Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 201
Filtrar
1.
Sci Rep ; 14(1): 14693, 2024 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926545

RESUMO

Our research aimed to elucidate the mechanism by which aurintricarboxylic acid (ATA) inhibits plasma membrane Ca2+-ATPase (PMCA), a crucial enzyme responsible for calcium transport. Given the pivotal role of PMCA in cellular calcium homeostasis, understanding how it is inhibited by ATA holds significant implications for potentially regulating physiopathological cellular processes in which this pump is involved. Our experimental findings revealed that ATA employs multiple modes of action to inhibit PMCA activity, which are influenced by ATP but also by the presence of calcium and magnesium ions. Specifically, magnesium appears to enhance this inhibitory effect. Our experimental and in-silico results suggest that, unlike those reported in other proteins, ATA complexed with magnesium (ATA·Mg) is the molecule that inhibits PMCA. In summary, our study presents a novel perspective and establishes a solid foundation for future research efforts aimed at the development of new pharmacological molecules both for PMCA and other proteins.


Assuntos
Ácido Aurintricarboxílico , Cálcio , Magnésio , ATPases Transportadoras de Cálcio da Membrana Plasmática , Magnésio/metabolismo , Magnésio/farmacologia , Ácido Aurintricarboxílico/farmacologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/antagonistas & inibidores , Cálcio/metabolismo , Trifosfato de Adenosina/metabolismo , Membrana Celular/metabolismo , Membrana Celular/efeitos dos fármacos , Animais , Humanos
2.
Free Radic Res ; 55(6): 698-713, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33788639

RESUMO

Endothelial cell (EC) dysfunction is the underlying cause for the development of several pathologies, and the interdependency between the pancreatic ß-cells and ECs has been established in the pathophysiology of diabetes. ECs release several factors that govern the expression of genes involved in the proliferation, physiology, and survival of the ß-cells. Of the known factors that collapse this intricately balanced system, endothelial dysfunction is the crucial condition that manifests as the causative factor for micro and macrovascular diseases. Our earlier studies demonstrated that activation of nuclear factor erythroid-related factor (Nrf2) renders protection to the ECs experiencing ER stress. In this study, using a co-culture system, the crosstalk between pancreatic cells under ER stress and ECs and the effect of a novel Nrf2 activator Rosolic Acid (RA), on the crosstalk was investigated. ECs pre-treated with different concentrations RA and co-cultured with thapsigargin-induced ER stressed pancreatic ß-cells showed increased levels of Nrf2 and its downstream targets such as heme oxygenase-1 (HO-1) and NADPH-quinone oxidoreductase-1 (NQO-1), and reduction of ER stress evinced by the decreased levels of glucose-regulated protein (GRP) 78 and C/ERB homologous protein (CHOP). The sensitization of ECs using RA, offered protection to pancreatic cells against ER stress as displayed by increased intracellular insulin and upregulated expression of cell survival and proliferative genes BCl2 and PDX-1. In addition, RA treatment resulted in elevated levels of various angiogenic factors, while inflammatory (TNF-α and IL-1ß) and apoptotic markers (CXCL10 and CCL2) decreased. RA treatment normalized the levels of 115 proteins of the 277, which were differentially regulated as revealed by proteomic studies of ER stressed pancreatic ß-cells in co-culture conditions. These findings clearly indicate the role of small molecule activators of Nrf2 not only in restoring the functioning of pancreatic cells but also in increasing the cell mass. Further, the study impinges on the strategies that can be developed to balance the pancreatic microenvironment, leading to the restoration of ß-cell mass and their normophysiology in diabetic patients.


Assuntos
Ácido Aurintricarboxílico/análogos & derivados , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células Endoteliais/metabolismo , Pâncreas/efeitos dos fármacos , Animais , Ácido Aurintricarboxílico/farmacologia , Ácido Aurintricarboxílico/uso terapêutico , Humanos , Camundongos , Pâncreas/patologia , Microambiente Tumoral
3.
Osteoarthritis Cartilage ; 28(5): 685-697, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31981738

RESUMO

OBJECTIVE: Innate immune response and particularly terminal complement complex (TCC) deposition are thought to be involved in the pathogenesis of posttraumatic osteoarthritis. However, the possible role of TCC in regulated cell death as well as chondrocyte hypertrophy and senescence has not been unraveled so far and was first addressed using an ex vivo human cartilage trauma-model. DESIGN: Cartilage explants were subjected to blunt impact (0.59 J) and exposed to human serum (HS) and cartilage homogenate (HG) with or without different potential therapeutics: RIPK1-inhibitor Necrostatin-1 (Nec), caspase-inhibitor zVAD, antioxidant N-acetyl cysteine (NAC) and TCC-inhibitors aurintricarboxylic acid (ATA) and clusterin (CLU). Cell death and hypertrophy/senescence-associated markers were evaluated on mRNA and protein level. RESULTS: Addition of HS resulted in significantly enhanced TCC deposition on chondrocytes and decrease of cell viability after trauma. This effect was potentiated by HG and was associated with expression of RIPK3, MLKL and CASP8. Cytotoxicity of HS could be prevented by heat-inactivation or specific inhibitors, whereby combination of Nec and zVAD as well as ATA exhibited highest cell protection. Moreover, HS+HG exposition enhanced the gene expression of CXCL1, IL-8, RUNX2 and VEGFA as well as secretion of IL-6 after cartilage trauma. CONCLUSIONS: Our findings imply crucial involvement of the complement system and primarily TCC in regulated cell death and phenotypic changes of chondrocytes after cartilage trauma. Inhibition of TCC formation or downstream signaling largely modified serum-induced pathophysiologic effects and might therefore represent a therapeutic target to maintain the survival and chondrogenic character of cartilage cells.


Assuntos
Morte Celular/genética , Condrócitos/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/genética , Hipertrofia/genética , Osteoartrite/genética , Ferimentos não Penetrantes/genética , Acetilcisteína/farmacologia , Idoso , Idoso de 80 Anos ou mais , Ácido Aurintricarboxílico/farmacologia , Cartilagem Articular/citologia , Morte Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Clusterina/farmacologia , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/efeitos dos fármacos , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Sequestradores de Radicais Livres/farmacologia , Humanos , Imidazóis/farmacologia , Imunidade Inata/genética , Indóis/farmacologia , Masculino , Pessoa de Meia-Idade , Oligopeptídeos/farmacologia , Osteoartrite/etiologia , Osteoartrite/metabolismo , Osteoartrite/patologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ferimentos não Penetrantes/complicações , Ferimentos não Penetrantes/metabolismo
4.
Biol Blood Marrow Transplant ; 25(6): 1062-1074, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30668984

RESUMO

Despite recent advances in therapy, allogeneic hematopoietic stem cell transplantation (HSCT) remains the only curative option for a range of high-risk hematologic malignancies. However, acute graft-versus-host disease (aGVHD) continues to limit the long-term success of HSCT, and new therapies are still needed. We previously demonstrated that aGVHD depends on the ability of donor conventional T cells (Tcons) to express the lymph node trafficking receptor, CC-Chemokine Receptor 7 (CCR7). Consequently, we examined the ability of cosalane, a recently identified CCR7 small-molecule antagonist, to attenuate aGVHD in mouse HSCT model systems. Here we show that the systemic administration of cosalane to transplant recipients after allogeneic HSCT did not prevent aGVHD. However, we were able to significantly reduce aGVHD by briefly incubating donor Tcons with cosalane ex vivo before transplantation. Cosalane did not result in Tcon toxicity and did not affect their activation or expansion. Instead, cosalane prevented donor Tcon trafficking into host secondary lymphoid tissues very early after transplantation and limited their subsequent accumulation within the liver and colon. Cosalane did not appear to impair the intrinsic ability of donor Tcons to produce inflammatory cytokines. Furthermore, cosalane-treated Tcons retained their graft-versus-leukemia (GVL) potential and rejected a murine P815 inoculum after transplantation. Collectively, our data indicate that a brief application of cosalane to donor Tcons before HSCT significantly reduces aGVHD in relevant preclinical models while generally sparing beneficial GVL effects, and that cosalane might represent a viable new approach for aGVHD prophylaxis.


Assuntos
Ácido Aurintricarboxílico/análogos & derivados , Doença Enxerto-Hospedeiro/genética , Efeito Enxerto vs Leucemia/genética , Transplante de Células-Tronco Hematopoéticas/métodos , Receptores CCR7/metabolismo , Linfócitos T/metabolismo , Condicionamento Pré-Transplante/métodos , Doença Aguda , Animais , Ácido Aurintricarboxílico/farmacologia , Ácido Aurintricarboxílico/uso terapêutico , Humanos , Camundongos , Doadores de Tecidos
5.
J Med Chem ; 62(3): 1677-1683, 2019 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-30562026

RESUMO

We identify three submicromolar inhibitors with new chemical scaffolds for cystathionine γ-lyase (CSE) by a tandem-well-based high-throughput assay. NSC4056, the most potent inhibitor with an IC50 of 0.6 µM, which is also known as aurintricarboxylic acid, selectively binds to Arg and Tyr residues of CSE active site and preferably inhibits the CSE activity in cells rather than cystathionine ß-synthase (CBS), the other H2S-generating enzyme. Moreover, NSC4056 effectively rescues hypotension in hemorrhagic shock rats.


