Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Biol Chem ; 400(6): 765-776, 2019 05 27.
Article in English | MEDLINE | ID: mdl-30763256

ABSTRACT

The Nm23 gene has been acknowledged to play a crucial role in lung cancer metastasis inhibitory cascades controlled by multiple factors. Low expression or allelic deletion of nm23-H1 is strongly linked to widespread metastasis and poor differentiation of non-small cell lung cancer (NSCLC). In this study, nm23-H1 was down regulated in epithelial-mesenchymal transition (EMT) and stemness enhancement under cobalt chloride (CoCl2)-induced hypoxia in NSCLC cells. Moreover, knocking down of nm23-H1 by shRNA apparently promoted hypoxia induced EMT and stemness, which was entirely suppressed via over expression of nm23-H1. Mechanistically, the Wnt/ß-catenin signaling pathway was found to participate in the nm23-H1-mediated process. Besides, XAV939 prohibited cell EMT and stemness which could be impaired by knocking down of nm23-H1, while stable transfection of nm23-H1 attenuated hypoxia phonotype induced by lithium chloride (LiCl). Generally, our experiment provided evidence that nm23-H1 can reverse hypoxia induced EMT and stemness through the inhibition of the Wnt/ß-catenin pathway, which may furnish a deeper perspective into the better treatment or prognosis for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Hypoxia , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/pathology , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/physiology , Neoplastic Stem Cells/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Humans , Lung Neoplasms/metabolism , Wnt Signaling Pathway
2.
BMC Cancer ; 18(1): 710, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29970055

ABSTRACT

BACKGROUND: Although originally identified as a putative metastasis suppressor, increasing studies have confirmed a possible role for Nm23-H1 in DNA repair, through the base excision repair and nucleotide excision repair pathways. In this study, we explored whether Nm23-H1 was also involved in double-strand break repair (DSBR). METHODS AND RESULTS: We constructed a stable A549-shNm23-H1 cell line with doxycycline-regulated expression of Nm23-H1, and a A549-nNm23-H1 cell line that over expressed a nucleus-localized version of Nm23-H1. Results from both lines confirmed that Nm23-H1 participated in the repair of double-strand breaks induced by X-rays, using Comet and γ-H2AX foci assays. Subsequent studies showed that Nm23-H1 activated the phosphorylation of checkpoint-related proteins including ATM serine/threonine kinase (on S1981), tumor protein p53 (on S15), and checkpoint kinase 2 (Chk2) (on T68). We also detected interactions between Nm23-H1 and the MRE11-RAD50-NBS1 (MRN) complex, as well as Ku80. Moreover, NBS1 and Ku80 levels were comparably higher in Nm23-H1 overexpressing cells than in control cells (t = 14.462, p < 0.001 and t = 5.347, p = 0.006, respectively). As Ku80 is the keystone of the non-homologous end joining (NHEJ) pathway, we speculate that Nm23-H1 promotes DSBR through NHEJ. CONCLUSIONS: The results indicate that Nm23-H1 participates in multiple steps of DSBR.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Lung Neoplasms/radiotherapy , NM23 Nucleoside Diphosphate Kinases/physiology , A549 Cells , DNA End-Joining Repair , Humans , Phosphorylation , Recombinational DNA Repair , X-Rays
3.
Lab Invest ; 98(5): 571-581, 2018 05.
Article in English | MEDLINE | ID: mdl-29449633

ABSTRACT

The understanding of protein-protein interactions is crucial in order to generate a second level of functional genomic analysis in human disease. Within a cellular microenvironment, protein-protein interactions generate new functions that can be defined by single or multiple modes of protein interactions. We outline here the clinical importance of targeting of the Nme-1 (NDPK-A)-Prune-1 protein complex in cancer, where an imbalance in the formation of this protein-protein complex can result in inhibition of tumor progression. We discuss here recent functional data using a small synthetic competitive cell-permeable peptide (CPP) that has shown therapeutic efficacy for impairing formation of the Nme-1-Prune-1 protein complex in mouse preclinical xenograft tumor models (e.g., breast, prostate, colon, and neuroblastoma). We thus believe that further discoveries in the near future related to the identification of new protein-protein interactions will have great impact on the development of new therapeutic strategies against various cancers.


Subject(s)
Carrier Proteins/physiology , Cell-Penetrating Peptides/pharmacology , NM23 Nucleoside Diphosphate Kinases/physiology , Neoplasms/drug therapy , Carrier Proteins/chemistry , Cell-Penetrating Peptides/therapeutic use , Humans , NM23 Nucleoside Diphosphate Kinases/chemistry , Neoplasms/pathology , Phosphoric Monoester Hydrolases , Phosphorylation , Tumor Suppressor Protein p53/physiology
4.
Lab Invest ; 98(5): 602-608, 2018 05.
Article in English | MEDLINE | ID: mdl-29339833

ABSTRACT

Nucleoside diphosphate kinases (NDPKs/NDK/NME) are a multifunctional class of proteins conserved throughout evolution. Whilst many of the functions of NDPKs have been identified as intracellular, extracellular eukaryotic and prokaryotic NDPK proteins are also detected in multiple systems and have been implicated in both normal physiology and disease. This review provides an overview of where the field stands on our developing understanding of how NDPK proteins get out of cells, the physiological role of extracellular NDPKs, and how extracellular NDPKs may signal to cells. We will also discuss some of the unanswered questions, the 'known-unknowns' that particularly warrant further investigation.


Subject(s)
NM23 Nucleoside Diphosphate Kinases/physiology , Animals , Hematologic Neoplasms/etiology , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Receptors, Cell Surface/physiology
5.
Exp Dermatol ; 24(6): 455-61, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25808322

ABSTRACT

Expression of the metastasis suppressor NME1 in melanoma is associated with reduced cellular motility and invasion in vitro and metastasis in vivo, but the underlying molecular mechanisms are not completely understood. Herein, we report a novel mechanism through which NME1 controls melanoma cell morphology via upregulation of the extracellular matrix (ECM) protein fibronectin. Expression of NME1 strongly suppressed cell motility in melanoma cell lines 1205LU and M14. The resulting sedentary phenotype was associated with a more flattened appearance and marked increases in actin stress fibre and focal adhesion formation. NME1-induced focal adhesions were colocalized with dense deposits of fibronectin, which were absent or minimal in the corresponding NME1-deficient parental lines. NME1 was a strong inducer of fibronectin mRNA and protein expression, shown with reciprocal approaches of forced NME1 expression and shRNA-mediated knock-down. Increased synthesis and ECM deposition of fibronectin was necessary for NME1-induced cell spreading, as knock-down of fibronectin opposed the effects of NME1 on cell morphology. Fibronectin knock-down also reversed the ability of NME1 to promote aggregation when cells were plated on a non-adherent substratum. Similarly, inhibiting activation of the fibronectin receptor integrin α4ß1 with an anti-α4 antibody reversed the motility-suppressing effect of NME1. A positive correlation was observed between NME1 and fibronectin mRNA in clinical biopsies of normal skin, benign nevi and primary melanomas, but not in metastatic forms, suggesting the NME1/fibronectin axis represents a barrier to melanoma progression. In summary, these findings indicate fibronectin is an important effector of the motility-suppressing function of NME1 in melanoma cells.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Fibronectins/physiology , Melanoma/pathology , NM23 Nucleoside Diphosphate Kinases/physiology , Skin Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Extracellular Matrix/pathology , Extracellular Matrix/physiology , Fibronectins/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , In Vitro Techniques , Melanoma/physiopathology , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasm Metastasis/pathology , Neoplasm Metastasis/physiopathology , RNA, Messenger/genetics , RNA, Messenger/physiology , Signal Transduction/physiology , Skin Neoplasms/physiopathology
6.
PLoS One ; 10(2): e0115968, 2015.
Article in English | MEDLINE | ID: mdl-25700270

ABSTRACT

Gastric cancer is one of the most common malignancies and has a high rate of metastasis. We hypothesize that NME2 (Nucleoside Diphosphate Kinase 2), which has previously been considered as an anti-metastatic gene, plays a role in the invasiveness of gastric cancer cells. Using a tissue chip technology and immunohistochemistry, we demonstrated that NME2 expression was associated with levels of differentiation of gastric cancer cells and their metastasis into the lymph nodes. When the NME2 gene product was over-expressed by ;in vitro stable transfection, cells from BGC823 and MKN45 gastric cancer cell lines had reduced rates of proliferation, migration, and invasion through the collagen matrix, suggesting an inhibitory activity of NME2 in the propagation and invasion of gastric cancer. NME2 could, therefore, severe as a risk marker for gastric cancer invasiveness and a potential new target for gene therapy to enhance or induce NME2 expression.


Subject(s)
Adenocarcinoma/enzymology , NM23 Nucleoside Diphosphate Kinases/physiology , Stomach Neoplasms/enzymology , Adenocarcinoma/secondary , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Stomach Neoplasms/pathology
7.
Hum Reprod ; 28(10): 2822-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23856325

ABSTRACT

STUDY QUESTION: Is Nometastatic gene 23-H1 (NME1, also known as nm23-H1) involved in regulating the biological behavior of endometrial stromal cells (ESCs), and does it participate in the pathogenesis of endometriosis? SUMMARY ANSWER: NME1 suppression induces ESC dysfunction in the endometriotic milieu. WHAT IS KNOWN ALREADY: NME1 is a wide-spectrum tumor metastasis suppressor gene that plays an important role in suppressing the invasion and metastasis of tumor cells. STUDY DESIGN, SIZE, DURATION: An in vitro investigation of the effect of NME1 on the proliferation, adhesion and invasion of eutopic ESCs from patients with endometriosis. PARTICIPANTS/MATERIALS, SETTING, METHODS: Primary ESCs were prepared from 12 samples of ectopic endometrial tissue (6 peritoneal and 6 ovarian lesions), 18 samples of eutopic endometrial tissues (16 from women with ovarian and 2 from women with pelvic endometriomas) and 12 samples of normal endometrial tissue from women without endometriosis, after the tissues had been analyzed histologically. The growth, invasiveness and adhesion of ESCs were studied by the 5-bromo-2'-deoxyuridine cell proliferation assay and by the Matrigel invasion and adhesion assay. Additionally, the effects of NME1 on the activation or expression of related regulatory proteins were investigated by in-cell Western and flow cytometry assays. MAIN RESULTS AND THE ROLE OF CHANCE: Expression of NME1 in ESCs derived from eutopic or ectopic endometrium from women with endometriosis is lower than in ESCs from women without endometriosis. Estrogen could down-regulate NME1 expression in ESCs. Silencing NME1 in ESCs promoted the expression of proliferating cell nuclear antigen (PCNA), the anti-apoptotic molecule, survivin, and the adhesion-related molecules, integrin ß1 and integrin ανß3. Silencing NME1 also stimulated ESC proliferation, adhesion and invasion but these effects were inhibited by MAPK/Erk and/or Akt blockers. LIMITATIONS, REASONS FOR CAUTION: Further studies are needed to examine the regulatory mechanism of estrogen on NME1 expression of ESCs. WIDER IMPLICATIONS OF THE FINDINGS: Abnormally low expression of NME1 in ESCs may be involved in the pathogenesis of endometriosis by up-regulating growth, adhesion and invasion of ESCs via activating the Akt and MAPK/Erk1/2 signal pathways. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by National Natural Science Foundation of China (NSFC) (31270969, 31101064 and 81270677) and Program for ZhouXue of Fudan University. None of the authors has any conflict of interest to declare.


Subject(s)
Endometriosis/pathology , Endometrium/pathology , NM23 Nucleoside Diphosphate Kinases/physiology , Oncogene Protein v-akt/metabolism , Cell Adhesion , Cell Proliferation , Down-Regulation/drug effects , Endometriosis/metabolism , Endometrium/metabolism , Estrogens/pharmacology , Female , Flow Cytometry , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , MAP Kinase Signaling System , NM23 Nucleoside Diphosphate Kinases/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Survivin
8.
Biochem Biophys Res Commun ; 434(3): 541-6, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23583378

ABSTRACT

Persistent STAT3 activation is a critical event in tumorigenesis and metastatic progression. Recent studies have found higher levels of STAT3 in metastatic tissues than in primary tumor tissues. We speculated that such increased STAT3 activity might be attributed to a loss of function or reduction in expression of metastasis inhibitory protein during cancer progression, and we therefore examined the role of tumor metastasis-suppressor nm23-H1 in the activation of STAT3 in the A549 lung cancer cell line. We found that IL-6-dependent induction of tyrosine phosphorylation and activation of STAT3 were influenced by nm23-H1 inhibition. IL-6-induced STAT3(Tyr705) phosphorylation was significantly enhanced in A549 cells transfected with siRNA specific for nm23-H1, and the effect of nm23-H1 depletion on IL-6-induced STAT3(Tyr705) phosphorylation was reversed by ectopic expression of shRNA-resistant nm23-H1 protein. Moreover, STAT3 directly bound to the STAT3 binding site on the nm23-H1 promoter and activated its expression. Thus, we have identified a new feedback mechanism that might provide insight into an in-built metastasis-suppression function in tumor cells and which could be a logical new target for treatment of early metastatic disease.


Subject(s)
NM23 Nucleoside Diphosphate Kinases/physiology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Base Sequence , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Primers , Feedback , Humans , Mutagenesis, Site-Directed , Phosphorylation , Real-Time Polymerase Chain Reaction
9.
Clin Exp Metastasis ; 30(1): 25-36, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22699362

ABSTRACT

Cutaneous malignant melanoma is the most lethal form of skin cancer, with 5-year survival rates of <5 % for patients presenting with metastatic disease. Mechanisms underlying metastatic spread of UVR-induced melanoma are not well understood, in part due to a paucity of animal models that accurately recapitulate the disease in its advanced forms. We have employed a transgenic mouse strain harboring a tandem deletion of the nm23-m1 and nm23-m2 genes to assess the combined contribution of these genes to suppression of melanoma metastasis. Crossing of the nm23-h1/nm23-h2 knockout in hemizygous-null form ([m1m2](+/-)) to a transgenic mouse strain (hepatocyte growth factor/scatter factor-overexpressing, or HGF(+) strain) vulnerable to poorly-metastatic, UVR-induced melanomas resulted in UVR-induced melanomas with high metastatic potential. Metastasis to draining lymph nodes was seen in almost all cases of back skin melanomas, while aggressive metastasis to lung, thoracic cavity, liver and bone also occurred. Interestingly, no differences were observed in the invasive characteristics of primary melanomas of HGF(+) and HGF(+) × [m1m2](+/-) strains, with both exhibiting invasion into the dermis and subcutis, indicating factors other than simple invasive activity were responsible for metastasis of HGF(+) × [m1m2](+/-) melanomas. Stable cell lines were established from the primary and metastatic melanoma lesions from these mice, with HGF(+) × [m1m2](+/-) lines exhibiting increased single cell migration and genomic instability. These studies demonstrate for the first time in vivo a potent metastasis suppressor activity of NM23 in UVR-induced melanoma, and have provided new tools for identifying molecular mechanisms that underlie melanoma metastasis.


Subject(s)
Disease Models, Animal , Genomic Instability , Melanoma/etiology , NM23 Nucleoside Diphosphate Kinases/physiology , Skin Neoplasms/etiology , Ultraviolet Rays/adverse effects , Animals , Cell Movement , DNA Damage/genetics , DNA Repair/genetics , Female , Hepatocyte Growth Factor/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Male , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Skin Neoplasms/secondary , Tumor Cells, Cultured , Wound Healing
10.
J Biol Chem ; 287(40): 33389-400, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22869372

ABSTRACT

Cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel mutations cause cystic fibrosis lung disease. A better understanding of CFTR regulatory mechanisms could suggest new therapeutic strategies. AMP-activated protein kinase (AMPK) binds to and phosphorylates CFTR, attenuating PKA-activated CFTR gating. However, the requirement for AMPK binding to CFTR and the potential role of other proteins in this regulation are unclear. We report that nucleoside diphosphate kinase A (NDPK-A) interacts with both AMPK and CFTR in overlay blots of airway epithelial cell lysates. Binding studies in Xenopus oocytes and transfected HEK-293 cells revealed that a CFTR peptide fragment that binds AMPK (CFTR-1420-57) disrupted the AMPK-CFTR interaction. Introduction of CFTR-1420-57 into human bronchial Calu-3 cells enhanced forskolin-stimulated whole cell conductance in patch clamp measurements. Similarly, injection of CFTR-1420-57 into Xenopus oocytes blocked the inhibition of cAMP-stimulated CFTR conductance by AMPK in two-electrode voltage clamp studies. AMPK also inhibited CFTR conductance with co-expression of WT NDPK-A in two-electrode voltage clamp studies, but co-expression of a catalytically inactive H118F mutant or various Ser-120 NDPK-A mutants prevented this inhibition. In vitro phosphorylation of WT NDPK-A was enhanced by purified active AMPK, but phosphorylation was prevented in H118F and phosphomimic Ser-120 NDPK-A mutants. AMPK does not appear to phosphorylate NDPK-A directly but rather promotes an NDPK-A autophosphorylation event that involves His-118 and Ser-120. Taken together, these results suggest that NDPK-A exists in a functional cellular complex with AMPK and CFTR in airway epithelia, and NDPK-A catalytic function is required for the AMPK-dependent regulation of CFTR.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gene Expression Regulation , NM23 Nucleoside Diphosphate Kinases/physiology , Animals , Bronchi/cytology , Catalysis , Cell Line , Epithelial Cells/cytology , HEK293 Cells , Humans , Ions/chemistry , Models, Biological , Mutation , Oocytes/cytology , Patch-Clamp Techniques , Phosphorylation , Protein Binding , Xenopus laevis
11.
Cancer Metastasis Rev ; 31(3-4): 593-603, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22706779

ABSTRACT

Metastatic disease is the major cause of death among cancer patients. A class of genes, named metastasis suppressors, has been described to specifically regulate the metastatic process. The metastasis suppressor genes are downregulated in the metastatic lesion compared to the primary tumor. In this review, we describe the body of research surrounding the first metastasis suppressor identified, Nm23. Nm23 overexpression in aggressive cancer cell lines reduced their metastatic potential in vivo with no significant reduction in primary tumor size. A complex mechanism of anti-metastatic action is unfolding involving several known Nm23 enzymatic activities (nucleotide diphosphate kinase, histidine kinase, and 3'-5' exonuclease), protein-protein interactions, and downstream gene regulation properties. Translational approaches involving Nm23 have progressed to the clinic. The upregulation of Nm23 expression by medroxyprogesterone acetate has been tested in a phase II trial. Other approaches with significant preclinical success include gene therapy using traditional or nanoparticle delivery, and cell permeable Nm23 protein. Recently, based on the inverse correlation of Nm23 and LPA1 expression, a LPA1 inhibitor has been shown to both inhibit metastasis and induce metastatic dormancy.


Subject(s)
Genes, Tumor Suppressor , NM23 Nucleoside Diphosphate Kinases/physiology , Neoplasm Metastasis/prevention & control , Animals , Humans , Mice , Mice, Transgenic , NM23 Nucleoside Diphosphate Kinases/genetics , Receptors, Lysophosphatidic Acid/physiology , Transfection
12.
Cancer Res ; 72(1): 133-43, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22080566

ABSTRACT

Reduced expression of the metastasis suppressor NM23-H1 is associated with aggressive forms of multiple cancers. Here, we establish that NM23-H1 (termed H1 isoform in human, M1 in mouse) and two of its attendant enzymatic activities, the 3'-5' exonuclease and nucleoside diphosphate kinase, are novel participants in the cellular response to UV radiation (UVR)-induced DNA damage. NM23-H1 deficiency compromised the kinetics of repair for total DNA polymerase-blocking lesions and nucleotide excision repair of (6-4) photoproducts in vitro. Kinase activity of NM23-H1 was critical for rapid repair of both polychromatic UVB/UVA-induced (290-400 nm) and UVC-induced (254 nm) DNA damage, whereas its 3'-5' exonuclease activity was dominant in the suppression of UVR-induced mutagenesis. Consistent with its role in DNA repair, NM23-H1 rapidly translocated to sites of UVR-induced (6-4) photoproduct DNA damage in the nucleus. In addition, transgenic mice hemizygous-null for nm23-m1 and nm23-m2 exhibited UVR-induced melanoma and follicular infundibular cyst formation, and tumor-associated melanocytes displayed invasion into adjacent dermis, consistent with loss of invasion-suppressing activity of NM23 in vivo. Taken together, our data show a critical role for NM23 isoforms in limiting mutagenesis and suppressing UVR-induced melanomagenesis.


Subject(s)
DNA Damage , Melanoma, Experimental/prevention & control , NM23 Nucleoside Diphosphate Kinases/physiology , Neoplasms, Radiation-Induced/prevention & control , Ultraviolet Rays , Animals , Cell Line, Tumor , Hypoxanthine Phosphoribosyltransferase/genetics , Melanoma, Experimental/etiology , Mice , Mice, Inbred C57BL , Mutation , NM23 Nucleoside Diphosphate Kinases/genetics
13.
Front Biosci (Elite Ed) ; 4(5): 1888-97, 2012 01 01.
Article in English | MEDLINE | ID: mdl-22202005

ABSTRACT

Metastases, rather than the primary tumors from which these malignant growths are spawned, are culpable for greater than 90 % of human cancer-associated mortality. Metastases arise through the completion of a series of cell-biological events - collectively termed "the invasion-metastasis cascade" - which involve the dissemination of tumor cells to distant organ sites and their subsequent adaptation to these foreign microenvironments. Importantly, a number of endogenous mechanisms exist that serve to prevent metastatic progression. These safeguards must be overcome by incipient metastatic tumor cells in order for them to generate detectable metastases. Here, I highlight four endogenous mechanisms that protect against the development of metastatic disease in breast carcinomas. I discuss how the expression of these genes are dampened during malignant progression, the downstream responses they orchestrate, and clinical opportunities to therapeutically target these mechanisms. Indeed, one potentially effective strategy for the remediation of metastatic disease involves the reactivation of endogenous anti-metastasis mechanisms. Therefore, knowledge regarding endogenous anti-metastasis mechanisms may both further our comprehension of the basic etiology of metastasis and also guide the treatment of human tumors.


Subject(s)
Neoplasm Metastasis , Animals , Cadherins/physiology , Humans , MicroRNAs/physiology , NM23 Nucleoside Diphosphate Kinases/physiology , Neoplasm Invasiveness
14.
Naunyn Schmiedebergs Arch Pharmacol ; 384(4-5): 351-62, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21713383

ABSTRACT

Nm23-H1, also known as NDPK-A, was the first of a class of metastasis suppressor genes to be identified. Overexpression of Nm23-H1 in metastatic cell lines (melanoma, breast carcinoma, prostate, colon, hepatocellular, and oral squamous cell carcinoma) reduced cell motility in in vitro assays and metastatic potential in xenograft models, without a significant effect on primary tumor size. The mechanism of Nm23-H1 suppression of metastasis, however, is incompletely understood. Nm23-H1 has been reported to bind proteins, including those in small G-protein complexes, transcriptional complexes, the Map kinase, the TGF-ß signaling pathways and the cytoskeleton. Evidence supporting these associations is presented together with evidence of resultant biochemical and phenotypic consequences of association. Cumulatively, the data suggest that part of the anti-metastatic function of Nm23-H1 lies in pathways that it interrupts via binding and inactivation of proteins.


Subject(s)
NM23 Nucleoside Diphosphate Kinases/physiology , Neoplasm Metastasis , Animals , Gene Expression Regulation, Neoplastic , Humans , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasm Metastasis/genetics , Neoplasm Metastasis/prevention & control , Protein Binding , Signal Transduction
16.
Naunyn Schmiedebergs Arch Pharmacol ; 384(4-5): 397-406, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21556888

ABSTRACT

Non-metastatic 23 [NM23/nucleoside diphosphate kinases (NDPK)] genes are the first discovered metastasis suppressor genes. More than two decades of research has demonstrated their roles in a variety of biological processes with NME1 and NME2 being most studied in the context of metastasis suppression. Although NME1 and NME2 share >85% homology at amino acid level, they show redundant as well as unique molecular functions. Phenotypic analyses of knockout (KO) mice for NM23 members (NDPK-A, B) and compound KO (A as well as B) showed requirement of both proteins in hematopoiesis suggesting shared functions in development disease. Several reviews have discussed NME1, however the role of NME2 appears to be relatively less understood in the context of metastasis suppression. Here, we focus on NME2 and by meta-analysis of gene expression from multiple tumor types, and survey of in vivo and vitro studies, suggest the possibility that NME2 may be one of the key factors in metastasis. This along with the relevance of normal physiological functions of NME2 in the context of metastasis is discussed. We further examined the genetic and epigenetic features of NME2 and NME1 gene promoters and found aspects of transcription control that could be unique to NME2/NME1. Findings on signaling pathways and small molecules which regulate the expression of NME2 that could be therapeutically important are also discussed.


Subject(s)
NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/physiology , Neoplasms/pathology , Animals , Binding Sites , Cell Line, Tumor , Gene Expression , Humans , Neoplasm Metastasis , Neoplasms/enzymology , Neoplasms/genetics , Transcription Factors/metabolism
17.
Naunyn Schmiedebergs Arch Pharmacol ; 384(4-5): 421-31, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21562815

ABSTRACT

The human NME gene family (also known as NM23) comprises ten genes that are involved in diverse physiological and pathological processes including proliferation, differentiation, development, ciliary functions, and metastasis. For the moment, only the NME1, NME2, and NME7 genes have been inactivated in transgenic knockout mice, as well as a double NME1-NME2 gene knockout. Mice lacking NME1 or NME2 grow to adulthood without health problems, although NME1 (-/-) mice have modest growth retardation. Double knockout NME1 (-/-)-NME2 (-/-) mice, by contrast, are highly hypotrophic and die at birth from profound anemia due to impaired erythroblast development. Evidence for a metastasis suppressor function of NME1 in vivo comes from crossing NME1 (-/-) mice with mice prone to develop hepatocellular carcinoma; the double transgenic mice present a higher incidence of lung metastases. Silencing of NME1 by siRNA interference has confirmed this function by conferring a "metastatic phenotype" on non-invasive human epithelial cancer cell lines. This function is specific to NME1 and is not observed when the NME2 is silenced. The data indicate that NME1 loss is causally involved at the early stages of the metastatic cascade. NME2 (-/-) mice and NME2 silencing experiments reveal a specific role of NME2 in activation of heterotrimeric G proteins and of KCa3.1 channel in T cells, pointing to a role of NME2 as a histidine phosphotransferase. Regarding NME7, consistent with its expression in axonemal structures, NME7 (-/-) mice present lesions similar to primary ciliary dyskinesia. This review summarizes the recent data obtained by knockout and silencing of NME/NM23 genes that provide mechanistic insights into their respective roles in physiology and pathology.


Subject(s)
Gene Silencing , Models, Genetic , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/physiology , Animals , Cell Line, Tumor , Humans , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Protein Subunits
18.
Naunyn Schmiedebergs Arch Pharmacol ; 384(4-5): 407-19, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21541759

ABSTRACT

NME/NDPK family proteins are involved in the control of intracellular nucleotide homeostasis as well as in both physiological and pathological cellular processes, such as proliferation, differentiation, development, apoptosis, and metastasis dissemination, through mechanisms still largely unknown. One family member, NME1/NDPK-A, is a metastasis suppressor, yet the primary physiological functions of this protein are still missing. The purpose of this study was to identify new NME1/NDPK-A-dependent biological functions and pathways regulated by this gene in the liver. We analyzed the proteomes of wild-type and transgenic NME1-null mouse livers by combining two-dimensional gel electrophoresis and mass spectrometry (matrix-assisted laser desorption/ionization time of flight and liquid chromatography-tandem mass spectrometry). We found that the levels of three proteins, namely, phenylalanine hydroxylase, annexin IV, and elongation factor 1 Bα (EF-1Bα), were strongly reduced in the cytosolic fraction of NME1(-/-) mouse livers when compared to the wild type. This was confirmed by immunoblotting analysis. No concomitant reduction in the corresponding messenger RNAs or of total protein level was observed, however, suggesting that NME1 controls annexin IV and EF-1Bα amounts by post-translational mechanisms. NME1 deletion induced a change in the subcellular location of annexin IV in hepatocytes resulting in enrichment of this protein at the plasma membrane. We also observed a redistribution of EF-1Bα in NME1(-/-) hepatocytes to an intracytoplasmic compartment that colocalized with a marker of the reticulum endoplasmic. Finally, we found reduced expression of annexin IV coincident with decreased NME1 expression in a panel of different carcinoma cell lines. Taken together, our data suggest for the first time that NME1 might regulate the subcellular trafficking of annexin IV and EF-1Bα. The potential role of these proteins in metastatic dissemination is discussed.


Subject(s)
Annexin A4/metabolism , Liver/enzymology , NM23 Nucleoside Diphosphate Kinases/physiology , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Protein Processing, Post-Translational , Animals , Annexin A4/genetics , Blotting, Western , Cell Line, Tumor , Cytosol/enzymology , Cytosol/metabolism , Electrophoresis, Gel, Two-Dimensional , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Humans , Liver/metabolism , Male , Mice , Mice, Knockout , NM23 Nucleoside Diphosphate Kinases/genetics , Protein Transport , Proteomics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
19.
Clin Cancer Res ; 17(9): 2893-9, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21478336

ABSTRACT

PURPOSE: We examined whether nm23-H1 is a prognostic factor of peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS). EXPERIMENTAL DESIGN: We studied 102 consecutive, untreated PTCL-NOS patients from 1998 to 2008. The expression of nm23-H1 and TIA-1 was studied by immunohistochemistry. RESULTS: nm23-H1 was positive in 44.1% and TIA-1 in 78.4% of the PTCL-NOS patients. nm23-H1 expression was not correlated with age, performance status (PS), lactate dehydrogenase (LDH) level, or stage but was significantly correlated with the prognostic index for T-cell lymphoma. The serum nm23-H1 level was 43.44 ng/mL in the cytoplasmic nm23-H1 strongly positive, 24.32 ng/mL in the cytoplasmic nm23-H1 moderately positive, and 13.64 ng/mL in the cytoplasmic nm23-H1-negative patients. The nm23-H1-positive group had significantly shorter overall survival (OS). TIA-1 had no prognostic impact on 5-year OS rates. OS was significantly shorter in patients with the following clinicopathologic features: age 60 or more years, PS of 2 to 4, LDH level greater than normal, bone marrow involvement, or nm23-H1-positive lymphoma. Multivariate analysis confirmed nm23-H1 expression to be an independent prognostic factor. CONCLUSIONS: The nm23-H1 protein may be an important prognostic factor in PTCL-NOS. Because our results suggested that nm23-HI is produced by lymphoma cells, we expect to see the development of new treatments targeting nm23 overexpression.


Subject(s)
Lymphoma, T-Cell, Peripheral/diagnosis , NM23 Nucleoside Diphosphate Kinases/metabolism , Adult , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , Cytoplasm/metabolism , Cytoplasm/pathology , Humans , Lymphoma, T-Cell, Peripheral/blood , Lymphoma, T-Cell, Peripheral/metabolism , Lymphoma, T-Cell, Peripheral/mortality , Middle Aged , NM23 Nucleoside Diphosphate Kinases/analysis , NM23 Nucleoside Diphosphate Kinases/blood , NM23 Nucleoside Diphosphate Kinases/physiology , Prognosis , Survival Analysis
20.
Naunyn Schmiedebergs Arch Pharmacol ; 384(4-5): 461-72, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21409430

ABSTRACT

Caveolae are flask-shaped invaginations in the plasma membrane that serve to compartmentalize and organize signal transduction processes, including signals mediated by G protein-coupled receptors and heterotrimeric G proteins. Herein we report evidence for a close association of the nucleoside diphosphate kinase B (NDPK B) and caveolin proteins which is required for G protein scaffolding and caveolae formation. A concomitant loss of the proteins NDPK B, caveolin isoforms 1 (Cav1) and 3, and heterotrimeric G proteins occurred when one of these proteins was specifically depleted in zebrafish embryos. Co-immunoprecipitation of Cav1 with the G protein Gß-subunit and NDPK B from zebrafish lysates corroborated the direct association of these proteins. Similarly, in embryonic fibroblasts from the respective knockout (KO) mice, the membrane content of the Cav1, Gß, and NDPK B was found to be mutually dependent on one another. A redistribution of Cav1 and Gß from the caveolae containing fractions of lower density to other membrane compartments with higher density could be detected by means of density gradient fractionation of membranes derived from NDPK A/B KO mouse embryonic fibroblasts (MEFs) and after shRNA-mediated NDPK B knockdown in H10 cardiomyocytes. This redistribution could be visualized by confocal microscopy analysis showing a decrease in the plasma membrane bound Cav1 in NDPK A/B KO cells and vice versa and a decrease in the plasma membrane pool of NDPK B in Cav1 KO cells. Consequently, ultrastructural analysis revealed a reduction of surface caveolae in the NDPK A/B KO cells. To prove that the disturbed subcellular localization of Cav1 in NDPK A/B KO MEFs as well as NDPK B in Cav1 KO MEFs is a result of the loss of NDPK B and Cav1, respectively, we performed rescue experiments. The adenoviral re-expression of NDPK B in NDPK A/B KO MEFs rescued the protein content and the plasma membrane localization of Cav1. The expression of an EGFP-Cav1 fusion protein in Cav1-KO cells induced a restoration of NDPK B expression levels and its appearance at the plasma membrane. We conclude from these findings that NDPK B, heterotrimeric G proteins, and caveolins are mutually dependent on each other for stabile localization and caveolae formation at the plasma membrane. The data point to a disturbed transport of caveolin/G protein/NDPK B complexes from intracellular membrane compartments if one of the components is missing.


Subject(s)
Caveolae , Caveolins/metabolism , GTP-Binding Proteins/metabolism , NM23 Nucleoside Diphosphate Kinases/physiology , Protein Multimerization , Animals , Caveolae/enzymology , Caveolae/metabolism , Caveolae/ultrastructure , Caveolins/genetics , Cell Line , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Immunoblotting , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Microscopy, Fluorescence , NM23 Nucleoside Diphosphate Kinases/genetics , Rats , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...