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 31 毫秒
1.
The MN1 oncogene is deregulated in human acute myeloid leukemia and its overexpression induces proliferation and represses myeloid differentiation of primitive human and mouse hematopoietic cells, leading to myeloid leukemia in mouse models. To delineate the sequences within MN1 necessary for MN1-induced leukemia, we tested the transforming capacity of in-frame deletion mutants, using retroviral transduction of mouse bone marrow. We found that integrity of the regions between amino acids 12 to 458 and 1119 to 1273 are required for MN1’s in vivo transforming activity, generating myeloid leukemia with some mutants also producing T-cell lympho-leukemia and megakaryocytic leukemia. Although both full length MN1 and a mutant that lacks the residues between 12–228 (Δ12–228 mutant) repressed myeloid differentiation and increased myeloproliferative activity in vitro, the mutant lost its transforming activity in vivo. Both MN1 and Δ12–228 increased the frequency of common myeloid progentiors (CMP) in vitro and microarray comparisons of purified MN1-CMP and Δ12–228-CMP cells showed many differentially expressed genes including Hoxa9, Meis1, Myb, Runx2, Cebpa, Cebpb and Cebpd. This collection of immediate MN1-responsive candidate genes distinguishes the leukemic activity from the in vitro myeloproliferative capacity of this oncoprotein.  相似文献   

2.
Over the last decade, comprehensive genome-wide sequencing studies have enabled us to find out unexpected genetic alterations of metabolism in cancer. An example is the identification of arginine missense mutations of isocitrate dehydrogenases-1 and -2 (IDH1/2) in glioma, acute myeloid leukemia (AML), chondrosarcomas, and cholangiocarcinoma. These alterations are closely associated with the production of a new stereospecific metabolite, (R)-2-hydroxyglutarate (R-2HG). A large number of follow-up studies have been performed to address the molecular mechanisms of IDH1/2 mutations underlying how these events contribute to malignant transformation. In the meanwhile, the development of selective mutant IDH1/2 chemical inhibitors is being actively pursued in the scientific community and pharmaceutical industry. The present review article briefly discusses the important findings that highlight the molecular mechanisms of IDH1/2 mutations in cancer and provides a current status for development of selective mutant IDH1/2 chemical inhibitors. [BMB Reports 2015; 48(5): 266-270]  相似文献   

3.
Mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) are key events in the development of glioma, acute myeloid leukemia (AML), chondrosarcoma, intrahepatic cholangiocarcinoma (ICC), and angioimmunoblastic T-cell lymphoma. They also cause D-2-hydroxyglutaric aciduria and Ollier and Maffucci syndromes. IDH1/2 mutations are associated with prolonged survival in glioma and in ICC, but not in AML. The reason for this is unknown. In their wild-type forms, IDH1 and IDH2 convert isocitrate and NADP+ to α-ketoglutarate (αKG) and NADPH. Missense mutations in the active sites of these enzymes induce a neo-enzymatic reaction wherein NADPH reduces αKG to D-2-hydroxyglutarate (D-2HG). The resulting D-2HG accumulation leads to hypoxia-inducible factor 1α degradation, and changes in epigenetics and extracellular matrix homeostasis. Such mutations also imply less NADPH production capacity. Each of these effects could play a role in cancer formation. Here, we provide an overview of the literature and discuss which downstream molecular effects are likely to be the drivers of the oncogenic and survival-prolonging properties of IDH1/2 mutations. We discuss interactions between mutant IDH1/2 inhibitors and conventional therapies. Understanding of the biochemical consequences of IDH1/2 mutations in oncogenesis and survival prolongation will yield valuable information for rational therapy design: it will tell us which oncogenic processes should be blocked and which “survivalogenic” effects should be retained.  相似文献   

4.
5.
Isocitrate dehydrogenase (IDH) is one of the key enzymes in the tricarboxylic acid cycle, and IDH mutations have been associated with many cancers, including glioblastoma, sarcoma, acute myeloid leukemia, etc. Three natural steroids 13 from Ganoderma sinense, a unique and rare edible-medicinal fungi in China, were found as potential IDH1 inhibitors by virtual ligand screening method. Among the three compounds, 3 showed the highest binding affinity to IDH1 with significant calculated binding free energy. Enzymatic kinetics demonstrated that 3 inhibited mutant enzyme in a noncompetitive manner. The half effective concentration of 3 for reducing the concentration of D-2HG in HT1080 cells was 35.97 μM. The levels of histone H3K9me3 methylation in HT1080 cells were reduced by treating with 3. Furthermore, knockdown of mutant IDH1 in HT1080 cells decreased the anti-proliferative sensitivity to 3. In short, our findings highlight that compound 3 may have clinical potential in tumor therapies as an effective inhibitor of mutant IDH1.  相似文献   

