Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nature ; 547(7664): 453-457, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28678785

ABSTRACT

Plasticity of the cell state has been proposed to drive resistance to multiple classes of cancer therapies, thereby limiting their effectiveness. A high-mesenchymal cell state observed in human tumours and cancer cell lines has been associated with resistance to multiple treatment modalities across diverse cancer lineages, but the mechanistic underpinning for this state has remained incompletely understood. Here we molecularly characterize this therapy-resistant high-mesenchymal cell state in human cancer cell lines and organoids and show that it depends on a druggable lipid-peroxidase pathway that protects against ferroptosis, a non-apoptotic form of cell death induced by the build-up of toxic lipid peroxides. We show that this cell state is characterized by activity of enzymes that promote the synthesis of polyunsaturated lipids. These lipids are the substrates for lipid peroxidation by lipoxygenase enzymes. This lipid metabolism creates a dependency on pathways converging on the phospholipid glutathione peroxidase (GPX4), a selenocysteine-containing enzyme that dissipates lipid peroxides and thereby prevents the iron-mediated reactions of peroxides that induce ferroptotic cell death. Dependency on GPX4 was found to exist across diverse therapy-resistant states characterized by high expression of ZEB1, including epithelial-mesenchymal transition in epithelial-derived carcinomas, TGFß-mediated therapy-resistance in melanoma, treatment-induced neuroendocrine transdifferentiation in prostate cancer, and sarcomas, which are fixed in a mesenchymal state owing to their cells of origin. We identify vulnerability to ferroptic cell death induced by inhibition of a lipid peroxidase pathway as a feature of therapy-resistant cancer cells across diverse mesenchymal cell-state contexts.


Subject(s)
Glutathione Peroxidase/metabolism , Lipid Peroxidation/drug effects , Neoplasms/drug therapy , Neoplasms/enzymology , Cadherins/metabolism , Cell Death , Cell Line, Tumor , Cell Lineage , Cell Transdifferentiation , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition , Humans , Iron/metabolism , Lipid Peroxides/metabolism , Male , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/metabolism , Melanoma/pathology , Mesoderm/drug effects , Mesoderm/enzymology , Mesoderm/metabolism , Mesoderm/pathology , Neoplasms/genetics , Neoplasms/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteomics , Proto-Oncogene Proteins B-raf/genetics , Reproducibility of Results , Zinc Finger E-box-Binding Homeobox 1/genetics
2.
J Biomed Opt ; 22(12): 1-10, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29222855

ABSTRACT

Chemical sun filters are commonly used as active ingredients in sunscreens due to their efficient absorption of ultraviolet (UV) radiation. Yet, it is known that these compounds can photochemically react with UV light and generate reactive oxygen species and oxidative stress in vitro, though this has yet to be validated in vivo. One label-free approach to probe oxidative stress is to measure and compare the relative endogenous fluorescence generated by cellular coenzymes nicotinamide adenine dinucleotides and flavin adenine dinucleotides. However, chemical sun filters are fluorescent, with emissive properties that contaminate endogenous fluorescent signals. To accurately distinguish the source of fluorescence in ex vivo skin samples treated with chemical sun filters, fluorescence lifetime imaging microscopy data were processed on a pixel-by-pixel basis using a non-Euclidean separation algorithm based on Mahalanobis distance and validated on simulated data. Applying this method, ex vivo samples exhibited a small oxidative shift when exposed to sun filters alone, though this shift was much smaller than that imparted by UV irradiation. Given the need for investigative tools to further study the clinical impact of chemical sun filters in patients, the reported methodology may be applied to visualize chemical sun filters and measure oxidative stress in patients' skin.


Subject(s)
Microscopy, Fluorescence , Oxidative Stress/drug effects , Skin/drug effects , Sunscreening Agents/pharmacology , Humans , Skin/pathology
3.
Cell Rep ; 19(11): 2177-2184, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28614705

ABSTRACT

The presence of dark melanin (eumelanin) within human epidermis represents one of the strongest predictors of low skin cancer risk. Topical rescue of eumelanin synthesis, previously achieved in "redhaired" Mc1r-deficient mice, demonstrated significant protection against UV damage. However, application of a topical strategy for human skin pigmentation has not been achieved, largely due to the greater barrier function of human epidermis. Salt-inducible kinase (SIK) has been demonstrated to regulate MITF, the master regulator of pigment gene expression, through its effects on CRTC and CREB activity. Here, we describe the development of small-molecule SIK inhibitors that were optimized for human skin penetration, resulting in MITF upregulation and induction of melanogenesis. When topically applied, pigment production was induced in Mc1r-deficient mice and normal human skin. These findings demonstrate a realistic pathway toward UV-independent topical modulation of human skin pigmentation, potentially impacting UV protection and skin cancer risk.


Subject(s)
Melanins/metabolism , Skin/metabolism , Ultraviolet Rays/adverse effects , Administration, Topical , Animals , Humans , Melanins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
Sci Rep ; 6: 37986, 2016 11 28.
Article in English | MEDLINE | ID: mdl-27892516

ABSTRACT

Melanoma is the most deadly form of skin cancer with a yearly global incidence over 232,000 patients. Individuals with fair skin and red hair exhibit the highest risk for developing melanoma, with evidence suggesting the red/blond pigment known as pheomelanin may elevate melanoma risk through both UV radiation-dependent and -independent mechanisms. Although the ability to identify, characterize, and monitor pheomelanin within skin is vital for improving our understanding of the underlying biology of these lesions, no tools exist for real-time, in vivo detection of the pigment. Here we show that the distribution of pheomelanin in cells and tissues can be visually characterized non-destructively and noninvasively in vivo with coherent anti-Stokes Raman scattering (CARS) microscopy, a label-free vibrational imaging technique. We validated our CARS imaging strategy in vitro to in vivo with synthetic pheomelanin, isolated melanocytes, and the Mc1re/e, red-haired mouse model. Nests of pheomelanotic melanocytes were observed in the red-haired animals, but not in the genetically matched Mc1re/e; Tyrc/c ("albino-red-haired") mice. Importantly, samples from human amelanotic melanomas subjected to CARS imaging exhibited strong pheomelanotic signals. This is the first time, to our knowledge, that pheomelanin has been visualized and spatially localized in melanocytes, skin, and human amelanotic melanomas.


Subject(s)
Melanins/analysis , Melanocytes/metabolism , Melanoma, Amelanotic/metabolism , Molecular Imaging/methods , Spectrum Analysis, Raman/methods , Animals , Ear/diagnostic imaging , Flow Cytometry/methods , Humans , Melanins/metabolism , Mice, Mutant Strains , Mice, Transgenic , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Receptor, Melanocortin, Type 1/genetics , Skin/diagnostic imaging , Skin/metabolism , Skin Neoplasms/metabolism
5.
Cancer Cell ; 28(6): 773-784, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26678339

ABSTRACT

Heterozygous mutation of IDH1 in cancers modifies IDH1 enzymatic activity, reprogramming metabolite flux and markedly elevating 2-hydroxyglutarate (2-HG). Here, we found that 2-HG depletion did not inhibit growth of several IDH1 mutant solid cancer types. To identify other metabolic therapeutic targets, we systematically profiled metabolites in endogenous IDH1 mutant cancer cells after mutant IDH1 inhibition and discovered a profound vulnerability to depletion of the coenzyme NAD+. Mutant IDH1 lowered NAD+ levels by downregulating the NAD+ salvage pathway enzyme nicotinate phosphoribosyltransferase (Naprt1), sensitizing to NAD+ depletion via concomitant nicotinamide phosphoribosyltransferase (NAMPT) inhibition. NAD+ depletion activated the intracellular energy sensor AMPK, triggered autophagy, and resulted in cytotoxicity. Thus, we identify NAD+ depletion as a metabolic susceptibility of IDH1 mutant cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cytokines/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Isocitrate Dehydrogenase/genetics , Mutation , NAD/deficiency , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/drug effects , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Cytokines/metabolism , Energy Metabolism/drug effects , Enzyme Activation , Female , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Glutarates/metabolism , HEK293 Cells , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/metabolism , Metabolomics/methods , Mice, SCID , Molecular Targeted Therapy , Nicotinamide Phosphoribosyltransferase/metabolism , Pentosyltransferases/metabolism , Signal Transduction/drug effects , Spheroids, Cellular , Time Factors , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
J Invest Dermatol ; 134(1): 16-17, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24352080

ABSTRACT

In the current issue, two articles highlight the impact of melanocyte transcription factor (MITF) on melanoma development. In the first, Lister et al. reveal in vivo proof of MITF directly regulating tumor development in BRAF(V600E) melanomas. In the second, Sturm et al. present a clinical trial that emphasizes the importance of the recently discovered E318K MITF germline mutation in patients with multiple primary melanomas.


Subject(s)
Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Nevus/genetics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Zebrafish Proteins/genetics , Animals , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL