543 research outputs found

    Epidermal stem cell dynamics

    Get PDF
    Wong and Reiter have explored the possibility that hair follicle stem cells can give rise to basal cell carcinoma (BCC). They expressed in mice an inducible human BCC-derived oncogenic allele of Smoothened, SmoM2, under the control of either the cytokeratin 14 (K14) or cytokeratin 15 (K15) promoter. Smoothened encodes a G-protein-coupled receptor protein in the hedgehog pathway, the misregulation of which is implicated in BCC and other human cancers. Chronic injury is thought to be a contributing factor. The authors used K14 as a marker for stem cells in the basal layer of the epidermis and K15 as a marker for epidermal stem cells in the bulge of hair follicles. Upon activation, K14 construct-bearing mice readily formed BCC-like tumours, whereas this was not the case in K15:SmoM2-carrying mice. Upon wounding the epidermis, however, there was widespread BCC-like tumour formation in the skin of K15:SmoM2 mice. The authors conclude that wounding recruited bulge epidermal stem cells to the surface, allowing the cells to escape quiescence in the stem cell niche and to arrive in an environment where the hedgehog pathway becomes activated and therefore tumorigenesis is elicited. While this is a provocative result and the authors' conclusion may well be correct, there are alternative explanations

    The neural crest and neural crest defects

    Get PDF
    The neural crest is a fascinating embryonic tissue for more than one reason. In the adult organism it gives rise to an array of distinct cell types and tissues. It is responsible for many birth defects, familial diseases and malignancies, and it is amenable to the elucidation of mechanisms that regulate stem cell differentiation. Subsequent to an epithelial-to-mesenchymal transformation, neural crest cells emigrate from the dorsal aspect of the neural tube into the embryo, stop in different places, and eventually give rise to the autonomic and enteric nervous systems, most primary sensory neurons, endocrine cells, and melanocytes of the skin and internal organs. Furthermore, neural crest cells are involved in the septation of the cardiac outflow tract and they form the cranial mesenchyme, which gives rise to bone, cartilage, and connective tissue of the face and ventral neck. Environmental insults can lead to neural crest defects, including cleft lip/cleft palate and fetal alcohol syndrome. Familial diseases that affect neural crest derivatives include Hischsprung's disease and albinism, whereas well-known neural crest-related malignancies include melanoma, neuroblastoma, neurofibromatosis and pheochromocytoma. Migratory neural crest cells form a heteroge­neous population of cells that includes stem cells, cells with restricted developmental potentials, and cells that are committed to a particular lineage. Growth factors play important roles in the survival, proliferation and differentiation of neural crest cells. In particular, neurotrophin-3 (NTS), the ligand of the tyrosine kinase receptor, TrkC, promotes the survival of proliferating neural crest stem cells. TrkC-deficient mice develop cardiac outflow tract defects that resemble human birth defects, including persistent truncus arteriosus and transposition of the great vessels. In these animals, cardiac neural crest stem cells become fate-restricted precociously. Action of stem cell factor (SCF), the ligand of the tyrosine kinase receptor c-kit, affects multiple systems. Heterozygous c-kit deficient mice, termed 'Dominant spotting' (W), have anemia, are sterile and show changes in coat color (white spotting) due to defects in the hemopoietic system, germ cell line and melanogenesis, respectively. Inactivation of the human c-kit gene causes piebaldism, which is characterized by a white forelock, patchy hypopigmentation of the skin and rare sensoryneural deafness. In the quail neural crest, SCF supports the survival of neural crest stem cells, promotes their dif­ferentiation into small diameter sensory neurons, and, together with a neurotrophin, supports survival of me lanocyte precursors. In c-kit deficient newborn mice, up to one third of substance P-immunoreactive nociceptive sensory neurons are missing, thus confirming across species that SCF signaling is essential for the development of small diameter sensory neurons. In addition, the number of calcitonin gene-related-peptide (CGRP)-immunoreactive putative visceral afferent neurons in the dorsal root ganglion is diminished in these mice. The norepinephrine transporter (NET) is expressed in many embryonic tissues, including premigratory and migratory neural crest cells. Norepinephrine (NE) uptake by neural crest cells promotes their differentiation into noradrenergic neuroblasts in vitro. In contrast, NE uptake inhibitors, such as tricyclic antidepressants and the drug of abuse, cocaine, inhibit noradrenergic differentiation in vitro and in vivo, suggesting that these drugs can be teratogenic. Since NET is expressed in many embryonic tissues, NE transport may have functions also in non-neural cells during embryonic development. In summary, growth factors, alone and synergistically as well as NEplay multiple roles in neural crest development.Biomedical Reviews 2002; 13: 29-37

    Epidermal Neural Crest Stem Cell (EPI-NCSC)—Mediated Recovery of Sensory Function in a Mouse Model of Spinal Cord Injury

    Get PDF
    Here we show that epidermal neural crest stem cell (EPI-NCSC) transplants in the contused spinal cord caused a 24% improvement in sensory connectivity and a substantial recovery of touch perception. Furthermore we present a novel method for the ex vivo expansion of EPI-NCSC into millions of stem cells that takes advantage of the migratory ability of neural crest stem cells and is based on a new culture medium and the use of microcarriers. Functional improvement was shown by two independent methods, spinal somatosensory evoked potentials (SpSEP) and the Semmes-Weinstein touch test. Subsets of transplanted cells differentiated into myelinating oligodendrocytes. Unilateral injections of EPI-NCSC into the lesion of midline contused mouse spinal cords elicited bilateral improvements. Intraspinal EPI-NCSC did not migrate laterally in the spinal cord or invade the spinal roots and dorsal root ganglia, thus implicating diffusible factors. EPI-NCSC expressed neurotrophic factors, angiogenic factors, and metalloproteases. The strength of EPI-NCSC thus is that they can exert a combination of pertinent functions in the contused spinal cord, including cell replacement, neuroprotection, angiogenesis and modulation of scar formation. EPI-NCSC are uniquely qualified for cell-based therapy in spinal cord injury, as neural crest cells and neural tube stem cells share a higher order stem cell and are thus ontologically closely related

    Human Epidermal Neural Crest Stem Cells (hEPI-NCSC)—Characterization and Directed Differentiation into Osteocytes and Melanocytes

    Get PDF
    Here we describe the isolation, characterisation and ex-vivo expansion of human epidermal neural crest stem cells (hEPI-NCSC) and we provide protocols for their directed differentiation into osteocytes and melanocytes. hEPI-NCSC are neural crest-derived multipotent stem cells that persist into adulthood in the bulge of hair follicles. Multipotency and self-renewal were determined by in vitro clonal analyses. hEPI-NCSC generate all major neural crest derivatives, including bone/cartilage cells, neurons, Schwann cells, myofibroblasts and melanocytes. Furthermore, hEPI-NCSC express additional neural crest stem cell markers and global stem cell genes. To variable degrees and in a donor-dependent manner, hEPI-NCSC express the six essential pluripotency genes C-MYC, KLF4, SOX2, LIN28, OCT-4/POU5F1 and NANOG. hEPI-NCSC can be expanded ex vivo into millions of stem cells that remain mulitpotent and continue to express stem cell genes. The novelty of hEPI-NCSC lies in the combination of their highly desirable traits. hEPI-NCSC are embryonic remnants in a postnatal location, the bulge of hair follicles. Therefore they are readily accessible in the hairy skin by minimal invasive procedure. hEPI-NCSC are multipotent somatic stem cells that can be isolated reproducibly and with high yield. By taking advantage of their migratory ability, hEPI-NCSC can be isolated as a highly pure population of stem cells. hEPI-NCSC can undergo robust ex vivo expansion and directed differentiation. As somatic stem cells, hEPI-NCSC are conducive to autologous transplantation, which avoids graft rejection. Together, these traits make hEPI-NCSC novel and attractive candidates for future cell-based therapies and regenerative medicine

    Epidermal stem cells in orthopaedic regenerative medicine.

    Get PDF
    PMC3709750In the last decade, great advances have been made in epidermal stem cell studies at the cellular and molecular level. These studies reported various subpopulations and differentiations existing in the epidermal stem cell. Although controversies and unknown issues remain, epidermal stem cells possess an immune-privileged property in transplantation together with easy accessibility, which is favorable for future clinical application. In this review, we will summarize the biological characteristics of epidermal stem cells, and their potential in orthopedic regenerative medicine. Epidermal stem cells play a critical role via cell replacement, and demonstrate significant translational potential in the treatment of orthopedic injuries and diseases, including treatment for wound healing, peripheral nerve and spinal cord injury, and even muscle and bone remodeling.JH Libraries Open Access Fun

