Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Mol Biol Cell ; 33(8): ar72, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35476603

ABSTRACT

Chromosome structure and nuclear organization are important factors in the regulation of gene expression. Transcription of a gene is influenced by local and global chromosome features such as chromatin condensation status. The relationship between the 3D position of a gene in the nucleus and its activity is less clear. Here we used high-throughput imaging to perform a large-scale analysis of the spatial location of nearly 100 hypoxia-responsive genes to determine whether their location and activity state are correlated. Radial distance analysis demonstrated that the majority of Hypoxia-Inducible Factor (HIF)- and CREB-dependent hypoxia-responsive genes are located in the intermediate region of the nucleus, and some of them changed their radial position in hypoxia. Analysis of the relative distances among a subset of HIF target genes revealed that some gene pairs altered their relative location to each other on hypoxic treatment, suggesting higher-order chromatin rearrangements. While these changes in location occurred in response to hypoxic activation of the target genes, they did not correlate with the extent of their activation. These results suggest that induction of the hypoxia-responsive gene expression program is accompanied by spatial alterations of the genome, but that radial and relative gene positions are not directly related to gene activity.


Subject(s)
Chromatin , Hypoxia , Cell Hypoxia , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Transcriptional Activation
3.
Breast Cancer ; 28(5): 1062-1071, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34047951

ABSTRACT

BACKGROUND: Previously, we reported that Wnt5a-positive breast cancer can be classified as estrogen receptor (ER)-positive breast cancer; its prognosis is worse than that of Wnt5a-negative breast cancer. This study aimed to investigate the mechanisms underlying the poor prognosis in Wnt5a-positive breast cancer patients. METHODS: In total, 151 consecutive ER-positive breast cancer patients who underwent resection between January 2011 and February 2014 were enrolled. DNA microarray and pathway analyses were conducted using MCF-7 cells stably expressing Wnt5a [MCF-7/Wnt5a (+)]. Based on the outcomes, cell viability/drug sensitivity assays, and mutation analysis were performed using cell cultures and breast cancer tissues. The relationship between Wnt5a and the PI3K-AKT-mTOR signaling pathway was also examined. RESULTS: The relapse-free survival rate in patients with Wnt5a-positive breast cancer was significantly lower than that in patients with Wnt5a-negative breast cancer (P = 0.047). DNA microarray data suggest that only the cytochrome P450 (CYP) pathway was significantly upregulated in MCF-7/Wnt5a (+) cells (P = 0.0440). Additionally, MCF-7/Wnt5a (+) cells displayed reduced sensitivity to the metabolic substrates of CYP, tamoxifen (P < 0.001), paclitaxel (P < 0.001), and cyclophosphamide (P < 0.001). Of note, PIK3CA mutations were not associated with the expression of Wnt5a in breast cancer tissue and culture cells. CONCLUSIONS: In ER-positive breast cancer, Wnt5a upregulates the CYP metabolic pathway and suppresses tamoxifen, paclitaxel, and cyclophosphamide resistance, all of the three, standard treatment methods for ER-positive breast cancer. Wnt5a is thus potentially involved in the poor prognosis of ER-positive breast cancer independently of the PI3K-AKT-mTOR signaling pathway.


Subject(s)
Breast Neoplasms/genetics , Receptors, Estrogen/antagonists & inhibitors , Wnt-5a Protein/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Longitudinal Studies , MCF-7 Cells , Middle Aged , Progression-Free Survival , Receptor, ErbB-2 , Retrospective Studies , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Up-Regulation
4.
Biochem Biophys Res Commun ; 520(1): 128-135, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31582221