Assuntos
Ácido Aurintricarboxílico/farmacologia , Cistationina gama-Liase/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Animais , Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/metabolismo , Domínio Catalítico/efeitos dos fármacos , Cistationina gama-Liase/química , Cistationina gama-Liase/metabolismo , Descoberta de Drogas , Inibidores Enzimáticos/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Simulação de Acoplamento Molecular , Estrutura Molecular , Nitroquinolinas/farmacologia , Ligação Proteica , Células RAW 264.7 , Ratos Sprague-Dawley , Relação Estrutura-Atividade
6.
Virus Res ; 256: 209-218, 2018 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-29958924

RESUMO

Alphavirus non-structural protein, nsP1 has a distinct molecular mechanism of capping the viral RNAs than the conventional capping mechanism of host. Thus, alphavirus capping enzyme nsP1 is a potential drug target. nsP1 catalyzes the methylation of guanosine triphosphate (GTP) by transferring the methyl group from S-adenosylmethionine (SAM) to a GTP molecule at its N7 position with the help of nsP1 methyltransferase (MTase) followed by guanylylation (GT) reaction which involves the formation of m7GMP-nsP1 covalent complex by nsP1 guanylyltransferase (GTase). In subsequent reactions, m7GMP moiety is added to the 5' end of the viral ppRNA by nsP1 GTase resulting in the formation of cap0 structure. In the present study, chikungunya virus (CHIKV) nsP1 MTase and GT reactions were confirmed by an indirect non-radioactive colorimetric assay and western blot assay using an antibody specific for the m7G cap, respectively. The purified recombinant CHIKV nsP1 has been used for the development of a rapid and sensitive non-radioactive enzyme linked immunosorbent assay (ELISA) to identify the inhibitors of CHIKV nsP1. The MTase reaction is followed by GT reaction and resulted in m7GMP-nsP1 covalent complex formation. The developed ELISA nsP1 assay measures this m7GMP-nsP1 complex by utilizing anti-m7G cap monoclonal antibody. The mutation of a conserved residue Asp63 to Ala revealed its role in nsP1 enzyme reaction. Inductively coupled plasma mass spectroscopy (ICP-MS) was used to determine the presence of magnesium ions (Mg2+) in the purified nsP1 protein. The divalent metal ion selectivity and investigation show preference for Mg2+ ion by CHIKV nsP1. Additionally, using the developed ELISA nsP1 assay, the inhibitory effects of sinefungin, aurintricarboxylic acid (ATA) and ribavirin were determined and the IC50 values were estimated to be 2.69 µM, 5.72 µM and 1.18 mM, respectively.


Assuntos
Antivirais/farmacologia , Vírus Chikungunya/enzimologia , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática/métodos , Metiltransferases/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores , Adenosina/análogos & derivados , Adenosina/farmacologia , Ácido Aurintricarboxílico/farmacologia , Cátions Bivalentes/metabolismo , Coenzimas/metabolismo , Concentração Inibidora 50 , Magnésio/metabolismo , Ribavirina/farmacologia
7.
J Cell Mol Med ; 22(2): 861-872, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29193716

RESUMO

Cardiovascular disease is the world's leading cause of morbidity and mortality, with high blood pressure (BP) contributing to increased severity and number of adverse outcomes. Plasma membrane calcium ATPase 4 (PMCA4) has been previously shown to modulate systemic BP. However, published data are conflicting, with both overexpression and inhibition of PMCA4 in vivo shown to increase arterial contractility. Hence, our objective was to determine the role of PMCA4 in the regulation of BP and to further understand how PMCA4 functionally regulates BP using a novel specific inhibitor to PMCA4, aurintricarboxylic acid (ATA). Our approach assessed conscious BP and contractility of resistance arteries from PMCA4 global knockout (PMCA4KO) mice compared to wild-type animals. Global ablation of PMCA4 had no significant effect on BP, arterial structure or isolated arterial contractility. ATA treatment significantly reduced BP and arterial contractility in wild-type mice but had no significant effect in PMCA4KO mice. The effect of ATAin vivo and ex vivo was abolished by the neuronal nitric oxide synthase (nNOS) inhibitor Vinyl-l-NIO. Thus, this highlights differences in the effects of PMCA4 ablation and acute inhibition on the vasculature. Importantly, for doses here used, we show the vascular effects of ATA to be specific for PMCA4 and that ATA may be a further experimental tool for elucidating the role of PMCA4.


