Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 362
1.
Cell Signal ; 120: 111203, 2024 Aug.
Article En | MEDLINE | ID: mdl-38723736

Metastasis is a key determinant in cancer mortality which is often associated with decreased levels of Nm23-H1, a well-established metastasis suppressor. Despite lacking a secretion signal peptide, Nm23-H1 has been reported to be present in the extracellular space and enclosed within extracellular vesicles (EVs). While the presence of Nm23-H1 proteins in EVs released by cancer cells has been observed through proteomics profiling, the role of vesicular Nm23-H1 remains unclear. Here, we investigated the function of vesicular Nm23-H1 using MDA-MB-231 (highly metastatic, low Nm23-H1) and MCF-7 (low/non-metastatic, high Nm23-H1) breast cancer cell models. Our findings confirm that Nm23-H1 is indeed encapsulated within EVs, and its levels can be manipulated through overexpression and knockdown approaches. Functional assays revealed that EVs derived from MDA-MB-231 cells that contained high levels of Nm23-H1 exhibit impaired pro-migratory properties, suggesting that vesicular Nm23-H1 may act as a metastasis suppressor. Furthermore, EVs with increased levels of Nm23-H1 altered the transcript levels of multiple cancer-related genes in recipient cells and stimulated type I interferon signaling through STAT1 phosphorylation. These results suggest the existence of an unconventional signaling pathway mediated by the uptake of EVs enriched with Nm23-H1, which may contribute to the anti-metastatic effect of Nm23-H1 in the tumor microenvironment. Additionally, our study demonstrates that elevated Nm23-H1 levels can impact the abundance of various other proteins encapsulated within breast cancer cell-derived EVs, such as SUSD2 (Sushi Domain Containing 2) which can also modulate metastasis.


Breast Neoplasms , Cell Movement , Extracellular Vesicles , NM23 Nucleoside Diphosphate Kinases , Humans , Extracellular Vesicles/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , NM23 Nucleoside Diphosphate Kinases/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Female , Cell Line, Tumor , STAT1 Transcription Factor/metabolism , MCF-7 Cells
2.
Cancer Metastasis Rev ; 43(2): 755-775, 2024 Jun.
Article En | MEDLINE | ID: mdl-38180572

We describe here the molecular basis of the complex formation of PRUNE1 with the tumor metastasis suppressors NME1 and NME2, two isoforms appertaining to the nucleoside diphosphate kinase (NDPK) enzyme family, and how this complex regulates signaling the immune system and energy metabolism, thereby shaping the tumor microenvironment (TME). Disrupting the interaction between NME1/2 and PRUNE1, as suggested, holds the potential to be an excellent therapeutic target for the treatment of cancer and the inhibition of metastasis dissemination. Furthermore, we postulate an interaction and regulation of the other Class I NME proteins, NME3 and NME4 proteins, with PRUNE1 and discuss potential functions. Class I NME1-4 proteins are NTP/NDP transphosphorylases required for balancing the intracellular pools of nucleotide diphosphates and triphosphates. They regulate different cellular functions by interacting with a large variety of other proteins, and in cancer and metastasis processes, they can exert pro- and anti-oncogenic properties depending on the cellular context. In this review, we therefore additionally discuss general aspects of class1 NME and PRUNE1 molecular structures as well as their posttranslational modifications and subcellular localization. The current knowledge on the contributions of PRUNE1 as well as NME proteins to signaling cascades is summarized with a special regard to cancer and metastasis.


Energy Metabolism , NM23 Nucleoside Diphosphate Kinases , Neoplasm Metastasis , Neoplasms , Signal Transduction , Humans , Neoplasms/pathology , Neoplasms/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Animals , Nucleoside-Diphosphate Kinase/metabolism , Acid Anhydride Hydrolases/metabolism , Tumor Microenvironment , Phosphoric Monoester Hydrolases
3.
Med Princ Pract ; 33(1): 56-65, 2024.
Article En | MEDLINE | ID: mdl-38029727

PURPOSE: The expression and regulatory mechanism of NSUN6 in lung cancer are still unclear. Our study explored whether NSUN6 mediates progression of lung cancer by affecting NM23-H1 expression in an m5C-dependent manner. METHODS: qRT-PCR, CCK-8, colony formation, transwell, and Western blot analysis were employed to probe the impact of NSUN6 on lung cancer cell proliferation, migration, and epithelial-mesenchymal transition (EMT). RMVar database was utilized to forecast the downstream genes of NSUN6. The mode of interaction between NSUN6 and NM23-H1 was determined by dot blot, luciferase assay, m5C RIP, and cell function assays. The effect of NSUN6 expression on tumor growth was verified in vivo. RESULTS: Expression of NSUN6 was reduced in lung cancer cells, and over-expression of NSUN6 restricted the proliferation of lung cancer cells, migration, and EMT. NSUN6 regulated NM23-H1 expression by modifying the 3'-UTR of NM23-H1 mRNA through m5C and inhibited lung cancer cell proliferation, migration, and EMT. In vivo experiments also showed that over-expression of NSUN6 inhibited the occurrence of lung cancer. CONCLUSION: NSUN6 regulates NM23-H1 expression in an m5C-dependent manner to affect EMT in lung cancer. Thus, NSUN6 may be considered as a potential therapeutic target for lung cancer.


Epithelial-Mesenchymal Transition , Lung Neoplasms , tRNA Methyltransferases , Humans , Cell Line, Tumor , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , tRNA Methyltransferases/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism
4.
Cancer Metastasis Rev ; 42(4): 1169-1188, 2023 12.
Article En | MEDLINE | ID: mdl-37749308

Since the identification of NM23 (now called NME1) as the first metastasis suppressor gene (MSG), a small number of other gene products and non-coding RNAs have been identified that suppress specific parameters of the metastatic cascade, yet which have little or no ability to regulate primary tumor initiation or maintenance. MSG can regulate various pathways or cell biological functions such as those controlling mitogen-activated protein kinase pathway mediators, cell-cell and cell-extracellular matrix protein adhesion, cytoskeletal architecture, G-protein-coupled receptors, apoptosis, and transcriptional complexes. One defining facet of this gene class is that their expression is typically downregulated, not mutated, in metastasis, such that any effective therapeutic intervention would involve their re-expression. This review will address the therapeutic targeting of MSG, once thought to be a daunting task only facilitated by ectopically re-expressing MSG in metastatic cells in vivo. Examples will be cited of attempts to identify actionable oncogenic pathways that might suppress the formation or progression of metastases through the re-expression of specific metastasis suppressors.


Genes, Tumor Suppressor , NM23 Nucleoside Diphosphate Kinases , Humans , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplasm Metastasis
5.
J Cell Biol ; 222(10)2023 10 02.
Article En | MEDLINE | ID: mdl-37584589

Mitochondria are dynamic organelles regulated by fission and fusion processes. The fusion of membranes requires elaborative coordination of proteins and lipids and is particularly crucial for the function and quality control of mitochondria. Phosphatidic acid (PA) on the mitochondrial outer membrane generated by PLD6 facilitates the fusion of mitochondria. However, how PA promotes mitochondrial fusion remains unclear. Here, we show that a mitochondrial outer membrane protein, NME3, is required for PLD6-induced mitochondrial tethering or clustering. NME3 is enriched at the contact interface of two closely positioned mitochondria depending on PLD6, and NME3 binds directly to PA-exposed lipid packing defects via its N-terminal amphipathic helix. The PA binding function and hexamerization confer NME3 mitochondrial tethering activity. Importantly, nutrient starvation enhances the enrichment efficiency of NME3 at the mitochondrial contact interface, and the tethering ability of NME3 contributes to fusion efficiency. Together, our findings demonstrate NME3 as a tethering protein promoting selective fusion between PLD6-remodeled mitochondria for quality control.


Mitochondria , NM23 Nucleoside Diphosphate Kinases , Phosphatidic Acids , Phospholipase D , Humans , Mitochondria/metabolism , Mitochondrial Dynamics , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Phosphatidic Acids/metabolism , Phospholipase D/metabolism
6.
Cancer Metastasis Rev ; 42(4): 1155-1167, 2023 12.
Article En | MEDLINE | ID: mdl-37353690

