Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters

Publication year range
1.
Mol Cell ; 82(3): 542-554.e6, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35081364

ABSTRACT

Non-covalent complexes of glycolytic enzymes, called metabolons, were postulated in the 1970s, but the concept has been controversial. Here we show that a c-Myc-responsive long noncoding RNA (lncRNA) that we call glycoLINC (gLINC) acts as a backbone for metabolon formation between all four glycolytic payoff phase enzymes (PGK1, PGAM1, ENO1, and PKM2) along with lactate dehydrogenase A (LDHA). The gLINC metabolon enhances glycolytic flux, increases ATP production, and enables cell survival under serine deprivation. Furthermore, gLINC overexpression in cancer cells promotes xenograft growth in mice fed a diet deprived of serine, suggesting that cancer cells employ gLINC during metabolic reprogramming. We propose that gLINC makes a functional contribution to cancer cell adaptation and provide the first example of a lncRNA-facilitated metabolon.


Subject(s)
Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Glycolysis , Membrane Proteins/metabolism , Neoplasms/enzymology , Phosphoglycerate Kinase/metabolism , Phosphoglycerate Mutase/metabolism , Phosphopyruvate Hydratase/metabolism , RNA, Long Noncoding/metabolism , Thyroid Hormones/metabolism , Tumor Suppressor Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Biomarkers, Tumor/genetics , Carrier Proteins/genetics , Cell Proliferation , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Membrane Proteins/genetics , Mice, Nude , Multienzyme Complexes , Neoplasms/genetics , Neoplasms/pathology , Phosphoglycerate Kinase/genetics , Phosphoglycerate Mutase/genetics , Phosphopyruvate Hydratase/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/genetics , Serine/deficiency , Thyroid Hormones/genetics , Tumor Burden , Tumor Suppressor Proteins/genetics , Thyroid Hormone-Binding Proteins
2.
EMBO J ; 41(14): e109777, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35670107

ABSTRACT

Autophagy represents a fundamental mechanism for maintaining cell survival and tissue homeostasis in response to physiological and pathological stress. Autophagy initiation converges on the FIP200-ATG13-ULK1 complex wherein the serine/threonine kinase ULK1 plays a central role. Here, we reveal that the E3 ubiquitin ligase TRIM27 functions as a negative regulatory component of the FIP200-ATG13-ULK1 complex. TRIM27 directly polyubiquitinates ULK1 at K568 and K571 sites with K48-linked ubiquitin chains, with proteasomal turnover maintaining control over basal ULK1 levels. However, during starvation-induced autophagy, TRIM27 catalyzes non-degradative K6- and K11-linked ubiquitination of the serine/threonine kinase 38-like (STK38L) kinase. In turn, STK38L ubiquitination promotes its activation and phosphorylation of ULK1 at Ser495, rendering ULK1 in a permissive state for TRIM27-mediated hyper-ubiquitination of ULK1. This cooperative mechanism serves to restrain the amplitude and duration of autophagy. Further evidence from mouse models shows that basal autophagy levels are increased in Trim27 knockout mice and that Trim27 differentially regulates tumorigenesis and metastasis. Our study identifies a key role of STK38L-TRIM27-ULK1 signaling axis in negatively controlling autophagy with relevance established in human breast cancer.


Subject(s)
Autophagy , Protein Serine-Threonine Kinases , Animals , Autophagy-Related Protein-1 Homolog/genetics , Carcinogenesis/genetics , DNA-Binding Proteins , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Nuclear Proteins , Protein Serine-Threonine Kinases/genetics , Serine , Transcription Factors , Ubiquitin-Protein Ligases
3.
Proc Natl Acad Sci U S A ; 119(49): e2208904119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36445966

ABSTRACT

The protooncoprotein N-Myc, which is overexpressed in approximately 25% of neuroblastomas as the consequence of MYCN gene amplification, has long been postulated to regulate DNA double-strand break (DSB) repair in neuroblastoma cells, but experimental evidence of this function is presently scant. Here, we show that N-Myc transcriptionally activates the long noncoding RNA MILIP to promote nonhomologous end-joining (NHEJ) DNA repair through facilitating Ku70-Ku80 heterodimerization in neuroblastoma cells. High MILIP expression was associated with poor outcome and appeared as an independent prognostic factor in neuroblastoma patients. Knockdown of MILIP reduced neuroblastoma cell viability through the induction of apoptosis and inhibition of proliferation, retarded neuroblastoma xenograft growth, and sensitized neuroblastoma cells to DNA-damaging therapeutics. The effect of MILIP knockdown was associated with the accumulation of DNA DSBs in neuroblastoma cells largely due to decreased activity of the NHEJ DNA repair pathway. Mechanistical investigations revealed that binding of MILIP to Ku70 and Ku80 increased their heterodimerization, and this was required for MILIP-mediated promotion of NHEJ DNA repair. Disrupting the interaction between MILIP and Ku70 or Ku80 increased DNA DSBs and reduced cell viability with therapeutic potential revealed where targeting MILIP using Gapmers cooperated with the DNA-damaging drug cisplatin to inhibit neuroblastoma growth in vivo. Collectively, our findings identify MILIP as an N-Myc downstream effector critical for activation of the NHEJ DNA repair pathway in neuroblastoma cells, with practical implications of MILIP targeting, alone and in combination with DNA-damaging therapeutics, for neuroblastoma treatment.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Neuroblastoma , RNA, Long Noncoding , Humans , DNA/genetics , DNA End-Joining Repair/genetics , DNA Repair/genetics , Neuroblastoma/drug therapy , Neuroblastoma/genetics , RNA, Long Noncoding/genetics
4.
Semin Cancer Biol ; 75: 72-83, 2021 10.
Article in English | MEDLINE | ID: mdl-32927018

ABSTRACT

The TP53 gene is arguably the most important tumor suppressor gene known, contributing multifaceted roles to the process of tumor development. Its protein product p53, is a crucial sequence-specific transcription factor which regulates the expression of a large network of protein-coding genes, as well as thousands of noncoding RNAs (ncRNAs), notably microRNAs and long ncRNAs (lncRNAs). Through a variety of direct and indirect mechanisms, ncRNAs in turn modulate p53 levels and activity. Here the numbers of studies are steadily building which link the contributions of dysregulated ncRNAs to tumorigenesis via their participation throughout the p53 regulatory network. In this review, we will examine how the principal forms of ncRNAs, namely microRNAs, lncRNAs and circular RNAs (circRNAs) function as either effectors or regulators amongst the diversity of p53's cellular responses. We first discuss the more recently discovered connections between miRNAs and p53 signaling before focusing on the remarkable diversity of crosstalk evident between lncRNAs and p53, and subsequently, developing reports linking circRNAs to p53. Highlighted throughout the review are the mechanistic impacts of dysregulated ncRNAs on p53 functions as well as the possible prognostic implications of these interactions. We also describe the emerging connections between ncRNAs and the often-perplexing functions of mutant p53. Finally, in the context of p53 therapeutic approaches, we describe some of the challenges in ncRNA research and their potential for translation.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Mutation , Neoplasms/pathology , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Biomarkers, Tumor/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics
5.
Int J Hyperthermia ; 39(1): 358-371, 2022.
Article in English | MEDLINE | ID: mdl-35184661

ABSTRACT

BACKGROUND: Hyperthermia is a widely used adjunct treatment for different cancers including nasopharyngeal carcinoma (NPC). The protooncogene c-Myc is up-regulated in NPC and its expression is associated with poor prognosis. OBJECTIVE: We hypothesized that c-Myc constitutes an important hyperthermia treatment target, and we investigated its contribution to hyperthermia responses in NPC. METHODS: The growth of the human NPC cell lines CNE1 and CNE2 was analyzed using CCK-8 and clonogenicity assays after 43 °C hyperthermia, knockdown or overexpression of c-Myc. Flow cytometry measurements assessed cell cycle parameters and apoptosis, while levels of c-Myc together with key transcriptional targets were determined using qPCR and Western blotting. Parallel experiments were undertaken using NPC xenografts in nude mice and lastly, global transcriptomic changes were determined using 'RNAseq'. RESULTS: Hyperthermia increased the ubiquitination and proteasomal destruction of c-Myc, causing a rapid decline in c-Myc protein levels in NPC cells. Similar to c-Myc knockdown, NPC cells treated with hyperthermia showed growth inhibition associated with the downregulation of c-Myc target genes. Moreover, low levels of c-Myc could be sustainably repressed in NPC cells through repeated hyperthermia treatments. Importantly, the key findings of growth inhibition and decreased c-Myc protein levels were reproduced in NPC tumor xenografts. Bioinformatic analyses showed that downregulation of c-Myc constituted a central node in the hyperthermia response of NPC cells. CONCLUSION: Our study reveals that hyperthermia can readily destabilize c-Myc levels in NPC cells and inhibit tumor growth. This proposes new strategies for implementing hyperthermia to target c-Myc-driven cancers to improve therapeutic efficacy.


Subject(s)
Hyperthermia, Induced , Nasopharyngeal Neoplasms , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Mice , Mice, Nude , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/therapy
6.
Nucleic Acids Res ; 48(6): 3089-3102, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32030426

ABSTRACT

Long non-coding RNAs (lncRNAs) have emerged as important biological tuners. Here, we reveal the role of an uncharacterized lncRNA we call SENEBLOC that is expressed by both normal and transformed cells under homeostatic conditions. SENEBLOC was shown to block the induction of cellular senescence through dual mechanisms that converge to repress the expression of p21. SENEBLOC facilitates the association of p53 with MDM2 by acting as a scaffold to promote p53 turnover and decrease p21 transactivation. Alternatively, SENEBLOC was shown to affect epigenetic silencing of the p21 gene promoter through regulation of HDAC5. Thus SENEBLOC drives both p53-dependent and p53-independent mechanisms that contribute to p21 repression. Moreover, SENEBLOC was shown to be involved in both oncogenic and replicative senescence, and from the perspective of senolytic agents we show that the antagonistic actions of rapamycin on senescence are dependent on SENEBLOC expression.


Subject(s)
Aging/genetics , Neoplasms/genetics , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , Heterografts , Histone Deacetylases/genetics , Humans , Mice , Protein Binding/genetics , Signal Transduction/genetics
7.
Proc Natl Acad Sci U S A ; 115(7): E1465-E1474, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29378948

ABSTRACT

The oncoprotein c-Myc plays an important role in regulating glycolysis under normoxia; yet, in cancer cells, HIF1α, which is essential for driving glycolysis under hypoxia, is often up-regulated even in the presence of oxygen. The relationship between these two major regulators of the Warburg effect remains to be fully defined. Here we demonstrate that regulation of a long noncoding RNA (lncRNA), named IDH1-AS1, enables c-Myc to collaborate with HIF1α in activating the Warburg effect under normoxia. c-Myc transcriptionally repressed IDH1-AS1, which, upon expression, promoted homodimerization of IDH1 and thus enhanced its enzymatic activity. This resulted in increased α-KG and decreased ROS production and subsequent HIF1α down-regulation, leading to attenuation of glycolysis. Hence, c-Myc repression of IDH1-AS1 promotes activation of the Warburg effect by HIF1α. As such, IDH1-AS1 overexpression inhibited cell proliferation, whereas silencing of IDH1-AS1 promoted cell proliferation and cancer xenograft growth. Restoring IDH1-AS1 expression may therefore represent a potential metabolic approach for cancer treatment.


Subject(s)
Glycolysis/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isocitrate Dehydrogenase/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Antisense/genetics , RNA, Long Noncoding/genetics , Uterine Cervical Neoplasms/prevention & control , Animals , Cell Proliferation , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Ketoglutaric Acids/metabolism , Mice , Mice, Nude , Proto-Oncogene Proteins c-myc/genetics , Transcriptional Activation , Tumor Cells, Cultured , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Proc Natl Acad Sci U S A ; 115(50): E11661-E11670, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30478051

ABSTRACT

Long noncoding RNAs (lncRNAs) function through a diverse array of mechanisms that are not presently fully understood. Here, we sought to find lncRNAs differentially regulated in cancer cells resistant to either TNF-related apoptosis-inducing ligand (TRAIL) or the Mcl-1 inhibitor UMI-77, agents that act through the extrinsic and intrinsic apoptotic pathways, respectively. This work identified a commonly up-regulated lncRNA, ovarian adenocarcinoma-amplified lncRNA (OVAAL), that conferred apoptotic resistance in multiple cancer types. Analysis of clinical samples revealed OVAAL expression was significantly increased in colorectal cancers and melanoma in comparison to the corresponding normal tissues. Functional investigations showed that OVAAL depletion significantly inhibited cancer cell proliferation and retarded tumor xenograft growth. Mechanically, OVAAL physically interacted with serine/threonine-protein kinase 3 (STK3), which, in turn, enhanced the binding between STK3 and Raf-1. The ternary complex OVAAL/STK3/Raf-1 enhanced the activation of the RAF protooncogene serine/threonine-protein kinase (RAF)/mitogen-activated protein kinase kinase 1 (MEK)/ERK signaling cascade, thus promoting c-Myc-mediated cell proliferation and Mcl-1-mediated cell survival. On the other hand, depletion of OVAAL triggered cellular senescence through polypyrimidine tract-binding protein 1 (PTBP1)-mediated p27 expression, which was regulated by competitive binding between OVAAL and p27 mRNA to PTBP1. Additionally, c-Myc was demonstrated to drive OVAAL transcription, indicating a positive feedback loop between c-Myc and OVAAL in controlling tumor growth. Taken together, these results reveal that OVAAL contributes to the survival of cancer cells through dual mechanisms controlling RAF/MEK/ERK signaling and p27-mediated cell senescence.


Subject(s)
Cellular Senescence/genetics , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , MAP Kinase Signaling System , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Cell Survival/genetics , Cell Survival/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Heterografts , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine-Threonine Kinase 3 , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism
9.
Histochem Cell Biol ; 153(2): 77-87, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31834485

ABSTRACT

CD44 is a transmembrane receptor that acts as adhesion protein, fundamentally recognizing hyaluronan, an essential component of the extracellular matrix. It has a well-established functional association with cancer metastasis, particularly the CD44 variant forms which are considered essential markers of cancer stem cells. CD44 itself lacks intrinsic kinase activity but rather engages in signalling through specific interactions with kinases and other signalling components. Proteolysis within its transmembrane region also leads to release of the CD44 cytoplasmic domain, which can translocate to the nucleus and regulate transcription. A third signalling modality has been reported where the intact CD44 receptor translocates to the nucleus. Here, we investigated the latter using imaging techniques together with biochemical analyses. Our findings support observations where CD44 is cleaved prior to nuclear translocation and challenges the evidence for the presence of intact CD44 receptors in the cell nucleus. Conclusions regarding the presence of intact CD44 in the cell nucleus as a signalling modality, therefore, require re-evaluation. We highlight artefacts and common technical issues associated with these experiments that can lead to misinterpretation.


Subject(s)
Cell Nucleus/metabolism , Hyaluronan Receptors/analysis , Receptors, Cell Surface/metabolism , HT29 Cells , Humans , Hyaluronan Receptors/metabolism , Signal Transduction
10.
Mol Ther ; 27(2): 365-379, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30341010

ABSTRACT

Laryngeal squamous cell carcinoma (LSCC) is a common form of head and neck cancer with poor prognosis. However, the mechanism underlying the pathogenesis of LSCC remains unclear. Here, we demonstrated increased expression of fascin actin-bundling protein 1 (FSCN1) and decreased expression of microRNA-145-5p (miR-145-5p) in a clinical cohort of LSCC. Luciferase assay revealed that miR-145-5p is a negative regulator of FSCN1. Importantly, low miR-145-5p expression was correlated with TNM (tumor, node, metastasis) status and metastasis. Moreover, cases with low miR-145-5p/high FSCN1 expression showed poor prognosis, and these characteristics together served as independent prognostic indicators of survival. Gain- and loss-of-function studies showed that miR-145-5p overexpression or FSCN1 knockdown inhibited LSCC migration, invasion, and growth by suppressing the epithelial-mesenchymal transition along with inducing cell-cycle arrest and apoptosis. Additionally, hypermethylation of the miR-145-5p promoter suggested that repression of miR-145-5p arises through epigenetic inactivation. LSCC tumor growth in vivo could be inhibited by using miR-145-5p agomir or FSCN1 small interfering RNA (siRNA), which highlights the potential for clinical translation. Collectively, our findings indicate that miR-145-5p plays critical roles in inhibiting the progression of LSCC by suppressing FSCN1. Both miR-145-5p and FSCN1 are important potential prognostic markers and therapeutic targets for LSCC.


Subject(s)
Carrier Proteins/metabolism , DNA Methylation/physiology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/metabolism , MicroRNAs/genetics , Microfilament Proteins/metabolism , Promoter Regions, Genetic/genetics , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carrier Proteins/genetics , Cell Line , Cell Line, Tumor , DNA Methylation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/physiology , Microfilament Proteins/genetics
11.
Proteomics ; 19(21-22): e1900020, 2019 11.
Article in English | MEDLINE | ID: mdl-31169343

ABSTRACT

This study intends to investigate the transcriptional regulatory role of miR-145-5p in laryngeal squamous cell carcinoma (LSCC). LSCC cell line TU-177 is transfected with miR-145-5p mimics, generating miR-145-5p-overexpression LSCC cells. Whole transcriptome microarrays are used to investigate the differentially expressed lncRNAs, circRNAs, mRNAs, and miRNAs. The target genes of miRNAs are predicted and performed functional annotation. Additionally, the circRNAs, lncRNAs, and mRNAs that interact with miRNAs are predicted, and then the competing endogenous RNAs (ceRNAs) are predicted. Microarray analysis identifies 26 miRNAs, 248 mRNAs, 1118 lncRNAs, and 382 circRNAs differentially expressed in miR-145-5p overexpressed LSCC cells. Overall, 675 target genes are identified for the differentially expressed miRNAs, which involved in cell adhesion associated gene ontology (GO) terms, and MAPK and FoxO signaling pathways. The up-regulated mRNAs involved in the pathway of ABC transporters, while the down-regulated mRNAs involved in pathway of olfactory transduction. Moreover, 149 ceRNAs are predicted, which are associated with apoptosis, Wnt pathway, and metabolic pathway. Furthermore, qPCR results confirm that miR-145-5p affects expression of lncRNAs, miRNAs, mRNAs, and circRNAs in LSCC cells. Collectively, miR-145-5p may be inhibits LSCC progression via ceRNA-mediated pathways, such as WNT2B-miR-145-5p-NONHSAT127539.2, CASP10-miR-145-5p-NONHSAT127539.2, CASP10-miR-145-5p-circ_0003519, and TPO-miR-145-5p-circ_0003519.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Regulatory Networks , Laryngeal Neoplasms/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Molecular Sequence Annotation , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Proteomics ; 19(21-22): e1900059, 2019 11.
Article in English | MEDLINE | ID: mdl-31287215

ABSTRACT

Dysregulation of fascin actin-bundling protein 1 (FSCN1) enhances cell proliferation, invasion, and motility in laryngeal squamous cell carcinoma (LSCC), while the mechanism remains unclear. Here, co-immunoprecipitation and mass spectrometry is utilized to identify potential FSCN1-binding proteins. Functional annotation of FSCN1-binding proteins are performed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis. Furthermore, the protein-protein interaction network of FSNC1-binding proteins is constructed and the interactions between FSCN1 and novel identified interacting proteins AIMP1 and LTA4H are validated. Moreover, the expression and functional role of AIMP1 and LTA4H in LSCC are investigated. A total of 123 proteins are identified as potential FSCN1-binding proteins, and functional annotation shows that FSCN1-binding proteins are significantly enriched in carcinogenic processes, such as filopodium assembly-regulation and GTPase activity. Co-IP/western blotting and immunofluorescence confirm that AIMP1 and LTA4H bind and colocalize with FSCN1. Furthermore, both AIMP1 and LTA4H are upregulated in LSCC tissues, and knockdown of AIMP1 or LTA4H inhibits LSCC cell proliferation, migration, and invasion. Collectively, the identification of FSCN1-binding partners enhances understanding of the mechanism of FSCN1-mediated malignant phenotypes, and these findings indicate that FSCN1 binds to AIMP1 and LTA4H might promote the progression of LSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carrier Proteins/genetics , Cytokines/genetics , Epoxide Hydrolases/genetics , Laryngeal Neoplasms/genetics , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , RNA-Binding Proteins/genetics , Carcinoma, Squamous Cell/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Laryngeal Neoplasms/pathology , Mass Spectrometry , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Protein Binding/genetics , Protein Interaction Maps/genetics
13.
Biochem Biophys Res Commun ; 514(3): 625-631, 2019 06 30.
Article in English | MEDLINE | ID: mdl-31076104

ABSTRACT

Fat1 cadherin is broadly expressed throughout the nervous system and has been implicated in neuronal differentiation. Here we examined the functional contribution of FAT1 during neuronal differentiation of the Ntera2 cell line model. FAT1 expression was increased during the retinoic acid (RA)-induced differentiation of NTera2 cells. Depletion of FAT1 with siRNA decreased the number of neurites produced after RA treatment. Moreover, FAT1 silencing also led to decreased Ser127-phosphorylation of YAP along with transcriptional increases in the Hippo target genes CTGF and ANKRD1, suggesting FAT1 alters Hippo signalling during differentiation. In the context of the Ntera2 model, FAT1 is required for efficient neuritogenesis, acting as a regulator of neurite formation during the early stages of differentiation.


Subject(s)
Cadherins/metabolism , Cell Differentiation , Neurites/metabolism , Neurogenesis , Animals , Cell Line, Tumor , Computer Simulation , Gene Knockdown Techniques , Gene Silencing , Hippo Signaling Pathway , Humans , Mice , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Tretinoin/pharmacology
14.
Am J Pathol ; 188(1): 229-241, 2018 01.
Article in English | MEDLINE | ID: mdl-29037860

ABSTRACT

Neurotrophin receptors are emerging targets in oncology, but their clinicopathologic significance in thyroid cancer is unclear. In this study, the neurotrophin tyrosine receptor kinase TrkA (also called NTRK1), the common neurotrophin receptor p75NTR, and the proneurotrophin receptor sortilin were analyzed with immunohistochemistry in a cohort of thyroid cancers (n = 128) and compared with adenomas and normal thyroid tissues (n = 62). TrkA was detected in 20% of thyroid cancers, compared with none of the benign samples (P = 0.0007). TrkA expression was independent of histologic subtypes but associated with lymph node metastasis (P = 0.0148), suggesting the involvement of TrkA in tumor invasiveness. Nerves in the tumor microenvironment were positive for TrkA. p75NTR was overexpressed in anaplastic thyroid cancers compared with papillary and follicular subtypes (P < 0.0001). Sortilin was overexpressed in thyroid cancers compared with benign thyroid tissues (P < 0.0001). Neurotrophin receptor expression was confirmed in a panel of thyroid cancer cell lines at the mRNA and protein levels. Functional investigations using the anaplastic thyroid cancer cell line CAL-62 found that siRNA against TrkA, p75NTR, and sortilin decreased cell survival and cell migration through decreased SRC and ERK activation. Together, these data reveal TrkA, p75NTR, and sortilin as potential therapeutic targets in thyroid cancer.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Adenocarcinoma, Follicular/metabolism , Carcinoma, Papillary/metabolism , Nerve Tissue Proteins/metabolism , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/metabolism , Thyroid Gland/metabolism , Thyroid Neoplasms/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adenocarcinoma, Follicular/pathology , Carcinoma, Papillary/pathology , Cell Line, Tumor , Cell Movement , Cell Survival , Humans , Lymphatic Metastasis/pathology , Nerve Tissue Proteins/genetics , RNA, Small Interfering , Receptor, trkA/genetics , Receptors, Nerve Growth Factor/genetics , Thyroid Neoplasms/pathology , Tumor Microenvironment
15.
Adv Exp Med Biol ; 1206: 127-149, 2019.
Article in English | MEDLINE | ID: mdl-31776983

ABSTRACT

The tumor suppressor gene Tp53 encodes p53, a pivotal transcription factor with a broad target gene repertoire. Induction and stabilization of p53 during DNA damage and oncogene activation function to induce cell cycle arrest, apoptosis, or senescence. These actions are a failsafe to counteract carcinogenesis but Tp53 also plays a key role in regulating different aspects of cell metabolism including autophagy. Autophagy or cellular "self-eating" involves the dismantling and remodeling of cellular components, activities which are fundamental in maintaining cellular homeostasis and in supporting cell growth. After providing an historical overview of Tp53 research, the purpose of this chapter is to review the different mechanistic aspects of Tp53's role in autophagy and to highlight the key challenges which lie ahead. Tp53 functions are regulated by tight control of its cellular levels and notably, Tp53 can be both an activator or inhibitor of autophagy. Under stress conditions such as nutrient depletion or hypoxia, Tp53 contributes to autophagic activation by inhibiting mTOR signaling. Alternatively, p53 can interact with death-associated protein kinase 1 (DAPK1), acting to stabilize nuclear p53 amongst other functions including activation of the key autophagic mediator, Beclin-1. Under normal physiological conditions, Tp53 can inhibit autophagosome formation but stress conditions can also result in Tp53-mediated promotion of autophagy, demonstrating that Tp53 actions are highly context dependent. Tp53 target genes also play key opposing roles in autophagy induction or inhibition such as DRAM and TIGAR, respectively. Finally, the role of Tp53 mutants in autophagy regulation are discussed.


Subject(s)
Apoptosis Regulatory Proteins , Autophagy , Membrane Proteins , Tumor Suppressor Protein p53 , Animals , Apoptosis Regulatory Proteins/metabolism , Humans , Membrane Proteins/metabolism , Phosphoric Monoester Hydrolases , Signal Transduction , Tumor Suppressor Protein p53/metabolism
16.
Nature ; 539(7630): 503-505, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27828945
17.
Cell Mol Life Sci ; 72(23): 4653-69, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26104008

ABSTRACT

The Hippo pathway is emerging as a critical nexus that balances self-renewal of progenitors against differentiation; however, upstream elements in vertebrate Hippo signalling are poorly understood. High expression of Fat1 cadherin within the developing neuroepithelium and the manifestation of severe neurological phenotypes in Fat1-knockout mice suggest roles in neurogenesis. Using the SH-SY5Y model of neuronal differentiation and employing gene silencing techniques, we show that FAT1 acts to control neurite outgrowth, also driving cells towards terminal differentiation via inhibitory effects on proliferation. FAT1 actions were shown to be mediated through Hippo signalling where it activated core Hippo kinase components and antagonised functions of the Hippo effector TAZ. Suppression of FAT1 promoted the nucleocytoplasmic shuttling of TAZ leading to enhanced transcription of the Hippo target gene CTGF together with accompanying increases in nuclear levels of Smad3. Silencing of TAZ reversed the effects of FAT1 depletion thus connecting inactivation of TAZ-TGFbeta signalling with Hippo signalling mediated through FAT1. These findings establish FAT1 as a new upstream Hippo element regulating early stages of differentiation in neuronal cells.


Subject(s)
Cadherins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/cytology , Protein Serine-Threonine Kinases/metabolism , Active Transport, Cell Nucleus , Cadherins/genetics , Cell Differentiation , Cell Line , Cell Proliferation , Gene Knockdown Techniques , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/genetics , Neurites/physiology , Neurons/physiology , Signal Transduction , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins
18.
Am J Pathol ; 184(12): 3156-62, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25285721

ABSTRACT

Nerve infiltration is essential to prostate cancer progression, but the mechanism by which nerves are attracted to prostate tumors remains unknown. We report that the precursor of nerve growth factor (proNGF) is overexpressed in prostate cancer and involved in the ability of prostate cancer cells to induce axonogenesis. A series of 120 prostate cancer and benign prostate hyperplasia (BPH) samples were analyzed by IHC for proNGF. ProNGF was mainly localized in the cytoplasm of epithelial cells, with marked expression in cancer compared with BPH. Importantly, the proNGF level positively correlated with the Gleason score (n = 104, τB = 0.51). A higher level of proNGF was observed in tumors with a Gleason score of ≥8 compared with a Gleason score of 7 and 6 (P < 0.001). In vitro, proNGF was detected in LNCaP, DU145, and PC-3 prostate cancer cells and BPH-1 cells but not in RWPE-1 immortalized nontumorigenic prostate epithelial cells or primary normal prostate epithelial cells. Co-culture of PC12 neuronal-like cells or 50B11 neurons with PC-3 cells resulted in neurite outgrowth in neuronal cells that was inhibited by blocking antibodies against proNGF, indicating that prostate cancer cells can induce axonogenesis via secretion of proNGF. These data reveal that ProNGF is a biomarker associated with high-risk prostate cancers and a potential driver of infiltration by nerves.


Subject(s)
Biomarkers, Tumor/metabolism , Nerve Growth Factor/metabolism , Prostate/innervation , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Axons/metabolism , Cell Line, Tumor , Coculture Techniques , Cytoplasm/metabolism , Epithelial Cells/metabolism , Humans , Immunohistochemistry , Male , Neoplasm Grading , Neurons/metabolism , Prostatic Hyperplasia/metabolism
19.
Brain Behav Immun ; 44: 235-46, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25449583

ABSTRACT

The immune and nociceptive systems are shaped during the neonatal period where they undergo fine-tuning and maturation. Painful experiences during this sensitive period of development are known to produce long-lasting effects on the immune and nociceptive responses. It is less clear, however, whether inflammatory pain responses are primed by neonatal exposure to mild immunological stimuli, such as with lipopolysaccharide (LPS). Here, we examine the impact of neonatal LPS exposure on inflammatory pain responses, peripheral and hippocampal interleukin-1ß (IL-1ß), as well as mast cell number and degranulation in preadolescent and adult rats. Wistar rats were injected with LPS (0.05mg/kg IP, Salmonella enteritidis) or saline on postnatal days (PNDs) 3 and 5 and later subjected to the formalin test at PNDs 22 and 80-97. At both time-points, and one-hour after formalin injection, blood and hippocampus were collected for measuring circulating and central IL-1ß levels using ELISA and Western blot, respectively. Paw tissue was also isolated to assess mast cell number and degree of degranulation using Toluidine Blue staining. Behavioural analyses indicate that at PND 22, LPS-challenged rats displayed enhanced flinching (p<.01) and licking (p<.01) in response to formalin injection. At PNDs 80-97, LPS-challenged rats exhibited increased flinching (p<.05), an effect observed in males only. Furthermore, neonatal LPS exposure enhanced circulating IL-1ß and mast cell degranulation in preadolescent but not adult rats following formalin injection. Hippocampal IL-1ß levels were increased in LPS-treated adult but not preadolescent rats in response to formalin injection. These data suggest neonatal LPS exposure produces developmentally regulated changes in formalin-induced behavioural responses, peripheral and central IL-1ß levels, as well as mast cell degranulation following noxious stimulation later in life. These findings highlight the importance of immune activation during the neonatal period in shaping immune response and pain sensitivity later in life. This is of clinical relevance given the high prevalence of bacterial infection during the neonatal period, particularly in the vulnerable population of preterm infants admitted to neonatal intensive care units.


Subject(s)
Encephalitis/immunology , Nociception/physiology , Pain/immunology , Animals , Animals, Newborn , Cell Count , Cell Degranulation , Encephalitis/chemically induced , Encephalitis/metabolism , Female , Formaldehyde , Hippocampus/immunology , Hippocampus/metabolism , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/blood , Interleukin-1beta/immunology , Lipopolysaccharides , Male , Mast Cells/immunology , Mast Cells/physiology , Motor Activity , Pain/chemically induced , Pain Measurement , Rats , Rats, Wistar
20.
Exp Cell Res ; 323(1): 41-55, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24560745

ABSTRACT

Fat1 is a single pass transmembrane protein and the largest member of the cadherin superfamily. Mouse knockout models and in vitro studies have suggested that Fat1 influences cell polarity and motility. Fat1 is also an upstream regulator of the Hippo pathway, at least in lower vertebrates, and hence may play a role in growth control. In previous work we have established that FAT1 cadherin is initially cleaved by proprotein convertases to form a noncovalently linked heterodimer prior to expression on the cell surface. Such processing was not a requirement for cell surface expression, since melanoma cells expressed both unprocessed FAT1 and the heterodimer on the cell surface. Here we further establish that the site 1 (S1) cleavage step to promote FAT1 heterodimerisation is catalysed by furin and we identify the cleavage site utilised. For a number of other transmembrane receptors that undergo heterodimerisation the S1 processing step is thought to occur constitutively but the functional significance of heterodimerisation has been controversial. It has also been generally unclear as to the significance of receptor heterodimerisation with respect to subsequent post-translational proteolysis that often occurs in transmembrane proteins. Exploiting the partial deficiency of FAT1 processing in melanoma cells together with furin-deficient LoVo cells, we manipulated furin expression to demonstrate that only the heterodimer form of FAT1 is subject to cleavage and subsequent release of the extracellular domain. This work establishes S1-processing as a clear functional prerequisite for ectodomain shedding of FAT1 with general implications for the shedding of other transmembrane receptors.


Subject(s)
Cadherins/metabolism , Furin/metabolism , Protein Multimerization , Cadherins/biosynthesis , Cell Line, Tumor , Furin/genetics , Humans , Keratinocytes , Melanoma/metabolism , Proprotein Convertases/genetics , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proteolysis , RNA Interference , RNA, Small Interfering , Serine Endopeptidases/genetics , Subtilisins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL