Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Sci Rep ; 11(1): 491, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436746

RESUMO

Tumor metastasis remains an obstacle in cancer treatment and is responsible for most cancer-related deaths. Nm23-H1 is one of the first metastasis suppressor proteins discovered with the ability to inhibit metastasis of many cancers including breast, colon, and liver cancer. Although loss of Nm23-H1 is observed in aggressive cancers and correlated with metastatic potential, little is known regarding the mechanisms that regulate its cellular level. Here, we examined the mechanisms that control Nm23-H1 expression in breast cancer cells. Initial studies in aggressive MDA-MB-231 cells (expressing low Nm23-H1) and less invasive MCF-7 cells (expressing high Nm23-H1) revealed that mRNA levels correlated with protein expression, suggesting that transcriptional mechanisms may control Nm23-H1 expression. Truncational analysis of the Nm23-H1 promoter revealed a proximal and minimal promoter that harbor putative binding sites for transcription factors including CTCF and EGR1. CTCF and EGR1 induced Nm23-H1 expression and reduced cell migration of MDA-MB-231 cells. Moreover, CTCF and EGR1 were recruited to the Nm23-H1 promoter in MCF-7 cells and their expression correlated with Nm23-H1 levels. This study indicates that loss of Nm23-H1 in aggressive breast cancer is apparently caused by downregulation of CTCF and EGR1, which potentially drive Nm23-H1 expression to promote a less invasive phenotype.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Fator de Ligação a CCCTC/metabolismo , Movimento Celular , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação Neoplásica da Expressão Gênica , Nucleosídeo NM23 Difosfato Quinases/genética , Apoptose , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Fator de Ligação a CCCTC/genética , Proliferação de Células , Proteína 1 de Resposta de Crescimento Precoce/genética , Feminino , Humanos , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Células Tumorais Cultivadas
2.
Molecules ; 25(21)2020 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-33172018

RESUMO

Adult neurogenesis is modulated by many Gi-coupled receptors but the precise mechanism remains elusive. A key step for maintaining the population of neural stem cells in the adult is asymmetric cell division (ACD), a process which entails the formation of two evolutionarily conserved protein complexes that establish the cell polarity and spindle orientation. Since ACD is extremely difficult to monitor in stratified tissues such as the vertebrate brain, we employed human neural progenitor cell lines to examine the regulation of the polarity and spindle orientation complexes during neuronal differentiation. Several components of the spindle orientation complex, but not those of the polarity complex, were upregulated upon differentiation of ENStem-A and ReNcell VM neural progenitor cells. Increased expression of nuclear mitotic apparatus (NuMA), Gαi subunit, and activators of G protein signaling (AGS3 and LGN) coincided with the appearance of a neuronal marker (ß-III tubulin) and the concomitant loss of neural progenitor cell markers (nestin and Sox-2). Co-immunoprecipitation assays demonstrated that both Gαi3 and NuMA were associated with AGS3 in differentiated ENStem-A cells. Interestingly, AGS3 appeared to preferentially interact with Gαi3 in ENStem-A cells, and this specificity for Gαi3 was recapitulated in co-immunoprecipitation experiments using HEK293 cells transiently overexpressing GST-tagged AGS3 and different Gαi subunits. Moreover, the binding of Gαi3 to AGS3 was suppressed by GTPγS and pertussis toxin. Disruption of AGS3/Gαi3 interaction by pertussis toxin indicates that AGS3 may recognize the same site on the Gα subunit as G protein-coupled receptors. Regulatory mechanisms controlling the formation of spindle orientation complex may provide novel means to manipulate ACD which in turn may have an impact on neurogenesis.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Diferenciação Celular , Linhagem Celular , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Inibidores de Dissociação do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Células-Tronco Neurais , Regulação para Cima
3.
Mol Cell Biochem ; 474(1-2): 95-112, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32705629

RESUMO

The Nm23 metastasis suppressor family is involved in a variety of physiological and pathological processes including cell proliferation, differentiation, tumorigenesis, and metastasis. Given that Nm23 proteins may function as hexamers composed of different members of the family, especially Nm23-H1 and H2 isoforms, it is pertinent to assess the importance of interface and surface residues in defining the functional characteristics of Nm23 proteins. Using molecular modeling to identify clusters of residues that may affect dimer formation and isoform specificity, mutants of Nm23-H1 were constructed and assayed for their ability to modulate cell migration. Mutations of dimer interface residues Gly22 and Lys39 affected the expression level of Nm23-H1, without altering the transcript level. The reduced protein expression was not due to increased protein degradation or altered subcellular distribution. Substitution of the surface residues of Nm23-H1 with Nm23-H2-specific Ser131 and/or Lys124/135 affected the electrophoretic mobility of the protein. Moreover, in cell migration assays, several mutants with altered surface residues exhibited impaired ability to suppress the mobility of MDA-MB-231 cells. Collectively, the study suggests that disrupting the dimer interface may affect the expression of Nm23-H1, while the residues at α-helix and ß-sheet on the surface of Nm23-H1 may contribute to its metastasis suppressive function.


Assuntos
Neoplasias da Mama/patologia , Movimento Celular , Proliferação de Células , Mutação , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Sequência de Aminoácidos , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Feminino , Humanos , Modelos Moleculares , Nucleosídeo NM23 Difosfato Quinases/química , Nucleosídeo NM23 Difosfato Quinases/genética , Metástase Neoplásica , Multimerização Proteica , Células Tumorais Cultivadas
4.
Biochem Pharmacol ; 177: 114020, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32389636

RESUMO

Melatonin MT1 and MT2 receptors represent attractive drug targets for the treatment of various disorders. However, the high conservation of the melatonin binding pocket has hindered the development of subtype-selective compounds. By leveraging on the recently resolved crystal structures of MT1 and MT2 receptors, this study aims to elucidate the structural basis of MT2-selectivity of a panel of isoquinolinone derivatives. Molecular modelling and ligand docking approaches were employed to predict residues involved in forming interactions with the MT2-selective isoquinolinones. Seven conserved residues (Asn175, His208, Trp264, Asn268, Gly271, Tyr294 and Tyr298) were selected as targets for site-directed mutagenesis. Ca2+ mobilization, cAMP inhibition, phosphorylation of extracellular signal-regulated kinase, and ligand binding assays were performed to functionally characterize the receptor mutants in transfected CHO cells. Unlike melatonin, isoquinolinones bearing a 3-methoxybenzyloxyl substituent were unaffected by alanine substitution at His208 of MT2. Although alanine substitutions at Tyr294 or Tyr298 reduced the potency of melatonin and some isoquinolinones on MT2, similar mutations on MT1 allowed five hitherto ineffective isoquinolinones to act as agonists. An isoquinolinone antagonist bearing a 4-methoxybenzyloxyl moiety turned into an agonist at MT2 mutants with alanine substitutions at His208, Tyr294 or Tyr298. A subset of residues is apparently involved in forming a hydrophobic binding cavity to confer selectivity upon the aromatic substituent of isoquinolinone compounds. Two conserved tyrosine residues on transmembrane helix 7 may confer ligand selectivity at MT1 and MT2 receptors, while a conserved histidine on transmembrane helix 5 is apparently involved in receptor activation.


Assuntos
Quinolonas/química , Quinolonas/farmacologia , Receptor MT2 de Melatonina/química , Receptor MT2 de Melatonina/metabolismo , Alanina , Substituição de Aminoácidos , Animais , Ligação Competitiva , Células CHO , Cálcio/metabolismo , Cricetulus , AMP Cíclico/metabolismo , Humanos , Melatonina/metabolismo , Simulação de Acoplamento Molecular , Mutação , Quinolonas/metabolismo , Receptor MT2 de Melatonina/genética
5.
Curr Med Chem ; 26(32): 5876-5899, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29788870

RESUMO

It is well-recognized that the majority of cancer-related deaths is attributed to metastasis, which can arise from virtually any type of tumor. Metastasis is a complex multistep process wherein cancer cells must break away from the primary tumor, intravasate into the circulatory or lymphatic systems, extravasate, proliferate and eventually colonize secondary sites. Since these molecular processes involve the coordinated actions of numerous proteins, targeted disruptions of key players along these pathways represent possible therapeutic interventions to impede metastasis formation and reduce cancer mortality. A diverse group of proteins with demonstrated ability to inhibit metastatic colonization have been identified and they are collectively known as metastasis suppressors. Given that the metastasis suppressors are often downregulated in tumors, drug-induced re-expression or upregulation of these proteins represents a promising approach to limit metastasis. Indeed, over 40 compounds are known to exhibit efficacy in upregulating the expression of metastasis suppressors via transcriptional or post-transcriptional mechanisms, and the most promising ones are being evaluated for their translational potentials. These small molecules range from natural products to drugs in clinical use and they apparently target different molecular pathways, reflecting the diverse nature of the metastasis suppressors. In this review, we provide an overview of the different classes of compounds known to possess the ability to upregulate one or more metastasis suppressors, with an emphasis on their mechanisms of action and therapeutic potentials.


Assuntos
Neoplasias/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas Supressoras de Tumor/antagonistas & inibidores , Regulação para Cima/efeitos dos fármacos , Animais , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Bibliotecas de Moléculas Pequenas/química , Proteínas Supressoras de Tumor/metabolismo
6.
Oncotarget ; 8(41): 69945-69960, 2017 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-29050254

RESUMO

The Nm23 metastasis suppressor family is involved in physiological and pathological processes including tumorigenesis and metastasis. Although the inverse correlation of Nm23 level with tumor metastasis potential has been widely observed, the mechanisms that regulate the expression of Nm23 remain poorly understood. Our previous studies have revealed that Nm23-H1/2 isoforms are upregulated by RGS19, a regulator of G protein signaling (RGS) protein which accelerates the termination of Gi signals. Here, we examined the ability of RGS19 to stimulate transcriptional regulation of Nm23 by screening a panel of luciferase reporter genes. Transient and stable overexpression of RGS19 upregulated the Nm23-H1/2 protein levels and activated several transcription factors including CREB, AP-1 and SRE in HEK293 cells. Interestingly, agents that increase the intracellular cAMP level and the phosphorylation of CREB (e.g., adrenergic receptor agonist, forskolin, and cAMP analogues) upregulated the expression of Nm23-H1/2 in HEK293 cells and several cancer cell lines including A549, HeLa, MDA-MB-231, and MDA-MB-435s cells. Conversely, inhibition of protein kinase A (PKA) by H-89 suppressed the phosphorylation of CREB and reduced the expression of Nm23-H1/2. Furthermore, activation of PKA attenuated cancer cell migration in wound healing and transwell assays. Collectively, these results revealed a PKA-dependent mechanism for controlling Nm23-H1/2 expression.

7.
Molecules ; 22(7)2017 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-28737723

RESUMO

Cognition and other higher brain functions are known to be intricately associated with the capacity of neural circuits to undergo structural reorganization. Structural remodelling of neural circuits, or structural plasticity, in the hippocampus plays a major role in learning and memory. Dynamic modifications of neuronal connectivity in the form of dendritic spine morphology alteration, as well as synapse formation and elimination, often result in the strengthening or weakening of specific neural circuits that determine synaptic plasticity. Changes in dendritic complexity and synapse number are mediated by cellular processes that are regulated by extracellular signals such as neurotransmitters and neurotrophic factors. As many neurotransmitters act on G protein-coupled receptors (GPCRs), it has become increasingly apparent that GPCRs can regulate structural plasticity through a myriad of G protein-dependent pathways and non-canonical signals. A thorough understanding of how GPCRs exert their regulatory influence on dendritic spine morphogenesis may provide new insights for treating cognitive impairment and decline in various age-related diseases. In this article, we review the evidence of GPCR-mediated regulation of structural plasticity, with a special emphasis on the involvement of common as well as distinct signalling pathways that are regulated by major neurotransmitters.


Assuntos
Cognição/fisiologia , Hipocampo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sinapses/metabolismo , Animais , Humanos , Plasticidade Neuronal/fisiologia
8.
Cell Signal ; 28(11): 1663-72, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27495875

RESUMO

Several RGS (regulator of G protein signaling) proteins are known to be upregulated in a variety of tumors but their roles in modulating tumorigenesis remain undefined. Since the expression of RGS20 is elevated in metastatic melanoma and breast tumors, we examined the effects of RGS20 overexpression and knockdown on the cell mobility and adhesive properties of different human cancer cell lines, including cervical cancer HeLa, breast adenocarcinoma MDA-MB-231, and non-small cell lung carcinoma H1299 and A549 cells. Expression of RGS20 enhanced cell aggregation, migration, invasion and adhesion as determined by hanging drop aggregation, wound healing, transwell chamber migration and invasion assays. Conversely, shRNA-mediated knockdown of endogenous RGS20 impaired these responses. In addition, RGS20 elevated the expression of vimentin (a mesenchymal cell marker) but down-regulated the expression of E-cadherin, two indicators commonly associated with metastasis. These results suggest that the expression of RGS20 may promote metastasis of tumor cells.


Assuntos
Movimento Celular , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas RGS/metabolismo , Animais , Antígenos CD , Caderinas/metabolismo , Adesão Celular , Agregação Celular , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Células NIH 3T3 , Invasividade Neoplásica , Vimentina/metabolismo
9.
J Mol Biol ; 428(19): 3869-84, 2016 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-27040396

RESUMO

CXCL8 (also known as interleukin-8 or IL-8) is a proinflammatory chemokine that not only modulates the inflammatory and immune responses, but whose upregulation is often associated with diseases including various types of cancer. Although numerous ligands for G protein-coupled receptors (GPCRs) have been shown to stimulate the production of CXCL8, the specificity of the G protein signal remains undefined. By expressing the constitutively active Gα subunits in HEK293 cells, CXCL8 production was herein demonstrated to be most effectively stimulated by Gαq family members, while those of Gαs and Gα12 elicited much weaker activities, and Gαi being totally ineffective. However, in cell lines such as HepG2, HeLa, and MCF-7 that endogenously express Gßγ-responsive phospholipase Cß isoforms (PLCß2/3), activation of the Gi-coupled α2-adrenoceptor significantly stimulated CXCL8 production. This Gi-induced CXCL8 production was apparently mediated via specific Gßγ dimers and required the presence of PLCß2/3. Co-activation of Gi-coupled α2-adrenoceptor and Gq-coupled bradykinin receptor resulted in a synergistic CXCL8 production, with Gßγ-responsive PLCß2/3, Src, ERK, and STAT3 serving as critical signaling intermediates. The treatment of HepG2 and B-10 endothelial cells with bradykinin stimulated CXCL8 production and cell proliferation. Interestingly, the latter response was driven by CXCL8 autocrine signaling because it was abolished by SB225002, an antagonist that prevents CXCL8 from binding to CXCR2. Collectively, our results provide a mechanistic basis for various G protein subfamilies to regulate the production of CXCL8, which may then lead to paracrine and/or autocrine signaling with major implications in both normal physiology and pathophysiological conditions.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Interleucina-8/biossíntese , Subunidades Proteicas/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Linhagem Celular , Proliferação de Células , Humanos
10.
Chem Biol Interact ; 259(Pt B): 295-300, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27019979

RESUMO

Flavonoids, a group of natural compounds mainly derived from plants, are known to possess osteogenic effects in bone cells. Here, we aimed to test if flavonoid could induce a cholinergic enzyme, acetylcholinesterase (AChE), as well as bone differentiation. In cultured rat osteoblasts, twenty flavonoids, deriving from Chinese herbs and having known induction of alkaline phosphatase (ALP1) expression, were tested for its induction activity on AChE expression. Eleven flavonoids showed the induction, and five of them had robust activation of AChE expression, including baicalin, calycosin, genistin, hyperin and pratensein: the induction of AChE included the levels of mRNA, protein and enzymatic activity. Moreover, the flavonoid-induced AChE expression in cultured osteoblast was in proline-rich membrane anchor (PRiMA)-linked tetrameric globular form (G4) only. In parallel, the expression of PRiMA was also induced by the application of flavonoids. The flavonoid-induced AChE in the cultures was not affected by estrogen receptor blocker, ICI 182,780. Taken together, the induction of PRiMA-linked AChE in osteoblast should be independent to classical estrogen signaling pathway.


Assuntos
Acetilcolinesterase/metabolismo , Flavonoides/farmacologia , Osteoblastos/enzimologia , Fosfatase Alcalina/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Humanos , Osteoblastos/efeitos dos fármacos , Multimerização Proteica , Ratos , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/metabolismo
11.
BMC Cancer ; 15: 775, 2015 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-26497576

RESUMO

BACKGROUND: G proteins are known to modulate various growth signals and are implicated in the regulation of tumorigenesis. The tumor suppressor Fhit is a newly identified interaction partner of Gq proteins that typically stimulate the phospholipase C pathway. Activated Gαq subunits have been shown to interact directly with Fhit, up-regulate Fhit expression and enhance its suppressive effect on cell growth and migration. Other signaling molecules may be involved in modulating Gαq/Fhit interaction. METHODS: To test the relationship of PLCß with the interaction between Gαq and Fhit, co-immunoprecipication assay was performed on HEK293 cells co-transfected with different combinations of Flag-Fhit, Gα16, Gα16QL, pcDNA3 vector, and PLCß isoforms. Possible associations of Fhit with other effectors of Gαq were also demonstrated by co-immunoprecipitation. The regions of Gαq for Fhit interaction and PLCß stimulation were further evaluated by inositol phosphates accumulation assay using a series of Gα16/z chimeras with discrete regions of Gα16 replaced by those of Gαz. RESULTS: PLCß1, 2 and 3 interacted with Fhit regardless of the expression of Gαq. Expression of PLCß increased the affinities of Fhit for both wild-type and activated Gαq. Swapping of the Fhit-interacting α2-ß4 region of Gαq with Gαi eliminated the association of Gαq with Fhit without affecting the ability of the mutant to stimulate PLCß. Other effectors of Gαq including RGS2 and p63RhoGEF were unable to interact with Fhit. CONCLUSIONS: PLCß may participate in the regulation of Fhit by Gq in a unique way. PLCß interacts with Fhit and increases the interaction between Gαq and Fhit. The Gαq/PLCß/Fhit complex formation points to a novel signaling pathway that may negatively regulate tumor cell growth.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfolipase C beta/metabolismo , Western Blotting , Células HEK293 , Humanos , Imunoprecipitação , Fosfolipase C beta/fisiologia , Ligação Proteica , Transdução de Sinais/fisiologia , Regulação para Cima
12.
BMC Struct Biol ; 15: 18, 2015 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-26377666

RESUMO

BACKGROUND: Stimulation of phospholipase Cß (PLCß) by the activated α-subunit of Gq (Gαq) constitutes a major signaling pathway for cellular regulation, and structural studies have recently revealed the molecular interactions between PLCß and Gαq. Yet, most of the PLCß-interacting residues identified on Gαq are not unique to members of the Gαq family. Molecular modeling predicts that the core PLCß-interacting residues located on the switch regions of Gαq are similarly positioned in Gαz which does not stimulate PLCß. Using wild-type and constitutively active chimeras constructed between Gαz and Gα14, a member of the Gαq family, we examined if the PLCß-interacting residues identified in Gαq are indeed essential. RESULTS: Four chimeras with the core PLCß-interacting residues composed of Gαz sequences were capable of binding PLCß2 and stimulating the formation of inositol trisphosphate. Surprisingly, all chimeras with a Gαz N-terminal half failed to functionally associate with PLCß2, despite the fact that many of them contained the core PLCß-interacting residues from Gα14. Further analyses revealed that the non-PLCß2 interacting chimeras were capable of interacting with other effector molecules such as adenylyl cyclase and tetratricopeptide repeat 1, indicating that they could adopt a GTP-bound active conformation. CONCLUSION: Collectively, our study suggests that the previously identified PLCß-interacting residues are insufficient to ensure productive interaction of Gα14 with PLCß, while an intact N-terminal half of Gα14 is apparently required for PLCß interaction.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/química , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Fosfolipase C beta/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Células HEK293 , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
13.
Cancer Lett ; 361(2): 207-17, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25748386

