Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
2.
bioRxiv ; 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38106189

ABSTRACT

Cutaneous melanomas are clinically and histologically heterogeneous. Most display activating mutations in Braf or Nras and complete loss of function of one or more tumor suppressor genes. Mouse models that replicate such mutations produce fast-growing, pigmented tumors. However, mice that combine Braf activation with only heterozygous loss of Pten also produce tumors and, as we show here, in an Albino background this occurs even with Braf activation alone. Such tumors arise rarely, grow slowly, and express low levels of pigmentation genes. The timing of their appearance was consistent with a single step stochastic event, but no evidence could be found that it required de novo mutation, suggesting instead the involvement of an epigenetic transition. Single-cell transcriptomic analysis revealed such tumors to be heterogeneous, including a minor cell type we term LNM ( L ow-pigment, N eural- and extracellular M atrix-signature) that displays gene expression resembling "neural crest"-like cell subsets detected in the fast-growing tumors of more heavily-mutated mice, as well as in human biopsy and xenograft samples. We provide evidence that LNM cells pre-exist in normal skin, are expanded by Braf activation, can transition into malignant cells, and persist with malignant cells through multiple rounds of transplantation. We discuss the possibility that LNM cells not only serve as a pre-malignant state in the production of some melanomas, but also as an important intermediate in the development of drug resistance.

3.
bioRxiv ; 2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37645823

ABSTRACT

Punch grafting procedures, where small pieces of normal skin are transplanted into stable vitiligo patches, results in repigmentation in only half of patients treated, yet the factors that determine whether a patient responds to treatment or not are still unknown. Reflectance confocal microscopy (RCM) is adept at visualizing melanocyte migration and epidermal changes over large areas while multiphoton microscopy (MPM) can capture metabolic changes in keratinocytes. With the overall goal of identifying optical biomarkers for early treatment response, we followed 12 vitiligo lesions undergoing punch grafting. Dendritic melanocytes adjacent to the graft site were observed before clinical evidence of repigmentation in treatment responsive patients but not in treatment non-responsive patients, suggesting that the early visualization of melanocytes is indicative of a therapeutic response. Keratinocyte metabolic changes in vitiligo skin adjacent to the graft site also correlated with treatment response, indicating that a keratinocyte microenvironment that more closely resembles normal skin is more hospitable for migrating melanocytes. Taken together, these studies suggest that successful melanocyte transplantation requires both the introduction of new melanocytes and modulation of the local tissue microenvironment.

4.
Nature ; 618(7966): 808-817, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37344645

ABSTRACT

Niche signals maintain stem cells in a prolonged quiescence or transiently activate them for proper regeneration1. Altering balanced niche signalling can lead to regenerative disorders. Melanocytic skin nevi in human often display excessive hair growth, suggesting hair stem cell hyperactivity. Here, using genetic mouse models of nevi2,3, we show that dermal clusters of senescent melanocytes drive epithelial hair stem cells to exit quiescence and change their transcriptome and composition, potently enhancing hair renewal. Nevus melanocytes activate a distinct secretome, enriched for signalling factors. Osteopontin, the leading nevus signalling factor, is both necessary and sufficient to induce hair growth. Injection of osteopontin or its genetic overexpression is sufficient to induce robust hair growth in mice, whereas germline and conditional deletions of either osteopontin or CD44, its cognate receptor on epithelial hair cells, rescue enhanced hair growth induced by dermal nevus melanocytes. Osteopontin is overexpressed in human hairy nevi, and it stimulates new growth of human hair follicles. Although broad accumulation of senescent cells, such as upon ageing or genotoxic stress, is detrimental for the regenerative capacity of tissue4, we show that signalling by senescent cell clusters can potently enhance the activity of adjacent intact stem cells and stimulate tissue renewal. This finding identifies senescent cells and their secretome as an attractive therapeutic target in regenerative disorders.


