Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Metab ; 5(8): 1303-1318, 2023 08.
Article in English | MEDLINE | ID: mdl-37580540

ABSTRACT

The genomic landscape of colorectal cancer (CRC) is shaped by inactivating mutations in tumour suppressors such as APC, and oncogenic mutations such as mutant KRAS. Here we used genetically engineered mouse models, and multimodal mass spectrometry-based metabolomics to study the impact of common genetic drivers of CRC on the metabolic landscape of the intestine. We show that untargeted metabolic profiling can be applied to stratify intestinal tissues according to underlying genetic alterations, and use mass spectrometry imaging to identify tumour, stromal and normal adjacent tissues. By identifying ions that drive variation between normal and transformed tissues, we found dysregulation of the methionine cycle to be a hallmark of APC-deficient CRC. Loss of Apc in the mouse intestine was found to be sufficient to drive expression of one of its enzymes, adenosylhomocysteinase (AHCY), which was also found to be transcriptionally upregulated in human CRC. Targeting of AHCY function impaired growth of APC-deficient organoids in vitro, and prevented the characteristic hyperproliferative/crypt progenitor phenotype driven by acute deletion of Apc in vivo, even in the context of mutant Kras. Finally, pharmacological inhibition of AHCY reduced intestinal tumour burden in ApcMin/+ mice indicating its potential as a metabolic drug target in CRC.


Subject(s)
Colorectal Neoplasms , Animals , Humans , Mice , Adenosylhomocysteinase/genetics , Adenosylhomocysteinase/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Metabolomics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics
3.
Elife ; 102021 12 13.
Article in English | MEDLINE | ID: mdl-34895463

ABSTRACT

Increased protein synthesis supports the rapid cell proliferation associated with cancer. The Rpl24Bst mutant mouse reduces the expression of the ribosomal protein RPL24 and has been used to suppress translation and limit tumorigenesis in multiple mouse models of cancer. Here, we show that Rpl24Bst also suppresses tumorigenesis and proliferation in a model of colorectal cancer (CRC) with two common patient mutations, Apc and Kras. In contrast to previous reports, Rpl24Bst mutation has no effect on ribosomal subunit abundance but suppresses translation elongation through phosphorylation of eEF2, reducing protein synthesis by 40% in tumour cells. Ablating eEF2 phosphorylation in Rpl24Bst mutant mice by inactivating its kinase, eEF2K, completely restores the rates of elongation and protein synthesis. Furthermore, eEF2K activity is required for the Rpl24Bst mutant to suppress tumorigenesis. This work demonstrates that elevation of eEF2 phosphorylation is an effective means to suppress colorectal tumorigenesis with two driver mutations. This positions translation elongation as a therapeutic target in CRC, as well as in other cancers where the Rpl24Bst mutation has a tumour suppressive effect in mouse models.


Subject(s)
Colorectal Neoplasms/genetics , Elongation Factor 2 Kinase/metabolism , Mutation , Ribosomal Proteins/genetics , Signal Transduction , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Phosphorylation , Ribosomal Proteins/metabolism
4.
EMBO J ; 40(20): e107680, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34532864

ABSTRACT

Cell plasticity is a crucial hallmark leading to cancer metastasis. Upregulation of Rho/ROCK pathway drives actomyosin contractility, protrusive forces, and contributes to the occurrence of highly invasive amoeboid cells in tumors. Cancer stem cells are similarly associated with metastasis, but how these populations arise in tumors is not fully understood. Here, we show that the novel oncogene RASSF1C drives mesenchymal-to-amoeboid transition and stem cell attributes in breast cancer cells. Mechanistically, RASSF1C activates Rho/ROCK via SRC-mediated RhoGDI inhibition, resulting in generation of actomyosin contractility. Moreover, we demonstrate that RASSF1C-induced amoeboid cells display increased expression of cancer stem-like markers such as CD133, ALDH1, and Nanog, and are accompanied by higher invasive potential in vitro and in vivo. Further, RASSF1C-induced amoeboid cells employ extracellular vesicles to transfer the invasive phenotype to target cells and tissue. Importantly, the underlying RASSF1C-driven biological processes concur to explain clinical data: namely, methylation of the RASSF1C promoter correlates with better survival in early-stage breast cancer patients. Therefore, we propose the use of RASSF1 gene promoter methylation status as a biomarker for patient stratification.