RESUMO

Nm23H1 and H2 are prototypical metastasis suppressors with diverse functions, but recent studies suggest that they may also regulate tumorigenesis. Here, we employed both cellular and in vivo assays to examine the effect of Nm23H1 and H2 on tumorigenesis induced by oncogenic Ras and/or p53 deficiency. Co-expression of Nm23H1 but not H2 in NIH3T3 cells effectively suppressed neoplastic transformation and tumorigenesis induced by the oncogenic H-Ras G12V mutant. Overexpression of Nm23H1 but not H2 also inhibited tumorigenesis by human cervical cancer HeLa cells with p53 deficiency. However, in human non-small-cell lung carcinoma H1299 cells harboring N-Ras Q61K oncogenic mutation and p53 deletion, overexpression of Nm23H1 did not affect tumorigenesis in nude mice assays, while overexpression of Nm23H2 enhanced tumor growth with elevated expression of the c-Myc proto-oncogene. Collectively, these results suggest that Nm23H1 and H2 have differential abilities to modulate tumorigenesis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Animais , Carcinogênese , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Transformação Celular Neoplásica/genética , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células NIH 3T3 , Nucleosídeo NM23 Difosfato Quinases/genética , Metástase Neoplásica , Proto-Oncogene Mas , Transfecção
14.
Naunyn Schmiedebergs Arch Pharmacol ; 388(2): 243-56, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25413836

RESUMO

Substantial effort has been directed at elucidating the functions of the products of the Nm23 tumor metastasis suppressor genes over the past two decades, with the ultimate goal of exploring their translational potentials in changing cancer patients' outcomes. Much attention has been focused on the better-known Nm23-H1, but despite having high sequence similarity, Nm23-H2 functions differently in many aspects. Besides acting as a metastasis suppressor, compelling data suggest that Nm23-H2 may modulate various tumor-associated biological events to enhance tumorigenesis in human solid tumors and hematological malignancies. Linkage to tumorigenesis may occur through the ability of Nm23-H2 to regulate transcription, cell proliferation, apoptosis, differentiation, and telomerase activity. In this review, we examine the linkages of Nm23-H2 to tumorigenesis in terms of its biochemical and structural properties and discuss its potential role in various tumor-associated events.


Assuntos
Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neoplasias/metabolismo , Animais , Diferenciação Celular , Genes myc , Humanos , Células-Tronco Multipotentes/fisiologia , Nucleosídeo NM23 Difosfato Quinases/química , Conformação Proteica , Telomerase/metabolismo
15.
Mol Cell Biochem ; 401(1-2): 27-38, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25480567

RESUMO

Activator of G protein signaling 3 (AGS3) is a guanine nucleotide dissociation inhibitor (GDI) which stabilizes the Gα(i/o) subunits as an AGS3/Gα(i/o)-GDP complex. It has recently been demonstrated in reconstitution experiments that the AGS3/Gα(i/o)-GDP complex may act as a substrate of resistance to inhibitors of cholinesterase 8A (Ric-8A), a guanine exchange factor (GEF) for heterotrimeric Gα proteins. Since the ability of Ric-8A to activate Gα(i/o) subunits that are bound to AGS3 in a cellular environment has not been confirmed, we thus examined the effect of Ric-8A on cAMP accumulation in HEK293 cells expressing different forms of AGS3 and Gα(i3). Co-immunoprecipitation assays indicate that full-length AGS3 and its N- and C-terminal truncated mutants can interact with Ric-8A in HEK293 cells. Yeast two-hybrid assay further confirmed that Ric-8A can directly bind to AGS3S, a short form of AGS3 which is endogenously expressed in heart. However, Ric-8A failed to facilitate Gα(i)-induced suppression of adenylyl cyclase, suggesting that it may not serve as a GEF for AGS3/Gα(i/o)-GDP complex in a cellular environment.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Guanosina Difosfato/metabolismo , Adenilil Ciclases/metabolismo , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Técnicas do Sistema de Duplo-Híbrido
16.
PLoS One ; 9(12): e113638, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25479338