Assuntos
Pressão Sanguínea , Artérias Mesentéricas/fisiopatologia , Óxido Nítrico Sintase Tipo I/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/antagonistas & inibidores , Animais , Ácido Aurintricarboxílico/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Cálcio/metabolismo , Estado de Consciência , Técnicas In Vitro , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Camundongos Knockout , Modelos Biológicos , Peptídeos/farmacologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo
8.
Anticancer Res ; 37(9): 4799-4806, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28870898

RESUMO

BACKGROUND: Rapidly-dividing cancer cells have higher requirement for iron compared to non-transformed cells, making iron chelating a potential anticancer strategy. In the present study we compared the anticancer activity of uncommon iron chelator aurintricarboxylic acid (ATA) with the known deferoxamine (DFO). MATERIALS AND METHODS: We investigated the impact of ATA and DFO on the viability and proliferation of MCF-7 cancer cells. Moreover we performed enzymatic activity assays and computational analysis of the ATA and DFO effects on pro-oncogenic phosphatases PTP1B and SHP2. RESULTS: ATA and DFO decrease the viability and proliferation of breast cancer cells, but only ATA considerably reduces the activity of PTP1B and SHP2 phosphatases. Our studies indicated that ATA strongly inactivates and binds in the PTP1B and SHP2 active site, interacting with arginine residue essential for enzyme activity. CONCLUSION: We confirmed that iron chelating can be considered as a potential strategy for the adjunctive treatment of breast cancer.


Assuntos
Ácido Aurintricarboxílico/farmacologia , Neoplasias da Mama/enzimologia , Desferroxamina/farmacologia , Quelantes de Ferro/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Ácido Aurintricarboxílico/química , Sítios de Ligação , Neoplasias da Mama/patologia , Catalase/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Desferroxamina/química , Feminino , Humanos , Concentração Inibidora 50 , Células MCF-7 , Simulação de Acoplamento Molecular , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo
9.
J Mol Cell Cardiol ; 109: 38-47, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28684310

RESUMO

AIMS: Ischaemic cardiovascular disease is a major cause of morbidity and mortality worldwide. Despite promising results from pre-clinical animal models, VEGF-based strategies for therapeutic angiogenesis have yet to achieve successful reperfusion of ischaemic tissues in patients. Failure to restore efficient VEGF activity in the ischaemic organ remains a major problem in current pro-angiogenic therapeutic approaches. Plasma membrane calcium ATPase 4 (PMCA4) negatively regulates VEGF-activated angiogenesis via inhibition of the calcineurin/NFAT signalling pathway. PMCA4 activity is inhibited by the small molecule aurintricarboxylic acid (ATA). We hypothesize that inhibition of PMCA4 with ATA might enhance VEGF-induced angiogenesis. METHODS AND RESULTS: We show that inhibition of PMCA4 with ATA in endothelial cells triggers a marked increase in VEGF-activated calcineurin/NFAT signalling that translates into a strong increase in endothelial cell motility and blood vessel formation. ATA enhances VEGF-induced calcineurin signalling by disrupting the interaction between PMCA4 and calcineurin at the endothelial-cell membrane. ATA concentrations at the nanomolar range, that efficiently inhibit PMCA4, had no deleterious effect on endothelial-cell viability or zebrafish embryonic development. However, high ATA concentrations at the micromolar level impaired endothelial cell viability and tubular morphogenesis, and were associated with toxicity in zebrafish embryos. In mice undergoing experimentally-induced hindlimb ischaemia, ATA treatment significantly increased the reperfusion of post-ischaemic limbs. CONCLUSIONS: Our study provides evidence for the therapeutic potential of targeting PMCA4 to improve VEGF-based pro-angiogenic interventions. This goal will require the development of refined, highly selective versions of ATA, or the identification of novel PMCA4 inhibitors.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Membrana Celular/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Ácido Aurintricarboxílico/farmacologia , ATPases Transportadoras de Cálcio/genética , Membrana Celular/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
10.
Oncotarget ; 8(7): 12234-12246, 2017 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-28103571

RESUMO

The survival of patients diagnosed with glioblastoma (GBM), the most deadly form of brain cancer, is compromised by the proclivity for local invasion into the surrounding normal brain, which prevents complete surgical resection and contributes to therapeutic resistance. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor (TNF) superfamily, can stimulate glioma cell invasion and survival via binding to fibroblast growth factor-inducible 14 (Fn14) and subsequent activation of the transcription factor NF-κB. To discover small molecule inhibitors that disrupt the TWEAK-Fn14 signaling axis, we utilized a cell-based drug-screening assay using HEK293 cells engineered to express both Fn14 and a NF-κB-driven firefly luciferase reporter protein. Focusing on the LOPAC1280 library of 1280 pharmacologically active compounds, we identified aurintricarboxylic acid (ATA) as an agent that suppressed TWEAK-Fn14-NF-κB dependent signaling, but not TNFα-TNFR-NF-κB driven signaling. We demonstrated that ATA repressed TWEAK-induced glioma cell chemotactic migration and invasion via inhibition of Rac1 activation but had no effect on cell viability or Fn14 expression. In addition, ATA treatment enhanced glioma cell sensitivity to both the chemotherapeutic agent temozolomide (TMZ) and radiation-induced cell death. In summary, this work reports a repurposed use of a small molecule inhibitor that targets the TWEAK-Fn14 signaling axis, which could potentially be developed as a new therapeutic agent for treatment of GBM patients.


Assuntos
Ácido Aurintricarboxílico/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Necrose Tumoral/metabolismo , Animais , Antineoplásicos Alquilantes/farmacologia , Ácido Aurintricarboxílico/química , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Citocina TWEAK , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Sinergismo Farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Células HEK293 , Humanos , Estimativa de Kaplan-Meier , Camundongos Nus , Estrutura Molecular , Interferência de RNA , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais/genética , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Receptor de TWEAK , Temozolomida , Fatores de Necrose Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
11.
Nat Commun ; 7: 11074, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-27020607

RESUMO

The heart responds to pathological overload through myocyte hypertrophy. Here we show that this response is regulated by cardiac fibroblasts via a paracrine mechanism involving plasma membrane calcium ATPase 4 (PMCA4). Pmca4 deletion in mice, both systemically and specifically in fibroblasts, reduces the hypertrophic response to pressure overload; however, knocking out Pmca4 specifically in cardiomyocytes does not produce this effect. Mechanistically, cardiac fibroblasts lacking PMCA4 produce higher levels of secreted frizzled related protein 2 (sFRP2), which inhibits the hypertrophic response in neighbouring cardiomyocytes. Furthermore, we show that treatment with the PMCA4 inhibitor aurintricarboxylic acid (ATA) inhibits and reverses cardiac hypertrophy induced by pressure overload in mice. Our results reveal that PMCA4 regulates the development of cardiac hypertrophy and provide proof of principle for a therapeutic approach to treat this condition.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Cardiomegalia/patologia , Membrana Celular/enzimologia , Fibroblastos/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Transdução de Sinais , Animais , Animais Recém-Nascidos , Aorta/patologia , Ácido Aurintricarboxílico/farmacologia , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/deficiência , Cardiomegalia/complicações , Membrana Celular/efeitos dos fármacos , Constrição Patológica , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Deleção de Genes , Proteínas de Membrana/metabolismo , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Pressão , Transdução de Sinais/efeitos dos fármacos
12.
Oncotarget ; 6(21): 18364-73, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-26286963

RESUMO

YopH is a bacterial protein tyrosine phosphatase, which is essential for the viability and pathogenic virulence of the plague-causing Yersinia sp. bacteria. Inactivation of YopH activity would lead to the loss of bacterial pathogenicity. We have studied the inhibitory properties of aurintricarboxylic acid (ATA) against YopH phosphatase and found that at nanomolar concentrations ATA reversibly decreases the activity of YopH. Computational docking studies indicated that in all binding poses ATA binds in the YopH active site. Molecular dynamics simulations showed that in the predicted binding pose, ATA binds to the essential Cys403 and Arg409 residues in the active site and has a stronger binding affinity than the natural substrate (pTyr). The cyclic voltammetry experiments suggest that ATA reacts remarkably strongly with molecular oxygen. Additionally, the electrochemical reduction of ATA in the presence of a negative potential from -2.0 to 2.5 V generates a current signal, which is observed for hydrogen peroxide. Here we showed that ATA indicates a unique mechanism of YopH inactivation due to a redox process. We proposed that the potent inhibitory properties of ATA are a result of its strong binding in the YopH active site and in situ generation of hydrogen peroxide near catalytic cysteine residue.


Assuntos
Ácido Aurintricarboxílico/química , Proteínas da Membrana Bacteriana Externa/química , Proteínas Tirosina Fosfatases/química , Fatores de Virulência/química , Algoritmos , Ácido Aurintricarboxílico/metabolismo , Ácido Aurintricarboxílico/farmacologia , Proteínas da Membrana Bacteriana Externa/antagonistas & inibidores , Proteínas da Membrana Bacteriana Externa/metabolismo , Humanos , Cinética , Conformação Molecular , Simulação de Dinâmica Molecular , Estrutura Molecular , Oxirredução , Peste/microbiologia , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Tirosina Fosfatases/metabolismo , Virulência , Fatores de Virulência/antagonistas & inibidores , Fatores de Virulência/metabolismo , Yersinia pestis/metabolismo , Yersinia pestis/patogenicidade , Yersinia pestis/fisiologia
13.
J Biomol Screen ; 19(8): 1147-53, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24793430

RESUMO

RNA-protein interactions are vital to the replication of the flaviviral genome. Discovery focused on small molecules that disrupt these interactions represent a viable path for identification of new inhibitors. The viral RNA (vRNA) cap methyltransferase (MTase) of the flaviviruses has been validated as a suitable drug target. Here we report the development of a high-throughput screen for the discovery of compounds that target the RNA binding site of flaviviral protein NS5A. The assay described here is based on displacement of an MT-bound polynucleotide aptamer, decathymidylate derivatized at its 5' end with fluorescein (FL-dT10). Based on the measurement of fluorescence polarization, FL-dT10 bound to yellow fever virus (YFV) MTase in a saturable manner with a Kd= 231 nM. The binding was reversed by a 250-nucleotide YFV messenger RNA (mRNA) transcript and by the triphenylmethane dye aurintricarboxylic acid (ATA). The EC50for ATA displacement was 1.54 µM. The MTase cofactors guanosine-5'-triphosphate and S-adenosyl-methionine failed to displace FL-dT10. Analysis by electrophoretic mobility shift assay (EMSA) suggests that ATA binds YFV MTase so as to displace the vRNA. The assay was determined to have a Z' of 0.83 and was successfully used to screen a library of known bioactives.


Assuntos
Aptâmeros de Nucleotídeos , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/farmacologia , Flavivirus/enzimologia , Metiltransferases/antagonistas & inibidores , Antivirais/química , Antivirais/farmacologia , Ácido Aurintricarboxílico/farmacologia , Sítios de Ligação , Inibidores Enzimáticos/química , Polarização de Fluorescência , Guanosina Trifosfato/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Terapia de Alvo Molecular/métodos , Oligodesoxirribonucleotídeos/metabolismo , Capuzes de RNA , RNA Mensageiro , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas não Estruturais Virais/metabolismo , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/metabolismo
14.
Biochemistry ; 52(36): 6151-9, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23947785

RESUMO

Aurintricarboxylic acid (ATA) is a potent inhibitor of many enzymes needed for cell and virus replication, such as polymerases, helicases, nucleases, and topoisomerases. This study examines how ATA interacts with the helicase encoded by the hepatitis C virus (HCV) and reveals that ATA interferes with both nucleic acid and ATP binding to the enzyme. We show that ATA directly binds HCV helicase to prevent the enzyme from interacting with nucleic acids and to modulate the affinity of HCV helicase for ATP, the fuel for helicase action. Amino acid substitutions in the helicase DNA binding cleft or its ATP binding site alter the ability of ATA to disrupt helicase-DNA interactions. These data, along with molecular modeling results, support the notion that an ATA polymer binds between Arg467 and Glu493 to prevent the helicase from binding either ATP or nucleic acids. We also characterize how ATA affects the kinetics of helicase-catalyzed ATP hydrolysis, and thermodynamic parameters describing the direct interaction between HCV helicase and ATA using microcalorimetry. The thermodynamics of ATA binding to HCV helicase reveal that ATA binding does not mimic nucleic acid binding in that ATA binding is driven by a smaller enthalpy change and an increase in entropy.


Assuntos
Trifosfato de Adenosina/metabolismo , Ácido Aurintricarboxílico/farmacologia , DNA/metabolismo , Proteínas não Estruturais Virais/efeitos dos fármacos , Substituição de Aminoácidos , Varredura Diferencial de Calorimetria , Hepacivirus/enzimologia , Modelos Moleculares , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
15.
J Mol Cell Cardiol ; 63: 57-68, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23880607

RESUMO

Isoform 4 of the plasma membrane calcium/calmodulin dependent ATPase (PMCA4) has recently emerged as an important regulator of several key pathophysiological processes in the heart, such as contractility and hypertrophy. However, direct monitoring of PMCA4 activity and assessment of calcium dynamics in its vicinity in cardiomyocytes are difficult due to the lack of molecular tools. In this study, we developed novel calcium fluorescent indicators by fusing the GCaMP2 calcium sensor to the N-terminus of PMCA4 to generate the PMCA4-GCaMP2 fusion molecule. We also identified a novel specific inhibitor of PMCA4, which might be useful for studying the role of this molecule in cardiomyocytes and other cell types. Using an adenoviral system we successfully expressed PMCA4-GCaMP2 in both neonatal and adult rat cardiomyocytes. This fusion molecule was correctly targeted to the plasma membrane and co-localised with caveolin-3. It could monitor signal oscillations in electrically stimulated cardiomyocytes. The PMCA4-GCaMP2 generated a higher signal amplitude and faster signal decay rate compared to a mutant inactive PMCA4(mut)GCaMP2 fusion protein, in electrically stimulated neonatal and adult rat cardiomyocytes. A small molecule library screen enabled us to identify a novel selective inhibitor for PMCA4, which we found to reduce signal amplitude of PMCA4-GCaMP2 and prolong the time of signal decay (Tau) to a level comparable with the signal generated by PMCA4(mut)GCaMP2. In addition, PMCA4-GCaMP2 but not the mutant form produced an enhanced signal in response to ß-adrenergic stimulation. Together, the PMCA4-GCaMP2 and PMCA4(mut)GCaMP2 demonstrate calcium dynamics in the vicinity of the pump under active or inactive conditions, respectively. In summary, the PMCA4-GCaMP2 together with the novel specific inhibitor provides new means with which to monitor calcium dynamics in the vicinity of a calcium transporter in cardiomyocytes and may become a useful tool to further study the biological functions of PMCA4. In addition, similar approaches could be useful for studying the activity of other calcium transporters during excitation-contraction coupling in the heart.


Assuntos
Calmodulina/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Miócitos Cardíacos/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Ácido Aurintricarboxílico/farmacologia , Cálcio/metabolismo , Sinalização do Cálcio , Calmodulina/genética , Cavéolas/metabolismo , Membrana Celular/metabolismo , Expressão Gênica , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Humanos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , ATPases Transportadoras de Cálcio da Membrana Plasmática/antagonistas & inibidores , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Transporte Proteico , Ratos , Proteínas Recombinantes de Fusão/genética
16.
Biotechnol Prog ; 28(6): 1566-74, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23011767

RESUMO

Dissociated primary neuron culture has been the most widely used model systems for neuroscience research. Most of these primary neurons are cultured on adhesion matrix-coated surface to provide a proper environment for cell anchorage under serum-free conditions. In this study, we provide an alternative technique to promote the adhesions of these neurons using aurintricarboxylic acid (ATA), a nonpeptide compound, without surface manipulations. We first demonstrated that ATA could promote Chinese hamster ovary cell attachment and proliferation in serum-free medium in a dosage-dependent manner. We later showed that ATA significantly enhanced the attachment of the retinoic acid differentiated P19 mouse embryonal carcinoma (P19) neurons, with an optimal concentration around 30 µg/mL. A similar result was seen in primary hippocampal neurons, with an optimal ATA concentration around 15 µg/mL. Further morphological assessments revealed that the average neurite length and neuronal polarization were almost identical to that obtained using a conventional method with poly-L-lysine surface. The advantages of using the ATA treatment technique for immunochemical analysis are discussed.


Assuntos
Ácido Aurintricarboxílico/farmacologia , Adesão Celular/efeitos dos fármacos , Técnicas de Cultura de Células/métodos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Actinas/metabolismo , Análise de Variância , Animais , Células CHO , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Meios de Cultura Livres de Soro , Camundongos , Camundongos Endogâmicos C57BL , Propriedades de Superfície , Tretinoína/farmacologia , Tubulina (Proteína)/metabolismo
17.
Adv Protein Chem Struct Biol ; 87: 219-47, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22607757

RESUMO

DNA methyltransferases (DNMTs) are promising epigenetic targets for the development of novel anticancer drugs and other diseases. Molecular modeling and experimental approaches are being used to identify and develop inhibitors of human DNMTs. Most of the computational efforts conducted so far with DNMT1 employ homology models of the enzyme. Recently, a crystallographic structure of the methyltransferase domain of human DNMT1 bound to unmethylated DNA was published. Following on our previous computational and experimental studies with DNMTs, we herein present molecular dynamics of the crystal structure of human DNMT1. Docking studies of established DNMT1 inhibitors with the crystal structure gave rise to a structure-based pharmacophore model that suggests key interactions of the inhibitors with the catalytic binding site. Results had a good agreement with the docking and pharmacophore models previously developed using a homology model of the catalytic domain of DNMT1. The docking protocol was able to distinguish active DNMT1 inhibitors from, for example, experimentally known inactive DNMT1 inhibitors. As part of our efforts to identify novel inhibitors of DNMT1, we conducted the experimental characterization of aurintricarboxylic acid (ATA) that in preliminary docking studies showed promising activity. ATA had a submicromolar inhibition (IC(50)=0.68 µM) against DNMT1. ATA was also evaluated for Dnmt3a inhibition showing an IC(50)=1.4 µM. This chapter illustrates the synergy from integrating molecular modeling and experimental methods to further advance the discovery of novel candidates for epigenetic therapies.


Assuntos
Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/farmacologia , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Cristalografia por Raios X , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Relação Estrutura-Atividade
18.
J Mol Model ; 18(4): 1583-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21805123

RESUMO

DNA methyltransferase 1 (DNMT1) is an emerging target for the treatment of cancer, brain disorders, and other diseases. Currently, there are only a few DNMT1 inhibitors with potential application as therapeutic agents or research tools. 5,5-Methylenedisalicylic acid is a novel scaffold previously identified by virtual screening with detectable although weak inhibitory activity of DNMT1 in biochemical assays. Herein, we report enzyme inhibition of a structurally related compound, trimethylaurintricarboxylic acid (NSC97317) that showed a low micromolar inhibition of DNMT1 (IC(50) = 4.79 µM). Docking studies of the new inhibitor with the catalytic domain of DNMT1 suggest that NSC97317 can bind into the catalytic site. Interactions with amino acid residues that participate in the mechanism of DNA methylation contribute to the binding recognition. In addition, NSC97317 had a good match with a structure-based pharmacophore model recently developed for inhibitors of DNMT1. Trimethylaurintricarboxylic acid can be a valuable biochemical tool to study DNMT1 inhibition in cancer and other diseases related to DNA methylation.