Subject(s)
Hair , Melanocytes , Signal Transduction , Animals , Mice , Hair/cytology , Hair/growth & development , Hair Follicle/cytology , Hair Follicle/physiology , Hyaluronan Receptors/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Nevus/metabolism , Nevus/pathology , Osteopontin/metabolism , Stem Cells/cytology
5.
J Med Chem ; 66(8): 5981-6001, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37026468

ABSTRACT

CDC42 GTPases (RHOJ, CDC42, and RHOQ) are overexpressed in multiple tumor types and activate pathways critical for tumor growth, angiogenesis, and metastasis. Recently, we reported the discovery of a novel lead compound, ARN22089, which blocks the interaction of CDC42 GTPases with specific downstream effectors. ARN22089 blocks tumor growth in BRAF mutant mouse melanoma models and patient-derived xenografts (PDXs) in vivo. ARN22089 also inhibits tumor angiogenesis in three-dimensional vascularized microtumor models in vitro. Notably, ARN22089 belongs to a novel class of trisubstituted pyrimidines. Based on these results, we describe an extensive structure-activity relationship of ∼30 compounds centered on ARN22089. We discovered and optimized two novel inhibitors (27, ARN25062, and 28, ARN24928), which are optimal back-up/follow-up leads with favorable drug-like properties and in vivo efficacy in PDX tumors. These findings further demonstrate the potential of this class of CDC42/RHOJ inhibitors for cancer treatment, with lead candidates ready for advanced preclinical studies.


Subject(s)
Neoplasms , rho GTP-Binding Proteins , Animals , Humans , Mice , Cell Line, Tumor , Neovascularization, Pathologic , p21-Activated Kinases/metabolism , Protein Binding
6.
J Am Acad Dermatol ; 88(2): 395-403, 2023 02.
Article in English | MEDLINE | ID: mdl-36370907

ABSTRACT

BACKGROUND: Vitiligo is a chronic autoimmune disorder characterized by depigmented patches of the skin. OBJECTIVE: To evaluate the efficacy and safety of ritlecitinib, an oral JAK3 (Janus kinase)/TEC (tyrosine kinase expressed in hepatocelluar carcinoma) inhibitor, in patients with active nonsegmental vitiligo in a phase 2b trial (NCT03715829). METHODS: Patients were randomized to once-daily oral ritlecitinib ± 4-week loading dose (200/50 mg, 100/50 mg, 30 mg, or 10 mg) or placebo for 24 weeks (dose-ranging period). Patients subsequently received ritlecitinib 200/50 mg daily in a 24-week extension period. The primary efficacy endpoint was percent change from baseline in Facial-Vitiligo Area Scoring Index at week 24. RESULTS: A total of 364 patients were treated in the dose-ranging period. Significant differences from placebo in percent change from baseline in Facial-Vitiligo Area Scoring Index were observed for the ritlecitinib 50 mg groups with (-21.2 vs 2.1; P < .001) or without (-18.5 vs 2.1; P < .001) a loading dose and ritlecitinib 30 mg group (-14.6 vs 2.1; P = .01). Accelerated improvement was observed after treatment with ritlecitinib 200/50 mg in the extension period (n = 187). No dose-dependent trends in treatment-emergent or serious adverse events were observed across the 48-week treatment. LIMITATIONS: Patients with stable vitiligo only were excluded. CONCLUSIONS: Oral ritlecitinib was effective and well tolerated over 48 weeks in patients with active nonsegmental vitiligo.


Subject(s)
Vitiligo , Humans , Vitiligo/drug therapy , Vitiligo/pathology , Double-Blind Method , Skin/pathology , Janus Kinases , Protein Kinase Inhibitors/adverse effects , Chronic Disease , Treatment Outcome
8.
J Chem Inf Model ; 62(12): 3023-3033, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35679463

ABSTRACT

Here, we show that alchemical free energy calculations can quantitatively compute the effect of mutations at the protein-protein interface. As a test case, we have used the protein complex formed by the small Rho-GTPase CDC42 and its downstream effector PAK1, a serine/threonine kinase. Notably, the CDC42/PAK1 complex offers a wealth of structural, mutagenesis, and binding affinity data because of its central role in cellular signaling and cancer progression. In this context, we have considered 16 mutations in the CDC42/PAK1 complex and obtained excellent agreement between computed and experimental data on binding affinity. Importantly, we also show that a careful analysis of the side-chain conformations in the mutated amino acids can considerably improve the computed estimates, solving issues related to sampling limitations. Overall, this study demonstrates that alchemical free energy calculations can conveniently be integrated into the design of experimental mutagenesis studies.