RESUMO

A series of substituted isoquinolinones were synthesized and their binding affinities and functional activities towards human melatonin MT1 and MT2 receptors were evaluated. Structure-activity relationship analysis revealed that substituted isoquinolinones bearing a 3-methoxybenzyloxyl group at C5, C6 or C7 position respectively (C5>C6>C7 in terms of their potency) conferred effective binding and selectivity toward the MT2 receptor, with 15b as the most potent compound. Most of the tested compounds were MT2-selective agonists as revealed in receptor-mediated cAMP inhibition, intracellular Ca2+ mobilization and phosphorylation of extracellular signal-regulated protein kinases. Intriguingly, compounds 7e and 7f bearing a 4-methoxybenzyloxyl group or 4-methylbenzyloxyl at C6 behaved as weak MT2-selective antagonists. These results suggest that substituted isoquinolinones represent a novel family of MT2-selective melatonin ligands. The position of the substituted benzyloxyl group, and the substituents on the benzyl ring appeared to dictate the functional characteristics of these compounds.


Assuntos
Isoquinolinas/química , Melatonina/metabolismo , Receptor MT1 de Melatonina/metabolismo , Receptor MT2 de Melatonina/metabolismo , Relação Estrutura-Atividade , Animais , Células CHO , Cricetulus , Desenho de Fármacos , Humanos , Isoquinolinas/síntese química , Ligantes , Melatonina/química , Fosforilação , Ligação Proteica , Receptor MT1 de Melatonina/antagonistas & inibidores , Receptor MT1 de Melatonina/química , Receptor MT2 de Melatonina/antagonistas & inibidores , Receptor MT2 de Melatonina/química
17.
Neuropharmacology ; 81: 267-73, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23823979

RESUMO

Angiotensin-(1-12) [Ang-(1-12)], a newer member of angiotensin peptides, is proposed to be converted enzymatically to angiotensin I (Ang I) and to angiotensin II (Ang II); the latter being the bioactive peptide. We studied the Ang-(1-12) and Ang II responses in COS-7 cells or CHO cells transfected with 5 µg AT1R by monitoring [Ca(2+)]i using the Fluo-4. Ang II (1 pM-1 µM) and Ang-(1-12) (5 pM-5 µM) increased [Ca(2+)]i with an EC50 of 0.19 nM and 24 nM in COS-7 cells; and 0.65 nM and 28.7 nM in CHO cells. The AT1R antagonist losartan (1 nM-10 µM) suppressed [Ca(2+)]i induced by Ang-(1-12) and Ang II. In CHO cells transfected with 5 µg AT2R, Ang II (1 pM-1 µM) increased [Ca(2+)]i, with an EC50 of 9.68 nM; whereas, Ang-(1-12) (5 pM-5 µM) failed to elicit a significant change in [Ca(2+)]i. In CHO cells transfected with AT1R, Ang-(1-12) stimulated ERK phosphorylation with a potency 300-fold less than that of Ang II. To evaluate the activity of Ang-(1-12) on native AT1R, whole cell patch recordings were made from neurons in the rat hypothalamic slices. Ang II or Ang-(1-12) ejected by pressure from a micropipette elicited a membrane depolarization; the latter was blocked by losartan (10 µM), and not affected by the AT2R antagonist PD123319 (10 µM), nor by the angiotensin converting enzyme inhibitor captopril (10 µM). Our result shows that Ang-(1-12) may produce its biological activity by acting directly on AT1R, albeit at a concentration higher than that of Ang II.


Assuntos
Angiotensinogênio/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptor Tipo 1 de Angiotensina/metabolismo , Angiotensina I/farmacologia , Angiotensina II/farmacologia , Animais , Animais Recém-Nascidos , Células CHO , Cricetulus , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética , Transfecção
18.
Cell Signal ; 25(12): 2440-52, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23993961

RESUMO

The tumor suppressor Fhit protein is defective or absent in many tumor cells due to methylation, mutation or deletion of the FHIT gene. Despite numerous attempts to unravel the functions of Fhit, the mechanisms by which the function and expression of Fhit are regulated remain poorly understood. We have recently shown that activated Gαq subunits interact directly with Fhit and enhance its inhibitory effect on cell growth. Here we investigated the regulation of Fhit expression by Gq. Our results showed that Fhit was up-regulated specifically by activating Gα subunits of the Gq subfamily but not by those of the other G protein subfamilies. This up-regulation effect was mediated by a PKC/MEK pathway independent of Src-mediated Fhit Tyr(114) phosphorylation. We further demonstrated that elevated Fhit expression was due to the specific regulation of Fhit protein synthesis in the ribosome by activated Gαq, where the regulations of cap-dependent protein synthesis were apparently not required. Moreover, we showed that activated Gαq could increase cell-cell adhesion through Fhit. These findings provide a possible handle to modulate the level of the Fhit tumor suppressor by manipulating the activity of Gq-coupled receptors.