ABSTRACT

Cells require proper regulation of energy metabolism to maintain cellular homeostasis. Pyruvate dehydrogenase (PDH) is a metabolic enzyme that converts pyruvate into acetyl-CoA, connecting glycolysis to the TCA cycle, thus regulating cellular energy metabolism. PDH is involved in multiple cellular processes, such as glucose metabolism, fatty acid synthesis, and protein acetylation, all of which are mediated by acetyl-CoA. We previously demonstrated that PDH-E1ß is downregulated in prolonged hypoxia and inhibits PDH activity, which serves as machinery to securely inhibit PDH activity together with PDH-E1α phosphorylation. PDH has been identified to localize to the nucleus in addition to mitochondria, but its precise regulatory mechanisms in the nucleus remain elusive. In the present study, we characterized nuclear PDH during prolonged hypoxia. Nuclear PDH complex was downregulated under hypoxic conditions, and PDH activity was reduced. Depletion of HIF-1α partly recovered nuclear levels of the PDH complex. Furthermore, decreased nuclear PDH activity resulted in reduced histone H3 acetylation, altering the gene expression profile of cells exposed to prolonged hypoxia. Taken together, these findings indicate that nuclear PDH complex is downregulated under prolonged hypoxic conditions and controls gene expression.


Subject(s)
Cell Nucleus/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Mitochondria/enzymology , Pyruvate Dehydrogenase Complex/metabolism , Cell Hypoxia , Cell Line, Tumor , Energy Metabolism , HeLa Cells , Histones/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MCF-7 Cells , Phosphorylation , RNA, Small Interfering/metabolism
5.
Int J Mol Sci ; 20(13)2019 Jul 03.
Article in English | MEDLINE | ID: mdl-31277312

ABSTRACT

Eukaryotes are often subjected to different kinds of stress. In order to adjust to such circumstances, eukaryotes activate stress-response pathways and regulate gene expression. Eukaryotic gene expression consists of many different steps, including transcription, RNA processing, RNA transport, and translation. In this review article, we focus on both transcriptional and post-transcriptional regulations of gene expression under hypoxic conditions. In the first part of the review, transcriptional regulations mediated by various transcription factors including Hypoxia-Inducible Factors (HIFs) are described. In the second part, we present RNA splicing regulations under hypoxic conditions, which are mediated by splicing factors and their kinases. This work summarizes and discusses the emerging studies of those two gene expression machineries under hypoxic conditions.


Subject(s)
Gene Expression Regulation , Hypoxia/genetics , Animals , Humans , Hypoxia/metabolism , RNA Splicing , Transcription Factors/metabolism , Transcription, Genetic
6.
Cancer Res ; 78(7): 1592-1603, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29436427

ABSTRACT

Downregulation of pyruvate dehydrogenase (PDH) is critical for the aberrant preferential activation of glycolysis in cancer cells under normoxic conditions. Phosphorylation-dependent inhibition of PDH is a relevant event in this process, but it is not durable as it relies on PDH kinases that are activated ordinarily under hypoxic conditions. Thus, it remains unclear how PDH is durably downregulated in cancer cells that are not hypoxic. Building on evidence that PDH activity depends on the stability of a multi-protein PDH complex, we found that the PDH-E1ß subunit of the PDH complex is downregulated to inhibit PDH activity under conditions of prolonged hypoxia. After restoration of normoxic conditions, reduced expression of PDH-E1ß was sustained such that glycolysis remained highly activated. Notably, PDH-E1ß silencing in cancer cells produced a metabolic state strongly resembling the Warburg effect, but inhibited tumor growth. Conversely, enforced exogenous expression of PDH-E1ß durably increased PDH activity and promoted the malignant growth of breast cancer cells in vivo Taken together, our results establish the specific mechanism through which PDH acts as an oncogenic factor by tuning glycolytic metabolism in cancer cells.Significance: This seminal study offers a mechanistic explanation for why glycolysis is aberrantly activated in normoxic cancer cells, offering insights into this long-standing hallmark of cancer termed the Warburg effect. Cancer Res; 78(7); 1592-603. ©2018 AACR.


Subject(s)
Cell Hypoxia/physiology , Glycolysis/physiology , Neoplasms/pathology , Pyruvate Dehydrogenase (Lipoamide)/biosynthesis , Pyruvate Dehydrogenase (Lipoamide)/genetics , Animals , Cell Line, Tumor , Disease Progression , Down-Regulation/genetics , Energy Metabolism/physiology , Female , HeLa Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/genetics , Oxygen Consumption/physiology
7.
Article in English | MEDLINE | ID: mdl-29183987

ABSTRACT

DNA fluorescence in situ hybridization (FISH) is the technique of choice to map the position of genomic loci in three-dimensional (3D) space at the single allele level in the cell nucleus. High-throughput DNA FISH methods have recently been developed using complex libraries of fluorescently labeled synthetic oligonucleotides and automated fluorescence microscopy, enabling large-scale interrogation of genomic organization. Although the FISH signals generated by high-throughput methods can, in principle, be analyzed by traditional spot-detection algorithms, these approaches require user intervention to optimize each interrogated genomic locus, making analysis of tens or hundreds of genomic loci in a single experiment prohibitive. We report here the design and testing of two separate machine learning-based workflows for FISH signal detection in a high-throughput format. The two methods rely on random forest (RF) classification or convolutional neural networks (CNNs), respectively. Both workflows detect DNA FISH signals with high accuracy in three separate fluorescence microscopy channels for tens of independent genomic loci, without the need for manual parameter value setting on a per locus basis. In particular, the CNN workflow, which we named SpotLearn, is highly efficient and accurate in the detection of DNA FISH signals with low signal-to-noise ratio (SNR). We suggest that SpotLearn will be useful to accurately and robustly detect diverse DNA FISH signals in a high-throughput fashion, enabling the visualization and positioning of hundreds of genomic loci in a single experiment.

8.
Int J Oncol ; 48(2): 657-69, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26691441

ABSTRACT

Identification and purification of cancer stem cells (CSCs) lead to the discovery of novel therapeutic targets; however, there has been no study on isolation of the CSC population among pancreatic neuroendocrine tumors (pNETs). This study aimed to identify pNET CSCs and to characterize a therapeutic candidate for pNET CSCs. We identified CSCs by aldehyde dehydrogenase (ALDH) activity in pNET clinical specimens and cell lines. We verified whether or not these cells have the stemness property in vivo and in vitro. ALDHhigh cells, but not control bulk cells, formed spheres, proliferated under hypoxic condition as well as normoxic condition and promoted cell motility, which are features of CSCs. Injection of as few as 10 ALDHhigh cells led to subcutaneous tumor formation, and 105 ALDHhigh cells, but not control bulk cells, established metastases in mice. Comprehensive gene expression analysis revealed that genes associated with mesenchymal stem cells, including CD73, were overexpressed in ALDHhigh cells. Additionally, the in vitro and in vivo effects of an inhibitor of CD73 were investigated. The CD73 inhibitor APCP significantly attenuated in vitro sphere formation and cell motility, as well as in vivo tumor growth observed for ALDHhigh cells. Finally, its expression was evaluated using clinical pNET tissue samples. Immunohistochemical analysis of clinical tissue samples demonstrated CD73 expression was significantly correlated with the invasion into adjacent organs. Since recent studies revealed CD73 as a potential biomarker of anti-PD-1 immune checkpoint therapy, CD73 might be a promising therapeutic target for pNET CSCs.


Subject(s)
5'-Nucleotidase/metabolism , Neoplastic Stem Cells/metabolism , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/physiology , Female , GPI-Linked Proteins/metabolism , Gene Expression/physiology , Humans , Mice
9.
Biochem Biophys Res Commun ; 469(2): 243-50, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26642955

ABSTRACT

Living cells are frequently exposed to various stresses. Hypoxic conditions induce endoplasmic reticulum (ER) stress, and activate the unfolded protein response (UPR) to maintain homeostasis. We previously reported that CREB has an important role in the proper response to prolonged hypoxia. To further understand the role of CREB in the hypoxic response, CREB stable knock-down (CREB-KD) cells were established from breast cancer MDA-MB231 cells and analyzed. CREB was activated by ER stress, and activation of CREB and the UPR pathway occurred in a coordinated manner in response to different stimuli, including ER stress-inducing chemicals, prolonged hypoxia, and oxygen-glucose deprivation (OGD). Depletion of CREB decreased the expression of IRE1α and PERK, two critical UPR signaling molecules. Promoter analysis and a chromatin immunoprecipitation assay indicated that CREB binds to the promoter region of these genes and regulates their expression. ER stress induced by hypoxia was reduced in CREB-KD cells, leading to reduced tumor metastasis to the lung. Finally, OGD strongly activated the UPR and induced cell death in control cells, whereas the UPR was moderately activated in CREB-KD cells, which were more resistant to cell death. This study demonstrates a new role for CREB as a regulator of ER stress, which is required to properly respond to stressful conditions, such as hypoxia.


Subject(s)
Breast Neoplasms/metabolism , CREB-Binding Protein/metabolism , Endoplasmic Reticulum/metabolism , Endoribonucleases/metabolism , Oxidative Stress/physiology , Protein Serine-Threonine Kinases/metabolism , eIF-2 Kinase/metabolism , CREB-Binding Protein/chemistry , Cell Line, Tumor , Endoribonucleases/chemistry , Gene Expression Regulation, Neoplastic , Humans , Protein Folding , Protein Serine-Threonine Kinases/chemistry , eIF-2 Kinase/chemistry
10.
Biochem Biophys Res Commun ; 451(2): 288-94, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-25088999

ABSTRACT

Cells are frequently exposed to hypoxia in physiological and pathophysiological conditions in organisms. Control of energy metabolism is one of the critical functions of the hypoxic response. Hypoxia-Inducible Factor (HIF) is a central transcription factor that regulates the hypoxic response. HIF prolyl-hydroxylase PHDs are the enzymes that hydroxylate the α subunit of HIF and negatively regulate its expression. To further understand the physiological role of PHD3, proteomics were used to identify PHD3-interacting proteins, and pyruvate dehydrogenase (PDH)-E1ß was identified as such a protein. PDH catalyzes the conversion of pyruvate to acetyl-coA, thus playing a key role in cellular energy metabolism. PDH activity was significantly decreased in PHD3-depleted MCF7 breast cancer cells and PHD3(-/-) MEFs. PHD3 depletion did not affect the expression of the PDH-E1α, E1ß, and E2 subunits, or the phosphorylation status of E1α, but destabilized the PDH complex (PDC), resulting in less functional PDC. Finally, PHD3(-/-) cells were resistant to cell death in prolonged hypoxia with decreased production of ROS. Taken together, the study reveals that PHD3 regulates PDH activity in cells by physically interacting with PDC.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Procollagen-Proline Dioxygenase/metabolism , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Animals , Cell Death , Cell Hypoxia , Cells, Cultured , Enzyme Stability , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , MCF-7 Cells , Mice , Mice, Knockout , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics , Protein Subunits , Pyruvate Dehydrogenase (Lipoamide)/chemistry , Pyruvate Dehydrogenase Complex/chemistry , RNA, Small Interfering/genetics
11.
J Biol Chem ; 288(31): 22584-95, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23775082

ABSTRACT

Responses to low levels of oxygen (hypoxia) are essential to maintain homeostasis. During the hypoxic response, gene expression is altered by various transcription factors. The transcription factor, hypoxia-inducible factor (HIF), plays a central role in the hypoxic response. The α subunit of HIF, which is actively degraded during normoxia, becomes stabilized during hypoxia, which leads to HIF activation. A microarray analysis of HeLa cells showed that expression of matrix metalloproteinase 1 (MMP1) was markedly induced during prolonged hypoxia. CREB and NF-κB binding sites were identified in the MMP1 promoter region between 1945 and 1896 nucleotides upstream of the transcription start site. Assays with luciferase reporters demonstrated that HIF activity was induced during the early phase of hypoxia, whereas CREB and NF-κB were activated during the later (prolonged) phase. Depletion of CREB and/or NF-κB reduced MMP1 induction during prolonged hypoxia both at the mRNA and protein levels. A chromatin immunoprecipitation assay demonstrated binding of CREB and NF-κB to the MMP1 promoter. Finally, cell migration and invasion on a collagen matrix and pulmonary metastasis in nude mice were inhibited after depletion of CREB and NF-κB in MDA-MB-231 cells. Taken together, these results suggest that the cooperative action of CREB and NF-κB plays an important role to induce MMP1 expression during prolonged hypoxia and regulates cell migration and invasion in cancer cells.


Subject(s)
CREB-Binding Protein/physiology , Hypoxia/metabolism , Matrix Metalloproteinase 1/metabolism , NF-kappa B/metabolism , HeLa Cells , Humans , Matrix Metalloproteinase 1/genetics , Promoter Regions, Genetic
12.
Biochem Biophys Res Commun ; 435(3): 447-53, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23665324

ABSTRACT

Muscarinic acetylcholine receptors (mAchRs) are critical components of the cholinergic system, which is the key regulator of both the central and peripheral nervous systems in mammals. Interestingly, several components of the cholinergic system, including mAchRs and choline acetyltransferase (ChAT), have recently been found to be expressed in mouse embryonic stem (ES) cells and human placenta. These results raise the intriguing possibility that mAchRs play physiological roles in the regulation of early embryogenesis. Early embryogenesis can be mimicked in vitro using an ES cell-based culture system in which the cells form a primitive streak-like structure and efficiently develop into mesodermal progenitors. Here we report that chemical inhibitors specifically targeting mAchRs suppressed the expression of genes essential for primitive streak formation, including Wnt3, and thereby blocked mesodermal progenitor differentiation. Interestingly, mAchR inhibitors also reduced the expression of Cyp26a1, an enzyme involved in the catabolism of retinoic acid (RA). RA is an important regulator of Wnt3 signaling. Our study presents evidence indicating that mAchRs influence RA signaling necessary for the induction of the primitive streak. To our knowledge, this is the first report showing that mAchRs have important functions not only in adult mammals but also during early mammalian embryogenesis.


Subject(s)
Embryoid Bodies/metabolism , Primitive Streak/metabolism , Receptors, Muscarinic/physiology , Animals , Cell Differentiation/drug effects , Dicyclomine/pharmacology , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Gene Expression Regulation, Developmental/drug effects , Humans , Mice , Muscarinic Antagonists/pharmacology , Phenylpropionates/pharmacology , Primitive Streak/cytology , Primitive Streak/drug effects , Tretinoin/physiology
14.
Hepatology ; 58(1): 218-28, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23447025

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is one of the most aggressive malignancies because of recurrence and/or metastasis even after curative resection. Emerging evidence suggests that tumor metastasis and recurrence might be driven by a small subpopulation of stemness cells, so-called cancer stem cells (CSCs). Previous investigations have revealed that glioma and breast CSCs exhibit intrinsically low proteasome activity and that breast CSCs also reportedly contain a lower reactive oxygen species (ROS) level than corresponding nontumorigenic cells. Here we visualized two stem cell features, low proteasome activity and low intracellular ROS, in HCC cells using two-color fluorescence activated cell sorting to isolate cells with stem cell features. These cells were then analyzed for their division behavior in normoxia and hypoxia, expression of stem cell markers, tumorigenicity, metastatic potential, specific gene expression signatures, and their clinical implications. A visualized small subpopulation of HCC cells demonstrated asymmetric divisions. Their remarkable tumorigenicity in nonobese diabetic/severe combined immunodeficient mice suggested the cancer initiation potential of these HCC CSCs. Comprehensive gene expression analysis revealed that chemokine-related genes were up-regulated in the CSCs subpopulation. Our identified HCC CSCs facilitated the migration of macrophages in vitro and demonstrated metastatic potential by way of recruitment of macrophages in vivo. In patients who undergo curative operation for HCC, the CSC-specific gene signature in the liver microenvironment significantly correlates with recurrence. CONCLUSION: Based on these findings, the stem cell feature monitoring system proposed here is a promising tool to analyze the in vivo significance of CSC microenvironments in human HCCs.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Carcinoma, Hepatocellular/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Female , Flow Cytometry , Humans , Liver Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Prognosis , Proteasome Endopeptidase Complex/metabolism , Reactive Oxygen Species/metabolism
15.
Cancer Cell ; 18(1): 23-38, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20609350

ABSTRACT

Neuroendocrine (NE) phenotype, seen in >30% of prostate adenocarcinomas (PCa), and NE prostate tumors are implicated in aggressive prostate cancer. Formation of NE prostate tumors in the TRAMP mouse model was suppressed in mice lacking the ubiquitin ligase Siah2, which regulates HIF-1alpha availability. Cooperation between HIF-1alpha and FoxA2, a transcription factor expressed in NE tissue, promotes recruitment of p300 to transactivate select HIF-regulated genes, Hes6, Sox9, and Jmjd1a. These HIF-regulated genes are highly expressed in metastatic PCa and required for hypoxia-mediated NE phenotype, metastasis in PCa, and the formation of NE tumors. Tissue-specific expression of FoxA2 combined with Siah2-dependent HIF-1alpha availability enables a transcriptional program required for NE prostate tumor development and NE phenotype in PCa.


Subject(s)
Adenocarcinoma/pathology , Hepatocyte Nuclear Factor 3-beta/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neuroendocrine Tumors/pathology , Neurosecretory Systems/pathology , Prostatic Neoplasms/pathology , Ubiquitin-Protein Ligases/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-beta/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/secondary , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Neurosecretory Systems/metabolism , Phenotype , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Signal Transduction , Transcriptional Activation
16.
Exp Cell Res ; 316(17): 2871-82, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20599946

ABSTRACT

Prolyl-hydroxylase PHDs are the key regulators of hypoxia-inducible factor (HIF) stability. PHD3 has been shown to form a large complex under hypoxic conditions. While attempting to characterize the complex by determining its components, we identified human PRP19. hPRP19 is a multi-functional protein that plays a role in splicing, ubiquitination, and cell growth. Here, we report that PHD3 efficiently forms a complex with hPRP19 under hypoxic conditions and prevents cell death under prolonged hypoxic conditions. hPRP19 interacts with PHD3 via its C-terminal WD40 region, and the interaction is enhanced under hypoxic conditions through the utilization of the N-terminal coiled-coil domain. Cell death observed under prolonged hypoxic conditions is suppressed by the forced expression of hPRP19 in PC12 and HEK293T cells. In contrast, hPRP19 silencing by siRNA increased the caspase activity and enhanced cell death under hypoxic conditions in HeLa cells. Further, silencing of both PHD3 and hPRP19 recovers the cell death induced by hPRP19 single siRNA. Taken together, the results of our study indicate that hPRP19 interacts with PHD3 to suppress the cell death under hypoxic conditions by limiting the function of PHD3 which leads to caspase activation.


Subject(s)
Cell Death , DNA Repair Enzymes/metabolism , Dioxygenases/metabolism , Hypoxia/etiology , Nuclear Proteins/metabolism , Animals , Caspases/metabolism , Cell Line , Dioxygenases/antagonists & inhibitors , Humans , Hypoxia/pathology , Hypoxia-Inducible Factor-Proline Dioxygenases , PC12 Cells , Procollagen-Proline Dioxygenase/metabolism , Protein Binding , RNA Splicing Factors , Rats
17.
Pigment Cell Melanoma Res ; 23(3): 338-51, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20230483

ABSTRACT

Basal cell carcinomas (BCC), squamous cell carcinoma (SCC), and melanomas are the major types of skin tumors. Despite being skin cancers, the characteristics of each cancer are widely varied. BCCs often do not proliferate rapidly, and rarely metastasize. Squamous cell carcinomas are more malignant and a certain subtype of SCC is highly metastatic. Melanomas are highly proliferative and invasive, and are most frequently metastatic. Ubiquitin and ubiquitin-related proteins post-translationally modify proteins and thereby alter the functions of their target proteins. The ubiquitination process is involved in various physiological responses, including cell growth, cell death, and DNA damage repair. Accumulating evidence suggests that ubiquitin pathways are involved in different types of cancers, including skin cancers. This review describes the major ubiquitin pathways in BCC, SCC, and melanoma. The ubiquitin pathways that are activated among the skin cancers are highly diverse, which might reflect the various characteristics of these three cancer types. Meanwhile, there are also common pathways between BCC, SCC, and melanoma. Therefore, examining the ubiquitin pathways will reveal the mechanisms of these three major skin cancer types and will suggest treatment options.


Subject(s)
Disease Progression , Melanoma/metabolism , Melanoma/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Ubiquitination , Animals , Cell Proliferation , Humans , Signal Transduction
18.
J Biochem ; 146(6): 757-65, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19864435

ABSTRACT

Cells induce the hypoxia responses to adapt to the environment when organisms are exposed to a low oxygen environment. The hypoxia response leads to the activation of multiple cellular signalling pathways involved in regulation of respiration, metabolism, cell survival and so forth. Hypoxia-Inducible-Factor (HIF) pathway plays a central role during the hypoxia response as its expression and activity are regulated in an oxygen-dependent manner and it also regulates the expression of multiple hypoxia responsive genes. The expression of HIF is regulated by proline hydroxylation, which is mediated by HIF prolyl-hydroxylase named PHD. The hydroxylated HIF-alpha subunit is degraded via the ubiquitin-proteasome pathway. The PHD activity needs to be strictly regulated to ensure the stabilization of HIF under hypoxic conditions, because PHD leads to HIF degradation. This review describes the regulatory mechanism of HIF stability and activity under normoxia and hypoxic conditions. Furthermore, the role of the HIF-independent pathways during the hypoxia response, which is as important as the HIF pathway, will also be described.


Subject(s)
Hypoxia , Signal Transduction , Animals , Cell Hypoxia , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
19.
Mol Cancer Res ; 7(4): 443-51, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19372575

ABSTRACT

Growing evidence indicates that ubiquitin ligases play a critical role in the hypoxia response. Among them, Siah2, a RING finger ligase, is an important regulator of pathways activated under hypoxia. Siah2 regulates prolyl hydroxylases PHD3 and 1 under oxygen concentration of 2% to 5%, thereby allowing accumulation of hypoxia-inducible factor (HIF)-1alpha, a master regulator of the hypoxia response within the range of physiological normoxic to mild hypoxic conditions. Growing evidence also indicates an important function for Siah2 in tumor development and progression based on pancreatic cancer, mammary tumor, and melanoma mouse models. This review summarizes our current understanding of Siah2 regulation and function with emphasis on hypoxia and tumorigenesis.


Subject(s)
Hypoxia/metabolism , Nuclear Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data
20.
Proc Natl Acad Sci U S A ; 105(43): 16713-8, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18946040

ABSTRACT

The ubiquitin ligase Siah2 has been shown to regulate prolyl hydroxylase 3 (PHD3) stability with concomitant effect on HIF-1alpha availability. Because HIF-1alpha is implicated in tumorigenesis and metastasis, we used SW1 mouse melanoma cells, which develop primary tumors with a propensity to metastasize, in a syngeneic mouse model to assess a possible role for Siah2 in these processes. Inhibiting Siah2 activity by expressing a peptide designed to outcompete association of Siah2-interacting proteins reduced metastasis through HIF-1alpha without affecting tumorigenesis. Conversely, inhibiting Siah2 activity by means of a dominant-negative Siah2 RING mutant primarily reduced tumorigenesis through the action of Sprouty 2, a negative regulator of Ras signaling. Consistent with our findings, reduced expression of PHD3 and Sprouty2 was observed in more advanced stages of melanoma tumors. Using complementary approaches, our data establish the role of Siah2 in tumorigenesis and metastasis by HIF-dependent and -independent mechanisms.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melanoma, Experimental/etiology , Membrane Proteins/metabolism , Neoplasm Metastasis , Ubiquitin-Protein Ligases/physiology , Adaptor Proteins, Signal Transducing , Animals , Intracellular Signaling Peptides and Proteins , Melanoma, Experimental/pathology , Membrane Proteins/analysis , Mice , Procollagen-Proline Dioxygenase/analysis , Procollagen-Proline Dioxygenase/metabolism , Protein Serine-Threonine Kinases , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...