6.
Tumor-associated mutations in the isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) genes result in the loss of normal catalytic activity, the production of α-ketoglutarate (α-KG), and gain of a new activity, the production of an oncometabolite, R-2-hydroxylglutarate (R-2-HG). New evidence supports previous findings that R-2-HG acts as an antagonist of α-KG to competitively inhibit the activity of multiple α-KG-dependent dioxygenases, including both histones and DNA demethylases involved in epigenetic control of gene expression and cell differentiation, and also reveals an intriguing new facet of R-2-HG in tumorigenesis.The NADP+-dependent isocitrate dehydrogenase IDH1 and IDH2 catalyze the oxidative decarboxylation of isocitrate to α-ketoglutarate (α-KG). IDH1 and IDH2 are localized in the cytoplasm and mitochondria, respectively, and represent by far the most frequently mutated metabolic enzymes in human cancer1. The tumor-derived mutants of both IDH1 and IDH2 lose their activity in producing α-KG2,3, and gain a surprising new catalytic activity, the production of R-2-hydroxyglutarate (R-2-HG) by reduction of α-KG4. Previous studies have shown that R-2-HG acts as an antagonist of α-KG to competitively inhibit a number of α-KG-dependent dioxygenases, including the JmjC domain-containing histone demethylases (KDMs) and the TET (ten-eleven translocation) family of DNA hydroxylases that catalyze the sequential oxidation of 5-methlycytosine (5mC) to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC), leading to eventual DNA demethylation (Figure 1)5,6. Three papers recently published in Nature provide additional evidence that α-KG-dependent dioxygenases are the pathophysiological targets of mutant IDH1/2, and further underscore the presumptive role of R-2-HG as the first oncometabolite in contributing to tumorigenesis after IDH1/2 mutations.Open in a separate windowFigure 1Summarization of reported mechanisms linking IDH mutation to tumorigensis. Regulation of α-KG-dependent dioxygenases by R-2-HG is likely to play a major role in the pathophysiology of tumors with IDH mutation.A subset of glioblastoma, known as the proneural subgroup, has previously found to display hypermethylation at a large number of loci and is enriched with IDH1 mutations7. In one of the three Nature papers, Turcan et al.8 determined whether IDH1 mutation alone is sufficient to cause the hypermethylation phenotype by ectopic expression of IDH1R132H mutant in immortalized primary human astrocytes, a cell type from which glioblastoma is believed to develop. The authors found that introduction of mutant IDH1 induced extensive DNA hypermethylation, altered the methylation of specific histones, and reshaped the methylome in a fashion that mirrors the changes observed in IDH1-mutated low-grade gliomas. The observed hypermethylation of DNA and histones can be explained by the direct inhibition of TET methylcytosine hydroxylases and JmjC family histone demethylases by R-2-HG, respectively. In keeping with the notion that TET hydroxylases directly regulate genomic DNA methylation levels and can be inhibited by the R-2-HG accumulated in IDH1/2-mutated cells, Turcan et al. also showed that ectopic expression of TET2 in cultured astrocytes decreased 5mC and increased 5hmC, and that both changes were inhibited by the co-expression of TET2 with mutant IDH1. These results are consistent with the findings made in acute myeloid leukemia (AML) in which IDH1/2 and TET2 genes are mutated in a mutually exclusive manner9. Moreover, Turcan et al. found that expression of wild-type IDH1 decreased the average DNA methylation level in the genome, supporting the notion that the concentration of α-KG may be a rate-limiting factor of TET-catalyzed DNA demethylation5.In the second paper, Lu et al.10 reported that ectopic expression of tumor-derived mutant IDH1/2 or feeding cells with cell-permeable R-2-HG increases histone demethylation and results in blockade of the differentiation of 3T3-L1 adipoblasts to adipocytes. These results indicate that mutation of IDH1/2 and accumulation of R-2-HG can broadly impair cell differentiation beyond the cell types in which IDH1/2 mutations are found to associate with tumorigenesis. The authors further confirmed that IDH1-mutated gliomas have elevated levels of histone methylation compared with gliomas retaining the wild-type IDH15,6. As previously reported5,6, multiple KDMs that are inhibited by 2-HG, including KDM4C/JMJD2C, which causes repressive histone H3K9 di- and trimethylation and, when suppressed by RNA interference, blocks the 3T3-L1 adipogenesis. It remains to be determined whether collective inhibition of multiple KDMs or a few individual ones, such as KDM4C, is responsible for altering cell differentiation in IDH1/2-mutated cells. The authors also noted that expression of mutant IDH1 increased histone methylation prior to the increase of DNA methylation, raising an intriguing possibility that histone methylation status may affect DNA methylation.In the third paper, Koivunen et al.11 proposed an enantiomer-specific mechanism of 2-HG in tumorigenesis. The authors reported two surprising findings. They showed first that immortalized human astrocytes stably expressing tumor-derived IDH1R132H mutant proliferate faster during late passages than those expressing either wild-type IDH1 or IDH1R132H/3DN mutant that lacks 2-HG-producing activity. Ectopic expression of R132H mutant IDH1 has previously been reported to decrease the growth of D54 glioblastoma cells12, raising an intriguing possibility that the mutation of IDH1/2 may exhibit different effects on cell growth in a cell context-dependent manner. More surprisingly, they found that R-2-HG, but not its enantiomer S-2-HG, substitutes for α-KG as a co-substrate, as opposed to an inhibitor, of EGLN, an α-KG-dependent prolylhydroxylase responsible for promoting the degradation of hypoxia inducible factor 1α (HIF-1α) (Figure 1). As the result of stimulating EGLN, accumulation of R-2-HG was found to associate with diminished, instead of increased, HIF-1α levels in cells expressing mutant IDH1/2. At first glance, these observations appear to be at odds with the generally accepted role of both enantiomers of 2-HG as inhibitors of α-KG-dependent dioxygenases, and HIF-1α as an oncogene in tumorigenesis, but may at least in part explain the apparent selection for IDH mutations to produce R-, but not S-2-HG in cancer. This data, also for the first time, reveals a qualitatively different property of two 2-HG enantiomers with respect to α-KG-dependent dioxygenases. It will be interesting to determine the strutural basis of this enantimoer-specific effect of 2-HG toward different α-KG-dependent dixoygenases. The observation that ectopic increase of R-2-HG reduces HIF-1α suggests that endogenous α-KG is limiting for HIF-1α hydroxylation by EGLN. The study by Koivunen et al. also suggests the complexity of EGLN regulation by R-2-HG and subsequent downregulation of HIF-1α. It remains to be determined genetically whether a reduction or fluctuation of HIF-1α levels contributes to gliomagenesis in IDH1/2-mutated cells, because elevated HIF-1α generally contributes to cancer development. The only piece of genetic evidece—IDH1/2 mutation occurs in a mutually exclusive manner with TET2 mutation in AML—supports the notion that epigenetic alteration plays a direct and perhaps a key role in IDH1/2 mutation-associated tumorigenesis.IDH1/2 mutation has rapidly emerged as a favorable diagnostic and prognostic marker for certain tumors, such as low-grade gliomas and benign cartilaginous tumors. While the full mechanism linking IDH mutation to tumorigenesis is incompletely understood, regulation of α-KG-dependent dioxygenases by 2-HG is likely to play a major role in the pathophysiology of tumors with IDH mutation. These recent reports also highlight the impact of altered metabolism and metabolites on the epigenetic modification of cell differentiation and tumorigenesis.  相似文献   

7.
As one of the best known cancer testis antigens, PRAME is overexpressed exclusively in germ line tissues such as the testis as well as in a variety of solid and hematological malignant cells including acute myeloid leukemia. Therefore, PRAME has been recognized as a promising target for both active and adoptive anti-leukemia immunotherapy. However, in most patients with PRAME-expressing acute myeloid leukemia, PRAME antigen-specific CD8+ CTL response are either undetectable or too weak to exert immune surveillance presumably due to the inadequate PRAME antigen expression and PRAME-specific antigen presentation by leukemia cells. In this study, we observed remarkably increased PRAME mRNA expression in human acute myeloid leukemia cell lines and primary acute myeloid leukemia cells after treatment with a novel subtype-selective histone deacetylase inhibitor chidamide in vitro. PRAME expression was further enhanced in acute myeloid leukemia cell lines after combined treatment with chidamide and DNA demethylating agent decitabine. Pre-treatment of an HLA-A0201+ acute myeloid leukemia cell line THP-1 with chidamide and/or decitabine increased sensitivity to purified CTLs that recognize PRAME100–108 or PRAME300–309 peptide presented by HLA-A0201. Chidamide-induced epigenetic upregulation of CD86 also contributed to increased cytotoxicity of PRAME antigen-specific CTLs. Our data thus provide a new line of evidence that epigenetic upregulation of cancer testis antigens by a subtype-selective HDAC inhibitor or in combination with hypomethylating agent increases CTL cytotoxicity and may represent a new opportunity in future design of treatment strategy targeting specifically PRAME-expressing acute myeloid leukemia.  相似文献   

8.
Disturbance of the dynamic balance between tyrosine phosphorylation and dephosphorylation of signaling molecules, controlled by protein tyrosine kinases and protein tyrosine phosphatases (PTPs), is known to lead to the development of cancer. While most approved targeted cancer therapies are tyrosine kinase inhibitors, PTPs have long been stigmatized as undruggable and have only recently gained renewed attention in drug discovery. One PTP target is the Src-homology 2 domain–containing phosphatase 2 (SHP2). SHP2 is implicated in tumor initiation, progression, metastasis, and treatment resistance, primarily because of its role as a signaling nexus of the extracellular signal–regulated kinase pathway, acting upstream of the small GTPase Ras. Efforts to develop small molecules that target SHP2 are ongoing, and several SHP2 allosteric inhibitors are currently in clinical trials for the treatment of solid tumors. However, while the reported allosteric inhibitors are highly effective against cells expressing WT SHP2, none have significant activity against the most frequent oncogenic SHP2 variants that drive leukemogenesis in several juvenile and acute leukemias. Here, we report the discovery of novel furanylbenzamide molecules as inhibitors of both WT and oncogenic SHP2. Importantly, these inhibitors readily cross cell membranes, bind and inhibit SHP2 under physiological conditions, and effectively decrease the growth of cancer cells, including triple-negative breast cancer cells, acute myeloid leukemia cells expressing either WT or oncogenic SHP2, and patient-derived acute myeloid leukemia cells. These novel compounds are effective chemical probes of active SHP2 and may serve as starting points for therapeutics targeting WT or mutant SHP2 in cancer.  相似文献   

9.
10.
Retinoids and 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) induce differentiation of myeloid leukemia cells into granulocyte and macrophage lineages, respectively. All-trans retinoic acid (ATRA), which is effective in the treatment of acute promyelocytic leukemia, can induce differentiation of other types of myeloid leukemia cells, and combined treatment with retinoid and 1,25(OH)2D3 effectively enhances the differentiation of leukemia cells into macrophage-like cells. Recent work has classified macrophages into M1 and M2 types. In this study, we investigated the effect of combined treatment with retinoid and 1,25(OH)2D3 on differentiation of myeloid leukemia THP-1 and HL60 cells. 9-cis Retinoic acid (9cRA) plus 1,25(OH)2D3 inhibited proliferation of THP-1 and HL60 cells and increased myeloid differentiation markers including nitroblue tetrazolium reducing activity and expression of CD14 and CD11b. ATRA and the synthetic retinoic acid receptor agonist Am80 exhibited similar effects in combination with 1,25(OH)2D3 but less effectively than 9cRA, while the retinoid X receptor agonist HX630 was not effective. 9cRA plus 1,25(OH)2D3 effectively increased expression of M2 macrophage marker genes, such as CD163, ARG1 and IL10, increased surface CD163 expression, and induced interleukin-10 secretion in myeloid leukemia cells, while 9cRA alone had weaker effects on these phenotypes and 1,25(OH)2D3 was not effective. Taken together, our results demonstrate selective induction of M2 macrophage markers in human myeloid leukemia cells by combined treatment with 9cRA and 1,25(OH)2D3.  相似文献   

11.

Background

Although Imatinib mesylate has revolutionized the treatment of chronic myeloid leukemia, some patients develop resistance with progression of leukemia. Alternative or additional targeting of signalling pathways deregulated in Bcr-Abl-driven chronic myeloid leukemia may provide a feasible option for improving clinical response and overcoming resistance.

Results

In this study, we investigate ability of CR8 isomers (R-CR8 and S-CR8) and MR4, three derivatives of the cyclin-dependent kinases (CDKs) inhibitor Roscovitine, to exert anti-leukemic activities against chronic myeloid leukemia in vitro and then, we decipher their mechanisms of action. We show that these CDKs inhibitors are potent inducers of growth arrest and apoptosis of both Imatinib-sensitive and –resistant chronic myeloid leukemia cell lines. CR8 and MR4 induce dose-dependent apoptosis through mitochondrial pathway and further caspases 8/10 and 9 activation via down-regulation of short-lived survival and anti-apoptotic factors Mcl-1, XIAP and survivin which are strongly implicated in survival of Bcr-Abl transformed cells.

Conclusions

These results suggest that CDK inhibitors may constitute a complementary approach to treat chronic myeloid leukemia.

Electronic supplementary material

The online version of this article (doi:10.1186/s12929-015-0163-x) contains supplementary material, which is available to authorized users.  相似文献   

12.

Background

The adult myeloid diseases, myelodysplastic syndrome and acute myeloid leukemia, have been reported to be associated with cigarette smoking, but the results have been conflicting. Previous studies may have ignored the relationship between myelodysplastic syndrome and acute myeloid leukemia, where approximately one-third of myelodysplastic syndrome cases will progress to acute myeloid leukemia, which could induce a serious bias in independent analyses. For the purposes of researching pathogenesis, we suggest that myelodysplastic syndrome and acute myeloid leukemia should be regarded as a single class of adult myeloid disease, and herein assessed the relationship between cigarette smoking and the risk of adult myeloid disease.