Subject(s)
Breast Neoplasms/genetics , Extracellular Vesicles/metabolism , Neoplastic Stem Cells/metabolism , Tumor Suppressor Proteins/genetics , rhoA GTP-Binding Protein/genetics , src-Family Kinases/genetics , AC133 Antigen/genetics , AC133 Antigen/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , CpG Islands , DNA Methylation , Extracellular Vesicles/chemistry , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Mice, SCID , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Survival Analysis , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
5.
Nat Commun ; 12(1): 3464, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34103493

ABSTRACT

Right-sided (proximal) colorectal cancer (CRC) has a poor prognosis and a distinct mutational profile, characterized by oncogenic BRAF mutations and aberrations in mismatch repair and TGFß signalling. Here, we describe a mouse model of right-sided colon cancer driven by oncogenic BRAF and loss of epithelial TGFß-receptor signalling. The proximal colonic tumours that develop in this model exhibit a foetal-like progenitor phenotype (Ly6a/Sca1+) and, importantly, lack expression of Lgr5 and its associated intestinal stem cell signature. These features are recapitulated in human BRAF-mutant, right-sided CRCs and represent fundamental differences between left- and right-sided disease. Microbial-driven inflammation supports the initiation and progression of these tumours with foetal-like characteristics, consistent with their predilection for the microbe-rich right colon and their antibiotic sensitivity. While MAPK-pathway activating mutations drive this foetal-like signature via ERK-dependent activation of the transcriptional coactivator YAP, the same foetal-like transcriptional programs are also initiated by inflammation in a MAPK-independent manner. Importantly, in both contexts, epithelial TGFß-receptor signalling is instrumental in suppressing the tumorigenic potential of these foetal-like progenitor cells.


Subject(s)
Carcinogenesis/metabolism , Colonic Neoplasms/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinogenesis/pathology , Cell Differentiation , Cell Survival , Colon/pathology , Colonic Neoplasms/genetics , Epithelial Cells/metabolism , Fetus/pathology , Inflammation/pathology , Kaplan-Meier Estimate , MAP Kinase Signaling System , Mice, Inbred C57BL , Mutation , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , YAP-Signaling Proteins
6.
Nat Genet ; 53(1): 16-26, 2021 01.
Article in English | MEDLINE | ID: mdl-33414552

ABSTRACT

Oncogenic KRAS mutations and inactivation of the APC tumor suppressor co-occur in colorectal cancer (CRC). Despite efforts to target mutant KRAS directly, most therapeutic approaches focus on downstream pathways, albeit with limited efficacy. Moreover, mutant KRAS alters the basal metabolism of cancer cells, increasing glutamine utilization to support proliferation. We show that concomitant mutation of Apc and Kras in the mouse intestinal epithelium profoundly rewires metabolism, increasing glutamine consumption. Furthermore, SLC7A5, a glutamine antiporter, is critical for colorectal tumorigenesis in models of both early- and late-stage metastatic disease. Mechanistically, SLC7A5 maintains intracellular amino acid levels following KRAS activation through transcriptional and metabolic reprogramming. This supports the increased demand for bulk protein synthesis that underpins the enhanced proliferation of KRAS-mutant cells. Moreover, targeting protein synthesis, via inhibition of the mTORC1 regulator, together with Slc7a5 deletion abrogates the growth of established Kras-mutant tumors. Together, these data suggest SLC7A5 as an attractive target for therapy-resistant KRAS-mutant CRC.


Subject(s)
Colorectal Neoplasms/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , 5' Untranslated Regions/genetics , Amino Acid Transport System ASC/metabolism , Animals , Carcinogenesis/pathology , Cell Proliferation , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glutamine/metabolism , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Kaplan-Meier Estimate , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Minor Histocompatibility Antigens/metabolism , Neoplasm Metastasis , Oncogenes , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
7.
J Cell Sci ; 134(1)2021 01 13.
Article in English | MEDLINE | ID: mdl-33441326

ABSTRACT

Cell division, differentiation and function are largely dependent on accurate proteome composition and regulated gene expression. To control this, protein synthesis is an intricate process governed by upstream signalling pathways. Eukaryotic translation is a multistep process and can be separated into four distinct phases: initiation, elongation, termination and recycling of ribosomal subunits. Translation initiation, the focus of this article, is highly regulated to control the activity and/or function of eukaryotic initiation factors (eIFs) and permit recruitment of mRNAs to the ribosomes. In this Cell Science at a Glance and accompanying poster, we outline the mechanisms by which tumour cells alter the process of translation initiation and discuss how this benefits tumour formation, proliferation and metastasis.