Metastatic progression is regulated by metastasis promoter and suppressor genes. NME1, the prototypic and first described metastasis suppressor gene, encodes a nucleoside diphosphate kinase (NDPK) involved in nucleotide metabolism; two related family members, NME2 and NME4, are also reported as metastasis suppressors. These proteins physically interact with members of the GTPase dynamin family, which have key functions in membrane fission and fusion reactions necessary for endocytosis and mitochondrial dynamics. Evidence supports a model in which NDPKs provide GTP to dynamins to maintain a high local GTP concentration for optimal dynamin function. NME1 and NME2 are cytosolic enzymes that provide GTP to dynamins at the plasma membrane, which drive endocytosis, suggesting that these NMEs are necessary to attenuate signaling by receptors on the cell surface. Disruption of NDPK activity in NME-deficient tumors may thus drive metastasis by prolonging signaling. NME4 is a mitochondrial enzyme that interacts with the dynamin OPA1 at the mitochondria inner membrane to drive inner membrane fusion and maintain a fused mitochondrial network. This function is consistent with the current view that mitochondrial fusion inhibits the metastatic potential of tumor cells whereas mitochondrial fission promotes metastasis progression. The roles of NME family members in dynamin-mediated endocytosis and mitochondrial dynamics and the intimate link between these processes and metastasis provide a new framework to understand the metastasis suppressor functions of NME proteins.


NM23 Nucleoside Diphosphate Kinases , Neoplasms , Humans , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Dynamins/metabolism , Neoplasms/pathology , Cell Membrane/metabolism , Guanosine Triphosphate
7.
J Inherit Metab Dis ; 46(2): 273-285, 2023 03.
Article En | MEDLINE | ID: mdl-36522796

Peroxisomes are essential organelles involved in lipid metabolisms including plasmalogen biosynthesis and ß-oxidation of very long-chain fatty acids. Peroxisomes proliferate by the growth and division of pre-existing peroxisomes. The peroxisomal membrane is elongated by Pex11ß and then divided by the dynamin-like GTPase, DLP1 (also known as DRP1 encoded by DNM1L gene), which also functions as a fission factor for mitochondria. Nucleoside diphosphate kinase 3 (NME3) localized in both peroxisomes and mitochondria generates GTP for DLP1 activity. Deficiencies of either of these factors induce abnormal morphology of peroxisomes and/or mitochondria, and are associated with central nervous system dysfunction. To investigate whether the impaired division of peroxisomes affects lipid metabolisms, we assessed the phospholipid composition of cells lacking each of the different division factors. In fibroblasts from the patients deficient in DLP1, NME3, or Pex11ß, docosahexaenoic acid (DHA, C22:6)-containing phospholipids were found to be decreased. Conversely, the levels of several fatty acids such as arachidonic acid (AA, C20:4) and oleic acid (C18:1) were elevated. Mouse embryonic fibroblasts from Drp1- and Pex11ß-knockout mice also showed a decrease in the levels of phospholipids containing DHA and AA. Collectively, these results suggest that the dynamics of organelle morphology exert marked effects on the fatty acid composition of phospholipids.


Docosahexaenoic Acids , Peroxisomes , Animals , Mice , Docosahexaenoic Acids/metabolism , Dynamins/metabolism , Fatty Acids/metabolism , Fibroblasts/metabolism , Morphogenesis , NM23 Nucleoside Diphosphate Kinases/metabolism , Peroxisomes/metabolism , Phospholipids/metabolism
8.
Medicine (Baltimore) ; 101(39): e30815, 2022 Sep 30.
Article En | MEDLINE | ID: mdl-36181032

BACKGROUND: The relationship between the expression of nm23-H1 and the invasion and prognosis of non-small cell lung cancer (NSCLC) is still controversial. Therefore, we conducted a meta-analysis to determine the prognostic value of nm23-H1 in patients with NSCLC. And to explore the relationship between the expression of nm23-H1 and clinicopathological features in patients with NSCLC. METHODS: Literature search in PubMed, EMBASE, Cochrane Library, CNKI, and WanFang database was performed up to June 14, 2021. Studies on the expression and clinical significance of nm23-H1 in NSCLC were included. According to the inclusion and exclusion criteria, 2 researchers independently screened the literatures, extracted the data, and evaluated the quality. Meta-analysis was performed using RevMan 5.4 software (Nordic Cochran Centre, Copenhagen, Denmark). RESULTS: Twenty-five studies met our inclusion criteria and were finally included for the analysis, involving 2198 participants. Our meta-analysis revealed that nm23-H1 expression was associated with tumor differentiation (OR = 0.54, 95% CI: 0.42-0.70, P < .00001), TNM stage (OR = 1.70, 95% CI: 1.23-2.34, P = .001), and lymph node status (OR = 0.26, 95% CI, 0.17-0.39, P < .00001), but have no associate with sex, age, pathological type, and T stages. Additionally, low nm23-H1 expression reduced the 3-year survival rate (OR = 2.74, 95% CI: 1.54-4.86, P = .0006) and 5-year survival rate (OR = 2.78, 95% CI: 1.36-5.69, P = .005). CONCLUSION: Nm23-H1 can be used as a biomarker to predict tumor invasiveness and evaluate the prognosis of patients with NSCLC.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/pathology , NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplasm Staging , Prognosis
9.
Clin Transl Med ; 12(9): e1037, 2022 09.
Article En | MEDLINE | ID: mdl-36116139

BACKGROUND: Colorectal cancer (CRC) has a high mortality rate, and therapeutic approaches to treat these cancers are varied and depend on the metabolic state of the tumour. Profiles of CRC tumours have identified several biomarkers, including microRNAs. microRNA-210 (miR-210) levels are directly correlated with CRC survival. miR-210 expression is higher in metastatic colon cancer cells versus non-metastatic and normal colon epithelium. Therefore, efficient methods to inhibit miR-210 expression in CRC may provide new advances in treatments. METHODS: Expression of miRs was determined in several metastatic and non-metastatic cell lines. miR-210 expression was inhibited using PMIS-miR-210 in transduced cells, which were transplanted into xenograft mice. In separate experiments, CRC tumours were allowed to grow in xenograft mice and treated with therapeutic injections of PMIS-miR-210. Molecular and biochemical experiments identified several new pathways targeted by miR-210 inhibition. RESULTS: miR-210 inhibition can significantly reduce tumour growth of implanted colon cancer cells in xenograft mouse models. The direct administration of PMIS-miR-210 to existing tumours can inhibit tumour growth in both NSG and Foxn1nu/j mouse models and is more efficacious than capecitabine treatments. Tumour cells further transfer the PMIS-miR-210 inhibitor to neighbouring cells by extracellular vesicles to inhibit miR-210 throughout the tumour. miR-210 inhibition activates the cleaved caspase 3 apoptotic pathway to reduce tumour formation. We demonstrate that the long non-coding transcript XIST is regulated by miR-210 correlating with decreased XIST expression in CRC tumours. XIST acts as a competing endogenous RNA for miR-210, which reduces XIST levels and miR-210 inhibition increases XIST transcripts in the nucleus and cytoplasm. The increased expression of NME1 is associated with H3K4me3 and H3K27ac modifications in the NME1 proximal promoter by XIST. CONCLUSION: Direct application of the PMIS-miR-210 inhibitor to growing tumours may be an effective colorectal cancer therapeutic.


Colonic Neoplasms , Extracellular Vesicles , MicroRNAs , NM23 Nucleoside Diphosphate Kinases , RNA, Long Noncoding , Animals , Apoptosis/genetics , Capecitabine , Caspase 3 , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
10.
Clin Exp Metastasis ; 39(5): 815-831, 2022 10.
Article En | MEDLINE | ID: mdl-35939247

Tumor-derived exosomes have documented roles in accelerating the initiation and outgrowth of metastases, as well as in therapy resistance. Little information supports the converse, that exosomes or similar vesicles can suppress metastasis. We investigated the NME1 (Nm23-H1) metastasis suppressor as a candidate for metastasis suppression by extracellular vesicles. Exosomes derived from two cancer cell lines (MDA-MB-231T and MDA-MB-435), when transfected with the NME1 (Nm23-H1) metastasis suppressor, secreted exosomes with NME1 as the predominant constituent. These exosomes entered recipient tumor cells, altered their endocytic patterns in agreement with NME1 function, and suppressed in vitro tumor cell motility and migration compared to exosomes from control transfectants. Proteomic analysis of exosomes revealed multiple differentially expressed proteins that could exert biological functions. Therefore, we also prepared and investigated liposomes, empty or containing partially purified rNME1. rNME1 containing liposomes recapitulated the effects of exosomes from NME1 transfectants in vitro. In an experimental lung metastasis assay the median lung metastases per histologic section was 158 using control liposomes and 15 in the rNME1 liposome group, 90.5% lower than the control liposome group (P = 0.016). The data expand the exosome/liposome field to include metastasis suppressive functions and describe a new translational approach to prevent metastasis.


Breast Neoplasms , Exosomes , Lung Neoplasms , NM23 Nucleoside Diphosphate Kinases , Cell Line, Tumor , Female , Humans , Liposomes , Lung Neoplasms/secondary , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplasm Metastasis , Proteomics
11.
Adv Clin Exp Med ; 31(6): 655-670, 2022 Jun.
Article En | MEDLINE | ID: mdl-35438846

BACKGROUND: High expression of NME1 is associated with hepatocellular carcinoma (HCC) progression and poor prognosis. However, there are few reports on the association between NME1 and microRNAs (miRNAs) in HCC progression. OBJECTIVES: To explore miRNAs that regulate NME1 expression in HCC. MATERIAL AND METHODS: Data from the Cancer Genome Atlas (TCGA), Human Protein Atlas (HPA), TargetScan, starBase, and mirDIP were used to analyze the expression pattern of NME1 in HCC tissues, the relationship between NME1 level and the progression of HCC or patient prognosis, miRNAs targeting NME1, and the biological processes that may be regulated by NME1. The regulation of miRNAs to NME1 was assessed using the dual-luciferase reporter assay, quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blotting. The cell cycle and cell proliferation were detected using propidium iodide (PI) staining and EdU assay, respectively. RESULTS: Highly expressed NME1 in HCC was associated with HCC progression and prognosis. The miR-139-5p and miR-335-5p were weakly expressed in HCC samples and negatively correlated with NME1. The downregulation of miR-139-5p in HCC patients resulted in worse overall survival (OS) and disease-free interval (DFI); however, the level of miR-335-5p was not significantly correlated with OS and DFI in patients with HCC. In vitro experiments verified that the level of miR-139-5p was lower and NME1 expression was higher in HCC cell lines compared to L-02. Moreover, miR-139-5p negatively regulates the expression of NME1 in HCC cell lines. The NME1 may regulate cell cycle, DNA replication, oxidative phosphorylation, and the pentose phosphate pathway. The miR-139-5p inhibited cell proliferation by negatively regulating NME1 expression. CONCLUSIONS: The upregulation of NME1 in HCC indicates a poor prognosis. The NME1 is negatively regulated by miR-139-5p to inhibit cell proliferation.


Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , NM23 Nucleoside Diphosphate Kinases , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism
12.
Microb Pathog ; 166: 105457, 2022 May.
Article En | MEDLINE | ID: mdl-35219843

Leishmania donovani pathogenicity is closely linked to its ability to live and replicate in the hostile environment of macrophages. All protozoan parasites, including Leishmania, are unable to synthesize purines de novo, and nucleoside diphosphate kinases (NDKs) are enzymes required to preserve the intracellular nucleoside phosphate equilibrium. For some pathogens, secretion of ATP-utilizing enzymes into the extracellular environment aids in pathogen survival via P2Z receptor mediated, ATP-induced death of infected macrophages. Here, Leishmanaia donovani nucleoside diphosphate kinase (LdNDKb) was cloned, expressed and purified by Ni2+-NTA affinity chromatography to elucidate its biological significance. The presence of secreted form of LdNDKb in the medium was confirmed by Western blot analysis. Interestingly, cellular localization by confocal microscopy showed that this protein was localized in the nucleus, inner leaflet of membrane and on the flagella of this parasite which indicates its multiple role in the life cycle of Leishmania donovani. Its possibility to bind with DNA was confirmed by gel retardation assay and electrophoretic mobility shift assay (EMSA) which show the binding with linear and supercoiled is not sequence specific. Further, treatment of J774 macrophages with recombinant LdNdKb and periodate oxidized ATP - a P2X7 receptor antagonist, inhibited ATP-induced cytolysis in vitro, as determined by lactate dehydrogenise release from J774 macrophages. Thus, LdNDKb prevents ATP-mediated host-cell plasma membrane permeabilization by hydrolyzing extracellular ATP, thereby, preserving the integrity of the host cells for the benefit of the parasite. This study indicates that LdNDKb could be explored for its potentiality as a drug/vaccine target against visceral leishmaniasis.


Leishmania donovani , Nucleoside-Diphosphate Kinase , Adenosine Triphosphate/metabolism , Cell Death , Macrophages/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Nucleoside-Diphosphate Kinase/genetics , Nucleoside-Diphosphate Kinase/metabolism
13.
Sci Rep ; 11(1): 23549, 2021 12 07.
Article En | MEDLINE | ID: mdl-34876614

Mitochondrial oxidative phosphorylation (OXPHOS) has become an attractive target in anti-cancer studies in recent years. In this study, we found that a small molecule phenylbutenoid dimer NMac1 (Nm23-H1 activator 1), (±)-trans-3-(3,4-dimethoxyphenyl)-4-[(E)-3,4-dimethoxystyryl]cyclohex-1-ene, a previously identified anti-metastatic agent, has novel anti-proliferative effect only under glucose starvation in metastatic breast cancer cells. NMac1 causes significant activation of AMPK by decreasing ATP synthesis, lowers mitochondrial membrane potential (MMP, ΔΨm), and inhibits oxygen consumption rate (OCR) under glucose starvation. These effects of NMac1 are provoked by a consequence of OXPHOS complex I inhibition. Through the structure-activity relationship (SAR) study of NMac1 derivatives, NMac24 was identified as the most effective compound in anti-proliferation. NMac1 and NMac24 effectively suppress cancer cell proliferation in 3D-spheroid in vivo-like models only under glucose starvation. These results suggest that NMac1 and NMac24 have the potential as anti-cancer agents having cytotoxic effects selectively in glucose restricted cells.


Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cyclohexenes/pharmacology , NM23 Nucleoside Diphosphate Kinases/drug effects , Styrenes/pharmacology , Adenosine Triphosphate/biosynthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclohexenes/chemistry , Electron Transport Complex I/antagonists & inhibitors , Enzyme Activators/chemistry , Enzyme Activators/pharmacology , Female , Gene Regulatory Networks/drug effects , Glucose/metabolism , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Metabolome/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Signal Transduction/drug effects , Structure-Activity Relationship , Styrenes/chemistry
14.
Cell Death Dis ; 12(10): 924, 2021 10 09.
Article En | MEDLINE | ID: mdl-34628473

Cancer stem cells (CSCs), which can self-renew and produce heterogeneous cancer cells, are the key factors during tumorigenesis. Transcription factors take essential effects on CSCs. However, the role of transcription factors in regulating the stemness of gastric cancer stem-like cells has not been well explored. In this investigation, it was found that transcription factor NME2 (NME/NM23 nucleoside diphosphate kinase 2) was upregulated in gastric cancer stem-like cells that sorted from the solid tumors of patients with gastric cancer and gastric cancer cell lines. NME2 could preserve the stemness of gastric cancer stem-like cells via suppressing their apoptosis. In vitro and in vivo data revealed that NME2 was crucial for maintaining the stemness of gastric cancer stem cells by enhancing the expression of anti-apoptosis genes. Consequently, our data contributed a new perspective to the relationship between transcription factor and the stemness maintenance of gastric cancer stem cells.


NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplastic Stem Cells/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Base Sequence , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Mice, Inbred NOD , Mice, SCID , Models, Biological , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplastic Stem Cells/pathology , Up-Regulation/genetics
15.
BMC Biol ; 19(1): 228, 2021 10 21.
Article En | MEDLINE | ID: mdl-34674701

BACKGROUND: Mitochondrial nucleoside diphosphate kinase (NDPK-D, NME4, NM23-H4) is a multifunctional enzyme mainly localized in the intermembrane space, bound to the inner membrane. RESULTS: We constructed loss-of-function mutants of NDPK-D, lacking either NDP kinase activity or membrane interaction and expressed mutants or wild-type protein in cancer cells. In a complementary approach, we performed depletion of NDPK-D by RNA interference. Both loss-of-function mutations and NDPK-D depletion promoted epithelial-mesenchymal transition and increased migratory and invasive potential. Immunocompromised mice developed more metastases when injected with cells expressing mutant NDPK-D as compared to wild-type. This metastatic reprogramming is a consequence of mitochondrial alterations, including fragmentation and loss of mitochondria, a metabolic switch from respiration to glycolysis, increased ROS generation, and further metabolic changes in mitochondria, all of which can trigger pro-metastatic protein expression and signaling cascades. In human cancer, NME4 expression is negatively associated with markers of epithelial-mesenchymal transition and tumor aggressiveness and a good prognosis factor for beneficial clinical outcome. CONCLUSIONS: These data demonstrate NME4 as a novel metastasis suppressor gene, the first localizing to mitochondria, pointing to a role of mitochondria in metastatic dissemination.


Neoplasms , Nucleoside-Diphosphate Kinase , Animals , Intracellular Membranes , Mice , Mitochondria , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Nucleoside Diphosphate Kinase D/metabolism , Nucleoside-Diphosphate Kinase/genetics , Nucleoside-Diphosphate Kinase/metabolism
16.
Oncogene ; 40(23): 4019-4032, 2021 06.
Article En | MEDLINE | ID: mdl-34012098

Membrane Type 1 Matrix Metalloprotease (MT1-MMP) contributes to the invasive progression of breast cancers by degrading extracellular matrix tissues. Nucleoside diphosphate kinase, NME1/NM23-H1, has been identified as a metastasis suppressor; however, its contribution to local invasion in breast cancer is not known. Here, we report that NME1 is up-regulated in ductal carcinoma in situ (DCIS) as compared to normal breast epithelial tissues. NME1 levels drop in microinvasive and invasive components of breast tumor cells relative to synchronous DCIS foci. We find a strong anti-correlation between NME1 and plasma membrane MT1-MMP levels in the invasive components of breast tumors, particularly in aggressive histological grade III and triple-negative breast cancers. Knockout of NME1 accelerates the invasive transition of breast tumors in the intraductal xenograft model. At the mechanistic level, we find that MT1-MMP, NME1 and dynamin-2, a GTPase known to require GTP production by NME1 for its membrane fission activity in the endocytic pathway, interact in clathrin-coated vesicles at the plasma membrane. Loss of NME1 function increases MT1-MMP surface levels by inhibiting endocytic clearance. As a consequence, the ECM degradation and invasive potentials of breast cancer cells are enhanced. This study identifies the down-modulation of NME1 as a potent driver of the in situ-to invasive transition during breast cancer progression.


Breast Neoplasms/metabolism , Dynamin II/metabolism , Extracellular Matrix/metabolism , Matrix Metalloproteinase 14/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line , Cell Movement/physiology , Female , Humans , Matrix Metalloproteinase 14/genetics , Mice , Mice, Nude , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Xenograft Model Antitumor Assays
17.
Int J Mol Sci ; 22(7)2021 Mar 27.
Article En | MEDLINE | ID: mdl-33801585

The metastasis suppressor function of NM23 proteins is widely understood. Multiple enzymatic activities of NM23 proteins have also been identified. However, relatively less known interesting aspects are being revealed from recent developments that corroborate the telomeric interactions of NM23 proteins. Telomeres are known to regulate essential physiological events such as metastasis, ageing, and cellular differentiation via inter-connected signalling pathways. Here, we review the literature on the association of NM23 proteins with telomeres or telomere-related factors, and discuss the potential implications of emerging telomeric functions of NM23 proteins. Further understanding of these aspects might be instrumental in better understanding the metastasis suppressor functions of NM23 proteins.


Aging , Gene Expression Regulation, Neoplastic , NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/metabolism , Telomere/metabolism , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Cytoskeleton/metabolism , DNA/chemistry , G-Quadruplexes , Humans , Lymphocyte Activation , Mitochondria/metabolism , Nucleoside Diphosphate Kinase D/chemistry , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , T-Lymphocytes/cytology , Telomere/ultrastructure , Transcription Factors/metabolism
18.
Int J Mol Sci ; 22(7)2021 Apr 02.
Article En | MEDLINE | ID: mdl-33918324

Epithelial-mesenchymal transition (EMT) is important for the initial steps of metastasis. Although it is well accepted that the nucleoside diphosphate kinase NME1 is a metastasis suppressor, its effect on EMT remains poorly documented, as does that of its closely related isoform, NME2. Here, by using gene silencing, inactivation and overexpression strategies in a variety of cellular models of cancer, we show that NME1 is a powerful inhibitor of EMT. Genetic manipulation of NME2, by contrast, had no effect on the EMT phenotype of cancer cells, indicating a specific function of NME1 in EMT regulation. Loss of NME1 in epithelial cancer cells resulted in a hybrid phenotype intermediate between epithelial and mesenchymal cells, which is known to be associated with cells with a highly metastatic character. Conversely, overexpression of NME1 in mesenchymal cancer cells resulted in a more epithelial phenotype. We found that NME1 expression was negatively associated with EMT markers in many human cancers and was reduced in human breast tumor cell lines with the aggressive 'triple-negative' phenotype when compared to human breast tumor cell lines positive for estrogen receptor. We show that NME1, but not NME2, is an inhibitor of essential concerted intracellular signaling pathways involved in inducing EMT, including the AKT and MAPK (ERK, p38, and JNK) pathways. Additionally, NME1 depletion considerably altered the distribution of E-cadherin, a gatekeeper of the epithelial phenotype, shifting it from the plasma membrane to the cytosol and resulting in less E-cadherin on the cell surface than in control cells. Functional aggregation and dispersion assays demonstrated that inactivation of NME1 decreases E-cadherin-mediated cell-cell adhesion. We conclude that NME1, but not NME2, acts specifically to inhibit EMT and prevent the earliest stages of metastasis.


Epithelial-Mesenchymal Transition , NM23 Nucleoside Diphosphate Kinases/metabolism , Animals , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Female , Gene Editing , Humans , MAP Kinase Signaling System , Mice, Nude , Triple Negative Breast Neoplasms/metabolism
19.
Redox Biol ; 44: 101978, 2021 08.
Article En | MEDLINE | ID: mdl-33903070

The metastasis suppressor protein NME1 is an evolutionarily conserved and multifunctional enzyme that plays an important role in suppressing the invasion and metastasis of tumour cells. The nucleoside diphosphate kinase (NDPK) activity of NME1 is well recognized in balancing the intracellular pools of nucleotide diphosphates and triphosphates to regulate cytoskeletal rearrangement and cell motility, endocytosis, intracellular trafficking, and metastasis. In addition, NME1 was found to function as a protein-histidine kinase, 3'-5' exonuclease and geranyl/farnesyl pyrophosphate kinase. These diverse cellular functions are regulated at the level of expression, post-translational modifications, and regulatory interactions. The NDPK activity of NME1 has been shown to be inhibited in vitro and in vivo under oxidative stress, and the inhibitory effect mediated via redox-sensitive cysteine residues. In this study, affinity purification followed by mass spectrometric analysis revealed NME1 to be a major coenzyme A (CoA) binding protein in cultured cells and rat tissues. NME1 is also found covalently modified by CoA (CoAlation) at Cys109 in the CoAlome analysis of HEK293/Pank1ß cells treated with the disulfide-stress inducer, diamide. Further analysis showed that recombinant NME1 is efficiently CoAlated in vitro and in cellular response to oxidising agents and metabolic stress. In vitro CoAlation of recombinant wild type NME1, but not the C109A mutant, results in the inhibition of its NDPK activity. Moreover, CoA also functions as a competitive inhibitor of the NME1 NDPK activity by binding non-covalently to the nucleotide binding site. Taken together, our data reveal metastasis suppressor protein NME1 as a novel binding partner of the key metabolic regulator CoA, which inhibits its nucleoside diphosphate kinase activity via non-covalent and covalent interactions.


Coenzyme A , Neoplasms , Animals , HEK293 Cells , Humans , NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplasm Metastasis , Oxidation-Reduction , Rats
20.
Exp Mol Med ; 53(3): 346-357, 2021 03.
Article En | MEDLINE | ID: mdl-33753879

Non-metastatic protein 23 H1 (Nm23-H1), a housekeeping enzyme, is a nucleoside diphosphate kinase-A (NDPK-A). It was the first identified metastasis suppressor protein. Nm23-H1 prolongs disease-free survival and is associated with a good prognosis in breast cancer patients. However, the molecular mechanisms underlying the role of Nm23-H1 in biological processes are still not well understood. This is a review of recent studies focusing on controlling NDPK activity based on the redox regulation of Nm23-H1, structural, and functional changes associated with the oxidation of cysteine residues, and the relationship between NDPK activity and cancer metastasis. Further understanding of the redox regulation of the NDPK function will likely provide a new perspective for developing new strategies for the activation of NDPK-A in suppressing cancer metastasis.


Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cysteine/chemistry , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Female , Humans , Neoplasm Metastasis , Oxidation-Reduction
...