30 research outputs found

    Christopher Stanislaus Potten

    Get PDF

    Dermal Pericytes Exhibit Declined Ability to Promote Human Skin Regeneration with Ageing in 3D Organotypic Culture Models.

    Get PDF
    The well documented decline in the regenerative ability of ageing human skin has been attributed to many factors including genomic instability, telomere shortening, poor nutrient sensing, cellular senescence, and stem cell exhaustion. However, a role for the dermal cellular and molecular microenvironment in skin ageing is just emerging. We previously showed that dermal pericytes co-operate with fibroblasts to improve human skin regeneration in an organotypic skin culture model, and even do so in the absence of fibroblasts. Here, we report that the number of dermal cells, particularly pericytes, declines significantly in human skin of donors aged > 50 years. Notably, aged pericytes promoted epidermal regeneration of neonatal keratinocytes in organotypic cultures and the resulting epithelium exhibited a Ki67+/ΔNp63+ basal layer and terminal differentiation. However, the epithelium lacked several features of homeostasis displaying lower levels of ΔNp63 expression, decreased LAMA5 deposition at the dermo-epidermal junction, and the absence of basement membrane and hemi-desmosome assembly. We conclude that a decline in pericyte incidence and function contribute to an impaired epidermal microenvironment and poor skin regeneration with ageing in the human skin

    Dermal contributions to human interfollicular epidermal architecture and self-renewal

    Get PDF
    © 2015 by the authors; licensee MDPI, Basel, Switzerland. The human interfollicular epidermis is renewed throughout life by populations of proliferating basal keratinocytes. Though interfollicular keratinocyte stem cells have been identified, it is not known how self-renewal in this compartment is spatially organized. At the epidermal-dermal junction, keratinocytes sit atop a heterogeneous mix of dermal cells that may regulate keratinocyte self-renewal by influencing local tissue architecture and signalling microenvironments. Focusing on the rete ridges and complementary dermal papillae in human skin, we review the identity and organisation of abundant dermal cells types and present evidence for interactions between the dermal microenvironment and the interfollicular keratinocytes

    Pericytes promote skin regeneration by inducing epidermal cell polarity and planar cell divisions.

    Get PDF
    The cellular and molecular microenvironment of epithelial stem/progenitor cells is critical for their long-term self-renewal. We demonstrate that mesenchymal stem cell-like dermal microvascular pericytes are a critical element of the skin's microenvironment influencing human skin regeneration using organotypic models. Specifically, pericytes were capable of promoting homeostatic skin tissue renewal by conferring more planar cell divisions generating two basal cells within the proliferative compartment of the human epidermis, while ensuring complete maturation of the tissue both spatially and temporally. Moreover, we provide evidence supporting the notion that BMP-2, a secreted protein preferentially expressed by pericytes in human skin, confers cell polarity and planar divisions on epidermal cells in organotypic cultures. Our data suggest that human skin regeneration is regulated by highly conserved mechanisms at play in other rapidly renewing tissues such as the bone marrow and in lower organisms such as Drosophila. Our work also provides the means to significantly improve ex vivo skin tissue regeneration for autologous transplantation

    Identification of a Cell Surface Protein with a Role in Stimulating Human Keratinocyte Proliferation, Expressed During Development and Carcinogenesis

    Get PDF
    In an attempt to define cell surface molecules with an important role in the development of squamous cell carcinomas (SCCs), we generated monoclonal antibodies (MoAbs) to a human keratinocyte cell line (FEP18-11-T1) capable of giving rise to SCCs in nude mice. MoAb 10G7 was selected for further study because it bound to a cell surface component preferentially expressed by this cell line as compared with normal human foreskin keratinocytes. This MoAb recognizes a cell surface protein (10G7 antigen) that is not detectable on normal keratinocytes in the foreskin in vivo, but whose expression is induced when the keratinocytes are dissociated from this tissue and placed in culture. Interestingly, the 10G7 antigen is downregulated upon keratinocyte differentiation in vitro. Consistent with its expression in hyper-proliferative epithella in vitro, 10G7 antigen exhibited a classic oncofetal pattern of expression in vivo. Thus, although no reactivity was obtained with MoAb 10G7 in the epithelia of normal foreskin or cervical tissue, strong reactivity was detected in epithelia from genital lesions ranging from benign warts to invasive SCCs. Epidermis from developing fetal tissue also exhibited strong reactivity with MoAb 10G7. We have been able to demonstrate that this MoAb is capable of stimulating FEP18–11-T1 keratinocyte proliferation in vitro in a concentration-dependent manner in the absence of growth factors, suggesting that the 10G7 antigen may play an important role in regulating cellular proliferation during development and in carcinogenesis in epithelial tissues

    Qualitative in vivo Bioluminescence Imaging.

    No full text
    Bioluminescence imaging (BLI) technology is an advanced method of carrying out molecular imaging on live laboratory animals in vivo. This powerful technique is widely-used in studying a variety of biological processes, and it has been an ideal tool in exploring tumor growth and metastatic spread in real-time. This technique ensures the optimal use of laboratory animal resources, particularly the ethical principle of reduction in animal use, given its non-invasive nature, ensuring that ongoing biological processes can be studied over time in the same animal, without the need to euthanize groups of mice at specific time points. In this protocol, the luciferase imaging technique was developed to study the effect of co-inoculating pericytes (contractile, αSMA mesenchymal stem cell-like cells, located abluminally in microvessels) on the growth and metastatic spread of ovarian cancers using an aggressive ovarian cancer cell line–OVCAR-5–as an example
    corecore