Subject(s)
Protein Serine-Threonine Kinases , p21-Activated Kinases , Mutagenesis , Mutation , Proteins/genetics , p21-Activated Kinases/genetics
9.
JCI Insight ; 7(13)2022 07 08.
Article in English | MEDLINE | ID: mdl-35653192

ABSTRACT

Vitiligo is an autoimmune skin disease characterized by the destruction of melanocytes by autoreactive CD8+ T cells. Melanocyte destruction in active vitiligo is mediated by CD8+ T cells, but the persistence of white patches in stable disease is poorly understood. The interaction between immune cells, melanocytes, and keratinocytes in situ in human skin has been difficult to study due to the lack of proper tools. We combine noninvasive multiphoton microscopy (MPM) imaging and single-cell RNA-Seq (scRNA-Seq) to identify subpopulations of keratinocytes in stable vitiligo patients. We show that, compared with nonlesional skin, some keratinocyte subpopulations are enriched in lesional vitiligo skin and shift their energy utilization toward oxidative phosphorylation. Systematic investigation of cell-to-cell communication networks show that this small population of keratinocyte secrete CXCL9 and CXCL10 to potentially drive vitiligo persistence. Pseudotemporal dynamics analyses predict an alternative differentiation trajectory that generates this new population of keratinocytes in vitiligo skin. Further MPM imaging of patients undergoing punch grafting treatment showed that keratinocytes favoring oxidative phosphorylation persist in nonresponders but normalize in responders. In summary, we couple advanced imaging with transcriptomics and bioinformatics to discover cell-to-cell communication networks and keratinocyte cell states that can perpetuate inflammation and prevent repigmentation.


Subject(s)
Vitiligo , CD8-Positive T-Lymphocytes , Humans , Keratinocytes , Melanocytes , Skin
11.
Cell Rep ; 39(1): 110641, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35385746

ABSTRACT

CDC42 family GTPases (RHOJ, RHOQ, CDC42) are upregulated but rarely mutated in cancer and control both the ability of tumor cells to invade surrounding tissues and the ability of endothelial cells to vascularize tumors. Here, we use computer-aided drug design to discover a chemical entity (ARN22089) that has broad activity against a panel of cancer cell lines, inhibits S6 phosphorylation and MAPK activation, activates pro-inflammatory and apoptotic signaling, and blocks tumor growth and angiogenesis in 3D vascularized microtumor models (VMT) in vitro. Additionally, ARN22089 has a favorable pharmacokinetic profile and can inhibit the growth of BRAF mutant mouse melanomas and patient-derived xenografts in vivo. ARN22089 selectively blocks CDC42 effector interactions without affecting the binding between closely related GTPases and their downstream effectors. Taken together, we identify a class of therapeutic agents that influence tumor growth by modulating CDC42 signaling in both the tumor cell and its microenvironment.


Subject(s)
Endothelial Cells , Neoplasms , Animals , Endothelial Cells/metabolism , Humans , Mice , Neoplasms/drug therapy , Neovascularization, Pathologic , Signal Transduction , Tumor Microenvironment , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
13.
Sci Rep ; 10(1): 18093, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093610

ABSTRACT

We introduce a compact, fast large area multiphoton exoscope (FLAME) system with enhanced molecular contrast for macroscopic imaging of human skin with microscopic resolution. A versatile imaging platform, FLAME combines optical and mechanical scanning mechanisms with deep learning image restoration to produce depth-resolved images that encompass sub-mm2 to cm2 scale areas of tissue within minutes and provide means for a comprehensive analysis of live or resected thick human skin tissue. The FLAME imaging platform, which expands on a design recently introduced by our group, also features time-resolved single photon counting detection to uniquely allow fast discrimination and 3D virtual staining of melanin. We demonstrate its performance and utility by fast ex vivo and in vivo imaging of human skin. With the ability to provide rapid access to depth resolved images of skin over cm2 area and to generate 3D distribution maps of key sub-cellular skin components such as melanocytic dendrites and melanin, FLAME is ready to be translated into a clinical imaging tool for enhancing diagnosis accuracy, guiding therapy and understanding skin biology.


Subject(s)
Image Processing, Computer-Assisted/methods , Melanins/metabolism , Melanocytes/cytology , Microscopy, Fluorescence, Multiphoton/methods , Skin/cytology , Humans , Melanocytes/metabolism , Skin/diagnostic imaging , Skin/metabolism
14.
Elife ; 92020 10 13.
Article in English | MEDLINE | ID: mdl-33047672

ABSTRACT

Mutational activation of the BRAF proto-oncogene in melanocytes reliably produces benign nevi (pigmented 'moles'), yet the same change is the most common driver mutation in melanoma. The reason nevi stop growing, and do not progress to melanoma, is widely attributed to a cell-autonomous process of 'oncogene-induced senescence'. Using a mouse model of Braf-driven nevus formation, analyzing both proliferative dynamics and single-cell gene expression, we found no evidence that nevus cells are senescent, either compared with other skin cells, or other melanocytes. We also found that nevus size distributions could not be fit by any simple cell-autonomous model of growth arrest, yet were easily fit by models based on collective cell behavior, for example in which arresting cells release an arrest-promoting factor. We suggest that nevus growth arrest is more likely related to the cell interactions that mediate size control in normal tissues, than to any cell-autonomous, 'oncogene-induced' program of senescence.


Melanocytes are pigment-producing cells found throughout the skin. Mutations that activate a gene called BRAF cause these cells to divide and produce melanocytic nevi, also known as "moles". These mutations are oncogenic, meaning they can cause cancer. Indeed, BRAF is the most commonly mutated gene in melanoma, a deadly skin cancer that arises from melanocytes. Yet, moles hardly ever progress to melanoma. A proposed explanation for this behavior is that, once activated, BRAF initiates a process called "oncogene-induced senescence" in each melanocyte. This process, likened to premature aging, is thought to be what causes cells in a mole to quit dividing. Although this hypothesis is widely accepted, it has proved difficult to test directly. To investigate this notion, Ruiz-Vega et al. studied mice with hundreds of moles created by the same BRAF mutation found in human moles. Analyzing the activity of genes in individual cells revealed that nevus melanocytes that have stopped growing are no more senescent than other skin cells, including non-mole melanocytes. Ruiz-Vega et al. then analyzed the sizes at which moles stopped growing, estimating the number of cells in each mole. The data were then compared with the results of a simulation and mathematical modeling. This revealed that any model based on the idea of cells independently shutting down after a number of random events could not reproduce the distribution of mole sizes that had been experimentally observed. On the other hand, models based on melanocytes acting collectively to shut down each other's growth fit the observed data much better. These findings suggest that moles do not stop growing as a direct result of the activation of BRAF, but because they sense and respond to their own overgrowth. The same kind of collective sensing is observed in normal tissues that maintain a constant size. Discovering that melanocytes do this not only sheds light on why moles stop growing, it could also help researchers devise new ways to prevent melanomas from forming.


Subject(s)
Cell Communication , Melanocytes/metabolism , Nevus, Pigmented/genetics , Animals , Mice , Nevus , Proto-Oncogene Mas , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
16.
Commun Biol ; 3(1): 453, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32814826

ABSTRACT

Intestinal stem cells are non-quiescent, dividing epithelial cells that rapidly differentiate into progenitor cells of the absorptive and secretory cell lineages. The kinetics of this process is rapid such that the epithelium is replaced weekly. To determine how the transcriptome and proteome keep pace with rapid differentiation, we developed a new cell sorting method to purify mouse colon epithelial cells. Here we show that alternative mRNA splicing and polyadenylation dominate changes in the transcriptome as stem cells differentiate into progenitors. In contrast, as progenitors differentiate into mature cell types, changes in mRNA levels dominate the transcriptome. RNA processing targets regulators of cell cycle, RNA, cell adhesion, SUMOylation, and Wnt and Notch signaling. Additionally, global proteome profiling detected >2,800 proteins and revealed RNA:protein patterns of abundance and correlation. Paired together, these data highlight new potentials for autocrine and feedback regulation and provide new insights into cell state transitions in the crypt.


Subject(s)
Cell Differentiation , Cell Self Renewal , Colon , Enterocytes/metabolism , Proteome , Stem Cells/metabolism , Transcriptome , Animals , Biomarkers , Cell Self Renewal/genetics , Computational Biology/methods , Enterocytes/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunophenotyping , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Proteomics , RNA Processing, Post-Transcriptional , Stem Cells/cytology
17.
Pigment Cell Melanoma Res ; 33(6): 869-877, 2020 11.
Article in English | MEDLINE | ID: mdl-32485062

ABSTRACT

Multiphoton microscopy (MPM) is a promising non-invasive imaging tool for discriminating benign nevi from melanoma. In this study, we establish a MPM morphologic catalogue of common nevi, information that will be critical in devising strategies to distinguish them from nevi that are evolving to melanoma that may present with more subtle signs of malignancy. Thirty common melanocytic nevi were imaged in vivo using MPM. Quantitative parameters that can distinguish between different types of nevi were developed and confirmed by examining the histology of eleven of the imaged nevi. MPM features of nevi examined included cytologic morphology of melanocytes in the epidermis and dermis, the size and distribution of nevomelanocytes both within and around nests, the size of rete ridges, and the presence of immune cells in the dermis. Distinguishing features include cytological morphology, the size of nevomelanocytes, the size of nevomelanocyte nests, and the distribution of nevomelanocytes. Notably, these distinguishing characteristics were not easily appreciated in fixed tissues, highlighting essential differences in the morphology of live skin. Taken together, this work provides a morphologic compendium of normal nevi, information that will be critical in future studies directed at identifying melanocytic nevi that are evolving to melanoma.


Subject(s)
Microscopy, Fluorescence, Multiphoton , Nevus, Pigmented/diagnostic imaging , Nevus, Pigmented/pathology , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/pathology , Adult , Aged, 80 and over , Biopsy , Cell Size , Female , Humans , Immunity , Male , Melanocytes/pathology , Middle Aged , Nevus, Pigmented/immunology , Skin Neoplasms/immunology , Young Adult
18.
J Gastrointest Oncol ; 11(1): 45-54, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32175104

ABSTRACT

BACKGROUND: Genetic analysis of gastrointestinal malignancies shows a great number of mutations. Most mutations found in gastric tumors are also found in colorectal and esophageal tumors. The challenge remains to identify mutations that distinguish gastric from colorectal and esophageal cancers. Using open-access cancer genomics data, we sought to identify mutations that accounted for the unique phenotypic features of gastric tumors. METHODS: Thirteen cancer genomics datasets with demographic, clinical, and genetic variables were analyzed. Pathologic stage and histology were compared between subjects with and without a specific mutated gene using two-sample t-tests, adjusted for multiple gene testing. Sequence convergence and functional impact of genetic mutations were analyzed using permutation test and PolyPhen-2 score. RESULTS: Analysis included 1,915 subjects with valid pathologic stage and histology. Mean age was 68 years (SD =10). About 54% were female. The most common race was Caucasian (37%) while minorities were rare with high rates of missing data (44%). Pathologic stage: 20% stage I, 35% stage II, 31% stage III, and 14% stage IV. Anatomical location: 30% gastric, 59% colorectal, and 11% esophageal. Histology of gastric cancer: 61% intestinal, 23% diffuse, 15% mixed, and 1% missing. Two mutated genes-CDH1, RHOA-distinguished gastric from colorectal and esophageal tumors. These mutations were highly specific to diffuse histology and advanced stages of gastric tumors and recurrent in transcribed regions known to impact protein functions. CONCLUSIONS: CDH1 and RHOA regulate cell-cell adhesion which is vital to cell growth and proliferation. Identification of these potential driver mutations is critical to better define therapeutic vulnerabilities for the rational design of gastric cancer therapies.

19.
Int J Dermatol ; 59(2): 253-256, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31286499

ABSTRACT

BACKGROUND: Clinical trial data for dupilumab, a monoclonal antibody against the interleukin-4 receptor (IL-4Rα), have shown that it is safe and effective for the treatment of moderate to severe atopic dermatitis in patients whose disease is resistant to other therapies. However, little real-world experience with dupilumab use has been reported thus far. The aim of this retrospective study was to assess overall outcomes in adult patients with atopic dermatitis (AD) treated with dupilumab. METHODS: A retrospective review of electronic medical records was conducted for patients treated with dupilumab in the Department of Dermatology at the University of California, Irvine. RESULTS: We analyzed the medical records of 77 AD patients who received dupilumab according to standard dosing and had at least one documented follow-up visit. In 66 patients (86%), dupilumab improved clinical disease severity, with 23 patients (30%) experiencing complete clearance on dupilumab. Dupilumab was generally well-tolerated and caused no serious adverse events. The most common side effects included dry eyes, conjunctivitis, and keratitis. The most common reason for discontinuation of treatment was lack of substantial clinical improvement or progression of disease severity, followed by ophthalmologic side effects. CONCLUSIONS: Overall, dupilumab was well-tolerated and resulted in clinical improvement in our patient population. These results provide additional important information on the safety and utility of dupilumab treatment for moderate to severe atopic dermatitis in the real-world clinical setting.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Dermatitis, Atopic/drug therapy , Dermatologic Agents/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Boron Compounds/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Calcineurin Inhibitors/therapeutic use , Combined Modality Therapy , Conjunctivitis/chemically induced , Dermatitis, Atopic/radiotherapy , Dermatologic Agents/adverse effects , Disease Progression , Drug Therapy, Combination , Dry Eye Syndromes/chemically induced , Female , Humans , Keratitis/chemically induced , Male , Middle Aged , Retrospective Studies , Severity of Illness Index , Treatment Outcome , Ultraviolet Therapy
20.
Pigment Cell Melanoma Res ; 33(2): 279-292, 2020 03.
Article in English | MEDLINE | ID: mdl-31562697

ABSTRACT

MITF, a gene that is mutated in familial melanoma and Waardenburg syndrome, encodes multiple isoforms expressed from alternative promoters that share common coding exons but have unique amino termini. It is not completely understood how these isoforms influence pigmentation in different tissues and how the expression of these independent isoforms of MITF is regulated. Here, we show that melanocytes express two isoforms of MITF, MITF-A and MITF-M. The expression of MITF-A is partially regulated by a newly identified retinoid enhancer element located upstream of the MITF-A promoter. Mitf-A knockout mice have only subtle changes in melanin accumulation in the hair and reduced Tyr expression in the eye. In contrast, Mitf-M-null mice have enlarged kidneys, lack neural crest-derived melanocytes in the skin, choroid, and iris stroma, yet maintain pigmentation within the retinal pigment epithelium and iris pigment epithelium of the eye. Taken together, these studies identify a critical role for MITF-M in melanocytes, a minor role for MITF-A in regulating pigmentation in the hair and Tyr expression in the eye, and a novel role for MITF-M in size control of the kidney.


Subject(s)
Homeostasis , Microphthalmia-Associated Transcription Factor/metabolism , Pigmentation , Animals , Binding Sites , Cell Line, Tumor , Eye/pathology , HEK293 Cells , Homeostasis/drug effects , Humans , Melanocytes/drug effects , Melanocytes/metabolism , Melanocytes/pathology , Mice, Inbred C57BL , Mice, Transgenic , Microphthalmia-Associated Transcription Factor/genetics , Phenotype , Pigmentation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Isoforms/metabolism , Retinoic Acid Receptor alpha/metabolism , Retinoids/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...