    Semiclassical Construction of Random Wave Functions for Confined Systems

    Get PDF
    We develop a statistical description of chaotic wavefunctions in closed systems obeying arbitrary boundary conditions by combining a semiclassical expression for the spatial two-point correlation function with a treatment of eigenfunctions as Gaussian random fields. Thereby we generalize Berry's isotropic random wave model by incorporating confinement effects through classical paths reflected at the boundaries. Our approach allows to explicitly calculate highly non-trivial statistics, such as intensity distributions, in terms of usually few short orbits, depending on the energy window considered. We compare with numerical quantum results for the Africa billiard and derive non-isotropic random wave models for other prominent confinement geometries.Comment: To be submitted to Physical Review Letter

    Monoclonal antibodies made to chick mesencephalic neural crest cells and to ciliary ganglion neurons identify a common antigen on the neurons and a neural crest subpopulation

    Full text link
    We previously reported the production of monoclonal antibodies (Mabs) that identified cell surface components of cultured chick and quail ciliary ganglion (CG) neurons and of a subpopulation of neural crest (NC) cells from 31-hr chick embryos (stage 9). Here we demonstrate that another Mab, CG-14, which was prepared to nitrocellulose-immobilized, lightly fixed (0.125% paraformaldehyde) mesencephalic NC cells from 31-hr (stage 9) chick embryos, labels the same antigen(s) recognized by CG-1 and CG-4 on both the CG neurons and the subpopulation of NC cells. All three Mabs label a polypeptide of 75 kD on Western blots of one-dimensional SDS-polyacrylamide gels. CG-14 blocked the binding of CG-1 and/or CG-4 to the 75 kD band on Western blots and blocked the binding of CG-1 and CG-4 to CG and NC cells. CG-1 and/or CG-4 antibodies, in turn, blocked the binding of CG-14 to Western blots, as well as NC and CG cells. We had previously shown that antibodies CG-1 and CG-4 were synergistically cytotoxic for the majority (95%) of cultured CG neurons in vitro in the presence of guinea pig complement. Here we show that the antibodies, which are both of the Γ2a subclass, are also cytotoxic for the NC cells that they label in vitro. After the cells are ablated in culture, no other cells bearing the antigen(s) recognized by any of the three Mabs appear over a 2.5-week period. CG-14, however, is not cytotoxic for either the CG or NC cell populations alone or in combinations with CG-1 or CG-4. These results confirm our original observation that cultured CG neurons and NC cells share cell-surface antigen(s). The antigen recognized by all three Mabs appears to be the same whether the immunogen used to produce the antibodies was CG neurons or NC cells. This finding encourages us to continue tests of the hypothesis that the subpopulation of mesencephalic neural crest cells contributes to the formation of the ciliary ganglion in the embryo. Further characterization of the antigen appears in the accompanying paper.Peer Reviewedhttp://deepblue.lib.umich.edu/bitstream/2027.42/50219/1/490210204_ftp.pd

    Human neural crest cells display molecular and phenotypic hallmarks of stem cells

    Get PDF
    The fields of both developmental and stem cell biology explore how functionally distinct cell types arise from a self-renewing founder population. Multipotent, proliferative human neural crest cells (hNCC) develop toward the end of the first month of pregnancy. It is assumed that most differentiate after migrating throughout the organism, although in animal models neural crest stem cells reportedly persist in postnatal tissues. Molecular pathways leading over time from an invasive mesenchyme to differentiated progeny such as the dorsal root ganglion, the maxillary bone or the adrenal medulla are altered in many congenital diseases. To identify additional components of such pathways, we derived and maintained self-renewing hNCC lines from pharyngulas. We show that, unlike their animal counterparts, hNCC are able to self-renew ex vivo under feeder-free conditions. While cross species comparisons showed extensive overlap between human, mouse and avian NCC transcriptomes, some molecular cascades are only active in the human cells, correlating with phenotypic differences. Furthermore, we found that the global hNCC molecular profile is highly similar to that of pluripotent embryonic stem cells when compared with other stem cell populations or hNCC derivatives. The pluripotency markers NANOG, POU5F1 and SOX2 are also expressed by hNCC, and a small subset of transcripts can unambiguously identify hNCC among other cell types. The hNCC molecular profile is thus both unique and globally characteristic of uncommitted stem cells
    corecore