Antler Stem Cells Sustain Regenerative Wound Healing in Deer and in Rats

Author(s):  
Chunyi Li

Deer antlers are unique mammalian appendages in that they can fully regenerate following lossfrom their pedicles (permanent bony protuberances). Antler regeneration starts from regenerative wound healing on top of a pedicle stump. A combination of tissue deletion and transplantation experiments showed that this type of regenerative healing is not skin-specific, but is bestowed by the pedicle periosteum (PP). PP cells express marker genes of both mesenchymal and embryonic stem cells, and can be induced to differentiate into multiple cell lineages in vitro. Therefore, PP cells are termed antler stem cells (AnSCs). Treatment of rats withfull-thickness cutaneous wounds (2 × 2 cm) through either direct injection of AnSCsinto the rats or topical application of conditioned medium of AnSCs on to the wounds can effectively induce regenerative wound healing. We believe our study has laid the foundations for developing an effective clinical therapy to achieve scar-less wound healing.

2009 ◽  
Vol 55 (3) ◽  
pp. 283-292 ◽  
Author(s):  
Takeshi TERAMURA ◽  
Yuta ONODERA ◽  
Hideki MURAKAMI ◽  
Syunsuke ITO ◽  
Toshihiro MIHARA ◽  
...  

2020 ◽  
Vol 15 (4) ◽  
pp. 301-307 ◽  
Author(s):  
Gaifang Wang ◽  
Maryam Farzaneh

Primary Ovarian Insufficiency (POI) is one of the main diseases causing female infertility that occurs in about 1% of women between 30-40 years of age. There are few effective methods for the treatment of women with POI. In the past few years, stem cell-based therapy as one of the most highly investigated new therapies has emerged as a promising strategy for the treatment of POI. Human pluripotent stem cells (hPSCs) can self-renew indefinitely and differentiate into any type of cell. Human Embryonic Stem Cells (hESCs) as a type of pluripotent stem cells are the most powerful candidate for the treatment of POI. Human-induced Pluripotent Stem Cells (hiPSCs) are derived from adult somatic cells by the treatment with exogenous defined factors to create an embryonic-like pluripotent state. Both hiPSCs and hESCs can proliferate and give rise to ectodermal, mesodermal, endodermal, and germ cell lineages. After ovarian stimulation, the number of available oocytes is limited and the yield of total oocytes with high quality is low. Therefore, a robust and reproducible in-vitro culture system that supports the differentiation of human oocytes from PSCs is necessary. Very few studies have focused on the derivation of oocyte-like cells from hiPSCs and the details of hPSCs differentiation into oocytes have not been fully investigated. Therefore, in this review, we focus on the differentiation potential of hPSCs into human oocyte-like cells.


Cells ◽  
2021 ◽  
Vol 10 (12) ◽  
pp. 3558
Author(s):  
Chih-Yu Yeh ◽  
Wei-Han Huang ◽  
Hung-Chi Chen ◽  
Yaa-Jyuhn James Meir

During the development of a multicellular organism, the specification of different cell lineages originates in a small group of pluripotent cells, the epiblasts, formed in the preimplantation embryo. The pluripotent epiblast is protected from premature differentiation until exposure to inductive cues in strictly controlled spatially and temporally organized patterns guiding fetus formation. Epiblasts cultured in vitro are embryonic stem cells (ESCs), which recapitulate the self-renewal and lineage specification properties of their endogenous counterparts. The characteristics of totipotency, although less understood than pluripotency, are becoming clearer. Recent studies have shown that a minor ESC subpopulation exhibits expanded developmental potential beyond pluripotency, displaying a characteristic reminiscent of two-cell embryo blastomeres (2CLCs). In addition, reprogramming both mouse and human ESCs in defined media can produce expanded/extended pluripotent stem cells (EPSCs) similar to but different from 2CLCs. Further, the molecular roadmaps driving the transition of various potency states have been clarified. These recent key findings will allow us to understand eutherian mammalian development by comparing the underlying differences between potency network components during development. Using the mouse as a paradigm and recent progress in human PSCs, we review the epiblast’s identity acquisition during embryogenesis and their ESC counterparts regarding their pluripotent fates and beyond.


2018 ◽  
Vol 38 (5) ◽  
Author(s):  
Chen Zhang ◽  
Fei Wang ◽  
Qisheng Zuo ◽  
Changhua Sun ◽  
Jing Jin ◽  
...  

Spermatogonial stem cells (SSCs) may apply to gene therapy, regenerative medicine in place of embryonic stem cells (ESCs). However, the application of SSCs was severely limited by the low induction efficiency and the lack of thorough analysis of the regulatory mechanisms of SSCs formation. Current evidences have demonstrated multiple marker genes of germ cells, while genes that specifically regulate the formation of SSCs have not been explored. In our study, cadherin-like and PC-esterase domain containing 1 (Cped1) expressed specifically in SSCs based on RNA-seq data analysis. To study the function of Cped1 in the formation of SSCs, we successfully established a CRISPR/Cas9 knockout system. The gene disruption frequency is 37% in DF1 and 25% in ESCs without off-target effects. Knockout of Cped1 could significantly inhibit the formation of SSCs in vivo and in vitro. The fragment of −1050 to −1 bp had the activity as Cped1 gene promoter. Histone acetylation could regulate the expression of Cped1. We added 5-azaeytidi (DNA methylation inhibitors) and TSA (histone deacetylase inhibitors) respectively during the cultivation of SSCs. TSA was validated to promote the transcription of Cped1. Dual-luciferase reporter assay revealed that active control area of the chicken Cped1 gene is −296 to −1 bp. There are Cebpb, Sp1, and Sox2 transcription factor binding sites in this region. Point-mutation experiment results showed that Sox2 negatively regulates the transcription of Cped1. Above results demonstrated that Cped1 is a key gene that regulates the formation of SSCs. Histone acetylation and transcription factor Sox2 participate in the regulation of Cped1.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 645-645
Author(s):  
Petter S. Woll ◽  
Colin H. Martin ◽  
Dan S. Kaufman

Abstract Derivation of multiple blood cell lineages from human embryonic stem cells (hESCs) clearly establishes these cells as an important model system to better characterize human hematopoietic ontogeny. Previously we have demonstrated development of myeloid, erythroid and lymphoid cells, as well as endothelial cells, from hESCs. However, the progenitor cells that give rise to these more mature cells remain poorly characterized. Here we use combined phenotypic and genetic analysis to discriminate between distinct progenitor cell populations. We also demonstrate that natural killer (NK) cells derived from these hESC-derived progenitors are capable of killing diverse tumors both in vitro and in vivo. By detailed flow cytometric analysis of differentiated hESCs over a defined time course, we identify development of two waves of CD34+ cells. The first wave consists of CD34brightCD31+Flk1+ cells, whereas the second wave consists of CD34dimCD45+ cells. Novel combined morphologic and phenotypic analysis by image scanning flow cytometry demonstrates these phenotypically different cell populations also are morphologically distinct. CD34brightCD31+ cells derived from hESCs appear to have a heterogeneous cell morphology, with an irregular cell shape, higher mean cell size and complex cytoplasmic organization. In contrast, CD34dimCD45+ cells are a more homogeneous cell population with a uniform spherical morphology and smaller cell size. Purified CD34brightCD31+Flk1+ cells express transcription factors associated with both the hematopoietic and endothelial lineages and can differentiate into both these lineages in vitro. In contrast, CD34dimCD45+ cells display a transcription profile suggestive of hematopoietic commitment and are significantly enriched for hematopoietic progenitors. Taken together, these results suggest that the CD34brightCD31+Flk1+ and CD34dimCD45+ cell populations are distinct cell lineages that represent early hemato-endothelial and hematopoietic precursors, respectively. Next, we better characterized the lymphoid developmental potential of the CD34dimCD45+ cells by the ability of these cells to differentiate into NK cells. Using a two-step differentiation process we find that hESC-derived NK cells express the wide repertoire of activating and inhibitory receptors similar to NK cells derived from other sources. We now demonstrate that these hESC-derived NK cells acquire cytolytic activity against breast cancer and glioma cell lines, as well as leukemia and lymphoma cells, in vitro. Finally, we have begun to test the in vivo efficacy of hESC-derived NK cells against established tumors. Here, luciferase (luc)-expressing K562 cells are inoculated into NOD/SCID mice, and several days later 2x106 NK cells derived from hESCs or from umbilical cord blood (UCB) cells are injected iv. The luc+ K562 cells allows serial bioluminescent imaging to follow growth of the tumor cells non-invasively over a prolonged time course. These studies demonstrate a beneficial effect of hESC-derived NK cells on tumor growth and metastasis, comparable to the effect of UCB-derived NK cells. Taken together, these studies characterize the earliest hematopoietic progenitors as they develop from hESCs during in vitro differentiation, providing a starting point to evaluate the effect of endogenous and exogenous factors on differentiation of distinct human hematopoietic lineages. This will translate into even more efficient derivation of NK cells from hESCs with a potential use for cancer immunotherapy.


2011 ◽  
Vol 437 (2) ◽  
pp. 345-355 ◽  
Author(s):  
Satoshi Yasuda ◽  
Tetsuya Hasegawa ◽  
Tetsuji Hosono ◽  
Mitsutoshi Satoh ◽  
Kei Watanabe ◽  
...  

An understanding of the mechanism that regulates the cardiac differentiation of pluripotent stem cells is necessary for the effective generation and expansion of cardiomyocytes as cell therapy products. In the present study, we have identified genes that modulate the cardiac differentiation of pluripotent embryonic cells. We isolated P19CL6 cell sublines that possess distinct properties in cardiomyogenesis and extracted 24 CMR (cardiomyogenesis-related candidate) genes correlated with cardiomyogenesis using a transcriptome analysis. Knockdown of the CMR genes by RNAi (RNA interference) revealed that 18 genes influence spontaneous contraction or transcript levels of cardiac marker genes in EC (embryonal carcinoma) cells. We also performed knockdown of the CMR genes in mouse ES (embryonic stem) cells and induced in vitro cardiac differentiation. Three CMR genes, AW551984, 2810405K02Rik (RIKEN cDNA 2810405K02 gene) and Cd302 (CD302 antigen), modulated the cardiac differentiation of both EC cells and ES cells. Depletion of AW551984 attenuated the expression of the early cardiac transcription factor Nkx2.5 (NK2 transcription factor related locus 5) without affecting transcript levels of pluripotency and early mesoderm marker genes during ES cell differentiation. Activation of Wnt/β-catenin signalling enhanced the expression of both AW551984 and Nkx2.5 in ES cells during embryoid body formation. Our findings indicate that AW551984 is a novel regulator of cardiomyogenesis from pluripotent embryonic cells, which links Wnt/β-catenin signalling to Nkx2.5 expression.


2017 ◽  
Vol 37 (4) ◽  
Author(s):  
Qisheng Zuo ◽  
Kai Jin ◽  
Yani Zhang ◽  
Jiuzhou Song ◽  
Bichun Li

The present study investigated the dynamic expression and regulatory mechanism of transforming growth factor β (TGF-β) signaling involved in embryonic stem cells (ESCs) differentiation into male germ cells. Candidate genes involved in TGF-β signaling pathway were screened from RNA-sequencing (RNA-seq), which were further validated by quantitative real-time PCR (qRT-PCR). Bone morphogenetic protein 4 (BMP4) was used to induce differentiation of ESCs in vitro. Inhibition of TGF-β signaling pathway was reflected by Western blot of SMAD2 and SMAD5 expression. Differentiating efficiency of germ cells was evaluated by immunofluorescence and fluorescence-activated cell sorting (FACS). Germ cell marker genes were assessed by qRT-PCR in the differentiation process, with activation or inhibition of TGF-β signaling pathway. In the process of in vitro induction, SMAD2 and SMAD5 were found to significantly up-regulated in BMP4 group versus the control and inhibition groups after 4 and 14 days. Expression of CKIT, CVH, DAZL, STRA8, and INTEGRIN α6 were significantly increased in the BMP4 group compared with the control group, while down-regulated in the inhibition groups. The proportion of germ cell-like cells was decreased from 17.9% to 2.2% after 4 days induction, and further decreased from 14.1% to 2.1% after 14 days induction. Correspondingly, expression of marker genes in germ cells was significantly lower. In vivo inhibition of TGF-β signaling pathway reduced germ cells formation from 5.5% to 1.6%, and down-regulated the expression of CKIT, CVH, DAZL, STRA8, and INTEGRIN α6. In conclusion, our study reveals the mechanism regulating spermatogonial stem cells (SSCs) and lays the basis for further understanding of the regulatory network.


Author(s):  
Kirsten Sjonnesen

Similar to embryonic stem cells (ESCs), iPSCs have the ability to differentiate into all three cell lineages, while not being subjected to the ethical complications attributed to ESCs. Our project goal was to develop a novel method of culturing murine iPSCs, reprogrammed from mouse embryonic fibroblasts, in large-scale quantities while maintaining their pluripotent characteristics and genomic integrity over a long term period. Stirred Suspension Bioreactors (SSBs) propose several benefits over static culture systems and facilitate the large-scale, economical expansion required for clinical studies. In our work with the SSB system, various techniques were used to promote and analyze the continued pluripotency of the murine iPSCs. Our results showed that iPSCs maintained their pluripotent state in SSBs and retained their ability to differentiate into different cell lineages after long term culture. miPSC were induced to differentiate into heart, bone and cartilage tissues at the end of maintenance period. The cumulative cell-fold expansion of iPSCs in suspension culture was 8.8×1010 cells, with 75% mean pluripotency at each passage. The cells expressed the major pluripotency markers as detected by RT-PCR. This study confirms the utility of the SSB system as a tool to cultivate large quantities of functional mouse iPSCs. SSBs have tremendous potential in future clinical applications and work has now begun to applying this system towards culturing human iPSCs. One potential application is the derivation of disease and patient derived iPSCs, which can recapitulate the disease phenotype for use as an in vitro model.


1993 ◽  
Vol 13 (1) ◽  
pp. 473-486
Author(s):  
G Keller ◽  
M Kennedy ◽  
T Papayannopoulou ◽  
M V Wiles

We report that embryonic stem cells efficiently undergo differentiation in vitro to mesoderm and hematopoietic cells and that this in vitro system recapitulates days 6.5 to 7.5 of mouse hematopoietic development. Embryonic stem cells differentiated as embryoid bodies (EBs) develop erythroid precursors by day 4 of differentiation, and by day 6, more than 85% of EBs contain such cells. A comparative reverse transcriptase-mediated polymerase chain reaction profile of marker genes for primitive endoderm (collagen alpha IV) and mesoderm (Brachyury) indicates that both cell types are present in the developing EBs as well in normal embryos prior to the onset of hematopoiesis. GATA-1, GATA-3, and vav are expressed in both the EBs and embryos just prior to and/or during the early onset of hematopoiesis, indicating that they could play a role in the early stages of hematopoietic development both in vivo and in vitro. The initial stages of hematopoietic development within the EBs occur in the absence of added growth factors and are not significantly influenced by the addition of a broad spectrum of factors, including interleukin-3 (IL-3), IL-1, IL-6, IL-11, erythropoietin, and Kit ligand. At days 10 and 14 of differentiation, EB hematopoiesis is significantly enhanced by the addition of both Kit ligand and IL-11 to the cultures. Kinetic analysis indicates that hematopoietic precursors develop within the EBs in an ordered pattern. Precursors of the primitive erythroid lineage appear first, approximately 24 h before precursors of the macrophage and definitive erythroid lineages. Bipotential neutrophil/macrophage and multilineage precursors appear next, and precursors of the mast cell lineage develop last. The kinetics of precursor development, as well as the growth factor responsiveness of these early cells, is similar to that found in the yolk sac and early fetal liver, indicating that the onset of hematopoiesis within the EBs parallels that found in the embryo.


Sign in / Sign up

Export Citation Format

Share Document