Assuntos
Hidrolases Anidrido Ácido/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/genética , Regulação para Cima , Hidrolases Anidrido Ácido/metabolismo , Linhagem Celular , Humanos , MAP Quinase Quinase Quinases/metabolismo , Proteínas de Neoplasias/metabolismo , Biossíntese de Proteínas , Proteína Quinase C/metabolismo , Transdução de Sinais
19.
Cell Commun Signal ; 11: 59, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23947369

RESUMO

BACKGROUND: The FHIT tumor suppressor gene is arguably the most commonly altered gene in cancer since it is inactivated in about 60% of human tumors. The Fhit protein is a member of the ubiquitous histidine triad proteins which hydrolyze dinucleoside polyphosphates such as Ap3A. Despite the fact that Fhit functions as a tumor suppressor, the pathway through which Fhit inhibits growth of cancer cells remains largely unknown. Phosphorylation by Src tyrosine kinases provides a linkage between Fhit and growth factor signaling. Since many G proteins can regulate cell proliferation through multiple signaling components including Src, we explored the relationship between Gα subunits and Fhit. RESULTS: Several members of the Gαq subfamily (Gα16, Gα14, and Gαq) were found to co-immunoprecipitate with Fhit in their GTP-bound active state in HEK293 cells. The binding of activated Gαq members to Fhit appeared to be direct and was detectable in native DLD-1 colon carcinoma cells. The use of Gα16/z chimeras further enabled the mapping of the Fhit-interacting domain to the α2-ß4 region of Gα16. However, Gαq/Fhit did not affect either Ap3A binding and hydrolysis by Fhit, or the ability of Gαq/16 to regulate downstream effectors including phospholipase Cß, Ras, ERK, STAT3, and IKK. Functional mutants of Fhit including the H96D, Y114F, L25W and L25W/I10W showed comparable abilities to associate with Gαq. Despite the lack of functional regulation of Gq signaling by Fhit, stimulation of Gq-coupled receptors in HEK293 and H1299 cells stably overexpressing Fhit led to reduced cell proliferation, as opposed to an enhanced cell proliferation typically seen with parental cells. CONCLUSIONS: Activated Gαq members interact with Fhit through their α2-ß4 region which may result in enhancement of the growth inhibitory effect of Fhit, thus providing a possible avenue for G protein-coupled receptors to modulate tumor suppression.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Hidrolases Anidrido Ácido/genética , Cálcio/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Fosfatos de Dinucleosídeos/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Humanos , Fosfatos de Inositol/metabolismo , Mutação , Proteínas de Neoplasias/genética , Fosforilação , RNA Interferente Pequeno/genética , Proteínas Supressoras de Tumor/genética , Quinases da Família src/metabolismo
20.
Cancer Lett ; 339(1): 33-41, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23911936

RESUMO

Regulator of G protein signaling 19 (RGS19) has recently been shown to inhibit Ras activation by upregulating the tumor metastasis suppressor Nm23. Here, we have examined the effect of RGS19 on Ras-induced oncogenesis. Coexpression of RGS19, but not RGS20, in NIH3T3 cells effectively suppressed neoplastic transformation and tumorigenesis induced by the oncogenic Ras(GV) mutant. In non-small cell lung carcinoma H1299 cells that harbor Ras mutations, shRNA-mediated knockdown of RGS19 facilitated tumorigenesis with the early appearance of large tumors in nude mice assays. Collectively, these results suggest that expression of RGS19 can suppress the oncogenic actions of Ras.


Assuntos
Transformação Celular Neoplásica/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas RGS/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Camundongos , Camundongos Nus , Células NIH 3T3 , Nucleosídeo NM23 Difosfato Quinases/genética , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas RGS/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA