follicle development, and all of them were upregulated in 12-m-XFWS compared with 12-m-TS and 3-m-XFWS. The genes identified have been reported to be involved in skin and hair follicle development, representing regulation of epidermal cell differentiation and migration (LAMA5, SRF, CDSN) [38,44] and regulation of hair follicle development (OVOL1) [45], stem cell proliferation and differentiation (DHCR24, NGFR, SMO) [46,47], and regulation of hair cycle (KDF1, HOXC13) [48]. Previously, researchers have reported that there are some genes responsible for the production of the "FGF" protein in the root of the hair [49]. ...
Article
Full-text available
The hair follicle is a complex mini-organ in the skin that undergoes organ induction, morphogenesis, and regeneration. However, the accurate molecular mechanism of skin and hair diversity regulation is still elusive. The sheep is an animal model that can be used to further explore the mechanisms of skin and hair diversity. In this study, we carried out a transcriptomic analysis of the mRNA expression in the skin of Xinji fine-wool sheep at different growth stages (3 and 12 months old) and 12-month-old Tan sheep and explored the transcripts’ relationship with hair follicle growth. A total of 1327 mRNAs and 67 transcription factors were identified to be differentially expressed in the different breeds and during different periods of skin development. The differentially expressed genes were enriched in GO terms represented by system development, multicellular organism development, animal organ development, and skin development, and three KEGG pathways typified those governing differences in skin structure. Combining protein–protein interaction networks of skin development (GO:0043588) and functional annotation, nine important candidate genes, namely, LAMA5, OVOL1, SRF, DHCR24, NGFR, SMO, CDSN, HOXC13, and KDF1, and many core genes with minor effects were confirmed to be associated with hair follicle development. Furthermore, members of the zf-C2H2 and homeobox transcription factor families, which were identified to play a crucial role in producing finer and denser wool, were mainly upregulated in 12-month-old Xinji fine-wool sheep when compared with expression in 12-month-old Tan sheep and 3-month-old Xinji fine-wool sheep. This study revealed the major–minor gene interactions in the developmental pathway and provided ideas for an in-depth understanding of the genetic structure and gene regulation in the skin/hair growth process.
... cultured Hair follicle stem cells were cultured from human scalp skin pieces, digested in 12.5mg/ml dispase for 24 hours at 4°C and cell suspension was made by treating the plucked hairs twice in 0.25 per cent trypsin/EDTA solution Yu et al. (2006). Gilanchi et al. (2014) performed explant culture for mouse hair follicle stem cell culture. Rochat et al. (1994) and Maleki et al. (2014) in human Kobayashi et al. (2010) in canine dissected the hair follicle from the skin tissue and then carefully cut into four fragments: the bulb was designated as P1; the lower intermediate fragment as P2; the upper intermediate fragment as P3 and uppermost fragment containing sebaceous glands as P4 and then incubated them in collagenase -dispase for a short period to facilitate the dissociation of dermal sheath and epithelial core. ...
... In 2014, many articles from different institutes in collaboration with researchers around the world were published. The results of these studies revealed that different synthetic and natural scaffolds such as human gingival [43], cellular dermal matrix [26,44], amniotic membrane [44][45][46][47][48][49], gelatin [50][51][52], collagen [53] chitosan [54,55] and cellulose membrane [56], used along with different cells seeded on these scaffolds, such as umbilical cord blood stem cells [57,58], menstrual blood stem cells [59], human endometrial stem cells [60] and even hair follicle stem cells [31,[61][62][63] could be effective in various skin diseases like melanoma [64], dystrophic epidermolysis bullos, chronic wounds and diabetic foot ulcers [65,66], respectively. Moravvej and colleagues published a report demonstrating the useful application of allogeneic cultured fibroblast on meshed split thickness skin grafts (STSGs) in 14 third-degree burn patients [67]. ...
Article
In the last decade, the Islamic Republic of Iran has witnessed significant improvement and growth in the field of interdisciplinary medicine and in its translation to patients, including the field of cell and stem cell therapy. The main aim of this report is to highlight various advances in regenerative medicine for skin and dermatology using stem cell technology, and its translation to clinic in the past two decades, in Iranian academic centers, clinical institutes and hospitals. While there have been numerous positive advances in clinical outcomes reported in Iran, there is no comparative analytical information on these studies. Here we present a historical overview of the progress and key advancements seen in skin regeneration in this country, review the research frameworks, regulatory approach and pathways and offer perspectives for the future.
... HFSCs were found to contribute to wound healing after skin injury by migration and differentiation into epidermal cells [7] . Studies have shown strong multipotency of bulge stem cells, where they were attracted to differentiate into neurons, glial cells, keratinocytes, melanocytes, and mesenchymal cells; also, they were detected to contribute to angiogenesis [8][9][10][11][12] . ...
... The use of stem cells to enhance tissue and organ regeneration is an important factor in tissue engineering [12,20] . HFSCs are very appropriate stem cells due to some factors such as multipotency and high proliferative potential, as well as ability to differentiate into neurons [10] , keratinocytes [21,22] , and endothelial cells [23] . Additionally, because many clinical applications of HFSCs are for the enhancement of wound healing [8] , promotion of nerve repair, and functional recovery of injured peripheral nerve and spinal cord [24] , it may even be the stem cell of choice in regenerative medicine in the future. ...
Article
Full-text available
Background: Hair follicle stem cells (HFSCs) located in the bulge area has shown to be highly proliferative and could differentiate into neurons, glia, smooth muscle cell, and melanocytes in vitro. Simvastatin is an HMG-CoA reductase inhibitor that exerts pleiotropic effects beyond simple low-density lipoprotein lowering and has a similar impact on the differentiation of bone marrow stromal cells and peripheral blood mononuclear cells. The present study examined the hypothesis that the application of simvastatin would induce the HFSCs differentiation into keratinocyte. Methods: The bulge of the hair follicle was anatomized, and HFSCs were cultivated. The flow cytometry and immunocytochemical staining for detection of nestin, CD34, and Kr15 biomarkers were performed before differentiation. In order to hasten the HFSCs differentiation to keratinocyte, HFSCs were treated with 1 µM, 2 µM, and 5 µM of simvastatin daily for a week. After differentiation, the flow cytometry and immunocytochemical staining were performed with Kr15 and Kr10 biomarkers, and the MTT assay was carried out as an index of cell viability and cell growth. Results: Our results showed that bulge of HFSCs were nestin and CD34 positive and Kr15 negative. Simvastatin significantly increased the viability of HFSCs (p < 0.05) at the concentration of 5 µM. In addition, the percentages of keratinocyte-differentiated cells treated with 5 µM of simvastatin showed a significant increase compared to all other treated groups (p < 0.05). Conclusion: Our findings demonstrate that 5 µM of simvastatin could induce HFSCs differentiation into keratinocyte.
... The rats were deeply anesthetized with chloroform, and the whisker follicles were dissected as described by Amoh et al. and Gilanchi et al. with modification [6,7,21]. After rinsing the animal's head, with betadine and 70% ethanol for 3 min, the upper lip was completely shaved and trimmed into small pieces. ...
... In recent years, most of researches have focused on the factors that affect the proliferation and differentiation of stem cells. In our previous studies the effects of silibinin, nt-3 and seladin-1 (DHCR24) on hair follicle stem cells differentiation into neural-like cells were evaluated [11,21,31]. Since keratinocytes play an important role in skin wound repair, directing stem cell differentiation into the keratinocyte is very considerable [38]. ...
Article
Full-text available
Background: Stem cells are characterized by self-renewal and differentiation capabilities. The bulge hair follicle stem cells (HFSCs) are able to convert to epithelial components. The active metabolite of vitamin D, 1,25(OH)2D3, plays important roles in this differentiation process. In the present study has found that 1,25(OH)2D3 induces the HFSCs differentiation into keratinocyte. Methods: HFSCs are isolated from rat whiskers and cultivated in DMEM medium. To isolate bulge stem cell population, flow cytometry and immunocytochemistry using K15, CD34 and nestin biomarkers were performed. In order to accelerate the HFSCs differentiation into eratinocyte, HFSCs were treated with 10−12 M, 1,25(OH)2D3 every 48 h for a week. Results: Immunocytochemistry results showed that bulge stem cells are nestin and CD34 positive but K15 negative before differentiation. Subsequently flow cytometry results, showed that the expression of nestin, CD34 and K15 were 70.96%, 93.03% and 6.88% respectively. After differentiation, the immunocytochemical and flow cytometry results indicated that differentiated cells have positive reaction to K15 with 68.94% expression level. Conclusion: It was concluded that 10−12 M, 1,25(OH)2D3 could induce the HFSCs differentiation into keratinocytes.
Article
Full-text available
Abstract Clinical applications of cell therapy and tissue regeneration under different conditions need a multiplicity of adult stem cell sources. Up to date, little is available on the comparative isolation, characterization, proliferation, rapid amplification, and osteogenic/adipogenic differentiation of rat mesenchymal stem cells (MSCs) isolated from living bulge cells of the hair follicle (HF) and bone marrow (BM) from the same animal. This work hopes to use HF-MSCs as an additional adult stem cell source for research and application. After reaching 80% confluence, the cell counting, viability %, and yields of HF-MSCs and BM-MSCs were nearly similar. The viability % was 91.41 ± 2.98 and 93.11 ± 3.06 while the cells yield of initial seeding was 33.15 ± 2.76 and 34.22 ± 3.99 and of second passage was 28.76 ± 1.01 and 29.56 ± 3.11 for HF-MSCs and BM-MSCs respectively. Clusters of differentiation (CDs) analysis revealed that HF-MSCs were positively expressed CD34, CD73 and CD200 and negatively expressed CD45. BM-MSCs were positively expressed CD73 and CD200 and negatively expressed of CD34 and CD45. The proliferation of HF-MSCs and BM-MSCs was determined by means of incorporation of Brd-U, population doubling time (PDT) assays and the quantity of formazan release. The percentage of Brd-U positive cells and PDT were relatively similar in both types of cells. The proliferation, as expressed by the quantity of formazan assay in confluent cells, revealed that the quantity of release by BM-MSCs was slightly higher than HF-MSCs. Adipogenic differentiated BM-MSCs showed moderate accumulation of oil red-O stained lipid droplets when compared to that of HF-MSCs which exhibited high stain. The total lipid concentration was significantly higher in adipogenic differentiated HF-MSCs than BM-MSCs (P
Article
An embryo has the capability to accept allo- or xeno-geneic cells, which probably makes it an ideal candidate for stem cell transplantation of various cerebral cortex abnormalities, such as cortical dysplasia. The aim of this study was to determine hair follicle-associated pluripotent (HAP) stem cells homing into various organs of mother and fetus. Cells were obtained, analyzed for immunophenotypic features, and then labelled with CM-Dil; nestin(+)HAP stem cells or media phosphate-buffered saline (PBS) were intravenously delivered on day 16 of gestation in BALB/c mice, which intraperitoneally received methylazoxymethanol (MAM) one day in advance, and homing was assessed at 24 h after cell injection. Flow cytometry and immunocytochemistry manifested positive expression of nestin in HAP stem cells. For both mother and fetus, brain, lungs, liver, and spleen were the host organs for cell implants. For the brain, the figure was considerably higher in fetus, 4.05 ± 0.5% (p ≤ 0.05 vs. mother). MAM-injected mice had a downward trend for SDF-1α and CXCR4 (p ≤ 0.05 vs. control), but HAP stem cells group showed an upward trend for CXCR4 (p ≤ 0.05 vs. MAM). We conclude the HAP stem cells show homing potential in experimental cortical dysplasia, which may permit these cells to be a target in future work on prenatal therapy of neural disorders.