Methods

The PubMed, Cochrane Library, EBSCO, and EMBASE databases were systematically searched for reports published from 1990 to 2015. Two authors independently assessed the methodological quality and the extracted data. The odds ratios and adjusted odds ratios (OR), a sensitivity analysis, and the publication bias were analyzed using the CMA v2 (Comprehensive Meta Analysis Version 2) software program.

Results

Twenty-five studies were included in this meta-analysis. The publication dates ranged from 1990 to 2014. The pooled OR in current smokers and ever-smokers showed an increased risk of adult myeloid disease, with ORs of 1.45 (95% CI, 1.30–1.62; p<0.001) and 1.23 (95% CI 1.15–1.32; p<0.001) versus non-smokers, respectively. In the subset analyses, the OR of adult myeloid disease was increased regardless of the form of disease, geographical region, NOS (Newcastle Ottawa Scale) score, and source of controls. The smoking status was divided into <20 and ≥20 cigarettes per day, and these groups had ORs of developing adult myeloid disease of 1.24 (95% CI, 1.09–1.40; p = 0.001) and 1.32 (95% CI, 1.14–1.53; p<0.001), respectively. In the groups divided based on the number of years the subjects had smoked (<20 and ≥20 years), the ORs were 1.05 (95% CI, 0.90–1.23; p = 0.25) and 1.30 (95% CI, 1.16–1.45; p<0.001), respectively. Similarly, <20 and ≥20 pack-years were associated with ORs of 1.15 (95% CI, 1.03–1.29; p = 0.017) and 1.34 (95% CI, 1.18–1.52; p<0.001), respectively.

Conclusions

This meta-analysis, for the first time, combined myelodysplastic syndrome with acute myeloid leukemia to assess the overall risk of adult myeloid disease, and it demonstrated that cigarette smoking is associated with a significantly increased risk of adult myeloid disease.  相似文献   

13.
Extensive molecular profiling of leukemias and preleukemic diseases has revealed that distinct clinical entities, like acute myeloid (AML) and T-lymphoblastic leukemia (T-ALL), share similar pathogenetic mutations. It is not well understood how the cell of origin, accompanying mutations, extracellular signals or structural differences in a mutated gene determine the phenotypic identity of leukemias. We dissected the functional aspects of different protein regions of the MN1 oncogene and their effect on the leukemic phenotype, building on the ability of MN1 to induce leukemia without accompanying mutations. We found that the most C-terminal region of MN1 was required to block myeloid differentiation at an early stage, and deletion of an extended C-terminal region resulted in loss of myeloid identity and cell differentiation along the T-cell lineage in vivo. Megakaryocytic/erythroid lineage differentiation was blocked by the N-terminal region. In addition, the N-terminus was required for proliferation and leukemogenesis in vitro and in vivo through upregulation of HoxA9, HoxA10 and Meis2. Our results provide evidence that a single oncogene can modulate cellular identity of leukemic cells based on its active gene regions. It is therefore likely that different mutations in the same oncogene may impact cell fate decisions and phenotypic appearance of malignant diseases.  相似文献   

14.
Oncoprotein E2a/Pbx1 is produced by the t(1;19) chromosomal translocation of human pre-B acute lymphoblastic leukemia. E2a/Pbx1 blocks differentiation of primary myeloid progenitors but, paradoxically, induces apoptosis in established pre-B-cell lines, and no transforming function of E2a/Pbx1 has been reported in cultured lymphoid progenitors. Here, we demonstrate that E2a/Pbx1 induces immortal proliferation of stem cell factor (SCF)-dependent pro-T thymocytes by a mechanism dependent upon both its transactivation and DNA-binding functions. E2a-Pbx1 cooperated with cytokines or activated signaling oncoproteins to induce cell division, as inactivation of conditional E2a/Pbx1 in either factor-dependent pro-T cells or pro-T cells made factor independent by expression of Bcr/Abl resulted in pro-T-cell quiescence, while reactivation of E2a/Pbx1 restored cell division. Infusion of E2a/Pbx1 pro-T cells in mice caused T lymphoblastic leukemia and, unexpectedly, acute myeloid leukemia. The acute lymphoblastic leukemia did not evidence further maturation, suggesting that E2a/Pbx1 establishes an early block in pro-T-cell development that cannot be overcome by marrow or thymic microenvironments. In an E2a/Pbx1 pro-T thymocyte clone that induced only pro-T acute lymphoblastic leukemia, coexpression of Bcr/Abl expanded its leukemic phenotype to include acute myeloid leukemia, suggesting that unique functions of cooperating signaling oncoproteins can influence the lymphoid versus myeloid character of E2a/Pbx1 leukemia and may cooperate with E2a/Pbx1 to dictate the pre-B-cell phenotype of human leukemia containing t(1;19).  相似文献   

15.
Autophagy is an intracellular bulk degradation process involved in cell survival upon stress induction, but also with a newly identified function in myeloid differentiation. The autophagy-related (ATG)8 protein family, including the GABARAP and LC3 subfamilies, is crucial for autophagosome biogenesis. In order to evaluate the significance of the GABARAPs in the pathogenesis of acute myeloid leukemia (AML), we compared their expression in primary AML patient samples, CD34+ progenitor cells and in granulocytes from healthy donors. GABARAPL1 and GABARAPL2/GATE-16, but not GABARAP, were significantly downregulated in particular AML subtypes compared to normal granulocytes. Moreover, the expression of GABARAPL1 and GATE-16 was significantly induced during ATRA-induced neutrophil differentiation of acute promyelocytic leukemia cells (APL). Lastly, knocking down GABARAPL2/GATE-16 in APL cells attenuatedneutrophil differentiation and decreased autophagic flux. In conclusion, low GABARAPL2/GATE-16 expression is associated with an immature myeloid leukemic phenotype and these proteins are necessary for neutrophil differentiation of APL cells.  相似文献   

16.
Somatic mutations in isocitrate dehydrogenase (IDH)-1 and -2 have recently been described in glioma. This mutation leads to a neomorphic enzymatic activity as the conversion of isocitrate to alpha ketoglutarate (αKG) is replaced by the conversion of αKG to D-2-hydroxyglutarate (D-2HG) with NADPH oxidation. It has been suggested that this oncometabolite D-2HG via inhibition of αKG-dioxygenases is involved in multiple functions such as epigenetic modifications or hypoxia responses. The present study is aimed at deciphering how the mutant IDH can affect cancer pathogenesis, in particular with respect to its associated oncometabolite D-2HG. We show that the overexpression of mutant IDH in glioma cells or treatment with D-2HG triggered an increase in cell proliferation. However, although mutant IDH reduced cell sensitivity to the apoptotic inducer etoposide, D-2HG exhibited no effect on apoptosis. Instead, we found that the apoptotic effect was mediated through the mitochondrial NADH pool reduction and could be inhibited by oxamate. These data show that besides D-2HG production, mutant IDH affects other crucial metabolite pools. These observations lead to a better understanding of the biology of IDH mutations in gliomas and their response to therapy.Gliomas are the most common type of human brain tumors and can be classified based on clinical and pathological criteria in four grades. The grade IV glioma, commonly known as glioblastoma multiforme (GBM), is the most invasive form and has a dismal prognosis with <5% patient survival at 5 years. These GBM can develop de novo (primary GBM) or through the progression from low-grade tumors (secondary GBM). Although these two types of GBM are histologically similar, primary and secondary GBM exhibit distinct genetic patterns. A recent integrated genome analysis of human GBM shows that 12% of these tumors have a mutation in the gene encoding isocitrate dehydrogenase 1 (IDH1) and to a lesser extent in IDH2 gene.1 This mutation is present in >90% secondary GBMs, whereas it is present in <5% primary GBMs.2 Mutations in IDH1 and IDH2 have also been identified in acute myeloid leukemia (AML)3 and chondrosarcomas.4 The occurrence of IDH mutations predicts a significantly longer survival for patients affected by GBM or grade III gliomas.1, 2 Whether this difference is driven by IDH mutations or reflects other fundamental biological differences between primary and secondary GBM is, as yet, unclear. For example, the prognostic significance of IDH mutations may be secondary to their prevalence among younger patients, as age is a well-known prognostic factor in gliomas.5 In AML, the prognostic significance of IDH mutations is more ambiguous. Several studies have reported that IDH mutations do not affect the prognosis in AML, whereas other studies have found that IDH mutations are associated with an increased or decreased risk of relapse when compared with IDH wild-type patients.6, 7The human genome has five IDH genes coding for three different IDH isoforms, the activities of which depend on either nicotinamide adenine dinucleotide (NAD+) for IDH3 or nicotinamide adenine dinucleotide phosphate (NADP+) for IDH1 and IDH2. Both IDH2 and IDH3 are located in the mitochondria where they participate in the TCA cycle, whereas IDH1 is mostly cytosolic.8 To date, all reported mutations are located in the IDH1 and IDH2 genes and result in an amino-acid substitution at residues located in the enzymatic active site, respectively, R132 for IDH1 and R140 or R172 for IDH2. This mutation disrupts the normal enzymatic function of IDH, that is, the conversion of isocitrate to alpha ketoglutarate (αKG) with the concomitant production of NADPH. Instead, mutant IDH displays a neomorphic activity converting αKG into D-2-hydroxyglutarate (D-2HG), although reducing NADPH.9 As a result, mutant IDH may alter the redox state of cells, modulate the activity of metabolic and epigenetic tumor suppressor enzymes that use αKG as a co-substrate.10 Loss of IDH function may also alter normal mitochondrial function and promote a metabolic switch in cancer cells to glycolysis.11, 12Mutant IDH is widely believed to have the ability to transform cells by modulating αKG-dependent enzymes. D-2HG and αKG are structurally similar suggesting that D-2HG may act as a competitive inhibitor of αKG-dioxygenases including prolyl hydroxylase involved in HIF-1α stability, histone demethylases and the Ten-Eleven Translocation (TET) family of 5-methylcytosine hydroxylases involved in epigenetic modifications of DNA.13, 14 In fact, IDH mutations lead to numerous metabolic abnormalities besides D-2HG production. Deciphering the relative importance of either D-2HG production, αKG or NADPH reduction in cancer pathogenesis remains to be determined. In this paper, we show that mutant IDH increases cell proliferation and reduces etoposide (ETO)-induced cell death through different metabolic pathways. Although cell proliferation changes are mediated through D-2HG, alteration in the mitochondrial NADH pool is involved in the response to apoptosis.  相似文献   

17.
The human CAN gene was first identified as a target of t(6;9)(p23;q34), associated with acute myeloid leukemia and myelodysplastic syndrome, which results in the expression of a DEK-CAN fusion gene. CAN, also called NUP214, is a nuclear pore complex (NPC) protein that contains multiple FG-peptide sequence motifs. It interacts at the NPC with at least two other proteins, the nucleoporin NUP88 and hCRM1 (exportin 1), which was recently shown to function as a nuclear export receptor. Depletion of CAN in knockout mouse embryonic cells results in cell cycle arrest in G2, followed by inhibition of nuclear protein import and a block of mRNA export. We overexpressed CAN and DEK-CAN in U937 myeloid precursor cells. DEK-CAN expression did not interfere with terminal myeloid differentiation of U937 cells, whereas CAN-overexpressing cells arrested in G0, accumulated mRNA in their nuclei, and died in an apoptotic manner. Interestingly, we found that hCRM1 and import factor p97/importin β colocalized with the ectopically expressed CAN protein, resulting in depletion of both factors from the NPC. Overexpression of the C-terminal FG-repeat region of CAN, which contains the binding site for hCRM1, caused sequestering of hCRM1 in the nucleoplasm and was sufficient to inhibit cell growth and to induce apoptosis. These results confirm that CAN plays a crucial role in nucleocytoplasmic transport and imply an essential role for hCRM1 in cell growth and survival.  相似文献   

18.
A series of 3-aryl-4-indolylmaleimide IDH1/R132H inhibitors with a novel structure was obtained by high-throughput screening and structure-based optimization. Most compounds such as 7a, 7d, 7h, 7i, 7k and 7o showed high inhibitory effects on IDH1/R132H and were highly selective against IDH1/WT, IDH2/WT, GDH, GK, and FBP. Evaluation of the biological activities and function at cellular level showed that compounds 7h, 7i and 7k could effectively suppress the production of 2-hydroxyglutaric acid in U87MG cells expressing IDH1/R132H. Additionally, 7h could reversed the differentiation block of the myeloid leukemic cell line, TF-1, caused by the overexpression of IDH1/R132H. We also explore the structure-activity relationship based on the experimental data, with an attempt to pave the way for future studies.  相似文献   

19.
The receptor-interacting protein kinase 3 (RIP3) associates with RIP1 in a necrosome complex that can induce necroptosis, apoptosis, or cell proliferation. We analyzed the expression of RIP1 and RIP3 in CD34+ leukemia cells from a cohort of patients with acute myeloid leukemia (AML) and CD34+ cells from healthy donors. RIP3 expression was significantly reduced in most AML samples, whereas the expression of RIP1 did not differ significantly. When re-expressed in the mouse DA1-3b leukemia cell line, RIP3 induced apoptosis and necroptosis in the presence of caspase inhibitors. Transfection of RIP3 in the WEHI-3b leukemia cell line or in the mouse embryonic fibroblasts also resulted in increased cell death. Surprisingly, re-expression of a RIP3 mutant with an inactive kinase domain (RIP3-kinase dead (RIP3-KD)) induced significantly more and earlier apoptosis than wild-type RIP3 (RIP3-WT), indicating that the RIP3 kinase domain is an essential regulator of apoptosis/necroptosis in leukemia cells. The induced in vivo expression of RIP3-KD but not RIP3-WT prolonged the survival of mice injected with leukemia cells. The expression of RIP3-KD induced p65/RelA nuclear factor-κB (NF-κB) subunit caspase-dependent cleavage, and a non-cleavable p65/RelA D361E mutant rescued these cells from apoptosis. p65/RelA cleavage appears to be at least partially mediated by caspase-6. These data indicate that RIP3 silencing in leukemia cells results in suppression of the complex regulation of the apoptosis/necroptosis switch and NF-κB activity.Impairment in cell death pathways represents a general characteristic of most cancer cells. Cells can die through several mechanisms; two such cell death pathways include apoptosis and necrosis, which display distinct characteristics.1 Necrosis can occur in either an incidental or intentional manner as a result of defined signals, and the term necroptosis has been proposed to describe this programmed necrosis.2 Activation of the receptor-interacting protein kinase 1 (RIP1) and 3 (RIP3) proteins in the necrosome complex can induce apoptosis, necroptosis, or cell proliferation after the activation of death receptors, including TNFR1, TRAIL, and FAS.3, 4 RIP1 and RIP3 are serine threonine kinases with strong homology.5 Both proteins are composed of a kinase domain at the N-terminus and a RIP homotypic interaction motif (RHIM) at the C-terminus of RIP3. The RIP1/RIP3 complex can induce necroptosis initiated by cell death receptors of the tumor necrosis factor family. RIP3 binds to RIP1 via their respective RHIM domains, and these proteins form a filamentous structure with characteristics similar to β-amyloids and can cross phosphorylate each other and several downstream targets involved in necroptosis, apoptosis, or nuclear factor-κB (NF-κB) activation.6The role of RIP3 in necroptosis and inflammation has been extensively studied, but its role in cancer remains poorly understood. A previous study in chronic lymphocytic leukemia (CLL) showed that malignant lymphoid cells were resistant to tumor necrosis factor-α (TNFα+Z-VAD-induced (carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone) necroptosis and expressed reduced levels of RIP3 and cylindromatosis (CYLD), which regulates RIP1.7 Another study on childhood acute lymphoblastic leukemia reported that RIP1 was necessary to mediate the inhibitor of apoptosis protein-mediated sensitization of blast cells to chemotherapy.8 Autocrine TNFα loops that activate NF-κB through RIP1 have also been described in various cancer cell lines.9, 10Here we report that the expression of RIP3 was decreased in the majority of acute myeloid leukemia (AML) patients examined, whereas the expression of RIP1 remained unaffected. The expression of a RIP3 mutant with an inactivated kinase domain (RIP3-kinase dead (RIP3-KD)) in myeloid cell lines resulted in massive and early apoptosis and the caspase-mediated cleavage of p65/RkelA at a caspase-6 putative consensus site. Moreover, only RIP3-KD prolonged the survival of leukemic mice. Our results show that RIP3 activity regulates the apoptosis/necroptosis switch via its kinase activity in leukemia cells, and that other functions of RIP3 that are independent of its kinase domain modulate apoptosis and NF-κB activity.  相似文献   

20.
Sirtuins, NAD-dependent protein deacetylases, play important roles in cellular functions such as metabolism and differentiation. Whether sirtuins function in tumorigenesis is still controversial, but sirtuins are aberrantly expressed in tumors, which may keep cancerous cells undifferentiated. Therefore, we investigated whether the inhibition of sirtuin family proteins induces cellular differentiation in leukemic cells. The sirtuin inhibitors tenovin-6 and BML-266 induce granulocytic differentiation in the acute promyelocytic leukemia (APL) cell line NB4. This differentiation is likely caused by an inhibition of SIRT2 deacetylase activity, judging from the accumulation of acetylated α-tubulin, a major SIRT2 substrate. Unlike the clinically used differentiation inducer all-trans retinoic acid, tenovin-6 shows limited effects on promyelocytic leukemia–retinoic acid receptor α (PML-RAR-α) stability and promyelocytic leukemia nuclear body formation in NB4 cells, suggesting that tenovin-6 does not directly target PML-RAR-α activity. In agreement with this, tenovin-6 induces cellular differentiation in the non-APL cell line HL-60, where PML-RAR-α does not exist. Knocking down SIRT2 by shRNA induces granulocytic differentiation in NB4 cells, which demonstrates that the inhibition of SIRT2 activity is sufficient to induce cell differentiation in NB4 cells. The overexpression of SIRT2 in NB4 cells decreases the level of granulocytic differentiation induced by tenovin-6, which indicates that tenovin-6 induces granulocytic differentiation by inhibiting SIRT2 activity. Taken together, our data suggest that targeting SIRT2 is a viable strategy to induce leukemic cell differentiation.  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