Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.355
Filter
1.
Cancer Metastasis Rev ; 43(2): 755-775, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38180572

ABSTRACT

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.


Subject(s)
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
2.
Proc Natl Acad Sci U S A ; 119(11): e2117013119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35259022

ABSTRACT

SignificanceThe study provided a long-sought molecular mechanism that could explain the link between fatty acid metabolism and cancer metastasis. Further understanding may lead to new strategies to inhibit cancer metastasis. The chemical proteomic approach developed here will be useful for discovering other regulatory mechanisms of protein function by small molecule metabolites.


Subject(s)
Acyl Coenzyme A/metabolism , NM23 Nucleoside Diphosphate Kinases/antagonists & inhibitors , Neoplasms/metabolism , Neoplasms/pathology , Breast Neoplasms , Endocytosis , Female , Humans , Neoplasm Metastasis , Neoplasms/etiology , Protein Binding , Proteome , Proteomics/methods
3.
Cancer Metastasis Rev ; 42(4): 1169-1188, 2023 12.
Article in English | MEDLINE | ID: mdl-37749308

ABSTRACT

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.


Subject(s)
Genes, Tumor Suppressor , NM23 Nucleoside Diphosphate Kinases , Humans , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplasm Metastasis
4.
Cancer Metastasis Rev ; 42(4): 1155-1167, 2023 12.
Article in English | MEDLINE | ID: mdl-37353690

ABSTRACT

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.


Subject(s)
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
5.
Med Princ Pract ; 33(1): 56-65, 2024.
Article in English | MEDLINE | ID: mdl-38029727

ABSTRACT

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.


Subject(s)
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
6.
Br J Cancer ; 129(10): 1679-1691, 2023 11.
Article in English | MEDLINE | ID: mdl-37731021

ABSTRACT

BACKGROUND: NME1 has been exploited as a potential translational target for decades. Substantial efforts have been made to upregulate the expression of NME1 and restore its anti-metastasis function in metastatic cancer. METHODS: Cycloheximide (CHX) chase assay was used to measure the steady-state protein stability of NME1 and HSP90α. The NME1-associating proteins were identified by immunoprecipitation combined with mass spectrometric analysis. Gene knockdown and overexpression were employed to examine the impact of HSP90AA1 on intracellular NME1 degradation. The motility and invasiveness of breast cancer cells were examined in vitro using wound healing and transwell invasion assays. The orthotopic spontaneous metastasis and intra-venous experimental metastasis assays were used to test the formation of metastasis in vivo, respectively. RESULTS: HSP90α interacts with NME1 and increases NME1 lifetime by impeding its ubiquitin-proteasome-mediated degradation. HSP90α overexpression significantly inhibits the metastatic potential of breast cancer cells in vitro and in vivo. A novel cell-permeable peptide, OPT22 successfully mimics the HSP90α function and prolongs the life span of endogenous NME1, resulting in reduced metastasis of breast cancer. CONCLUSION: These results not only reveal a new mechanism of NME1 degradation but also pave the way for the development of new and effective approaches to metastatic cancer therapy.


Subject(s)
Breast Neoplasms , Heat-Shock Proteins , Humans , Female , Heat-Shock Proteins/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Protein Processing, Post-Translational , HSP90 Heat-Shock Proteins/metabolism , Neoplasm Metastasis , NM23 Nucleoside Diphosphate Kinases/genetics
7.
RNA ; 27(6): 694-709, 2021 06.
Article in English | MEDLINE | ID: mdl-33795480

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs that function as critical posttranscriptional regulators in various biological processes. While most miRNAs are generated from processing of long primary transcripts via sequential Drosha and Dicer cleavage, some miRNAs that bypass Drosha cleavage can be transcribed as part of another small noncoding RNA. Here, we develop the target-oriented miRNA discovery (TOMiD) bioinformatic analysis method to identify Drosha-independent miRNAs from Argonaute crosslinking and sequencing of hybrids (Ago-CLASH) data sets. Using this technique, we discovered a novel miRNA derived from a primate specific noncoding RNA, the small NF90 associated RNA A (snaR-A). The miRNA derived from snaR-A (miR-snaR) arises independently of Drosha processing but requires Exportin-5 and Dicer for biogenesis. We identify that miR-snaR is concurrently up-regulated with the full snaR-A transcript in cancer cells. Functionally, miR-snaR associates with Ago proteins and targets NME1, a key metastasis inhibitor, contributing to snaR-A's role in promoting cancer cell migration. Our findings suggest a functional link between a novel miRNA and its precursor noncoding RNA.


Subject(s)
Computational Biology/methods , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , 3' Untranslated Regions , Cell Movement , DEAD-box RNA Helicases/metabolism , HCT116 Cells , HEK293 Cells , Humans , Karyopherins/metabolism , MCF-7 Cells , NM23 Nucleoside Diphosphate Kinases/antagonists & inhibitors , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasms/pathology , RNA, Long Noncoding/metabolism , Ribonuclease III/metabolism
8.
J Inherit Metab Dis ; 46(2): 273-285, 2023 03.
Article in English | MEDLINE | ID: mdl-36522796

ABSTRACT

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.


Subject(s)
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
9.
Int J Mol Sci ; 24(11)2023 May 27.
Article in English | MEDLINE | ID: mdl-37298313

ABSTRACT

Coenzyme A (CoA) is a key cellular metabolite which participates in diverse metabolic pathways, regulation of gene expression and the antioxidant defense mechanism. Human NME1 (hNME1), which is a moonlighting protein, was identified as a major CoA-binding protein. Biochemical studies showed that hNME1 is regulated by CoA through both covalent and non-covalent binding, which leads to a decrease in the hNME1 nucleoside diphosphate kinase (NDPK) activity. In this study, we expanded the knowledge on previous findings by focusing on the non-covalent mode of CoA binding to the hNME1. With X-ray crystallography, we solved the CoA bound structure of hNME1 (hNME1-CoA) and determined the stabilization interactions CoA forms within the nucleotide-binding site of hNME1. A hydrophobic patch stabilizing the CoA adenine ring, while salt bridges and hydrogen bonds stabilizing the phosphate groups of CoA were observed. With molecular dynamics studies, we extended our structural analysis by characterizing the hNME1-CoA structure and elucidating possible orientations of the pantetheine tail, which is absent in the X-ray structure due to its flexibility. Crystallographic studies suggested the involvement of arginine 58 and threonine 94 in mediating specific interactions with CoA. Site-directed mutagenesis and CoA-based affinity purifications showed that arginine 58 mutation to glutamate (R58E) and threonine 94 mutation to aspartate (T94D) prevent hNME1 from binding to CoA. Overall, our results reveal a unique mode by which hNME1 binds CoA, which differs significantly from that of ADP binding: the α- and ß-phosphates of CoA are oriented away from the nucleotide-binding site, while 3'-phosphate faces catalytic histidine 118 (H118). The interactions formed by the CoA adenine ring and phosphate groups contribute to the specific mode of CoA binding to hNME1.


Subject(s)
Nucleotides , Threonine , Humans , Crystallography, X-Ray , Binding Sites , Coenzyme A , Arginine , Adenine , NM23 Nucleoside Diphosphate Kinases/genetics
10.
Microb Pathog ; 166: 105457, 2022 May.
Article in English | MEDLINE | ID: mdl-35219843

ABSTRACT

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.


Subject(s)
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
11.
Proc Natl Acad Sci U S A ; 116(2): 566-574, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30587587

ABSTRACT

We report a patient who presented with congenital hypotonia, hypoventilation, and cerebellar histopathological alterations. Exome analysis revealed a homozygous mutation in the initiation codon of the NME3 gene, which encodes an NDP kinase. The initiation-codon mutation leads to deficiency in NME3 protein expression. NME3 is a mitochondrial outer-membrane protein capable of interacting with MFN1/2, and its depletion causes dysfunction in mitochondrial dynamics. Consistently, the patient's fibroblasts were characterized by a slow rate of mitochondrial dynamics, which was reversed by expression of wild-type or catalytic-dead NME3. Moreover, glucose starvation caused mitochondrial fragmentation and cell death in the patient's cells. The expression of wild-type and catalytic-dead but not oligomerization-attenuated NME3 restored mitochondrial elongation. However, only wild-type NME3 sustained ATP production and viability. Thus, the separate functions of NME3 in mitochondrial fusion and NDP kinase cooperate in metabolic adaptation for cell survival in response to glucose starvation. Given the critical role of mitochondrial dynamics and energy requirements in neuronal development, the homozygous mutation in NME3 is linked to a fatal mitochondrial neurodegenerative disorder.


Subject(s)
Adenosine Triphosphate , Energy Metabolism/genetics , Homozygote , Mitochondrial Dynamics/genetics , NM23 Nucleoside Diphosphate Kinases , Neurodegenerative Diseases , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Cell Line , Cell Survival , Female , Humans , Male , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/pathology , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology
12.
PLoS Genet ; 15(9): e1008378, 2019 09.
Article in English | MEDLINE | ID: mdl-31479451

ABSTRACT

Primary ciliary dyskinesia (PCD) is a hereditary defect of motile cilia in humans and several domestic animal species. Typical clinical findings are chronic recurrent infections of the respiratory tract and fertility problems. We analyzed an Alaskan Malamute family, in which two out of six puppies were affected by PCD. The parents were unaffected suggesting autosomal recessive inheritance. Linkage and homozygosity mapping defined critical intervals comprising ~118 Mb. Whole genome sequencing of one case and comparison to 601 control genomes identified a disease associated frameshift variant, c.43delA, in the NME5 gene encoding a sparsely characterized protein associated with ciliary function. Nme5-/- knockout mice exhibit doming of the skull, hydrocephalus and sperm flagellar defects. The genotypes at NME5:c.43delA showed the expected co-segregation with the phenotype in the Alaskan Malamute family. An additional unrelated Alaskan Malamute with PCD and hydrocephalus that became available later in the study was also homozygous mutant at the NME5:c.43delA variant. The mutant allele was not present in more than 1000 control dogs from different breeds. Immunohistochemistry demonstrated absence of the NME5 protein from nasal epithelia of an affected dog. We therefore propose NME5:c.43delA as the most likely candidate causative variant for PCD in Alaskan Malamutes. These findings enable genetic testing to avoid the unintentional breeding of affected dogs in the future. Furthermore, the results of this study identify NME5 as a novel candidate gene for unsolved human PCD and/or hydrocephalus cases.


Subject(s)
Ciliary Motility Disorders/genetics , NM23 Nucleoside Diphosphate Kinases/genetics , Animals , Breeding , Cilia/genetics , Ciliary Motility Disorders/physiopathology , Dogs/genetics , Female , Frameshift Mutation/genetics , Genetic Linkage/genetics , Genetic Testing , Genotype , Humans , Male , Mutation/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Phenotype , Whole Genome Sequencing
13.
BMC Biol ; 19(1): 228, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34674701

ABSTRACT

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.


Subject(s)
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
14.
Int J Mol Sci ; 23(20)2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36293204

ABSTRACT

The endothelial cells of the blood-brain barrier adhere closely, which is provided by tight junctions (TJs). The aim of the study was to assess the damage to the endothelial TJs in pregnancy, complicated by fetal growth restriction (FGR) and circulatory centralization (brain-sparing effect, BS). The serum concentrations of NR1 subunit of the N-methyl-D-aspartate receptor (NR1), nucleoside diphosphate kinase A (NME1), S100 calcium-binding protein B (S100B), occludin (OCLN), claudin-5 (CLN5), and zonula occludens protein - 1 (zo-1), and the placental expressions of OCLN, claudin-4 (CLN4), CLN5, and zo-1 were assessed with ELISA. The significantly higher serum NME1 concentrations and the serum CLN5/zo-1 index were observed in FGR pregnancy with BS, as compared to the FGR group without BS. The FGR newborns with BS were about 20 times more likely to develop an intraventricular hemorrhage (IVH) than the FGR infants without BS. The cerebroplacental ratio (CPR) allowed to predict the IVH in growth-restricted fetuses. The significantly lower placental CLN4 expression was observed in the FGR group with BS and who postnatally developed an IVH, as compared to the growth-restricted infants with BS without IVH signs. Pregnancy complicated by FGR and BS is associated with the destabilization of the fetal blood-brain barrier. The IVH in newborns is reflected in the inhibition of the placental CLN4 expression, which may be a useful marker in the prediction of an IVH among growth-restricted fetuses.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Infant, Newborn , Female , Humans , Pregnancy , Claudin-5 , Occludin , Claudin-4 , Receptors, N-Methyl-D-Aspartate , Placenta , Brain , Fetal Growth Retardation , Fetus , Cerebral Hemorrhage , S100 Proteins , NM23 Nucleoside Diphosphate Kinases
15.
Br J Cancer ; 124(1): 161-165, 2021 01.
Article in English | MEDLINE | ID: mdl-33024267

ABSTRACT

NME1 is a metastasis-suppressor gene (MSG), capable of suppressing metastatic activity in cell lines of melanoma, breast carcinoma and other cancer origins without affecting their growth in culture or as primary tumours. Herein, we selectively ablated the tandemly arranged Nme1 and Nme2 genes to assess their individual impacts on metastatic activity in a mouse model (HGF:p16-/-) of ultraviolet radiation (UVR)-induced melanoma. Metastatic activity was strongly enhanced in both genders of Nme1- and Nme2-null mice, with stronger activity in females across all genotypes. The study ascribes MSG activity to Nme2 for the first time in an in vivo model of spontaneous cancer, as well as a novel metastasis-suppressor function to Nme1 in the specific context of UVR-induced melanoma.


Subject(s)
Genes, Tumor Suppressor , Melanoma/genetics , Melanoma/pathology , NM23 Nucleoside Diphosphate Kinases/genetics , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ultraviolet Rays/adverse effects
16.
Int J Mol Sci ; 22(7)2021 Apr 02.
Article in English | MEDLINE | ID: mdl-33918324

ABSTRACT

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.


Subject(s)
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
17.
Int J Mol Sci ; 22(22)2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34830075

ABSTRACT

This study aimed to investigate the effects of the human macrophage (MP) secretome in cellular xenograft rejection. The role of human nucleoside diphosphate kinase A (hNME1), from the secretome of MPs involved in the neuronal differentiation of miniature pig adipose tissue-derived mesenchymal stem cells (mp AD-MSCs), was evaluated by proteomic analysis. Herein, we first demonstrate that hNME1 strongly binds to porcine ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 1 (pST8SIA1), which is a ganglioside GD3 synthase. When hNME1 binds with pST8SIA1, it induces degradation of pST8SIA1 in mp AD-MSCs, thereby inhibiting the expression of ganglioside GD3 followed by decreased neuronal differentiation of mp AD-MSCs. Therefore, we produced nanobodies (NBs) named NB-hNME1 that bind to hNME1 specifically, and the inhibitory effect of NB-hNME1 was evaluated for blocking the binding between hNME1 and pST8SIA1. Consequently, NB-hNME1 effectively blocked the binding of hNME1 to pST8SIA1, thereby recovering the expression of ganglioside GD3 and neuronal differentiation of mp AD-MSCs. Our findings suggest that mp AD-MSCs could be a potential candidate for use as an additive, such as an immunosuppressant, in stem cell transplantation.


Subject(s)
Cell Differentiation/drug effects , Gangliosides/biosynthesis , Mesenchymal Stem Cells/enzymology , NM23 Nucleoside Diphosphate Kinases/pharmacology , Neurons/enzymology , Sialyltransferases/antagonists & inhibitors , Animals , Humans , Sialyltransferases/metabolism , Swine , Swine, Miniature
18.
Int J Mol Sci ; 22(2)2021 Jan 09.
Article in English | MEDLINE | ID: mdl-33435297

ABSTRACT

Metastasis accounts for over 90% of cancer-related deaths, yet the mechanisms guiding this process remain unclear. Secreted nucleoside diphosphate kinase A and B (NDPK) support breast cancer metastasis. Proteomic evidence confirms their presence in breast cancer-derived extracellular vesicles (EVs). We investigated the role of EV-associated NDPK in modulating the host microenvironment in favor of pre-metastatic niche formation. We measured NDPK expression and activity in EVs isolated from triple-negative breast cancer (MDA-MB-231) and non-tumorigenic mammary epithelial (HME1) cells using flow cytometry, western blot, and ATP assay. We evaluated the effects of EV-associated NDPK on endothelial cell migration, vascular remodeling, and metastasis. We further assessed MDA-MB-231 EV-induced proteomic changes in support of pre-metastatic lung niche formation. NDPK-B expression and phosphotransferase activity were enriched in MDA-MB-231 EVs that promote vascular endothelial cell migration and disrupt monolayer integrity. MDA-MB-231 EV-treated mice demonstrate pulmonary vascular leakage and enhanced experimental lung metastasis, whereas treatment with an NDPK inhibitor or a P2Y1 purinoreceptor antagonist blunts these effects. We identified perturbations to the purinergic signaling pathway in experimental lungs, lending evidence to support a role for EV-associated NDPK-B in lung pre-metastatic niche formation and metastatic outgrowth. These studies prompt further evaluation of NDPK-mediated EV signaling using targeted genetic silencing approaches.


Subject(s)
Extracellular Vesicles/pathology , Lung Neoplasms/secondary , Receptors, Purinergic P2Y/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/pathology , Animals , Cell Line , Cell Line, Tumor , Cell Movement , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Vesicles/metabolism , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, SCID , NM23 Nucleoside Diphosphate Kinases/metabolism , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment
19.
Int J Mol Sci ; 22(7)2021 Mar 27.
Article in English | MEDLINE | ID: mdl-33801585

ABSTRACT

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.


Subject(s)
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
20.
Hum Mol Genet ; 27(2): 373-384, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29177465

ABSTRACT

Attempts have been made to treat nonsense-associated genetic disorders by chemical agents and hence an improved mechanistic insight into the decoding of readthrough signals is essential for the identification and characterisation of factors for the treatment of these disorders. To identify either novel compounds or genes that modulate translation readthrough, we have employed dual reporter-based high-throughput screens that use enzymatic and fluorescence activities and screened bioactive National Institute of Neurological Disease Syndrome (NINDS) compounds (n = 1000) and siRNA (n = 288) libraries. Whilst siRNAs targeting kinases such as CSNK1G3 and NME3 negatively regulate readthrough, neither the bioactive NINDS compounds nor PTC124 promote readthrough. Of note, PTC124 has previously been shown to promote readthrough. Furthermore, the impacts of G418 on the components of eukaryotic selenocysteine incorporation machinery have also been investigated. The selenocysteine machinery decodes the stop codon UGA specifying selenocysteine in natural selenoprotein genes. We have found that the eukaryotic SelC gene promotes the selenocysteine insertion sequence (SECIS)-mediated readthrough but inhibits the readthrough activity induced by G418. We have previously reported that SECIS-mediated readthrough at UGA codons follows a non-processive mechanism. Here, we show that G418-mediated promotion of readthrough also occurs through a non-processive mechanism which competes with translation termination. Based on our observations, we suggest that proteins generated through a non-processive mechanism may be therapeutically beneficial for the resolution of nonsense-associated genetic disorders.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Small Molecule Libraries/pharmacology , Aminoglycosides/metabolism , Base Sequence , Casein Kinase Ialpha/metabolism , Codon, Nonsense , Codon, Terminator , Humans , NM23 Nucleoside Diphosphate Kinases/metabolism , Nucleic Acid Conformation , Oxadiazoles/pharmacology , Peptide Chain Termination, Translational , Protein Biosynthesis , Protein Synthesis Inhibitors , RNA, Messenger/genetics , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL