Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
2.
Stem Cell Res Ther ; 15(1): 76, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38475896

ABSTRACT

BACKGROUND: A traditional view is that stem cells (SCs) divide slowly. Meanwhile, both embryonic and pluripotent SCs display a shorter cell cycle duration (CCD) in comparison to more committed progenitors (CPs). METHODS: We examined the in vitro proliferation and cycling behavior of somatic adult human cells using live cell imaging of passage zero keratinocytes and single-cell RNA sequencing. RESULTS: We found two populations of keratinocytes: those with short CCD and protracted near exponential growth, and those with long CCD and terminal differentiation. Applying the ergodic principle, the comparative numbers of cycling cells in S phase in an enriched population of SCs confirmed a shorter CCD than CPs. Further, analysis of single-cell RNA sequencing of cycling adult human keratinocyte SCs and CPs indicated a shortening of both G1 and G2M phases in the SC. CONCLUSIONS: Contrary to the pervasive paradigm, SCs progress through cell cycle more quickly than more differentiated dividing CPs. Thus, somatic human adult keratinocyte SCs may divide infrequently, but divide rapidly when they divide. Additionally, it was found that SC-like proliferation persisted in vitro.


Subject(s)
Pluripotent Stem Cells , Adult , Humans , Cell Proliferation , Cell Cycle , Cell Division , Cell Differentiation , Phenotype , Keratinocytes/metabolism
3.
Sci Immunol ; 7(70): eabl9165, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35427179

ABSTRACT

Inflammatory conditions represent the largest class of chronic skin disease, but the molecular dysregulation underlying many individual cases remains unclear. Single-cell RNA sequencing (scRNA-seq) has increased precision in dissecting the complex mixture of immune and stromal cell perturbations in inflammatory skin disease states. We single-cell-profiled CD45+ immune cell transcriptomes from skin samples of 31 patients (7 atopic dermatitis, 8 psoriasis vulgaris, 2 lichen planus (LP), 1 bullous pemphigoid (BP), 6 clinical/histopathologically indeterminate rashes, and 7 healthy controls). Our data revealed active proliferative expansion of the Treg and Trm components and universal T cell exhaustion in human rashes, with a relative attenuation of antigen-presenting cells. Skin-resident memory T cells showed the greatest transcriptional dysregulation in both atopic dermatitis and psoriasis, whereas atopic dermatitis also demonstrated recurrent abnormalities in ILC and CD8+ cytotoxic lymphocytes. Transcript signatures differentiating these rash types included genes previously implicated in T helper cell (TH2)/TH17 diatheses, segregated in unbiased functional networks, and accurately identified disease class in untrained validation data sets. These gene signatures were able to classify clinicopathologically ambiguous rashes with diagnoses consistent with therapeutic response. Thus, we have defined major classes of human inflammatory skin disease at the molecular level and described a quantitative method to classify indeterminate instances of pathologic inflammation. To make this approach accessible to the scientific community, we created a proof-of-principle web interface (RashX), where scientists and clinicians can visualize their patient-level rash scRNA-seq-derived data in the context of our TH2/TH17 transcriptional framework.


Subject(s)
Dermatitis, Atopic , Exanthema , Psoriasis , Skin Diseases , Exanthema/metabolism , Exanthema/pathology , Humans , Skin , Skin Diseases/metabolism , Skin Diseases/pathology
4.
Aging Cell ; 20(2): e13310, 2021 02.
Article in English | MEDLINE | ID: mdl-33524216

ABSTRACT

With age, the epidermis becomes hypoplastic and hypoproliferative. Hypoproliferation due to aging has been associated with decreased stem cell (SC) self-renewal in multiple murine tissues. The fate of SC self-renewal divisions can be asymmetric (one SC, one committed progenitor) or symmetric (two SCs). Increased asymmetric SC self-renewal has been observed in inflammatory-mediated hyperproliferation, while increased symmetric SC self-renewal has been observed in cancers. We analyzed SC self-renewal divisions in aging human epidermis to better understand the role of SCs in the hypoproliferation of aging. In human subjects, neonatal to 78 years, there was an age-dependent decrease in epidermal basal layer divisions. The balance of SC self-renewal shifted toward symmetric SC self-renewal, with a decline in asymmetric SC self-renewal. Asymmetric SC divisions maintain epidermal stratification, and this decrease may contribute to the hypoplasia of aging skin. P53 decreases in multiple tissues with age, and p53 has been shown to promote asymmetric SC self-renewal. Fewer aged than adult ALDH+CD44+ keratinocyte SCs exhibited p53 expression and activity and Nutlin-3 (a p53 activator) returned p53 activity as well as asymmetric SC self-renewal divisions to adult levels. Nutlin-3 increased Notch signaling (NICD, Hes1) and DAPT inhibition of Notch activation prevented Nutlin-3 (p53)-induced asymmetric SC self-renewal divisions in aged keratinocytes. These studies indicate a role for p53 in the decreased asymmetric SC divisions with age and suggest that in aged keratinocytes, Notch is required for p53-induced asymmetric SC divisions.


Subject(s)
Cellular Senescence , Epidermis/metabolism , Tumor Suppressor Protein p53/metabolism , Asymmetric Cell Division , Cell Self Renewal , Humans , Imidazoles/pharmacology , Piperazines/pharmacology , Tumor Suppressor Protein p53/genetics
5.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33602818

ABSTRACT

Pruritus is a common symptom of inflammatory skin conditions, including atopic dermatitis (AD). Although primary sensory neurons that transmit pruritic signals are well-cataloged, little is known about the neuronal alterations that occur as a result of skin disruption in AD. To address this question, we examined the molecular and behavioral consequences of challenging Grhl3PAR2/+ mice, which overexpress PAR2 in suprabasal keratinocytes, with serial topical application of the environmental allergen house dust mite (HDM). We monitored behavior and used RNA sequencing, qPCR, and in situ hybridization to evaluate gene expression in trigeminal ganglia (TG), before and after HDM. We found that neither Grhl3PAR2/+ nor wild-type (WT) mice exhibited spontaneous scratching, and pruritogen-induced acute scratching did not differ. In contrast, HDM exacerbated scratching in Grhl3PAR2/+ mice. Despite the absence of scratching in untreated Grhl3PAR2/+ mice, several TG genes in these mice were up-regulated compared to WT. HDM treatment of the Grhl3PAR2/+ mice enhanced up-regulation of this set of genes and induced additional genes, many within the subset of TG neurons that express TRPV1. The same set of genes was up-regulated in HDM-treated Grhl3PAR2/+ mice that did not scratch, but at lesser magnitude. Finally, we recorded comparable transcriptional changes in IL31Tg mice, demonstrating that a common genetic program is induced in two AD models. Taken together, we conclude that transcriptional changes that occur in primary sensory neurons in dermatitis-susceptible animals underlie a genetic priming that not only sensitizes the animal to chronic allergens but also contributes to pruritus in atopic skin disease.


Subject(s)
Allergens/toxicity , DNA-Binding Proteins/physiology , Dermatitis, Atopic/pathology , Receptor, PAR-2/metabolism , Sensory Receptor Cells/pathology , Skin/pathology , Transcription Factors/physiology , Animals , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/metabolism , Disease Models, Animal , Mice , Mice, Transgenic , RNA-Seq , Receptor, PAR-2/genetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Skin/drug effects , Skin/innervation , Skin/metabolism
8.
iScience ; 23(10): 101582, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33205009

ABSTRACT

Inflammatory response heterogeneity has impeded high-resolution dissection of diverse immune cell populations during activation. We characterize mouse cutaneous immune cells by single-cell RNA sequencing, after inducing inflammation using imiquimod and oxazolone dermatitis models. We identify 13 CD45+ subpopulations, which broadly represent most functionally characterized immune cell types. Oxazolone pervasively upregulates Jak2/Stat3 expression across T cells and antigen-presenting cells (APCs). Oxazolone also induces Il4/Il13 expression in newly infiltrating basophils, and Il4ra and Ccl24, most prominently in APCs. In contrast, imiquimod broadly upregulates Il17/Il22 and Ccl4/Ccl5. A comparative analysis of single-cell inflammatory transcriptional responses reveals that APC response to oxazolone is tightly restricted by cell identity, whereas imiquimod enforces shared programs on multiple APC populations in parallel. These global molecular patterns not only contrast immune responses on a systems level but also suggest that the mechanisms of new sources of inflammation can eventually be deduced by comparison to known signatures.

9.
BMC Dermatol ; 19(1): 2, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670022

ABSTRACT

BACKGROUND: A defective skin barrier and bacterial colonization are two important factors in maintenance and progression of atopic dermatitis and chronic allergic/irritant hand dermatitis. A water-based lipid delivery system containing physiologic lipids was previously shown to be a useful adjunct in the treatment of hand dermatitis. We tested the ability of this formulation to penetrate into the viable epidermis and in addition assessed its antibacterial properties. METHODS: Epidermal penetration of the product was assessed by fluorescence microscopy. Recovery of Escherichia coli and Staphylococcus aureus MRSA from skin treated with Neosalus® foam was quantified. RESULTS: Components of Neosalus® penetrated the stratum corneum and were distributed throughout the viable epidermis. Neosalus® significantly decreased recovery of both Staphylococcus aureus and Escherichia coli from the skin surface. CONCLUSIONS: The ability of components of Neosalus® to be taken up into the viable epidermis and potentially made available for incorporation into the barrier lipids, combined with antibacterial properties, indicate that this formulation may be valuable not only in chronic hand dermatitis, but also in various other forms of dermatitis. TRIAL REGISTRATION: Current Controlled Trials ISRCTN18191379 , 28/12/2018, retrospectively registered.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Drug Delivery Systems/methods , Epidermis/drug effects , Adult , Anti-Bacterial Agents/pharmacokinetics , Dermatitis, Allergic Contact/drug therapy , Dermatitis, Allergic Contact/microbiology , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/microbiology , Emollients/administration & dosage , Emollients/pharmacokinetics , Epidermis/metabolism , Epidermis/microbiology , Escherichia coli/isolation & purification , Female , Healthy Volunteers , Humans , Lipids/chemistry , Male , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Permeability , Retrospective Studies , Skin Cream/administration & dosage , Skin Cream/pharmacokinetics , Treatment Outcome , Water/chemistry , Young Adult
10.
Cell Rep ; 25(4): 871-883, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30355494

ABSTRACT

Perturbations in the transcriptional programs specifying epidermal differentiation cause diverse skin pathologies ranging from impaired barrier function to inflammatory skin disease. However, the global scope and organization of this complex cellular program remain undefined. Here we report single-cell RNA sequencing profiles of 92,889 human epidermal cells from 9 normal and 3 inflamed skin samples. Transcriptomics-derived keratinocyte subpopulations reflect classic epidermal strata but also sharply compartmentalize epithelial functions such as cell-cell communication, inflammation, and WNT pathway modulation. In keratinocytes, ∼12% of assessed transcript expression varies in coordinate patterns, revealing undescribed gene expression programs governing epidermal homeostasis. We also identify molecular fingerprints of inflammatory skin states, including S100 activation in the interfollicular epidermis of normal scalp, enrichment of a CD1C+CD301A+ myeloid dendritic cell population in psoriatic epidermis, and IL1ßhiCCL3hiCD14+ monocyte-derived macrophages enriched in foreskin. This compendium of RNA profiles provides a critical step toward elucidating epidermal diseases of development, differentiation, and inflammation.


Subject(s)
Epidermis/metabolism , Epidermis/pathology , Inflammation/genetics , Inflammation/pathology , Single-Cell Analysis , Transcription, Genetic , Amphiregulin/pharmacology , Biomarkers/metabolism , Cell Aggregation/genetics , Cell Communication , Cell Differentiation , Cell Proliferation , Foreskin/cytology , Hair Follicle/metabolism , Humans , Inflammation/immunology , Keratinocytes/metabolism , Kinetics , Male , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , S100 Proteins/metabolism , Time Factors , Transcriptome/genetics , Wnt Proteins/metabolism
11.
Curr Probl Dermatol ; 54: 71-78, 2018.
Article in English | MEDLINE | ID: mdl-30130775

ABSTRACT

Intracellular pH influences proliferation and differentiation in a range of stem-like and progenitor cells, including embryonic stem cells, induced pluripotent stem cells, mesenchymal stem cells, and cancer stem cells. Sodium hydrogen exchanger (NHE1), a glycoprotein that plays a major role in regulating intracellular pH, has a major role in the proliferation and cell differentiation in multiple cell types. We review observations collected on the influence of pH on multiple stem-like cell populations. Altering pH, either intracellular or extracellular, can influence stem cell maintenance, self-renewal, differentiation, and pluripotency. Study of the influence of NHE1 and intracellular pH on epidermal stem cell behavior could lead to the discovery of new targets to use in order to manipulate stem cell divisions. This is highly relevant for skin conditions such as psoriasis, wound healing, and melanoma where stem cell proliferation and migration are key factors.


Subject(s)
Skin/cytology , Skin/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Hydrogen-Ion Concentration , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Skin/chemistry , Sodium-Hydrogen Exchanger 1/metabolism , Wound Healing/physiology
12.
Stem Cells ; 35(8): 2001-2007, 2017 08.
Article in English | MEDLINE | ID: mdl-28600817

ABSTRACT

The balance between asymmetric and symmetric stem cell (SC) divisions is key to tissue homeostasis, and dysregulation of this balance has been shown in cancers. We hypothesized that the balance between asymmetric cell divisions (ACDs) and symmetric cell divisions (SCDs) would be dysregulated in the benign hyperproliferation of psoriasis. We found that, while SCDs were increased in squamous cell carcinoma (SCC) (human and murine), ACDs were increased in the benign hyperproliferation of psoriasis (human and murine). Furthermore, while sonic hedgehog (linked to human cancer) and pifithrinα (p53 inhibitor) promoted SCDs, interleukin (IL)-1α and amphiregulin (associated with benign epidermal hyperproliferation) promoted ACDs. While there was dysregulation of the ACD:SCD ratio, no change in SC frequency was detected in epidermis from psoriasis patients, or in human keratinocytes treated with IL-1α or amphiregulin. We investigated the mechanism whereby immune alterations of psoriasis result in ACDs. IL17 inhibitors are effective new therapies for psoriasis. We found that IL17A increased ACDs in human keratinocytes. Additionally, studies in the imiquimod-induced psoriasis-like mouse model revealed that ACDs in psoriasis are IL17A-dependent. In summary, our studies suggest an association between benign hyperproliferation and increased ACDs. This work begins to elucidate the mechanisms by which immune alteration can induce keratinocyte hyperproliferation. Altogether, this work affirms that a finely tuned balance of ACDs and SCDs is important and that manipulating this balance may constitute an effective treatment strategy for hyperproliferative diseases. Stem Cells 2017;35:2001-2007.


Subject(s)
Asymmetric Cell Division , Interleukin-17/metabolism , Psoriasis/metabolism , Psoriasis/pathology , Aminoquinolines/pharmacology , Aminoquinolines/therapeutic use , Animals , Asymmetric Cell Division/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Homeostasis/drug effects , Humans , Imiquimod , Mice , Psoriasis/drug therapy
14.
Melanoma Res ; 25(2): 138-48, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25643237

ABSTRACT

Cells with aldehyde dehydrogenase activity (ALDH+) are the most tumorigenic cells in many cancers, including melanoma, making ALDH a candidate therapeutic target. We examined the effects of chemical inhibition of ALDH1 on the response of human melanoma xenografts to chemotherapy and the effects of ALDH1A1 RNA silencing on melanoma growth and metastasis. Addition of ALDH1 inhibitors (e.g. diethylaminobenzaldehyde) to dacarbazine chemotherapy, not only reduced tumor growth in vivo, but also resulted in a significant decrease in the number of residual cells capable of tumorigenesis. shRNA depletion of ALDH1A1 in melanoma cells resulted not only in a significant delay in appearance of xenograft melanomas and reduction in growth, but also significantly decreased the number of metastases and metastatic burden after lateral tail vein injections in mice. In summary, ALDH1 inhibition in combinatorial therapy with dacarbazine reduced the number of residual tumorigenic cells post-therapy and ALDH1A1 depletion had marked inhibitory effects on both melanoma growth and metastasis. These findings suggest that ALDH1 inhibition may not only be able to provide a therapeutic advantage in melanoma treatment, but may also prevent rapid relapse after therapy, as residual tumorigenic cells are fewer and metastatic ability is diminished.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Melanoma/therapy , RNAi Therapeutics , Retinal Dehydrogenase/antagonists & inhibitors , Skin Neoplasms/therapy , Aldehyde Dehydrogenase 1 Family , Animals , Cell Line, Tumor , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Melanoma/enzymology , Melanoma/genetics , Melanoma/secondary , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , RNA Interference , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Stem Cells ; 31(4): 786-99, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23335266

ABSTRACT

Like for other somatic tissues, isolation of a pure population of stem cells has been a primary goal in epidermal biology. We isolated discrete populations of freshly obtained human neonatal keratinocytes (HNKs) using previously untested candidate stem cell markers aldehyde dehydrogenase (ALDH) and CD44 as well as the previously studied combination of integrin α6 and CD71. An in vivo transplantation assay combined with limiting dilution analysis was used to quantify enrichment for long-term repopulating cells in the isolated populations. The ALDH(+) CD44(+) population was enriched 12.6-fold for long-term repopulating epidermal stem cells (EpiSCs) and the integrin α6(hi) CD71(lo) population was enriched 5.6-fold, over unfractionated cells. In addition to long-term repopulation, CD44(+) ALDH(+) keratinocytes exhibited other stem cell properties. CD44(+) ALDH(+) keratinocytes had self-renewal ability, demonstrated by increased numbers of cells expressing nuclear Bmi-1, serial transplantation of CD44(+) ALDH(+) cells, and holoclone formation in vitro. CD44(+) ALDH(+) cells were multipotent, producing greater numbers of hair follicle-like structures than CD44(-) ALDH(-) cells. Furthermore, 58% ± 7% of CD44(+) ALDH(+) cells exhibited label-retention. In vitro, CD44(+) ALDH(+) cells showed enhanced colony formation, in both keratinocyte and embryonic stem cell growth media. In summary, the CD44(+) ALDH(+) population exhibits stem cell properties including long-term epidermal regeneration, multipotency, label retention, and holoclone formation. This study shows that it is possible to quantify the relative number of EpiSCs in human keratinocyte populations using long-term repopulation as a functional test of stem cell nature. Future studies will combine isolation strategies as dictated by the results of quantitative transplantation assays, in order to achieve a nearly pure population of EpiSCs.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Epidermal Cells , Hyaluronan Receptors/metabolism , Keratinocytes/cytology , Stem Cells/cytology , Animals , Epidermis/metabolism , Flow Cytometry , Humans , In Vitro Techniques , Keratinocytes/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Regeneration/physiology , Stem Cells/metabolism
16.
J Invest Dermatol ; 132(11): 2522-33, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22763787

ABSTRACT

Maintenance, repair, and renewal of the epidermis are thought to depend on a pool of dedicated epidermal stem cells (EpiSCs). Like for many somatic tissues, isolation of a nearly pure population of stem cells is a primary goal in cutaneous biology. We used a quantitative transplantation assay, using injection of keratinocytes into subcutis combined with limiting dilution analysis, to assess the long-term repopulating ability of putative murine EpiSC populations. Putative EpiSC populations were isolated by FACS sorting. The CD133(+) population and the subpopulation of CD133(+) cells that exhibits high mitochondrial membrane potential (DΨm(hi)) were enriched for long-term repopulating EpiSCs versus unfractionated cells (3.9- and 5.2-fold, respectively). Evidence for self-renewal capacity was obtained by serial transplantation of long-term epidermal repopulating units derived from CD133(+) and CD133(+)ΔΨm(hi) keratinocytes. CD133(+) keratinocytes were multipotent and produced significantly more hair follicles than CD133(-) cells. CD133(+) cells were a subset of the previously described integrin α6(+)CD34(+) bulge cell population, and 28.9±8.6% were label-retaining cells. Thus, murine keratinocytes within the CD133(+) and CD133(+)ΔΨm(hi) populations contain EpiSCs that regenerate the epidermis for the long term, are self-renewing, multipotent, and label-retaining cells.


Subject(s)
Antigens, CD/metabolism , Epidermal Cells , Epidermis/physiology , Glycoproteins/metabolism , Keratinocytes/cytology , Multipotent Stem Cells/cytology , Peptides/metabolism , AC133 Antigen , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cell Proliferation , Fibroblasts/cytology , Fibroblasts/physiology , Flow Cytometry , Green Fluorescent Proteins/genetics , Integrin alpha6/metabolism , Keratinocytes/physiology , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Multipotent Stem Cells/physiology , Regeneration/physiology , Skin Transplantation , Transplantation, Homologous
17.
J Invest Dermatol ; 132(3 Pt 2): 797-810, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22205306

ABSTRACT

This is a chronicle of concepts in the field of epidermal stem cell biology and a historic look at their development over time. The past 25 years have seen the evolution of epidermal stem cell science, from first fundamental studies to a sophisticated science. The study of epithelial stem cell biology was aided by the ability to visualize the distribution of stem cells and their progeny through lineage analysis studies. The excellent progress we have made in understanding epidermal stem cell biology is discussed in this article. The challenges we still face in understanding epidermal stem cells include defining molecular markers for stem and progenitor sub-populations, determining the locations and contributions of the different stem cell niches, and mapping regulatory pathways of epidermal stem cell proliferation and differentiation. However, our rapidly evolving understanding of epidermal stem cells has many potential uses that promise to translate into improved patient therapy.


Subject(s)
Cell Biology/history , Dermatology/history , Pluripotent Stem Cells/cytology , Stem Cell Research/history , Epidermal Cells , History, 20th Century , History, 21st Century , Keratinocytes/cytology
18.
J Surg Oncol ; 103(6): 555-7, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21480249

ABSTRACT

While many solid tumors have been reported to contain stem cell-like cells termed cancer stem cells, the case for a melanoma stem cell has been debated over the last few years. Herein, we summarize current knowledge of melanoma-initiating cells and provide an update on recently gained knowledge regarding cancer stem cells and melanoma.


Subject(s)
Melanoma/physiopathology , Melanoma/secondary , Neoplastic Cells, Circulating/metabolism , Neoplastic Stem Cells/metabolism , Aldehyde Dehydrogenase/metabolism , Biomarkers, Tumor/metabolism , Humans , Neoplasm Metastasis/physiopathology
19.
J Invest Dermatol ; 130(12): 2799-808, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20739950

ABSTRACT

Despite increasing knowledge regarding melanoma-initiating cells (MICs), questions persist regarding the number and phenotypic nature of cells with tumor-generating capability. Evidence for a phenotypically distinct human MIC has been found in NOD/SCID (non-obese diabetic/severe combined immunodeficiency) mice. However, a phenotypically distinct human MIC was not found in the NOD/SCIDIl2rg(-)/(-) (NSG) mouse model. The demonstration of a distinct population of human melanoma cells responsible for tumorigenesis and tumor cell self-renewal would provide an important target for new melanoma therapies. In this study, we show a 100-fold range in MIC frequency in human melanoma (1 in 18,000 to 1 in 1,851,000 cells) in the NOD/SCID mouse. In this model, human melanoma cells with high aldehyde dehydrogenase (ALDH) activity were enriched 16.8-fold in tumorigenic cells over unfractionated (UNF) cells, such that 1 in 21,000 cells was a MIC. In the NSG mouse, the ALDH expressing cell population was enriched 100-fold in tumorigenic cells over UNF cells, such that one in four cells was a MIC. Xenograft melanomas that developed from ALDH(+) cells displayed robust self-renewal, whereas those from ALDH(-) cells showed minimal self-renewal in vitro. Thus, ALDH(+) melanoma cells have enhanced tumorigenicity over ALDH(-) cells and superior self-renewal ability.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Biomarkers, Tumor/metabolism , Melanoma/metabolism , Melanoma/secondary , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Animals , Cell Division/physiology , Cell Line, Tumor , Cell Separation/methods , Disease Models, Animal , Humans , Lymphatic Metastasis , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Transplantation, Heterologous , Tumor Cells, Cultured
20.
Methods Mol Biol ; 585: 421-32, 2010.
Article in English | MEDLINE | ID: mdl-19908020

ABSTRACT

Epidermal stem cells are of major importance for tissue homeostasis, wound repair, tumor initiation, and gene therapy. Here we describe an in vivo regeneration assay to test for the ability of keratinocyte progenitors to maintain an epidermis over the long-term in vivo. Limiting dilution analysis of epidermal repopulating units in this in vivo regeneration assay at sequential time points allows the frequency of short-term (transit amplifying cell) and long-term (stem cell) repopulating cells to be quantified.


Subject(s)
Epidermal Cells , Keratinocytes/cytology , Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Lineage , Cell Proliferation , Epidermis/physiology , Mice , Mice, SCID , Regeneration
SELECTION OF CITATIONS
SEARCH DETAIL