Assuntos
Ácido Aurintricarboxílico/análogos & derivados , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/metabolismo , Ácido Aurintricarboxílico/farmacologia , Domínio Catalítico/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/química , Desenho de Fármacos , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Estrutura Terciária de Proteína
19.
Int J Cancer ; 130(5): 1060-70, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21400516

RESUMO

c-Met [the hepatocyte growth factor (HGF) receptor] is a receptor tyrosine kinase playing a role in various biological events. Overexpression of the receptor has been observed in a number of cancers, correlating with increased metastatic tendency and poor prognosis. Additionally, activating mutations in c-Met kinase domain have been reported in a subset of familial cancers causing resistance to treatment. Receptor trafficking, relying on the integrity of the microtubule network, plays an important role in activation of downstream targets and initiation of signalling events. Aurintricarboxylic acid (ATA) is a triphenylmethane derivative that has been reported to inhibit microtubule motor proteins kinesins. Additional reported properties of this inhibitor include inhibition of protein tyrosine phosphatases, nucleases and members of the Jak family. Here we demonstrate that ATA prevents HGF-induced c-Met phosphorylation, internalisation, subsequent receptor trafficking and degradation. In addition, ATA prevented HGF-induced downstream signalling which also affected cellular function, as assayed by collective cell migration of A549 cells. Surprisingly, the inhibitory effect of ATA on HGF-induced phosphorylation and signalling in vivo was associated with an increase in basal c-Met kinase activity in vitro. It is concluded that the inhibitory effects of ATA on c-Met in vivo is an allosteric effect mediated through the kinase domain of the receptor. As the currently tested adenosine triphosphate competitive tyrosine kinase inhibitors (TKIs) may lead to tumor resistance (McDermott U, et al., Cancer Res 2010;70:1625-34), our findings suggest that novel anti-c-Met therapies could be developed in the future for cancer treatment.


Assuntos
Ácido Aurintricarboxílico/farmacologia , Cinesinas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/metabolismo , Regulação Alostérica , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Regulação para Baixo , Células HeLa , Humanos , Neoplasias Pulmonares , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
20.
Blood Coagul Fibrinolysis ; 22(2): 132-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21245742

RESUMO

Activation of the vascular endothelium and increased adhesion of circulating leukocytes to the activated endothelium are important events in inflammation and coagulation. Aurintricarboxylic acid (ATA), a triphenylmethyl dye compound, is known to inhibit platelet adhesion by interfering with the binding of von Willebrand factor to platelet glycoprotein Ib. However, the effect of ATA on the inflammatory response of endothelial cells has not yet been investigated. Here, we investigated the functional role and molecular mechanism of ATA on the activation of human endothelial cells. ATA inhibited the expression of intercellular adhesion molecule-1 (ICAM-1), and endothelial cell selectin (E-selectin) was upregulated on human umbilical vein endothelial cells (HUVECs) in response to tumor necrosis factor-α or lipopolysaccharide (LPS). We also observed the inhibitory effect of ATA on LPS-induced mRNA expression of ICAM-1 and E-selectin. Furthermore, ATA inhibited the binding of leukocytes to activated HUVECs. ATA significantly inhibited the nuclear translocation of nuclear factor-κB (NF-κB) and degradation of IκB on activated HUVECs, suggesting that ATA inhibits NF-κB signaling. Finally, three NF-κB inhibitors effectively inhibited the expressions of ICAM-1 and E-selectin on activated endothelial cells. The present data suggest that ATA exerts beneficial effect in various inflammation conditions through inhibition of adhesion molecule expression in activated endothelial cells and the resulting inhibition of leukocytes tissue accumulation.


Assuntos
Ácido Aurintricarboxílico/farmacologia , Selectina E/metabolismo , Proteínas I-kappa B/antagonistas & inibidores , Molécula 1 de Adesão Intercelular/metabolismo , NF-kappa B/antagonistas & inibidores , Adesão Celular/efeitos dos fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Regulação para Baixo , Selectina E/genética , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Expressão Gênica , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Molécula 1 de Adesão Intercelular/genética , Leucócitos/metabolismo , Lipopolissacarídeos/farmacologia , Inibidor de NF-kappaB alfa , NF-kappa B/genética , NF-kappa B/metabolismo , RNA Mensageiro/biossíntese , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA