Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 5(7): 101611, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38942020

ABSTRACT

Resistance to targeted therapy remains a major clinical challenge in melanoma. To uncover resistance mechanisms, we perform single-cell RNA sequencing on fine-needle aspirates from resistant and responding tumors of patients undergoing BRAFi/MEKi treatment. Among the genes most prominently expressed in resistant tumors is POSTN, predicted to signal to a macrophage population associated with targeted therapy resistance (TTR). Accordingly, tumors from patients with fast disease progression after therapy exhibit high POSTN expression levels and high numbers of TTR macrophages. POSTN polarizes human macrophages toward a TTR phenotype and promotes resistance to targeted therapy in a melanoma mouse model, which is associated with a phenotype change in intratumoral macrophages. Finally, polarized TTR macrophages directly protect human melanoma cells from MEKi-induced killing via CD44 receptor expression on melanoma cells. Thus, interfering with the protective activity of TTR macrophages may offer a strategy to overcome resistance to targeted therapy in melanoma.


Subject(s)
Drug Resistance, Neoplasm , Macrophages , Melanoma , Melanoma/drug therapy , Melanoma/pathology , Melanoma/genetics , Melanoma/metabolism , Humans , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Animals , Macrophages/metabolism , Macrophages/drug effects , Mice , Cell Line, Tumor , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics
2.
J Biol Chem ; 300(1): 105480, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37992803

ABSTRACT

The bone-derived hormone fibroblast growth factor-23 (FGF23) has recently received much attention due to its association with chronic kidney disease and cardiovascular disease progression. Extracellular sodium concentration ([Na+]) plays a significant role in bone metabolism. Hyponatremia (lower serum [Na+]) has recently been shown to be independently associated with FGF23 levels in patients with chronic systolic heart failure. However, nothing is known about the direct impact of [Na+] on FGF23 production. Here, we show that an elevated [Na+] (+20 mM) suppressed FGF23 formation, whereas low [Na+] (-20 mM) increased FGF23 synthesis in the osteoblast-like cell lines UMR-106 and MC3T3-E1. Similar bidirectional changes in FGF23 abundance were observed when osmolality was altered by mannitol but not by urea, suggesting a role of tonicity in FGF23 formation. Moreover, these changes in FGF23 were inversely proportional to the expression of NFAT5 (nuclear factor of activated T cells-5), a transcription factor responsible for tonicity-mediated cellular adaptations. Furthermore, arginine vasopressin, which is often responsible for hyponatremia, did not affect FGF23 production. Next, we performed a comprehensive and unbiased RNA-seq analysis of UMR-106 cells exposed to low versus high [Na+], which revealed several novel genes involved in cellular adaptation to altered tonicity. Additional analysis of cells with Crisp-Cas9-mediated NFAT5 deletion indicated that NFAT5 controls numerous genes associated with FGF23 synthesis, thereby confirming its role in [Na+]-mediated FGF23 regulation. In line with these in vitro observations, we found that hyponatremia patients have higher FGF23 levels. Our results suggest that [Na+] is a critical regulator of FGF23 synthesis.


Subject(s)
Fibroblast Growth Factor-23 , Sodium , Humans , Fibroblast Growth Factor-23/genetics , Fibroblast Growth Factor-23/metabolism , Hyponatremia/physiopathology , Renal Insufficiency, Chronic/physiopathology , Sodium/metabolism , Sodium/pharmacology , Cell Line, Tumor , Cell Line , Animals , Mice , Mice, Inbred C57BL , Arginine Vasopressin/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Rats
3.
Sci Adv ; 9(2): eadc8825, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36638181

ABSTRACT

Metastatic disease is a major cause of death for patients with melanoma. Melanoma cells can become metastatic not only due to cell-intrinsic plasticity but also due to cancer-induced protumorigenic remodeling of the immune microenvironment. Here, we report that innate immune surveillance by natural killer (NK) cells is bypassed by human melanoma cells expressing the stem cell marker NGFR. Using in vitro and in vivo cytotoxic assays, we show that NGFR protects melanoma cells from NK cell-mediated killing and, furthermore, boosts metastasis formation in a mouse model with adoptively transferred human NK cells. Mechanistically, NGFR leads to down-regulation of NK cell activating ligands and simultaneous up-regulation of the fatty acid stearoyl-coenzyme A desaturase (SCD) in melanoma cells. Notably, pharmacological and small interfering RNA-mediated inhibition of SCD reverted NGFR-induced NK cell evasion in vitro and in vivo. Hence, NGFR orchestrates immune control antagonizing pathways to protect melanoma cells from NK cell clearance, which ultimately favors metastatic disease.


Subject(s)
Antineoplastic Agents , Melanoma , Mice , Animals , Humans , Cell Line, Tumor , Melanoma/pathology , Killer Cells, Natural , Lipids , Tumor Microenvironment , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism
4.
Dev Cell ; 57(19): 2249-2250, 2022 10 10.
Article in English | MEDLINE | ID: mdl-36220078

ABSTRACT

Neural crest cells arise in the neurectoderm of vertebrate embryos, but their developmental potential goes way beyond neurectodermal derivatives. In this issue of Developmental Cell, Hovland et al. reveal that neural crest cells re-employ embryonic stem cell factors in combination with specific transcription factors to enable their broad potential.


Subject(s)
Neural Crest , Pluripotent Stem Cells , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Stem Cell Factor , Transcription Factors , Vertebrates
5.
Cell Stem Cell ; 29(10): 1459-1474.e9, 2022 Oct 06.
Article in English | MEDLINE | ID: mdl-36113462

ABSTRACT

Fibrosis is the final path of nearly every form of chronic disease, regardless of the pathogenesis. Upon chronic injury, activated, fibrogenic fibroblasts deposit excess extracellular matrix, and severe tissue fibrosis can occur in virtually any organ. However, antifibrotic therapies that target fibrogenic cells, while sparing homeostatic fibroblasts in healthy tissues, are limited. We tested whether specific immunization against endogenous proteins, strongly expressed in fibrogenic cells but highly restricted in quiescent fibroblasts, can elicit an antigen-specific cytotoxic T cell response to ameliorate organ fibrosis. In silico epitope prediction revealed that activation of the genes Adam12 and Gli1 in profibrotic cells and the resulting "self-peptides" can be exploited for T cell vaccines to ablate fibrogenic cells. We demonstrate the efficacy of a vaccination approach to mount CD8+ T cell responses that reduce fibroblasts and fibrosis in the liver and lungs in mice. These results provide proof of principle for vaccination-based immunotherapies to treat fibrosis.


Subject(s)
Fibroblasts , Lung , Animals , Epitopes/metabolism , Fibroblasts/metabolism , Fibrosis , Immunotherapy , Liver/pathology , Lung/metabolism , Mice , Vaccination , Zinc Finger Protein GLI1/metabolism
6.
EMBO J ; 41(17): e111955, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35894449

ABSTRACT

Schwann cell precursors (SCPs) are transient glial progenitors that are important for the formation of late neural crest derivatives, yet their heterogeneity and developmental potential remain incompletely understood. In this issue, Kastriti, Faure, von Ahsen et al (2022) use comprehensive single-cell RNA sequencing analyses to identify a transient "hub" state common to SCPs and neural crest cells (NCCs), revealing a striking similarity of SCPs to late migrating NCCs. These results raise important questions about the potential role of such a state in adult tissue regeneration and tumourigenesis.


Subject(s)
Neural Crest , Schwann Cells , Cell Differentiation , Neurogenesis , Neuroglia
7.
Front Cell Dev Biol ; 10: 916033, 2022.
Article in English | MEDLINE | ID: mdl-35693944

ABSTRACT

Deregulation of cellular metabolism through metabolic rewiring and translational reprogramming are considered hallmark traits of tumor development and malignant progression. The transcription factor YY1 is a master regulator of metabolism that we have previously shown to orchestrate a metabolic program required for melanoma formation. In this study, we demonstrate that YY1, while being essential for primary melanoma formation, suppresses metastatic spreading. Its downregulation or loss resulted in the induction of an invasiveness gene program and sensitized melanoma cells for pro-invasive signaling molecules, such as TGF-ß. In addition, NGFR, a key effector in melanoma invasion and phenotype switching, was among the most upregulated genes after YY1 knockdown. High levels of NGFR were also associated with other metabolic stress inducers, further indicating that YY1 knockdown mimics a metabolic stress program associated with an increased invasion potential in melanoma. Accordingly, while counteracting tumor growth, loss of YY1 strongly promoted melanoma cell invasiveness in vitro and metastasis formation in melanoma mouse models in vivo. Thus, our findings show that the metabolic regulator YY1 controls phenotype switching in melanoma.

9.
Nat Commun ; 12(1): 5056, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34417458

ABSTRACT

Melanoma cells rely on developmental programs during tumor initiation and progression. Here we show that the embryonic stem cell (ESC) factor Sall4 is re-expressed in the Tyr::NrasQ61K; Cdkn2a-/- melanoma model and that its expression is necessary for primary melanoma formation. Surprisingly, while Sall4 loss prevents tumor formation, it promotes micrometastases to distant organs in this melanoma-prone mouse model. Transcriptional profiling and in vitro assays using human melanoma cells demonstrate that SALL4 loss induces a phenotype switch and the acquisition of an invasive phenotype. We show that SALL4 negatively regulates invasiveness through interaction with the histone deacetylase (HDAC) 2 and direct co-binding to a set of invasiveness genes. Consequently, SALL4 knock down, as well as HDAC inhibition, promote the expression of an invasive signature, while inhibition of histone acetylation partially reverts the invasiveness program induced by SALL4 loss. Thus, SALL4 appears to regulate phenotype switching in melanoma through an HDAC2-mediated mechanism.


Subject(s)
Epigenesis, Genetic , Melanoma/genetics , Melanoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Stem Cell Factor/metabolism , Transcription Factors/metabolism , Acetylation , Animals , Base Sequence , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Adhesion/genetics , Cell Line, Tumor , Cell Lineage , Cell Proliferation , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histone Deacetylase 2/metabolism , Histones/metabolism , Humans , Melanocytes/metabolism , Melanocytes/pathology , Mice, Nude , Mice, Transgenic , Neoplasm Invasiveness , Neoplasm Micrometastasis , Protein Binding , Tumor Burden
10.
Nat Commun ; 12(1): 4700, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34349124

ABSTRACT

During skin injury, immune response and repair mechanisms have to be coordinated for rapid skin regeneration and the prevention of microbial infections. Natural Killer (NK) cells infiltrate hypoxic skin lesions and Hypoxia-inducible transcription factors (HIFs) mediate adaptation to low oxygen. We demonstrate that mice lacking the Hypoxia-inducible factor (HIF)-1α isoform in NK cells show impaired release of the cytokines Interferon (IFN)-γ and Granulocyte Macrophage - Colony Stimulating Factor (GM-CSF) as part of a blunted immune response. This accelerates skin angiogenesis and wound healing. Despite rapid wound closure, bactericidal activity and the ability to restrict systemic bacterial infection are impaired. Conversely, forced activation of the HIF pathway supports cytokine release and NK cell-mediated antibacterial defence including direct killing of bacteria by NK cells despite delayed wound closure. Our results identify, HIF-1α in NK cells as a nexus that balances antimicrobial defence versus global repair in the skin.


Subject(s)
Killer Cells, Natural/immunology , Skin/immunology , Skin/microbiology , Wound Healing , Animals , Cell Hypoxia , Cytokines/metabolism , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Mice , Neovascularization, Physiologic , Skin/blood supply , Skin Diseases, Bacterial/prevention & control
12.
Stem Cells Transl Med ; 10(4): 522-533, 2021 04.
Article in English | MEDLINE | ID: mdl-33258291

ABSTRACT

Melanoma is the deadliest of all skin cancers due to its high metastatic potential. In recent years, advances in targeted therapy and immunotherapy have contributed to a remarkable progress in the treatment of metastatic disease. However, intrinsic or acquired resistance to such therapies remains a major obstacle in melanoma treatment. Melanoma disease progression, beginning from tumor initiation and growth to acquisition of invasive phenotypes and metastatic spread and acquisition of treatment resistance, has been associated with cellular dedifferentiation and the hijacking of gene regulatory networks reminiscent of the neural crest (NC)-the developmental structure which gives rise to melanocytes and hence melanoma. This review summarizes the experimental evidence for the involvement of NC stem cell (NCSC)-like cell states during melanoma progression and addresses novel approaches to combat the emergence of stemness characteristics that have shown to be linked with aggressive disease outcome and drug resistance.


Subject(s)
Melanoma , Neural Crest , Neural Stem Cells , Disease Progression , Drug Resistance, Neoplasm , Humans , Melanocytes , Melanoma/pathology , Melanoma/therapy , Neural Crest/cytology
13.
Nat Commun ; 10(1): 2192, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097699

ABSTRACT

The transcription factor Yin Yang 1 (YY1) plays an important role in human disease. It is often overexpressed in cancers and mutations can lead to a congenital haploinsufficiency syndrome characterized by craniofacial dysmorphisms and neurological dysfunctions, consistent with a role in brain development. Here, we show that Yy1 controls murine cerebral cortex development in a stage-dependent manner. By regulating a wide range of metabolic pathways and protein translation, Yy1 maintains proliferation and survival of neural progenitor cells (NPCs) at early stages of brain development. Despite its constitutive expression, however, the dependence on Yy1 declines over the course of corticogenesis. This is associated with decreasing importance of processes controlled by Yy1 during development, as reflected by diminished protein synthesis rates at later developmental stages. Thus, our study unravels a novel role for Yy1 as a stage-dependent regulator of brain development and shows that biosynthetic demands of NPCs dynamically change throughout development.


Subject(s)
Cerebral Cortex/growth & development , Gene Expression Regulation, Developmental/physiology , Neural Stem Cells/physiology , YY1 Transcription Factor/physiology , Animals , Cell Proliferation/genetics , Cell Survival/genetics , Cells, Cultured , Embryo, Mammalian , Female , G1 Phase Cell Cycle Checkpoints/genetics , Gene Knockout Techniques , Metabolic Networks and Pathways/physiology , Mice , Mice, Transgenic , Models, Animal , Primary Cell Culture , RNA, Small Interfering/metabolism
14.
J Clin Invest ; 129(7): 2702-2716, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31039140

ABSTRACT

The development of metastatic melanoma is thought to require the dynamic shifting of neoplastic cells between proliferative and invasive phenotypes. Contrary to this conventional "phenotype switching" model, we now show that disease progression can involve malignant melanoma cells simultaneously displaying proliferative and invasive properties. Using a genetic mouse model of melanoma in combination with in vitro analyses of melanoma cell lines, we found that conditional deletion of the downstream signaling molecule Smad4, which abrogates all canonical TGF-ß signaling, indeed inhibits both tumor growth and metastasis. Conditional deletion of the inhibitory signaling factor Smad7, however, generated cells that are both highly invasive and proliferative, indicating that invasiveness is compatible with a high proliferation rate. In fact, conditional Smad7 deletion led to sustained melanoma growth and at the same time promoted massive metastasis formation, a result consistent with data indicating that low SMAD7 levels in patient tumors are associated with a poor survival. Our findings reveal that modulation of SMAD7 levels can overcome the need for phenotype switching during tumor progression and may thus represent a novel therapeutic target in metastatic disease.


Subject(s)
Melanoma/metabolism , Signal Transduction , Smad7 Protein/metabolism , Animals , Disease-Free Survival , Humans , Melanoma/genetics , Melanoma/mortality , Melanoma/pathology , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplasm Metastasis , Smad4 Protein/genetics , Smad4 Protein/metabolism , Smad7 Protein/genetics , Survival Rate
15.
Cell Stem Cell ; 24(4): 637-653.e9, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30951662

ABSTRACT

Increasing evidence suggests that cancer cells highjack developmental programs for disease initiation and progression. Melanoma arises from melanocytes that originate during development from neural crest stem cells (NCSCs). Here, we identified the transcription factor Yin Yang 1 (Yy1) as an NCSCs regulator. Conditional deletion of Yy1 in NCSCs resulted in stage-dependent hypoplasia of all major neural crest derivatives due to decreased proliferation and increased cell death. Moreover, conditional ablation of one Yy1 allele in a melanoma mouse model prevented tumorigenesis, indicating a particular susceptibility of melanoma cells to reduced Yy1 levels. Combined RNA sequencing (RNA-seq), chromatin immunoprecipitation (ChIP)-seq, and untargeted metabolomics demonstrated that YY1 governs multiple metabolic pathways and protein synthesis in both NCSCs and melanoma. In addition to directly regulating a metabolic gene set, YY1 can act upstream of MITF/c-MYC as part of a gene regulatory network controlling metabolism. Thus, both NCSC development and melanoma formation depend on an intricate YY1-controlled metabolic program.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Neural Crest/cytology , Neural Crest/metabolism , YY1 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Humans , Mice , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , YY1 Transcription Factor/deficiency
16.
Cancer Cell ; 34(1): 69-84.e14, 2018 07 09.
Article in English | MEDLINE | ID: mdl-30008323

ABSTRACT

Human melanomas frequently harbor amplifications of EZH2. However, the contribution of EZH2 to melanoma formation has remained elusive. Taking advantage of murine melanoma models, we show that EZH2 drives tumorigenesis from benign BrafV600E- or NrasQ61K-expressing melanocytes by silencing of genes relevant for the integrity of the primary cilium, a signaling organelle projecting from the surface of vertebrate cells. Consequently, gain of EZH2 promotes loss of primary cilia in benign melanocytic lesions. In contrast, blockade of EZH2 activity evokes ciliogenesis and cilia-dependent growth inhibition in malignant melanoma. Finally, we demonstrate that loss of cilia enhances pro-tumorigenic WNT/ß-catenin signaling, and is itself sufficient to drive metastatic melanoma in benign cells. Thus, primary cilia deconstruction is a key process in EZH2-driven melanomagenesis.


Subject(s)
Cell Movement , Cell Proliferation , Cilia/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Melanocytes/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cilia/genetics , Cilia/pathology , Enhancer of Zeste Homolog 2 Protein/genetics , Female , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Lymphatic Metastasis , Male , Melanocytes/pathology , Melanoma/genetics , Melanoma/secondary , Membrane Proteins/genetics , Mice, Nude , Mice, Transgenic , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism
17.
Dev Biol ; 444 Suppl 1: S356-S365, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29778801

ABSTRACT

Multipotent neural crest cells can self-renew and give rise to a plethora of neural and non-neural cell types in the vertebrate embryo. Intriguingly, cells reminiscent of such neural crest stem cells (NCSCs) have also been isolated from various postnatal and adult neural crest (NC)-derived structures. However, it has been debated whether NCSC-like cells in the adult correspond to 'in vitro artefacts' emerging upon isolation or fulfil a physiological role in vivo. Here, we discuss recent findings indicating that in different adult NC derivatives, injury or stress responses induce a NCSC-like state, presumably by reprogramming differentiated cells such as Schwann cells. Thereby, injury or stress appear to endow NC-derived cells with the capacity to generate new cell types during the repair process; in addition, injury can activate a repair program in adult NC-derived cells, which promotes tissue repair or regeneration by paracrine signalling. Thus, there is increasing evidence that NCSC-like cells in NC derivatives represent an in vivo state implicated in distinct physiological functions in the adult organism.


Subject(s)
Neural Stem Cells/metabolism , Stress, Physiological/physiology , Wound Healing/physiology , Adult , Animals , Cell Differentiation , Cell Movement , Cell Proliferation/physiology , Cells, Cultured , Humans , Neural Crest/metabolism , Neural Crest/physiology , Neurogenesis/physiology , Schwann Cells , Wounds and Injuries/physiopathology
18.
Genesis ; 56(6-7): e23105, 2018 06.
Article in English | MEDLINE | ID: mdl-29673028

ABSTRACT

The neural crest is one of the embryonic structures with the broadest developmental potential in vertebrates. Morphologically, neural crest cells emerge during neurulation in the dorsal folds of the neural tube before undergoing an epithelial-to-mesenchymal transition (EMT), delaminating from the neural tube, and migrating to multiple sites in the growing embryo. Neural crest cells generate cell types as diverse as peripheral neurons and glia, melanocytes, and so-called mesectodermal derivatives that include craniofacial bone and cartilage and smooth muscle cells in cardiovascular structures. In mice, the fate of neural crest cells has been determined mainly by means of transgenesis and genome editing technologies. The most frequently used method relies on the Cre-loxP system, in which expression of Cre-recombinase in neural crest cells or their derivatives genetically enables the expression of a Cre-reporter allele, thus permanently marking neural crest-derived cells. Here, we provide an overview of the Cre-driver lines used in the field and discuss to what extent these lines allow precise neural crest stage and lineage-specific fate mapping.


Subject(s)
Cell Lineage/physiology , Neural Crest/embryology , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Chromosome Mapping/methods , Epithelial-Mesenchymal Transition/physiology , Integrases/metabolism , Mice , Neural Tube/embryology
19.
Nat Commun ; 9(1): 236, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29339718

ABSTRACT

Cutaneous wound healing is a complex process that aims to re-establish the original structure of the skin and its functions. Among other disorders, peripheral neuropathies are known to severely impair wound healing capabilities of the skin, revealing the importance of skin innervation for proper repair. Here, we report that peripheral glia are crucially involved in this process. Using a mouse model of wound healing, combined with in vivo fate mapping, we show that injury activates peripheral glia by promoting de-differentiation, cell-cycle re-entry and dissemination of the cells into the wound bed. Moreover, injury-activated glia upregulate the expression of many secreted factors previously associated with wound healing and promote myofibroblast differentiation by paracrine modulation of TGF-ß signalling. Accordingly, depletion of these cells impairs epithelial proliferation and wound closure through contraction, while their expansion promotes myofibroblast formation. Thus, injury-activated glia and/or their secretome might have therapeutic potential in human wound healing disorders.


Subject(s)
Cell Differentiation/physiology , Neuroglia/physiology , Skin/physiopathology , Wound Healing/physiology , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Profiling , Humans , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Myofibroblasts/metabolism , Myofibroblasts/physiology , Neuroglia/cytology , Neuroglia/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Signal Transduction/genetics , Skin/injuries , Skin/innervation , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Wound Healing/genetics
20.
Nat Commun ; 9(1): 314, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29358574

ABSTRACT

The originally published version of this Article was updated shortly after publication to add the words 'The' and 'affinity' to the title, following their inadvertent removal during the production process. This has now been corrected in both the PDF and HTML versions of the Article.

SELECTION OF CITATIONS
SEARCH DETAIL
...