Subject(s)
Neoplasms , Ribosomes , Eukaryotic Initiation Factors/metabolism , Humans , Neoplasms/genetics , Neoplasms/metabolism , Peptide Chain Initiation, Translational , Protein Biosynthesis , RNA, Messenger/metabolism , Ribosomes/genetics , Ribosomes/metabolism
8.
Cancer Discov ; 11(5): 1228-1247, 2021 05.
Article in English | MEDLINE | ID: mdl-33328217

ABSTRACT

KRAS-mutant colorectal cancers are resistant to therapeutics, presenting a significant problem for ∼40% of cases. Rapalogs, which inhibit mTORC1 and thus protein synthesis, are significantly less potent in KRAS-mutant colorectal cancer. Using Kras-mutant mouse models and mouse- and patient-derived organoids, we demonstrate that KRAS with G12D mutation fundamentally rewires translation to increase both bulk and mRNA-specific translation initiation. This occurs via the MNK/eIF4E pathway culminating in sustained expression of c-MYC. By genetic and small-molecule targeting of this pathway, we acutely sensitize KRASG12D models to rapamycin via suppression of c-MYC. We show that 45% of colorectal cancers have high signaling through mTORC1 and the MNKs, with this signature correlating with a 3.5-year shorter cancer-specific survival in a subset of patients. This work provides a c-MYC-dependent cotargeting strategy with remarkable potency in multiple Kras-mutant mouse models and metastatic human organoids and identifies a patient population that may benefit from its clinical application. SIGNIFICANCE: KRAS mutation and elevated c-MYC are widespread in many tumors but remain predominantly untargetable. We find that mutant KRAS modulates translation, culminating in increased expression of c-MYC. We describe an effective strategy targeting mTORC1 and MNK in KRAS-mutant mouse and human models, pathways that are also commonly co-upregulated in colorectal cancer.This article is highlighted in the In This Issue feature, p. 995.


Subject(s)
Colorectal Neoplasms/genetics , Eukaryotic Initiation Factor-4E/drug effects , Intracellular Signaling Peptides and Proteins/drug effects , MTOR Inhibitors/pharmacology , Protein Serine-Threonine Kinases/drug effects , Animals , Colorectal Neoplasms/metabolism , Disease Models, Animal , Eukaryotic Initiation Factor-4E/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Serine-Threonine Kinases/metabolism
9.
Dis Model Mech ; 13(3)2020 03 26.
Article in English | MEDLINE | ID: mdl-32298235

ABSTRACT

Regulation of protein synthesis makes a major contribution to post-transcriptional control pathways. During disease, or under stress, cells initiate processes to reprogramme protein synthesis and thus orchestrate the appropriate cellular response. Recent data show that the elongation stage of protein synthesis is a key regulatory node for translational control in health and disease. There is a complex set of factors that individually affect the overall rate of elongation and, for the most part, these influence either transfer RNA (tRNA)- and eukaryotic elongation factor 1A (eEF1A)-dependent codon decoding, and/or elongation factor 2 (eEF2)-dependent ribosome translocation along the mRNA. Decoding speeds depend on the relative abundance of each tRNA, the cognate:near-cognate tRNA ratios and the degree of tRNA modification, whereas eEF2-dependent ribosome translocation is negatively regulated by phosphorylation on threonine-56 by eEF2 kinase. Additional factors that contribute to the control of the elongation rate include epigenetic modification of the mRNA, coding sequence variation and the expression of eIF5A, which stimulates peptide bond formation between proline residues. Importantly, dysregulation of elongation control is central to disease mechanisms in both tumorigenesis and neurodegeneration, making the individual key steps in this process attractive therapeutic targets. Here, we discuss the relative contribution of individual components of the translational apparatus (e.g. tRNAs, elongation factors and their modifiers) to the overall control of translation elongation and how their dysregulation contributes towards disease processes.


Subject(s)
Disease , Health , Peptide Chain Elongation, Translational , Aminoacylation , Animals , Carcinogenesis/genetics , Humans , RNA, Transfer/genetics , RNA, Transfer/metabolism
10.
Nat Commun ; 9(1): 424, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382819

ABSTRACT

Transition from pluripotency to differentiation is a pivotal yet poorly understood developmental step. Here, we show that the tumour suppressor RASSF1A is a key player driving the early specification of cell fate. RASSF1A acts as a natural barrier to stem cell self-renewal and iPS cell generation, by switching YAP from an integral component in the ß-catenin-TCF pluripotency network to a key factor that promotes differentiation. We demonstrate that epigenetic regulation of the Rassf1A promoter maintains stemness by allowing a quaternary association of YAP-TEAD and ß-catenin-TCF3 complexes on the Oct4 distal enhancer. However, during differentiation, promoter demethylation allows GATA1-mediated RASSF1A expression which prevents YAP from contributing to the TEAD/ß-catenin-TCF3 complex. Simultaneously, we find that RASSF1A promotes a YAP-p73 transcriptional programme that enables differentiation. Together, our findings demonstrate that RASSF1A mediates transcription factor selection of YAP in stem cells, thereby acting as a functional "switch" between pluripotency and initiation of differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Embryonic Stem Cells/cytology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Protein p73/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins , Cell Differentiation , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/physiology , Female , Gene Expression Regulation, Developmental , Hippo Signaling Pathway , Humans , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction , TEA Domain Transcription Factors , Transcription Factors/metabolism , Tumor Protein p73/genetics , Tumor Suppressor Proteins/genetics , Wnt Proteins/metabolism , YAP-Signaling Proteins , beta Catenin/metabolism
11.
Sci Rep ; 7(1): 9408, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28842598

ABSTRACT

Pharmaceutical research requires pre-clinical testing of new therapeutics using both in-vitro and in-vivo models. However, the species specificity of non-human in-vivo models and the inadequate recapitulation of physiological conditions in-vitro are intrinsic weaknesses. Here we show that perfusion is a vital factor for engineered human tissues to recapitulate key aspects of the tumour microenvironment. Organotypic culture and human tumour explants were allowed to grow long-term (14-35 days) and phenotypic features of perfused microtumours compared with those in the static culture. Differentiation status and therapeutic responses were significantly different under perfusion, indicating a distinct biological response of cultures grown under static conditions. Furthermore, heterogeneous co-culture of tumour and endothelial cells demonstrated selective cell-killing under therapeutic perfusion versus episodic delivery. We present a perfused 3D microtumour culture platform that sustains a more physiological tissue state and increased viability for long-term analyses. This system has the potential to tackle the disadvantages inherit of conventional pharmaceutical models and is suitable for precision medicine screening of tumour explants, particularly in hard-to-treat cancer types such as brain cancer which suffer from a lack of clinical samples.


Subject(s)
Cell Culture Techniques/methods , Drug Screening Assays, Antitumor/methods , Perfusion/methods , Antineoplastic Agents/pharmacology , Cell Culture Techniques/instrumentation , Cell Differentiation , Cell Line, Tumor , Coculture Techniques , Drug Evaluation, Preclinical/methods , Endothelial Cells , Humans , Tumor Microenvironment
12.
Mol Cell ; 63(1): 156-66, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27292796

ABSTRACT

Epigenetic inactivation of the Hippo pathway scaffold RASSF1A is associated with poor prognosis in a wide range of sporadic human cancers. Loss of expression reduces tumor suppressor activity and promotes genomic instability, but how this pleiotropic biomarker is regulated at the protein level is unknown. Here we show that TGF-ß is the physiological signal that stimulates RASSF1A degradation by the ubiquitin-proteasome pathway. In response to TGF-ß, RASSF1A is recruited to TGF-ß receptor I and targeted for degradation by the co-recruited E3 ubiquitin ligase ITCH. RASSF1A degradation is necessary to permit Hippo pathway effector YAP1 association with SMADs and subsequent nuclear translocation of receptor-activated SMAD2. We find that RASSF1A expression regulates TGF-ß-induced YAP1/SMAD2 interaction and leads to SMAD2 cytoplasmic retention and inefficient transcription of TGF-ß targets genes. Moreover, RASSF1A limits TGF-ß induced invasion, offering a new framework on how RASSF1A affects YAP1 transcriptional output and elicits its tumor-suppressive function.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Lung Neoplasms/metabolism , Phosphoproteins/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Suppressor Proteins/metabolism , Active Transport, Cell Nucleus , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , DNA Methylation , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Invasiveness , Protein Serine-Threonine Kinases/metabolism , Proteolysis , RNA Interference , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors , Transcription, Genetic , Transfection , Transforming Growth Factor beta1/pharmacology , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , YAP-Signaling Proteins
13.
Curr Biol ; 25(23): 3019-34, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26549256

ABSTRACT

Tumor progression to invasive carcinoma is associated with activation of SRC family kinase (SRC, YES, FYN) activity and loss of cellular cohesion. The hippo pathway-regulated cofactor YAP1 supports the tumorigenicity of RAS mutations but requires both inactivation of hippo signaling and YES-mediated phosphorylation of YAP1 for oncogenic activity. Exactly how SRC kinases are activated and hippo signaling is lost in sporadic human malignancies remains unknown. Here, we provide evidence that hippo-mediated inhibition of YAP1 is lost upon promoter methylation of the RAS effector and hippo kinase scaffold RASSF1A. We find that RASSF1A promoter methylation reduces YAP phospho-S127, which derepresses YAP1, and actively supports YAP1 activation by switching RASSF1 transcription to the independently transcribed RASSF1C isoform that promotes Tyr kinase activity. Using affinity proteomics, proximity ligation, and real-time molecular visualization, we find that RASSF1C targets SRC/YES to epithelial cell-cell junctions and promotes tyrosine phosphorylation of E-cadherin, ß-catenin, and YAP1. RASSF1A restricts SRC activity, preventing motility, invasion, and tumorigenesis in vitro and in vivo, with epigenetic inactivation correlating with increased inhibitory pY527-SRC in breast tumors. These data imply that distinct RASSF1 isoforms have opposing functions, which provide a biomarker for YAP1 activation and explain correlations of RASSF1 methylation with advanced invasive disease in humans. The ablation of epithelial integrity together with subsequent YAP1 nuclear localization allows transcriptional activation of ß-catenin/TBX-YAP/TEAD target genes, including Myc, and an invasive phenotype. These findings define gene transcript switching as a tumor suppressor mechanism under epigenetic control.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Phosphoproteins/genetics , Signal Transduction , Tumor Suppressor Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Phosphoproteins/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Transcription Factors , Transcriptional Activation , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins
14.
FASEB J ; 24(10): 3869-81, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20522784

ABSTRACT

The hair follicle is a cyclic biological system that progresses through stages of growth, regression, and quiescence, which involves dynamic changes in a program of gene regulation. Micro-RNAs (miRNAs) are critically important for the control of gene expression and silencing. Here, we show that global miRNA expression in the skin markedly changes during distinct stages of the hair cycle in mice. Furthermore, we show that expression of miR-31 markedly increases during anagen and decreases during catagen and telogen. Administration of antisense miR-31 inhibitor into mouse skin during the early- and midanagen phases of the hair cycle results in accelerated anagen development, and altered differentiation of hair matrix keratinocytes and hair shaft formation. Microarray, qRT-PCR and Western blot analyses revealed that miR-31 negatively regulates expression of Fgf10, the components of Wnt and BMP signaling pathways Sclerostin and BAMBI, and Dlx3 transcription factor, as well as selected keratin genes, both in vitro and in vivo. Using luciferase reporter assay, we show that Krt16, Krt17, Dlx3, and Fgf10 serve as direct miR-31 targets. Thus, by targeting a number of growth regulatory molecules and cytoskeletal proteins, miR-31 is involved in establishing an optimal balance of gene expression in the hair follicle required for its proper growth and hair fiber formation.


Subject(s)
Hydrogen-Ion Concentration
SELECTION OF CITATIONS
SEARCH DETAIL
...