Hair follicle stem cells(HFSCs)in the bulge are a multipotent adult stem cell population.They can periodically give rise to new HFs and even regenerate the epidermis and sebaceous glands during wound healing.An increa...Hair follicle stem cells(HFSCs)in the bulge are a multipotent adult stem cell population.They can periodically give rise to new HFs and even regenerate the epidermis and sebaceous glands during wound healing.An increasing number of biomarkers have been used to isolate,label,and trace HFSCs in recent years.Considering more detailed data from single-cell transcriptomics technology,we mainly focus on the important HFSC molecular markers and their regulatory roles in this review.展开更多
Hair follicle-derived neural crest stem cells can be induced to differentiate into Schwann cells in vivo and in vitro. However, the underlying regulatory mechanism during cell differentiation remains poorly understood...Hair follicle-derived neural crest stem cells can be induced to differentiate into Schwann cells in vivo and in vitro. However, the underlying regulatory mechanism during cell differentiation remains poorly understood. This study isolated neural crest stem cells from human hair folli-cles and induced them to differentiate into Schwann cells. Quantitative RT-PCR showed that microRNA (miR)-21 expression was gradually increased during the differentiation of neural crest stem cells into Schwann cells. After transfection with the miR-21 agonist (agomir-21), the differentiation capacity of neural crest stem cells was enhanced. By contrast, after transfection with the miR-21 antagonist (antagomir-21), the differentiation capacity was attenuated. Further study results showed that SOX-2 was an effective target of miR-21. Without compromising SOX2 mRNA expression, miR-21 can down-regulate SOX protein expression by binding to the 3′-UTR of miR-21 mRNA. Knocking out the SOX2 gene from the neural crest stem cells significantly reversed the antagomir-21 inhibition of neural crest stem cells differentiating into Schwann cells. The results suggest that miR-21 expression was increased during the differentiation of neural crest stem cells into Schwann cells and miR-21 promoted the differentiation through down-regu-lating SOX protein expression by binding to the 3′-UTR of SOX2 mRNA.展开更多
Emerging studies of treating spinal cord injury (SCI) with adult stem cells led us to evaluate the effects of transplantation of hair follicle stem cells in rats with a compression-induced spinal cord lesion. Here, ...Emerging studies of treating spinal cord injury (SCI) with adult stem cells led us to evaluate the effects of transplantation of hair follicle stem cells in rats with a compression-induced spinal cord lesion. Here, we proposed a hypothesis that rat hair follicle stem cell transplantation can promote the recovery of injured spinal cord. Compression-induced spinal cord injury was induced in Wistar rats in this study. The bulge area of the rat vibdssa follicles was isolated, cultivated and characterized with nestin as a stem cell marker. 5-Bromo-2'-deoxyuridine (BrdU) labeled bulge stem cells were transplanted into rats with spinal cord injury. Immunohistochemical staining results showed that some of the grafted cells could survive and differentiate into oligodendrocytes (receptor-interacting protein positive cells) and neuronal-like cells (~lll-tubulin positive cells) at 3 weeks after transplantation. In addition, recovery of hind limb locomotor function in spinal cord injury rats at 8 weeks following cell transplantation was assessed using the Basso, Beattie and Bresnahan (BBB) locomotor rating scale. The results demon- strate that the grafted hair follicle stem cells can survive for a long time period in vivo and differentiate into neuronal- and glial-like cells. These results suggest that hair follicle stem cells can promote the recovery of spinal cord injury.展开更多
Hair follicles are easily accessible skin appendages that protect against cold and potential injuries.Hair follicles contain various pools of stem cells,such as epithelial,melanocyte,and mesenchymal stem cells(MSCs)th...Hair follicles are easily accessible skin appendages that protect against cold and potential injuries.Hair follicles contain various pools of stem cells,such as epithelial,melanocyte,and mesenchymal stem cells(MSCs)that continuously self-renew,differentiate,regulate hair growth,and maintain skin homeostasis.Recently,MSCs derived from the dermal papilla or dermal sheath of the human hair follicle have received attention because of their accessibility and broad differentiation potential.In this review,we describe the applications of human hair follicle-derived MSCs(hHF-MSCs)in tissue engineering and regenerative medicine.We have described protocols for isolating hHF-MSCs from human hair follicles and their culture condition in detail.We also summarize strategies for maintaining hHF-MSCs in a highly proliferative but undifferentiated state after repeated in vitro passages,including supplementation of growth factors,3D suspension culture technology,and 3D aggregates of MSCs.In addition,we report the potential of hHF-MSCs in obtaining induced smooth muscle cells and tissue-engineered blood vessels,regenerated hair follicles,induced red blood cells,and induced pluripotent stem cells.In summary,the abundance,convenient accessibility,and broad differentiation potential make hHF-MSCs an ideal seed cell source of regenerative medical and cell therapy.展开更多
Hair follicle stem cells(HFSCs) normally give rise to keratinocytes, sebocytes, and transient amplifying progenitor cells. Along with the capacity to proliferate rapidly, HFSCs provide the basis for establishing a put...Hair follicle stem cells(HFSCs) normally give rise to keratinocytes, sebocytes, and transient amplifying progenitor cells. Along with the capacity to proliferate rapidly, HFSCs provide the basis for establishing a putative source of stem cells for cell therapy. HFSCs are multipotent stem cells originating from the bulge area. The importance of these cells arises from two important characteristics, distinguishing them from all other adult stem cells. First, they are accessible and proliferate for long periods. Second, they are multipotent, possessing the ability to differentiate into mesodermal and ectodermal cell types. In addition to a developmental capacity in vitro, HFSCs display an ability to form differentiated cells in vivo. During the last two decades, numerous studies have led to the development of an appropriate culture condition for producing various cell lineages from HFSCs. Therefore, these stem cells are considered as a novel source for cell therapy of a broad spectrum of neurodegenerative disorders. This review presents the current status of human, rat, and mouse HFSCs from both the cellular and molecular biology and cell therapy perspectives. The first section of this review highlights the importance of HFSCs and in vitro differentiation, while the final section emphasizes the significance of cell differentiation in vivo.展开更多
Objective To explore the protective effect of NANOG against hydrogen peroxide(H_2O_2)-induced cell damage in the human hair follicle mesenchymal stem cells(hHF-MSCs). Methods NANOG was expressed from a lentiviral vect...Objective To explore the protective effect of NANOG against hydrogen peroxide(H_2O_2)-induced cell damage in the human hair follicle mesenchymal stem cells(hHF-MSCs). Methods NANOG was expressed from a lentiviral vector, pLVX-IRES-ZsGreen. NANOG hHF-MSCs and vector hHF-MSCs were treated with 400 μmol/L hydrogen peroxide(H_2O_2) for 2 h, the cell survival rate, cell morphology, ROS production, apoptosis and expression of AKT, ERK, and p21 were determined and compared. Results Our results showed that NANOG could activate AKT and upregulate the expression of p-AKT, but not p-ERK. When treated with 400 μmol/L H_2O_2, NANOG hHF-MSCs showed higher cell survival rate, lower ROS production and apoptosis, higher expression of p-AKT, higher ratio of p-AKT/AKT. Conclusion Our results suggest that NANOG could protect hHF-MSCs against cell damage caused by H_2O_2 through activating AKT signaling pathway.展开更多
In the bulge region of the hair follicle, a densely and concentrically packed cell mass is encircled by the arrector pili muscle (APM), which offers a specilized microenvironment (niche) for housing heterogeneous adul...In the bulge region of the hair follicle, a densely and concentrically packed cell mass is encircled by the arrector pili muscle (APM), which offers a specilized microenvironment (niche) for housing heterogeneous adult stem cells. However, the detailed histological architecture and the cellular composition of the bulge region warrants intensive study and may have implications for the regulation of hair follicle growth regulation. This study was designed to define the gene-expression pro-files of putative stem cells and lineage-specific precursors in the mid-portions of plucked hair follicles prepared according to the presence of detectable autofluorescence. The structure was also characterized by using a consecutive sectioning technique. The bulge region of the hair follicle with autofluorescence was precisely excised by employing a micro-dissection procedure. Semi-quantitative reverse transcription polymerase chain reaction (RT-PCR) was performed to identify the gene expression profiles specific for epithelial, melanocyte and stromal stem cells in the bulge region of the hair follicle visualized by autofluorescence. The morphology and its age-dependent changes of bulge region of the hair follicles with autofluorescence segment were also examined in 9 scalp skin specimens collected from patients aged 30 weeks to 75 years, by serial sectioning and immuno-staining. Gene expression profile analysis revealed that there were cells with mRNA transcripts of DctHiTyraseLo-Tyrp1LoMC1RLoMITFLo/K15Hi/NPNTHi in the bulge region of the hair follicle with autofluorescence segments, which differed from the patterns in hair bulbs. Small cell-protrusions that sprouted from the outer root sheath (ORS) were clearly observed at the APM inserting level in serial sections of hair follicles by immunohistological staining, which were characteristically replete with K15+/K19+expressing cells. Likewise, the muscle bundles of APM positive for smooth muscle actin intimately encircled these cell-protrusions, and the occurrence frequency of the cell-protrusions was increased in fetal scalp skin compared with adult scalp skin. This study provided the evidence that the cell-protrusions occurring at the ORS relative to the APM insertion are more likely to be characteristic of the visible niches that are filled with abundant stem cells. The occurrence frequency of these cell-protrusions was significantly increased in fetal scalp skin samples (128%) as compared with the scalp skins of younger (49.4%) and older (25.4%) adults (P<0.01), but difference in the frequency between the two adult groups were not significant. These results indicated that these cell-protrusions function as a niche house for the myriad stem cells and/or precursors to meet the needs of the development of hair follicles in an embryo. The micro-dissection used in this study was simple and reliable in excising the bulge region of the hair follicle with autofluorescence segments dependent on their autofluorescence is of value for the study of stem cell culture.展开更多
The skin contains various populaions of stem cells, but its characterization has been hampered by lack of markers and unclear location. The hair follicle has a niche for stem cells called a “bulge” which acts as a r...The skin contains various populaions of stem cells, but its characterization has been hampered by lack of markers and unclear location. The hair follicle has a niche for stem cells called a “bulge” which acts as a reservoir of multipotent stem cells. In the study reported here, an immunohistochemical and immunofluorescence analysis was performed on mouse and human tissues in order to determine the possible presence of stem cells of hair follicle through cytokeratin 15 (CK15), CD34, and CD200 markers identified as crucial to the stem cells and to identify the bulge region. Mouse (n = 7) and human (n = 7) skin samples were used. The expression of proteins was determined by the indirect immunoperoxidase technique and a secondary antibody bound to a fluorochrome. The specificity of staining was evaluated by negative controls. The results revealed that the stem cells associated with CD34 and CD200 antibodies were differentially expressed in the interfollicular epidermis, sebaceous glands, and bulge region, indicating that, in mice, CD34 and, in humans, CD200 are more specific than CK15 in detecting bulge cells. It also suggests that CD34 is specific for mouse bulge cells, while CD200 might have specificity for progenitor cells and partially differentiated cells in humans.展开更多
Skin contains various populations of stem cells (SCs). Among these are hair follicle stem cells (HFSCs) in the bulge region. The behavior of HFSCs deserves to be widely studied due to the benefits to be derived from t...Skin contains various populations of stem cells (SCs). Among these are hair follicle stem cells (HFSCs) in the bulge region. The behavior of HFSCs deserves to be widely studied due to the benefits to be derived from their identification, isolation, and amplification. Skin samples of newborn mice (n = 32) and human adults (n = 10) were used, and the bulge region was isolated and cultured. The isolation and characterization of cells were conducted through immunocytochemistry and immunofluorescence, using mainly CD34 and CD200 monoclonal antibodies. Initially, cells grew slowly from the explant around the bulge region, accruing cells with different morphology in both mouse and human, latter being mostly polygonal;the mouse cells reaching confluence faster (5 to 7 days) than the human (12 to 15 days). It was possible to isolate into subcultures cells with small size (10 - 13 μm diameter), round-shape, scant cytoplasm, central prominent nucleus and with nucleolus, which formed colonies, maintaining their phenotype in a high proportion (77% - 83% and 91% in mouse and human, respectively), without showing changes in their morphology during almost 7 months in the mouse cells, and a month and a half in the human. These results demonstrate that the selection, the isolation, and the conditioned mediums allowed population increases of bulge cells and indicate that cultured cells may retain their sternness in that they maintained their phenotypic characteristics, expressed specific markers for SCs, and showed a high proliferative capacity for long periods. Hair follicles, in mice and humans, are important repositories of multipotent stem cells, due to their tendency to differentiate into keratinocytes. Human HFSCs, obtained by depilation, preserve their potential for proliferation and prove to be easily accessible. This suggests that the bulge cells may present an alternative source of autologous stem cells for tissue engineering and regenerative medicine.展开更多
Objective: To establish the method of constructing skin-equivalents (SE) using hair follicle stem cells (HFSC). Methods: K19 positive cells derived from hair were cultivated using serum-free medium KGM and seede...Objective: To establish the method of constructing skin-equivalents (SE) using hair follicle stem cells (HFSC). Methods: K19 positive cells derived from hair were cultivated using serum-free medium KGM and seeded on dermal equivalents (DE). After the culture between the air-liquid interface for 14 days, SE were harvested and used for evaluation. Results: K19 positive cells chosen as HFSC were located in bulge of out root sheet in hair follicle. Cultivated HFSC could build a fully developed, multi-layered epidermis on the basis of DE, resembling the skin structure. Conclusion: HFSC located in out root sheet can differentiate into kerafinocyte in vitro and be used for SE construction.展开更多
Tissue engineering essentially refers to technology for growing new human tissue and is distinct from regenerative medicine. Currently, pieces of skin are already being fabricated for clinical use and many other tissu...Tissue engineering essentially refers to technology for growing new human tissue and is distinct from regenerative medicine. Currently, pieces of skin are already being fabricated for clinical use and many other tissue types may be fabricated in the future.Tissue engineering was first defined in 1987 by the United States National Science Foundation which critically discussed the future targets of bioengineering research and its consequences. The principles of tissue engineering are to initiate cell cultures in vitro, grow them on scaffolds in situ and transplant the composite into a recipient in vivo. From the beginning, scaffolds have been necessary in tissue engineering applications. Regardless, the latest technology has redirected established approaches by omitting scaffolds. Currently, scientists from diverse research institutes are engineering skin without scaffolds. Due to their advantageous properties, stem cells have robustly transformed the tissue engineering field as part of an engineered bilayered skin substitute that will later be discussed in detail. Additionally, utilizing biomaterials or skin replacement products in skin tissue engineering as strategy to successfully direct cell proliferation and differentiation as well as to optimize the safety of handling during grafting is beneficial. This approach has also led to the cells' application in developing the novel skin substitute that will be briefly explained in this review.展开更多
The interaction between flair folllcle dermis and its epidermis plays an important role in modulation of the growth and development of t he hair follicle. Stem cell factor (SCF ). which was found in recent years,is a ...The interaction between flair folllcle dermis and its epidermis plays an important role in modulation of the growth and development of t he hair follicle. Stem cell factor (SCF ). which was found in recent years,is a cytokine related to the survival . growl h and development of the hemopoietic stem cells and can exert important biological effects on the development of keratinocytes and melanocytes. In this study, the expression of SCF in the hair follicle spithelium was invesstigated with immunohistochemistry and in situ hybridization. It was found that the gene of the encoded SCF was strongly expressed al a limited area in the middle of the hair follicle epithelium. The protein of SCF was evenly expressed in each part of the hair follicle epithelium. The findings suggest that the expression of SCF in the hair follicle epithelium at the level of molecule is different from thai at the level of protein.展开更多
Background Sweat glands (SGs) can not regenerate after complete destruction in the severe skin injury, so it is important to find a ideal stem cell source in order to regenerate functional SGs. Hair follicle stem ce...Background Sweat glands (SGs) can not regenerate after complete destruction in the severe skin injury, so it is important to find a ideal stem cell source in order to regenerate functional SGs. Hair follicle stem cells (HFSCs) possess the obvious properties of the adult stem cells, which are multipotent and easily accessible. In this research, we attempted to direct the HFSCs suffered from the sweat gland cells (SGCs) special differentiation by a cooperative co- culture system in vitro. Methods The designed co-culture microenvironment in the transwell was consist of two critial factors: heat shocked SGCs and dermis-like mesenchymal tissue, which appeared independently in the two control groups; after induction, the purified induced SGC-like cells were transplanted into the full-thickness scalded wounds of the nude mice, after 4 weeks, the reconstructed SG-like structures were identified by immunohistochemical and immunofluorescence analysis. Results A part of HFSCs in experimental group finally expressed SGCs phenotypes, by contrast, the control group 1 which just containing dermis-like mesenchymal tissue failed and the control group 2 consisted of heat shocked SGCs was in a poor efficiency; by immunofluorescence staining and flow cytometry analysis, the expression of HFSCs special biomarkers was down regulated, instead of the positive efficiency of SGCs special antigens increased; besides, the induced SGCs displayed a high expression of ectodysplasin A (EDA) and ectodysplasin A receptor (EDAR) genes and proteins; after cell transplantation, the youngest SG-like structures formed and be positive in SGCs special antigens, which never happened in untreated wounds (P 〈0.05). Conclusion The HFSCs are multipotential and capable in differentiating into SGCs which promise a potential stem cells reservoir for future use; our special co-culture microenvironment is promising for HFSCs differentiating; the induced SGCs are functional and could work well in the regeneration of SGs.展开更多
Hair follicle stem cells(HFSCs)reside in the bulge region of the outer root sheath of the hair follicle.They are considered slow-cycling cells that are endowed with multilineage differentiation potential and superior ...Hair follicle stem cells(HFSCs)reside in the bulge region of the outer root sheath of the hair follicle.They are considered slow-cycling cells that are endowed with multilineage differentiation potential and superior proliferative capacity.The normal morphology and periodic growth of HFSCs play a significant role in normal skin functions,wound repair and skin regeneration.The HFSCs involved in these pathophysiological processes are regulated by a series of cell signal transduction pathways,such as lymphoid enhancer factor/T-cell factor,Wnt/β-catenin,transforming growth factor-β/bone morphogenetic protein,Notch and Hedgehog.The mechanisms of the interactions among these signaling pathways and their regulatory effects on HFSCs have been previously studied,but many mechanisms are still unclear.This article reviews the regulation of hair follicles,HFSCs and related signaling pathways,with the aims of summarizing previous research results,revealing the regulatory mechanisms of HFSC proliferation and differentiation and providing important references and new ideas for treating clinical diseases.展开更多
基金National Natural Science Foundation of China,No.82173446the Youth Training Program of the Army Medical University,No.2018XQN01.
文摘Hair follicle stem cells(HFSCs)in the bulge are a multipotent adult stem cell population.They can periodically give rise to new HFs and even regenerate the epidermis and sebaceous glands during wound healing.An increasing number of biomarkers have been used to isolate,label,and trace HFSCs in recent years.Considering more detailed data from single-cell transcriptomics technology,we mainly focus on the important HFSC molecular markers and their regulatory roles in this review.
基金supported by the National Natural Science Foundation of China,No.81070855
文摘Hair follicle-derived neural crest stem cells can be induced to differentiate into Schwann cells in vivo and in vitro. However, the underlying regulatory mechanism during cell differentiation remains poorly understood. This study isolated neural crest stem cells from human hair folli-cles and induced them to differentiate into Schwann cells. Quantitative RT-PCR showed that microRNA (miR)-21 expression was gradually increased during the differentiation of neural crest stem cells into Schwann cells. After transfection with the miR-21 agonist (agomir-21), the differentiation capacity of neural crest stem cells was enhanced. By contrast, after transfection with the miR-21 antagonist (antagomir-21), the differentiation capacity was attenuated. Further study results showed that SOX-2 was an effective target of miR-21. Without compromising SOX2 mRNA expression, miR-21 can down-regulate SOX protein expression by binding to the 3′-UTR of miR-21 mRNA. Knocking out the SOX2 gene from the neural crest stem cells significantly reversed the antagomir-21 inhibition of neural crest stem cells differentiating into Schwann cells. The results suggest that miR-21 expression was increased during the differentiation of neural crest stem cells into Schwann cells and miR-21 promoted the differentiation through down-regu-lating SOX protein expression by binding to the 3′-UTR of SOX2 mRNA.
基金financially supported by a grant from Iran University of Medical Sciences(Tehran–Iran),No.531
文摘Emerging studies of treating spinal cord injury (SCI) with adult stem cells led us to evaluate the effects of transplantation of hair follicle stem cells in rats with a compression-induced spinal cord lesion. Here, we proposed a hypothesis that rat hair follicle stem cell transplantation can promote the recovery of injured spinal cord. Compression-induced spinal cord injury was induced in Wistar rats in this study. The bulge area of the rat vibdssa follicles was isolated, cultivated and characterized with nestin as a stem cell marker. 5-Bromo-2'-deoxyuridine (BrdU) labeled bulge stem cells were transplanted into rats with spinal cord injury. Immunohistochemical staining results showed that some of the grafted cells could survive and differentiate into oligodendrocytes (receptor-interacting protein positive cells) and neuronal-like cells (~lll-tubulin positive cells) at 3 weeks after transplantation. In addition, recovery of hind limb locomotor function in spinal cord injury rats at 8 weeks following cell transplantation was assessed using the Basso, Beattie and Bresnahan (BBB) locomotor rating scale. The results demon- strate that the grafted hair follicle stem cells can survive for a long time period in vivo and differentiate into neuronal- and glial-like cells. These results suggest that hair follicle stem cells can promote the recovery of spinal cord injury.
基金National Natural Science Foundation of China,No.81573067the Joint Construction Project between Jilin Province and Provincial Colleges,No.SXGJQY2017-12+2 种基金the Jilin Province Science and Technology Development Plan,No.20190304044YYthe Innovative Special Industry Fund Project in Jilin Province,No.2018C049-2the Open Research Project of the State Key Laboratory of Industrial Control Technology,Zhejiang University,China,No.ICT1800381.
文摘Hair follicles are easily accessible skin appendages that protect against cold and potential injuries.Hair follicles contain various pools of stem cells,such as epithelial,melanocyte,and mesenchymal stem cells(MSCs)that continuously self-renew,differentiate,regulate hair growth,and maintain skin homeostasis.Recently,MSCs derived from the dermal papilla or dermal sheath of the human hair follicle have received attention because of their accessibility and broad differentiation potential.In this review,we describe the applications of human hair follicle-derived MSCs(hHF-MSCs)in tissue engineering and regenerative medicine.We have described protocols for isolating hHF-MSCs from human hair follicles and their culture condition in detail.We also summarize strategies for maintaining hHF-MSCs in a highly proliferative but undifferentiated state after repeated in vitro passages,including supplementation of growth factors,3D suspension culture technology,and 3D aggregates of MSCs.In addition,we report the potential of hHF-MSCs in obtaining induced smooth muscle cells and tissue-engineered blood vessels,regenerated hair follicles,induced red blood cells,and induced pluripotent stem cells.In summary,the abundance,convenient accessibility,and broad differentiation potential make hHF-MSCs an ideal seed cell source of regenerative medical and cell therapy.
文摘Hair follicle stem cells(HFSCs) normally give rise to keratinocytes, sebocytes, and transient amplifying progenitor cells. Along with the capacity to proliferate rapidly, HFSCs provide the basis for establishing a putative source of stem cells for cell therapy. HFSCs are multipotent stem cells originating from the bulge area. The importance of these cells arises from two important characteristics, distinguishing them from all other adult stem cells. First, they are accessible and proliferate for long periods. Second, they are multipotent, possessing the ability to differentiate into mesodermal and ectodermal cell types. In addition to a developmental capacity in vitro, HFSCs display an ability to form differentiated cells in vivo. During the last two decades, numerous studies have led to the development of an appropriate culture condition for producing various cell lineages from HFSCs. Therefore, these stem cells are considered as a novel source for cell therapy of a broad spectrum of neurodegenerative disorders. This review presents the current status of human, rat, and mouse HFSCs from both the cellular and molecular biology and cell therapy perspectives. The first section of this review highlights the importance of HFSCs and in vitro differentiation, while the final section emphasizes the significance of cell differentiation in vivo.
基金supported by the Jilin Province Science and Technology Development Plan [20190304044YY]the Innovative special industry fund project in Jilin province [2018C049-2]+2 种基金the Joint construction project between Jilin province and provincial colleges [SXGJQY2017-12]the Open Research Project of the State Key Laboratory of Industrial Control Technology,Zhejiang University,China [ICT1800381]the China Natural National Science Foundation [81573067]
文摘Objective To explore the protective effect of NANOG against hydrogen peroxide(H_2O_2)-induced cell damage in the human hair follicle mesenchymal stem cells(hHF-MSCs). Methods NANOG was expressed from a lentiviral vector, pLVX-IRES-ZsGreen. NANOG hHF-MSCs and vector hHF-MSCs were treated with 400 μmol/L hydrogen peroxide(H_2O_2) for 2 h, the cell survival rate, cell morphology, ROS production, apoptosis and expression of AKT, ERK, and p21 were determined and compared. Results Our results showed that NANOG could activate AKT and upregulate the expression of p-AKT, but not p-ERK. When treated with 400 μmol/L H_2O_2, NANOG hHF-MSCs showed higher cell survival rate, lower ROS production and apoptosis, higher expression of p-AKT, higher ratio of p-AKT/AKT. Conclusion Our results suggest that NANOG could protect hHF-MSCs against cell damage caused by H_2O_2 through activating AKT signaling pathway.
基金supported by grants from the National Natural Science Foundation of China (No. 8107138)a CMA-LOreal China Hair Grant (No. H2010040414)
文摘In the bulge region of the hair follicle, a densely and concentrically packed cell mass is encircled by the arrector pili muscle (APM), which offers a specilized microenvironment (niche) for housing heterogeneous adult stem cells. However, the detailed histological architecture and the cellular composition of the bulge region warrants intensive study and may have implications for the regulation of hair follicle growth regulation. This study was designed to define the gene-expression pro-files of putative stem cells and lineage-specific precursors in the mid-portions of plucked hair follicles prepared according to the presence of detectable autofluorescence. The structure was also characterized by using a consecutive sectioning technique. The bulge region of the hair follicle with autofluorescence was precisely excised by employing a micro-dissection procedure. Semi-quantitative reverse transcription polymerase chain reaction (RT-PCR) was performed to identify the gene expression profiles specific for epithelial, melanocyte and stromal stem cells in the bulge region of the hair follicle visualized by autofluorescence. The morphology and its age-dependent changes of bulge region of the hair follicles with autofluorescence segment were also examined in 9 scalp skin specimens collected from patients aged 30 weeks to 75 years, by serial sectioning and immuno-staining. Gene expression profile analysis revealed that there were cells with mRNA transcripts of DctHiTyraseLo-Tyrp1LoMC1RLoMITFLo/K15Hi/NPNTHi in the bulge region of the hair follicle with autofluorescence segments, which differed from the patterns in hair bulbs. Small cell-protrusions that sprouted from the outer root sheath (ORS) were clearly observed at the APM inserting level in serial sections of hair follicles by immunohistological staining, which were characteristically replete with K15+/K19+expressing cells. Likewise, the muscle bundles of APM positive for smooth muscle actin intimately encircled these cell-protrusions, and the occurrence frequency of the cell-protrusions was increased in fetal scalp skin compared with adult scalp skin. This study provided the evidence that the cell-protrusions occurring at the ORS relative to the APM insertion are more likely to be characteristic of the visible niches that are filled with abundant stem cells. The occurrence frequency of these cell-protrusions was significantly increased in fetal scalp skin samples (128%) as compared with the scalp skins of younger (49.4%) and older (25.4%) adults (P<0.01), but difference in the frequency between the two adult groups were not significant. These results indicated that these cell-protrusions function as a niche house for the myriad stem cells and/or precursors to meet the needs of the development of hair follicles in an embryo. The micro-dissection used in this study was simple and reliable in excising the bulge region of the hair follicle with autofluorescence segments dependent on their autofluorescence is of value for the study of stem cell culture.
文摘The skin contains various populaions of stem cells, but its characterization has been hampered by lack of markers and unclear location. The hair follicle has a niche for stem cells called a “bulge” which acts as a reservoir of multipotent stem cells. In the study reported here, an immunohistochemical and immunofluorescence analysis was performed on mouse and human tissues in order to determine the possible presence of stem cells of hair follicle through cytokeratin 15 (CK15), CD34, and CD200 markers identified as crucial to the stem cells and to identify the bulge region. Mouse (n = 7) and human (n = 7) skin samples were used. The expression of proteins was determined by the indirect immunoperoxidase technique and a secondary antibody bound to a fluorochrome. The specificity of staining was evaluated by negative controls. The results revealed that the stem cells associated with CD34 and CD200 antibodies were differentially expressed in the interfollicular epidermis, sebaceous glands, and bulge region, indicating that, in mice, CD34 and, in humans, CD200 are more specific than CK15 in detecting bulge cells. It also suggests that CD34 is specific for mouse bulge cells, while CD200 might have specificity for progenitor cells and partially differentiated cells in humans.
文摘Skin contains various populations of stem cells (SCs). Among these are hair follicle stem cells (HFSCs) in the bulge region. The behavior of HFSCs deserves to be widely studied due to the benefits to be derived from their identification, isolation, and amplification. Skin samples of newborn mice (n = 32) and human adults (n = 10) were used, and the bulge region was isolated and cultured. The isolation and characterization of cells were conducted through immunocytochemistry and immunofluorescence, using mainly CD34 and CD200 monoclonal antibodies. Initially, cells grew slowly from the explant around the bulge region, accruing cells with different morphology in both mouse and human, latter being mostly polygonal;the mouse cells reaching confluence faster (5 to 7 days) than the human (12 to 15 days). It was possible to isolate into subcultures cells with small size (10 - 13 μm diameter), round-shape, scant cytoplasm, central prominent nucleus and with nucleolus, which formed colonies, maintaining their phenotype in a high proportion (77% - 83% and 91% in mouse and human, respectively), without showing changes in their morphology during almost 7 months in the mouse cells, and a month and a half in the human. These results demonstrate that the selection, the isolation, and the conditioned mediums allowed population increases of bulge cells and indicate that cultured cells may retain their sternness in that they maintained their phenotypic characteristics, expressed specific markers for SCs, and showed a high proliferative capacity for long periods. Hair follicles, in mice and humans, are important repositories of multipotent stem cells, due to their tendency to differentiate into keratinocytes. Human HFSCs, obtained by depilation, preserve their potential for proliferation and prove to be easily accessible. This suggests that the bulge cells may present an alternative source of autologous stem cells for tissue engineering and regenerative medicine.
文摘Objective: To establish the method of constructing skin-equivalents (SE) using hair follicle stem cells (HFSC). Methods: K19 positive cells derived from hair were cultivated using serum-free medium KGM and seeded on dermal equivalents (DE). After the culture between the air-liquid interface for 14 days, SE were harvested and used for evaluation. Results: K19 positive cells chosen as HFSC were located in bulge of out root sheet in hair follicle. Cultivated HFSC could build a fully developed, multi-layered epidermis on the basis of DE, resembling the skin structure. Conclusion: HFSC located in out root sheet can differentiate into kerafinocyte in vitro and be used for SE construction.
基金Supported by Postgraduate Research Grant Scheme of Universiti Sains Malaysia,No.1001/PPSP/8144012Techno Fund grant from the Ministry of Science,Technology and Innovation of Malaysia,No.304/PPSP/6150101
文摘Tissue engineering essentially refers to technology for growing new human tissue and is distinct from regenerative medicine. Currently, pieces of skin are already being fabricated for clinical use and many other tissue types may be fabricated in the future.Tissue engineering was first defined in 1987 by the United States National Science Foundation which critically discussed the future targets of bioengineering research and its consequences. The principles of tissue engineering are to initiate cell cultures in vitro, grow them on scaffolds in situ and transplant the composite into a recipient in vivo. From the beginning, scaffolds have been necessary in tissue engineering applications. Regardless, the latest technology has redirected established approaches by omitting scaffolds. Currently, scientists from diverse research institutes are engineering skin without scaffolds. Due to their advantageous properties, stem cells have robustly transformed the tissue engineering field as part of an engineered bilayered skin substitute that will later be discussed in detail. Additionally, utilizing biomaterials or skin replacement products in skin tissue engineering as strategy to successfully direct cell proliferation and differentiation as well as to optimize the safety of handling during grafting is beneficial. This approach has also led to the cells' application in developing the novel skin substitute that will be briefly explained in this review.
文摘The interaction between flair folllcle dermis and its epidermis plays an important role in modulation of the growth and development of t he hair follicle. Stem cell factor (SCF ). which was found in recent years,is a cytokine related to the survival . growl h and development of the hemopoietic stem cells and can exert important biological effects on the development of keratinocytes and melanocytes. In this study, the expression of SCF in the hair follicle spithelium was invesstigated with immunohistochemistry and in situ hybridization. It was found that the gene of the encoded SCF was strongly expressed al a limited area in the middle of the hair follicle epithelium. The protein of SCF was evenly expressed in each part of the hair follicle epithelium. The findings suggest that the expression of SCF in the hair follicle epithelium at the level of molecule is different from thai at the level of protein.
基金This work was supported by grants from the National Basic Science and Development Programme (Nos. 2012CB518103 and 2012CB518105), the National Natural Science Foundation of China for Creative Research Groups (No. 81121004).
文摘Background Sweat glands (SGs) can not regenerate after complete destruction in the severe skin injury, so it is important to find a ideal stem cell source in order to regenerate functional SGs. Hair follicle stem cells (HFSCs) possess the obvious properties of the adult stem cells, which are multipotent and easily accessible. In this research, we attempted to direct the HFSCs suffered from the sweat gland cells (SGCs) special differentiation by a cooperative co- culture system in vitro. Methods The designed co-culture microenvironment in the transwell was consist of two critial factors: heat shocked SGCs and dermis-like mesenchymal tissue, which appeared independently in the two control groups; after induction, the purified induced SGC-like cells were transplanted into the full-thickness scalded wounds of the nude mice, after 4 weeks, the reconstructed SG-like structures were identified by immunohistochemical and immunofluorescence analysis. Results A part of HFSCs in experimental group finally expressed SGCs phenotypes, by contrast, the control group 1 which just containing dermis-like mesenchymal tissue failed and the control group 2 consisted of heat shocked SGCs was in a poor efficiency; by immunofluorescence staining and flow cytometry analysis, the expression of HFSCs special biomarkers was down regulated, instead of the positive efficiency of SGCs special antigens increased; besides, the induced SGCs displayed a high expression of ectodysplasin A (EDA) and ectodysplasin A receptor (EDAR) genes and proteins; after cell transplantation, the youngest SG-like structures formed and be positive in SGCs special antigens, which never happened in untreated wounds (P 〈0.05). Conclusion The HFSCs are multipotential and capable in differentiating into SGCs which promise a potential stem cells reservoir for future use; our special co-culture microenvironment is promising for HFSCs differentiating; the induced SGCs are functional and could work well in the regeneration of SGs.
基金supported by the National Natural Science Foundation of China(81772136,82172205,81902042)the Medical Scientific Research Foundation of Guangdong Province(A2018113)+2 种基金the Guangdong Basic and Applied Basic Research Foundation(2021A1515011453,2022A1515012160)the Special Fund of Foshan Summit plan(2019C002,2019D008,2019A006 and 2020A015)the Foundation of Foshan City(FS0AA-KJ218-1301-0034,2018AB003411).
文摘Hair follicle stem cells(HFSCs)reside in the bulge region of the outer root sheath of the hair follicle.They are considered slow-cycling cells that are endowed with multilineage differentiation potential and superior proliferative capacity.The normal morphology and periodic growth of HFSCs play a significant role in normal skin functions,wound repair and skin regeneration.The HFSCs involved in these pathophysiological processes are regulated by a series of cell signal transduction pathways,such as lymphoid enhancer factor/T-cell factor,Wnt/β-catenin,transforming growth factor-β/bone morphogenetic protein,Notch and Hedgehog.The mechanisms of the interactions among these signaling pathways and their regulatory effects on HFSCs have been previously studied,but many mechanisms are still unclear.This article reviews the regulation of hair follicles,HFSCs and related signaling pathways,with the aims of summarizing previous research results,revealing the regulatory mechanisms of HFSC proliferation and differentiation and providing important references and new ideas for treating clinical diseases.