首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
2.
3.
4.
The regulation of metabolism and growth must be tightly coupled to guarantee the efficient use of energy and anabolic substrates throughout the cell cycle. Fructose 2,6-bisphosphate (Fru-2,6-BP) is an allosteric activator of 6-phosphofructo-1-kinase (PFK-1), a rate-limiting enzyme and essential control point in glycolysis. The concentration of Fru-2,6-BP in mammalian cells is set by four 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (PFKFB1–4), which interconvert fructose 6-phosphate and Fru-2,6-BP. The relative functions of the PFKFB3 and PFKFB4 enzymes are of particular interest because they are activated in human cancers and increased by mitogens and low oxygen. We examined the cellular localization of PFKFB3 and PFKFB4 and unexpectedly found that whereas PFKFB4 localized to the cytoplasm (i.e. the site of glycolysis), PFKFB3 localized to the nucleus. We then overexpressed PFKFB3 and observed no change in glucose metabolism but rather a marked increase in cell proliferation. These effects on proliferation were completely abrogated by mutating either the active site or nuclear localization residues of PFKFB3, demonstrating a requirement for nuclear delivery of Fru-2,6-BP. Using protein array analyses, we then found that ectopic expression of PFKFB3 increased the expression of several key cell cycle proteins, including cyclin-dependent kinase (Cdk)-1, Cdc25C, and cyclin D3 and decreased the expression of the cell cycle inhibitor p27, a universal inhibitor of Cdk-1 and the cell cycle. We also observed that the addition of Fru-2,6-BP to HeLa cell lysates increased the phosphorylation of the Cdk-specific Thr-187 site of p27. Taken together, these observations demonstrate an unexpected role for PFKFB3 in nuclear signaling and indicate that Fru-2,6-BP may couple the activation of glucose metabolism with cell proliferation.Neoplastic transformation and growth require a massive increase in glucose uptake and glycolytic flux not only for energy production but also for the synthesis of nucleic acids, amino acids, and fatty acids. A central control point of glycolysis is the negative allosteric regulation of a rate-limiting enzyme, phosphofructokinase-1 (PFK-1),2 by ATP (i.e. the Pasteur effect) (1, 2). When intracellular ATP production exceeds usage, ATP inhibits PFK-1 and glycolytic flux. Fructose 2,6-bisphosphate (Fru-2,6-BP) is a potent allosteric activator of PFK-1 that overrides this inhibitory influence of ATP on PFK-1, allowing forward flux of the entire pathway (35).The steady-state cellular concentration of Fru-2,6-BP is dependent on the activities of bifunctional 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (PFKFB), which are encoded by four independent genes (PFKFB1–4) (6, 7). The PFKFB3 mRNA is distinguished by the presence of multiple copies of an AUUUA instability motif in its 3′-untranslated region and the PFKFB3 protein product has a high kinase:phosphatase activity ratio (740:1) (8). PFKFB3 mRNA is overexpressed by rapidly proliferating transformed cells and the PFKFB3 protein is highly expressed in solid tumors and leukemias (811). PFKFB3 expression is increased in response to several mitogenic stimuli, including progesterone, serum, and insulin (1214). These studies indicate that the PFKFB3 enzyme may serve an essential function in the regulation of glucose metabolism during cell proliferation.The PFKFB3 mRNA is spliced into several variants that encode distinct carboxyl-terminal domains (9, 15). Importantly, the functional consequences of the disparate carboxyl-terminal variants of PFKFB3 are unknown. The mRNA splice variant 5 is the dominant PFKFB3 mRNA in human brain, several transformed cells, and colon adenocarcinoma tissues (9, 10). In the following series of experiments, we present data that the carboxyl-terminal domain of PFKFB3 variant 5 localizes the enzyme to the nucleus where its product, Fru-2,6-BP, increases the expression and activity of cyclin-dependent kinase-1. These data demonstrate a heretofore unidentified function of the PFKFB3 enzyme that is distinct from glycolysis, and provide a potential mechanism for the coupling of metabolism and proliferation.  相似文献   

5.
6.
Hypoxic response to low oxygen levels is characteristic of most solid cancers. Hypoxia-inducible factors (HIFs) regulate cellular metabolism, survival, proliferation, and cancer stem cell growth during hypoxia. The genome-wide analysis identified HAT1, a type B histone acetyltransferase, as an upregulated and essential gene in glioblastoma (GBM). GSEA analysis of differentially regulated genes in HAT1 silenced cells identified significant depletion of “hypoxia” gene sets. Hypoxia conditions induced HIF2A, not HIF1A protein levels in glioma cells in a HAT1-dependent manner. HAT1 and HIF2A interacted with each other and occupied the promoter of VEGFA, a bonafide HIF1A/HIF2A target. Acetylation of K512 and K596 residues by HAT1 is essential for HIF2A stabilization under normoxia and hypoxia as HIF2A carrying acetylation mimic mutations at either of these residues (H512Q or K596Q) showed stable expression in HAT1 silenced cells under normoxia and hypoxia conditions. Finally, we demonstrate that the HAT1-HIF2A axis is essential for hypoxia-promoted cancer stem cell maintenance and reprogramming. Thus, our study identifies that the HAT1-dependent acetylation of HIF2A is vital to executing the hypoxia-induced cell survival and cancer stem cell growth, therefore proposing the HAT1-HIF2A axis as a potential therapeutic target.  相似文献   

7.
The control of glucose metabolism and the cell cycle must be coordinated in order to guarantee sufficient ATP and anabolic substrates at distinct phases of the cell cycle. The family of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (PFKFB1-4) are well established regulators of glucose metabolism via their synthesis of fructose-2,6-bisphosphate (F2,6BP), a potent allosteric activator of 6-phosphofructo-1-kinase (Pfk-1). PFKFB3 is overexpressed in human cancers, regulated by HIF-1α, Akt and PTEN, and required for the survival and growth of multiple cancer types. Although most functional studies of the role of PFKFB3 in cancer progression have invoked its well-recognized function in the regulation of glycolysis, recent observations have established that PFKFB3 also traffics to the nucleus and that its product, F2,6BP, activates cyclin-dependent kinases (Cdks). In particular, F2,6BP stimulates the Cdk-mediated phosphorylation of the Cip/Kip protein p27 (threonine 187), which in turn results in p27''s ubiquitination and proteasomal degradation. As p27 is a potent suppressor of the G1/S transition and activator of apoptosis, we hypothesized that the known requirement of PFKFB3 for cell cycle progression and prevention of apoptosis may be partly due to the ability of F2,6BP to activate Cdks. In this study, we demonstrate that siRNA silencing of endogenous PFKFB3 inhibits Cdk1 activity, which in turn stabilizes p27 protein levels causing cell cycle arrest at G1/S and increased apoptosis in HeLa cells. Importantly, we demonstrate that the increase in apoptosis and suppression of the G1/S transition caused by siRNA silencing of PFKFB3 expression is reversed by co-siRNA silencing of p27. Taken together with prior publications, these observations support a model whereby PFKFB3 and F2,6BP function not only as regulators of Pfk-1 but also of Cdk1 activity, and therefore serve to couple glucose metabolism with cell proliferation and survival in transformed cells.The homodimeric bifunctional 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (PFKFB) phosphorylate fructose 6-phosphate (F6P) to fructose-2,6-bisphosphate (F2,6BP), which in turn activates 6-phosphofructo-1-kinase and glycolytic flux to lactate.1 Of the four genes encoding distinct PFKFB isozymes (PFKFB1-4), PFKFB3 is distinguished by the presence of multiple copies of the AUUUA instability motif in its 3''untranslated region,2 a very high kinase:phosphatase activity ratio (740 : 1),3 increased protein expression in rapidly proliferating transformed cells,2 solid tumors and leukemias2, 4, 5 and regulation by several proteins essential for tumor progression (e.g. HIF-1α,6 Akt7 and PTEN8, 9). Not surprisingly, heterozygous genomic deletion of the pfkfb3 gene has been found to reduce both the glucose metabolism and growth of Ras-transformed tumors in syngeneic mice.10In recent studies, we unexpectedly observed that PFKFB3 trafficked to the nucleus of multiple cell lines via a highly conserved nuclear localization motif in the C-terminal domain.11 Although the precise role of nuclear PFKFB3 is unknown, ectopic expression of wild-type PFKFB3 in the nucleus was found to stimulate cellular proliferation without affecting glycolysis, suggesting a novel role for nuclear F2,6BP in regulating the cell cycle.11 Moreover, the addition of F2,6BP to total cell lysates was found to increase the cyclin-dependent kinase (Cdk)-dependent phosphorylation of its substrate p27 at threonine 187 (T187), a posttranslational modification that targets p27 for degradation (i.e. high Cdk activity suppresses p27 levels).11 Given that p27 can potently block the G1/S transition and stimulate apoptosis, these data indicated that PFKFB3-mediated production of F2,6BP in the nucleus may directly stimulate Cdks to phosphorylate T187-p27, targeting p27 for degradation by the proteasome and allowing cells to both proliferate and evade apoptosis. Furthermore, these data signified that PFKFB3 may not only be essential for the regulation of glycolysis in the cytoplasm but also for the control of the cell cycle in the nucleus.Based on these prior studies, we postulated that selective inhibition of PFKFB3 would suppress Cdk1 activity, which in turn would reduce the phosphorylation of T187-p27, resulting in increased p27 expression, reduced G1/S transition and increased apoptosis. We provide evidence to support this chain of biochemical and cellular events after PFKFB3 inhibition as well as direct verification that p27 itself is required for the simultaneous suppression of G1/S transition and induction of apoptosis caused by PFKFB3 inhibition. Given that PFKFB3 inhibitors are entering phase I trials for the treatment of advanced cancers,12 we believe that this new mechanism of action may facilitate the development of rational phase I/II trials that combine other apoptosis-activating agents that disrupt p27 function (e.g. Cdk1 inhibitors) as well as potential biomarkers such as p27 that may demonstrate the on-target effects of PFKFB3 inhibitors in biopsies and resected tumors. From a broader perspective, these data provide further support for the concept that PFKFB3 may be an essential coupler of glucose metabolism and cell cycle progression.  相似文献   

8.
9.
10.
11.
Hypoxic environment in solid tumor is known to favor cell survival and to initiate the formation of new capillaries. In this work, we identified by 2D gel analysis 94-kDa glucose-regulated protein (GRP94) as being upregulated in human endothelial cells in response to hypoxia. Three putative hypoxia responsive elements (HRE) were found in the GRP94 promoter. Competition experiments of HIF-1 DNA binding using specific probes containing each HRE sequence of the GRP94 promoter clearly evidenced that HIF-1 binds these sequences with high affinity. The human GRP94 promoter was then cloned upstream of the luciferase gene and showed enhanced activity in hypoxic conditions. Mutation of two of the three HREs present in this promoter completely inhibited the hypoxia-induced increase in luciferase activity.  相似文献   

12.
13.
The type II transmembrane serine protease matriptase-2 (TMPRSS6) down-regulates the expression of hepcidin, the main regulator of systemic iron homeostasis, and increases in this way iron plasma levels. Matriptase-2 is up-regulated under hypoxic conditions, providing a new link between hypoxia signaling and iron homeostasis. In this study, we have characterized the TMPRSS6 promoter region and identified a functional hypoxia-responsive element (HRE). Mutations of the hypoxia inducible factor (HIF)-binding site located within the HRE abrogate HIF-1α-dependent induction of TMPRSS6 expression. The action of HIF-1α on TMPRSS6 promoter activity reveals a new regulative element for the suppression of hepcidin synthesis.  相似文献   

14.
15.
16.
17.
18.
19.
A DNA fragment containing 1.5 kb of the 5'-flanking region of the human ubiquitous PFKFB3 gene, coding for 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase, was cloned and its promoter activity was examined. The 5' flanking region contains a TATA box-like and GC-rich sequences, yielding several potential Specific protein (Sp-1) and activator protein (AP)-2 binding sites. Putative regulatory motifs for E-box, nuclear factor (NF)-1 and progesterone response element were also found by computer assisted analysis. Transient expression assays of truncated promoter-reporter constructs in HeLa cells showed that this gene is induced by phorbol esters (PDB) and cyclic-AMP-dependent protein kinase signal activation. Furthermore, the genomic organization of the PFKFB3 gene is reported. This gene spans more than 26 kb containing at least 16 exons that accounts for the two reported isoforms, inducible and ubiquitous, generated through alternative splicing of exon 15.  相似文献   

20.
Wu DY  Yao Z 《Cell research》2006,16(3):319-322
  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号