Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.563
Filtrar
1.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34768947

RESUMO

Inhibition of phosphatidylcholine-specific phospholipase C (PC-PLC) has previously been shown to be a potential target for novel cancer therapeutics. One downstream consequence of PC-PLC activity is the activation of NF-κB, a nuclear transcription factor responsible for transcribing genes related to oncogenic traits, such as proliferation, angiogenesis, metastasis, and cancer cell survival. Another biological pathway linked to NF-κB is the exogenous delivery of nitric oxide (NO), which decreases NF-κB activity through an apparent negative-feedback loop. In this study, we designed and synthesised 13 novel NO-releasing derivatives of our previously reported class of PC-PLC inhibitors, 2-morpholinobenzoic acids. These molecules contained a secondary benzylamine group, which was readily nitrosylated and subsequently confirmed to release NO in vitro using a DAF-FM fluorescence-based assay. It was then discovered that these NO-releasing derivatives possessed significantly improved anti-proliferative activity in both MDA-MB-231 and HCT116 cancer cell lines compared to their non-nitrosylated parent compounds. These results confirmed that the inclusion of an exogenous NO-releasing functional group onto a known PC-PLC inhibitor enhances anti-proliferative activity and that this relationship can be exploited in order to further improve the anti-proliferative activity of current/future PC-PLC inhibitors.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Doadores de Óxido Nítrico/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Antineoplásicos/química , Benzilaminas/química , Benzilaminas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desenho de Fármacos , Inibidores Enzimáticos/química , Feminino , Células HCT116 , Células HEK293 , Humanos , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/química , Compostos Nitrosos/química , Compostos Nitrosos/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
2.
Sci Rep ; 11(1): 11273, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34050231

RESUMO

Nitric oxide (NO) is produced from endothelial cells and cardiomyocytes composing the myocardium and benefits cardiac function through both vascular-dependent and-independent effects. This study was purposed to investigate the possible adverse effect of NO focusing on the voltage-gated Na+ channel in cardiomyocytes. We carried out patch-clamp experiments on rat neonatal cardiomyocytes demonstrating that NOC-18, an NO donor, significantly reduced Na+ channel current in a dose-dependent manner by a long-term application for 24 h, accompanied by a reduction of Nav1.5-mRNA and the protein, and an increase of a transcription factor forkhead box protein O1 (FOXO1) in the nucleus. The effect of NOC-18 on the Na+ channel was blocked by an inhibitor of thiol oxidation N-ethylmaleimide, a disulfide reducing agent disulfide 1,4-Dithioerythritol, or a FOXO1 activator paclitaxel, suggesting that NO is a negative regulator of the voltage-gated Na+ channel through thiols in regulatory protein(s) for the channel transcription.


Assuntos
Miócitos Cardíacos/fisiologia , Óxido Nítrico/metabolismo , Canais de Sódio Disparados por Voltagem/metabolismo , Animais , Animais Recém-Nascidos , Núcleo Celular/metabolismo , Células Endoteliais/metabolismo , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Óxido Nítrico/fisiologia , Compostos Nitrosos/metabolismo , Compostos Nitrosos/farmacologia , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Transdução de Sinais , Sódio/metabolismo , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos
3.
Amino Acids ; 53(4): 563-573, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33586042

RESUMO

Nitrosylation of sulfhydryl (SH) groups of cysteine (Cys) moieties is an important post-translational modification (PTM), often on a par with phosphorylation. S-Nitrosoalbumin (ALB-Cys34SNO; SNALB) in plasma and S-nitrosohemoglobin (Hb-Cysß93SNO; HbSNO) in red blood cells are considered the most abundant high-molecular-mass pools of nitric oxide (NO) bioactivity in the human circulation. SNALB per se is not an NO donor. Yet, it acts as a vasodilator and an inhibitor of platelet aggregation. SNALB can be formed by nitrosation of the sole reduced Cys group of albumin (Cys34) by nitrosating species such as nitrous acid (HONO) and nitrous anhydride (N2O3), two unstable intermediates of NO autoxidation. SNALB can also be formed by the transfer (S-transnitrosylation) of the nitrosyl group (NO+) of a low-molecular-mass (LMM) S-nitrosothiol (RSNO) to ALB-Cys34SH. In the present study, the effects of LMM thiols on the inhibitory potential of ALB-Cys34SNO on human washed platelets were investigated. ALB-Cys34SNO was prepared by reacting n-butylnitrite with albumin after selective extraction from plasma of a healthy donor on HiTrapBlue Sepharose cartridges. ALB-Cys34SNO was used in platelet aggregation measurements after extended purification on HiTrapBlue Sepharose and enrichment by ultrafiltration (cutoff, 20 kDa). All tested LMM cysteinyl thiols (R-CysSH) including L-cysteine and L-homocysteine (at 10 µM) were found to mediate the collagen-induced (1 µg/mL) aggregation of human washed platelets by SNALB (range, 0-10 µM) by cGMP-dependent and cGMP-independent mechanisms. The LMM thiols themselves did not affect platelet aggregation. It is assumed that the underlying mechanism involves S-transnitrosylation of SH groups of the platelet surface by LMM RSNO formed through the reaction of SNALB with the thiols: ALB-Cys34SNO + R-CysSH ↔ ALB-Cys34SH + R-CysSNO. Such S-transnitrosylation reactions may be accompanied by release of NO finally resulting in cGMP-dependent and cGMP-independent mechanisms.


Assuntos
Plaquetas/efeitos dos fármacos , Compostos Nitrosos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Albumina Sérica Humana/farmacologia , Compostos de Sulfidrila/química , Plaquetas/metabolismo , Humanos , Óxido Nítrico/metabolismo , Compostos Nitrosos/química , Processamento de Proteína Pós-Traducional , S-Nitrosotióis/química , S-Nitrosotióis/farmacologia , Albumina Sérica Humana/química , Compostos de Sulfidrila/farmacologia
4.
Nitric Oxide ; 106: 24-34, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33098968

RESUMO

The aim of the current study is to report a simple and efficient method to chemically modify chitosan in order to form S-nitroso-chitosan for antibacterial applications. Firstly, commercial chitosan (CS) was modified to form thiolated chitosan (TCS) based on an easy and environmental-friendly method. TCS was featured based on physicochemical and morphological techniques. Results have confirmed that thiol groups in TCS formed after CS's primary amino groups were replaced with secondary amino groups. Free thiol groups in TCS were nitrosated to form S-nitrosothiol moieties covalently bond to the polymer backbone (S-nitroso-CS). Kinetic measurements have shown that S-nitroso-CS was capable of generating NO in a sustained manner at levels suitable for biomedical applications. The antibacterial activities of CS, TCS and S-nitroso-CS were evaluated based on the minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC) and time-kill curves determined for Escherichia coli, Staphylococcus aureus and Streptococcus mutans. MIC/MBC values reached 25/25, 0.7/0.7 and 3.1/3.1 µg mL-1 for CS/TCS and 3.1/3.1, 0.1/0.2, 0.1/0.2 µg mL-1 for S-nitroso-CS, respectively. Decreased MIC and MBC values have indicated that S-nitroso-CS has higher antibacterial activity than CS and TCS. Time-kill curves have shown that the bacterial cell viability decreased 5-fold for E. coli and 2-fold for S. mutans in comparison to their respective controls, after 0.5 h of incubation with S-nitroso-CS. Together, CS backbone chemically modified with S-nitroso moieties have yielded a polymer capable of generating therapeutic NO concentrations with strong antibacterial effect.


Assuntos
Antibacterianos/farmacologia , Quitosana/farmacologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico/farmacologia , Compostos Nitrosos/farmacologia , Antibacterianos/síntese química , Sobrevivência Celular/efeitos dos fármacos , Quitosana/síntese química , Liberação Controlada de Fármacos , Escherichia coli/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Óxido Nítrico/química , Doadores de Óxido Nítrico/síntese química , Compostos Nitrosos/síntese química , Staphylococcus aureus/efeitos dos fármacos , Streptococcus mutans/efeitos dos fármacos
5.
Anticancer Res ; 40(12): 6751-6763, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33288568

RESUMO

BACKGROUND/AIM: Chemoresistance is a major consequence of multicycle chemotherapy and can be attributed to constitutive activation of pro-survival signaling pathways. Nitric oxide is a ubiquitous signaling molecule which has been shown to inhibit several pathways involved with survival signaling in cancer cells. We have previously demonstrated the anti-tumor activity of a nitric oxide-donor, nitrosylcobalamin (NO-Cbl), mediated by increased expression of tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) and its receptors in human tumors. We also demonstrated that a functional Apo2L/TRAIL receptor is necessary for the induction of cell death by NO-Cbl and the Apo2L/TRAIL death receptor DR4 (TRAIL R1) is S-nitrosylated. The aim of the study was to examine the effects of nitric oxide (NO) on nuclear factor kappa B (NF-κB) and determine whether nitric oxide could sensitize drug-resistant melanomas to Apo2L/TRAIL via inhibition of NF-κB or inhibitor kappa B kinase (IKK). MATERIALS AND METHODS: Antiproliferative effects of NO-Cbl and Apo2L/TRAIL were assessed in malignant melanomas and non-tumorigenic melanocyte and fibroblast cell lines. Athymic nude mice bearing human melanoma A375 xenografts were treated with NO-Cbl and Apo2L/TRAIL. Apoptosis was measured by the TUNEL assay. The activation status of NF-κB was established by assaying luciferase reporter activity, the phosphorylation status of IκBα, and in vitro IKK activity. RESULTS: NO-Cbl sensitized Apo2L/TRAIL-resistant melanoma cell lines to growth inhibition by Apo2L/TRAIL, but had minimal effect on normal cell lines. NO-Cbl and Apo2L/TRAIL exerted synergistic anti-tumor activity against A375 xenografts. NO-Cbl suppressed Apo2L/TRAIL- and TNF-α-mediated activation of a transfected NF-κB-driven luciferase reporter. NO-Cbl inhibited IKK activation, characterized by decreased phosphorylation of IκBα. CONCLUSION: NO-Cbl treatment rendered Apo2L/TRAIL-resistant malignancies sensitive to the anti-tumor effects of Apo2L/TRAIL in vitro and in vivo. The use of nitric oxide to inhibit NF-κB and potentiate the effects of chemotherapeutic agents, such as Apo2L/TRAIL, represents a promising anti-cancer combination based on recent clinical investigations of anti-TRAIL antibodies for cancer treatment strategies.


Assuntos
NF-kappa B/metabolismo , Óxido Nítrico/farmacologia , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Masculino , Camundongos Nus , Inibidor de NF-kappaB alfa/metabolismo , Compostos Nitrosos/farmacologia , Vitamina B 12/análogos & derivados , Vitamina B 12/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Int J Mol Sci ; 21(21)2020 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-33158109

RESUMO

Traumatic brain injury (TBI) can cause physical, cognitive, social, and behavioral changes that can lead to permanent disability or death. After primary brain injury, translocated free zinc can accumulate in neurons and lead to secondary events such as oxidative stress, inflammation, edema, swelling, and cognitive impairment. Under pathological conditions, such as ischemia and TBI, excessive zinc release, and accumulation occurs in neurons. Based on previous research, it hypothesized that calcium as well as zinc would be influx into the TRPC5 channel. Therefore, we hypothesized that the suppression of TRPC5 would prevent neuronal cell death by reducing the influx of zinc and calcium. To test our hypothesis, we used a TBI animal model. After the TBI, we immediately injected NU6027 (1 mg/kg, intraperitoneal), TRPC5 inhibitor, and then sacrificed animals 24 h later. We conducted Fluoro-Jade B (FJB) staining to confirm the presence of degenerating neurons in the hippocampal cornus ammonis 3 (CA3). After the TBI, the degenerating neuronal cell count was decreased in the NU6027-treated group compared with the vehicle-treated group. Our findings suggest that the suppression of TRPC5 can open a new therapeutic window for a reduction of the neuronal death that may occur after TBI.


Assuntos
Lesões Encefálicas Traumáticas/patologia , Morte Celular/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Compostos Nitrosos/farmacologia , Pirimidinas/farmacologia , Animais , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/fisiopatologia , Contagem de Células , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Neurônios/patologia , Neurônios/fisiologia , Compostos Nitrosos/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Pirimidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPC/antagonistas & inibidores , Zinco/metabolismo
7.
J Biol Chem ; 295(45): 15342-15365, 2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-32868295

RESUMO

The contraction and relaxation of the heart is controlled by stimulation of the ß1-adrenoreceptor (AR) signaling cascade, which leads to activation of cAMP-dependent protein kinase (PKA) and subsequent cardiac protein phosphorylation. Phosphorylation is counteracted by the main cardiac protein phosphatases, PP2A and PP1. Both kinase and phosphatases are sensitive to intramolecular disulfide formation in their catalytic subunits that inhibits their activity. Additionally, intermolecular disulfide formation between PKA type I regulatory subunits (PKA-RI) has been described to enhance PKA's affinity for protein kinase A anchoring proteins, which alters its subcellular distribution. Nitroxyl donors have been shown to affect contractility and relaxation, but the mechanistic basis for this effect is unclear. The present study investigates the impact of several nitroxyl donors and the thiol-oxidizing agent diamide on cardiac myocyte protein phosphorylation and oxidation. Although all tested compounds equally induced intermolecular disulfide formation in PKA-RI, only 1-nitrosocyclohexalycetate (NCA) and diamide induced reproducible protein phosphorylation. Phosphorylation occurred independently of ß1-AR activation, but was abolished after pharmacological PKA inhibition and thus potentially attributable to increased PKA activity. NCA treatment of cardiac myocytes induced translocation of PKA and phosphatases to the myofilament compartment as shown by fractionation, immunofluorescence, and proximity ligation assays. Assessment of kinase and phosphatase activity within the myofilament fraction of cardiac myocytes after exposure to NCA revealed activation of PKA and inhibition of phosphatase activity thus explaining the increase in phosphorylation. The data suggest that the NCA-mediated effect on cardiac myocyte protein phosphorylation orchestrates alterations in the kinase/phosphatase balance.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Oxidantes/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Receptores Adrenérgicos beta 1/metabolismo , Transdução de Sinais , Acetatos/farmacologia , Animais , Bovinos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diamida/farmacologia , Humanos , Masculino , Camundongos , Miócitos Cardíacos/metabolismo , Compostos Nitrosos/farmacologia , Oxirredução , Fosfoproteínas Fosfatases/metabolismo , Fosforilação/efeitos dos fármacos , Coelhos , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
8.
Eur J Med Chem ; 208: 112768, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-32932211

RESUMO

4-Nitroso-3-trifluoromethyl-5-alkyl[(het)aryl]pyrazoles were synthesized via one-pot nitrosation of 1,3-diketones or their lithium salts followed by treatment of hydrazines. Reduction of nitroso-derivatives made it possible to obtain 4-amino-3-trifluoromethylpyrazoles chlorides. According to computer-aided calculations, all synthesized compounds are expected to have acceptable ADME profile for drug design. Tuberculostatic, antibacterial, antimycotic, antioxidant and cytotoxic activities of the compounds were evaluated in vitro, while their analgesic and anti-inflammatory action was tested in vivo along with acute toxicity studies. N-Unsubstituted 4-nitrosopyrazoles were the most effective tuberculostatics (MIC to 0.36 µg/ml) and antibacterial agents against Streptococcus pyogenes (MIC to 7.8 µg/ml), Staphylococcus aureus,S. aureus MRSA and Neisseria gonorrhoeae (MIC to 15.6 µg/ml). 4-Nitroso-1-methyl-5-phenylpyrazole had the pronounced antimycotic action against a wide range of fungi (Trichophytonrubrum, T. tonsurans, T. violaceum, T. interdigitale, Epidermophytonfloccosum, Microsporumcanis with MIC 0.38-12.5 µg/ml). N-Unsubstituted 4-aminopyrazoles shown high radical-scavenging activity in ABTS test, ORAC/AAPH and oxidative erythrocyte hemolysis assays. 1-Methyl-5-phenyl-3-trifluoromethylpyrazol-4-aminium chloride revealed potential anticancer activity against HeLa cells (SI > 1351). The pronounced analgesic activity was found for 4-nitroso- and 4-aminopyrazoles having phenyl fragment at the position 5 in "hot plate" test. The most of the obtained pyrazoles had a moderate acute toxicity.


Assuntos
Compostos Nitrosos/farmacologia , Pirazóis/farmacologia , Animais , Bactérias/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Epidermophyton/efeitos dos fármacos , Células HeLa , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Compostos Nitrosos/síntese química , Compostos Nitrosos/farmacocinética , Compostos Nitrosos/toxicidade , Pirazóis/síntese química , Pirazóis/farmacocinética , Pirazóis/toxicidade , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Trichophyton/efeitos dos fármacos
9.
Int J Food Microbiol ; 331: 108731, 2020 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-32535525

RESUMO

This study aims to enhance the color and microbiological qualities of a raw beef using natural ingredients. Nitroso-hemoglobin (NO-Hb) integrated with vitamin C (VC), calcium lactate, and ginger complexation were used as natural inhibitors against the growth of aerobic and pathogenic bacteria, namely (Escherichia coli (E. coli), Staphylococcus aureus (S. aureus), and Salmonella. NO-Hb inhibited E. coli, S. aureus, and Salmonella, and enhanced the color stability more than nitrite in the minced beef model. After the multiexponential analysis of relaxation decays, the water component (T2b) was analyzed using the low-field NMR. The results indicated that, at the 7th d of cold-storage the third component (T2) was detected. Significant correlations were observed between T21 and T22 relaxation times and water-holding capacity in minced beef, implying that the LF-NMR measurements could be an efficient method for the determination and prediction of beef freshness. NO-Hb- ginger mixture, as a novel ingredient, could be used instead of nitrite in terms of meat safety.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Microbiologia de Alimentos/métodos , Carne Vermelha/microbiologia , Animais , Bovinos , Cor , Hemoglobinas/farmacologia , Compostos Nitrosos/farmacologia , Extratos Vegetais/farmacologia
10.
Biol Pharm Bull ; 43(6): 1020-1022, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32201424

RESUMO

Glioblastoma comprises 54% of all the gliomas derived from glial cells and are lethally malignant tumors of the central nervous system (CNS). Glioma cells disrupt the blood-brain barrier, leading to access of circulating immune cells to the CNS. Blocking the interaction between programmed cell death 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) enhances T-cell responses against tumor cells, and inhibition of the PD-1/PD-L1 pathway is used as immunotherapy for cancer, including glioblastoma. Nitric oxide (NO) has multiple physiological roles, such as immune modulation and neural transmission in the CNS. Moreover, it has both tumor-promoting and tumor-suppressive functions. We examined the effects of NOC-18, an NO donor, on the expression of PD-L1 in A172 glioblastoma cells. NOC-18 increased PD-L1 expression in A172 glioblastoma cells. Moreover, this increase is regulated via the c-Jun N-terminal kinase pathway.


Assuntos
Antígeno B7-H1/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico/metabolismo , Compostos Nitrosos/farmacologia , Linhagem Celular Tumoral , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Neoplasias do Sistema Nervoso Central/genética , Neoplasias do Sistema Nervoso Central/metabolismo , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos
11.
Biochem Pharmacol ; 176: 113817, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31972169

RESUMO

Mechanical ventilation (MV) with supraphysiological levels of oxygen (hyperoxia) is a life-saving therapy for the management of patients with respiratory distress. However, a significant number of patients on MV develop ventilator-associated pneumonia (VAP). Previously, we have reported that prolonged exposure to hyperoxia impairs the capacity of macrophages to phagocytize Pseudomonas aeruginosa (PA), which can contribute to the compromised innate immunity in VAP. In this study, we show that the high mortality rate in mice subjected to hyperoxia and PA infection was accompanied by a significant decrease in the airway levels of nitric oxide (NO). Decreased NO levels were found to be, in part, due to a significant reduction in NO release by macrophages upon exposure to PA lipopolysaccharide (LPS). Based on these findings, we postulated that NO supplementation should restore hyperoxia-compromised innate immunity and decrease mortality by increasing the clearance of PA under hyperoxic conditions. To test this hypothesis, cultured macrophages were exposed to hyperoxia (95% O2) in the presence or absence of the NO donor, (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETA-NONOate/D-NO). Interestingly, D-NO (up to 37.5 µM) significantly attenuated hyperoxia-compromised macrophage migratory, phagocytic, and bactericidal function. To determine whether the administration of exogenous NO enhances the host defense in bacteria clearance, C57BL/6 mice were exposed to hyperoxia (99% O2) and intranasally inoculated with PA in the presence or absence of D-NO. D-NO (300 µM-800 µM) significantly increased the survival of mice inoculated with PA under hyperoxic conditions, and significantly decreased bacterial loads in the lung and attenuated lung injury. These results suggest the NO donor, D-NO, can improve the clinical outcomes in VAP by augmenting the innate immunity in bacterial clearance. Thus, provided these results can be extrapolated to humans, NO supplementation may represent a potential therapeutic strategy for preventing and treating patients with VAP.


Assuntos
Imunidade Inata/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Compostos Nitrosos/farmacologia , Pneumonia Associada à Ventilação Mecânica/tratamento farmacológico , Infecções por Pseudomonas/prevenção & controle , Pseudomonas aeruginosa/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Humanos , Hiperóxia/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/imunologia , Óxido Nítrico/metabolismo , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Pneumonia Associada à Ventilação Mecânica/imunologia , Pneumonia Associada à Ventilação Mecânica/microbiologia , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/fisiologia , Células RAW 264.7
12.
Biomed Pharmacother ; 120: 109468, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31605952

RESUMO

Development of resistance to chemotherapy drugs is a significant problem in treating human malignancies in the clinic. Overexpression of ABC transporter proteins, including P-170 glycoprotein (P-gp), and breast cancer resistance protein (BCRP, ABCG2) have been implicated in this multi-drug resistance (MDR). These ABC transporters are ATP-dependent efflux proteins. We have recently shown that nitric oxide (NO) inhibits the ATPase activities of P-gp, resulting in a significant enhancement of drug accumulation and the reversal of multi-drug resistance in NCI/ADR-RES cells, a P-gp-overexpressing human MDR cell line. In this study, we used [O2-(2,4-dinitrophenyl)-1-[(4-ethoxycarbonyl)-piperazin-1 yl]-diazene-1-ium-1-2-diolate] (JS-K), a tumor-specific NO-donor to study the reversal of drug resistance in both P-gp- and BCRP-overexpressing human tumor cells. We report here that while JS-K was extremely effective in reversing adriamycin resistance in the P-gp-overexpressing tumor cells (NCI/ADR-RES); it was significantly resistant to BCRP-overexpressing (MCF-7/MX) tumor cells, suggesting that JS-K may be a substrate for BCRP. Using another NO-donor (DETNO), we show that NO directly inhibits the ATP activities of BCRP, inducing significant increases in the accumulations of both Hoechst 33342 dye and topotecan, substrates for BCRP. Furthermore, NO treatment significantly reversed topotecan and mitoxantrone resistance to MCF-7/MX tumor cells. Molecular docking studies indicated that while DETNO and JS-K bind to ATP binding site in both ABC proteins, binding score was significantly reduced, compared to the ATP binding. Our results indicate that appropriately designed NO donors may find success in reversing multidrug resistance in the clinic.


Assuntos
Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Compostos Azo/farmacologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Óxido Nítrico/farmacologia , Piperazinas/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Compostos Azo/química , Linhagem Celular Tumoral , Sobrevivência Celular , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Humanos , Mitoxantrona/química , Mitoxantrona/farmacologia , Simulação de Acoplamento Molecular , Estrutura Molecular , Compostos Nitrosos/química , Compostos Nitrosos/farmacologia , Piperazinas/química , Topotecan/química , Topotecan/farmacologia
13.
Br J Cancer ; 121(7): 600-610, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31481733

RESUMO

BACKGROUND: Up to 40% of lung adenocarcinoma have been reported to lack ataxia-telangiectasia mutated (ATM) protein expression. We asked whether ATM-deficient lung cancer cell lines are sensitive to poly-ADP ribose polymerase (PARP) inhibitors and determined the mechanism of action of olaparib in ATM-deficient A549 cells. METHODS: We analysed drug sensitivity data for olaparib and talazoparib in lung adenocarcinoma cell lines from the Genomics of Drug Sensitivity in Cancer (GDSC) project. We deleted ATM from A549 lung adenocarcinoma cells using CRISPR/Cas9 and determined the effects of olaparib and the ATM/Rad3-related (ATR) inhibitor VE-821 on cell viability. RESULTS: IC50 values for both olaparib and talazoparib positively correlated with ATM mRNA levels and gene amplification status in lung adenocarcinoma cell lines. ATM mutation was associated with a significant decrease in the IC50 for olaparib while a similar trend was observed for talazoparib. A549 cells with deletion of ATM were sensitive to ionising radiation and olaparib. Olaparib induced phosphorylation of DNA damage markers and reversible G2 arrest in ATM-deficient cells, while the combination of olaparib and VE-821 induced cell death. CONCLUSIONS: Patients with tumours characterised by ATM-deficiency may benefit from treatment with a PARP inhibitor in combination with an ATR inhibitor.


Assuntos
Adenocarcinoma/tratamento farmacológico , Proteínas Mutadas de Ataxia Telangiectasia/deficiência , Neoplasias Pulmonares/tratamento farmacológico , Ftalazinas/farmacologia , Piperazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Pirazinas/farmacologia , Sulfonas/farmacologia , Adenocarcinoma/metabolismo , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Deleção de Genes , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Mutação , Compostos Nitrosos/farmacologia , Fosforilação , Pirimidinas/farmacologia , RNA Mensageiro/metabolismo , Proteína Supressora de Tumor p53/metabolismo
14.
Medicina (Kaunas) ; 55(10)2019 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-31554309

RESUMO

Background and objective: Nitric oxide (NO) is known to exert cardioprotective effects against heart ischemic damage and may be involved in ischemic pre- and postconditioning. NO-triggered cardioprotective mechanisms are not well understood but may involve regulation of mitochondrial permeability transition pore (mPTP). In this study, we aimed to identify differentially phosphorylated mitochondrial proteins possibly involved in the NO/protein kinase G (PKG)/mPTP signaling pathway that can increase the resistance of cardiomyocytes to ischemic damage. Materials and methods: Isolated hearts from Wistar rats were perfused with NO donor NOC-18 prior to induction of stop-flow ischemia. To quantify and characterize the phosphoproteins, mitochondrial proteins were resolved and analyzed by two-dimensional gel electrophoresis followed by Pro-Q Diamond phosphoprotein gel staining, excision, trypsin digestions, and mass spectrometry. Quantitative proteomic analysis coupled with liquid chromatography-tandem mass spectrometry was also performed. Results: Mitochondrial protein phosphorylation patterns in NOC-18-pretreated ischemic hearts versus ischemic hearts were compared. Pretreatment of hearts with NOC-18 caused changes in mitochondrial phosphoproteome after ischemia which involved modifications of 10 mitochondrial membrane-bound and 10 matrix proteins. Among them, α-subunit of ATP synthase and adenine nucleotide (ADP/ATP) translocase 1, both of which are considered as potential structural components of mPTP, were identified. We also found that treatment of isolated non-ischemic mitochondria with recombinant PKG did not cause the same protein phosphorylation as pretreatment of hearts with NOC-18. Conclusions: Our study suggests that pretreatment of hearts with NOC-18 causes changes in mitochondrial phosphoproteome after ischemia which involves modifications of certain proteins thought to be involved in the regulation of mPTP opening and intracellular redox state. These proteins may be potential targets for pharmacological preconditioning of the heart.


Assuntos
Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Isquemia Miocárdica/metabolismo , Doadores de Óxido Nítrico/farmacologia , Compostos Nitrosos/farmacologia , Fosfoproteínas/metabolismo , Proteoma/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Coração/efeitos dos fármacos , Masculino , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/efeitos dos fármacos , Poro de Transição de Permeabilidade Mitocondrial , Miocárdio/metabolismo , Fosforilação , Proteoma/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
15.
Nitric Oxide ; 93: 25-33, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31541732

RESUMO

Leishmaniasis is a neglected tropical disease that demands for new therapeutic strategies due to adverse side effects and resistance development promoted by current drugs. Nitric oxide (NO)-donors show potential to kill Leishmania spp. but their use is limited because of their instability. In this work, we synthesize, characterize, and encapsulate S-nitroso-mercaptosuccinic acid into chitosan nanoparticles (NONPs) and investigate their activity on promastigotes and intracellular amastigotes of Leishmania (Leishmania) amazonensis. Cytotoxicity on macrophages was also evaluated. We verified that NONPs reduced both forms of the parasite in a single treatment. We also noticed reduction of parasitophorous vacuoles as an evidence of inhibition of parasite growth and resolution of infection. No substantial cytotoxicity was detected on macrophages. NONPs were able to provide a sustained parasite killing for both L. (L.) amazonensis infective stages with no toxicity on macrophages, representing a promising nanoplatform for cutaneous leishmaniasis.


Assuntos
Quitosana/química , Leishmania/efeitos dos fármacos , Nanopartículas/química , Doadores de Óxido Nítrico/farmacologia , Compostos Nitrosos/farmacologia , Tiomalatos/farmacologia , Animais , Quitosana/toxicidade , Cinética , Macrófagos/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Nanopartículas/toxicidade , Óxido Nítrico/química , Doadores de Óxido Nítrico/química , Doadores de Óxido Nítrico/toxicidade , Compostos Nitrosos/química , Compostos Nitrosos/toxicidade , Tiomalatos/química , Tiomalatos/toxicidade , Tripanossomicidas
16.
Cell Signal ; 64: 109411, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31491460

RESUMO

Induction of nitrosative stress has been observed in various cancer types and in tumor environment. However, it is still unclear how cancer cells combat the effect of nitrosative stress. The main targets of nitrosative stress in cells are cellular lipids, proteins and DNA. Autophagy or self-cleaning generates energy for cell survival under stress conditions. In the present study we investigated the role of autophagy under nitrosative stress in MCF7, a breast cancer cell line. Interestingly, we observed induction of autophagy associated with cell death when MCF7 cells were treated with NO donor compound DETA-NONOate for eight hours. While investigating the mode of cell death under nitrosative stress in MCF7 cells, it was found that it was neither apoptotic nor necrotic. Moreover, nitrosative stress did not alter mitochondrial membrane potential and cellular redox status in MCF7 cells. But we observed an increase in NAD+/NADH and a drop in NADH level in MCF7 cells following NO donor treatment. Sirtuins having NAD+ dependent deacetylase activity, play an important role in cell survival mechanisms. So we further checked the status of SIRT1 under nitrosative stress in MCF7 cells. Surprisingly, we observed an induction of SIRT1, phospho-AMPK and p53 in MCF7 cells under nitrosative stress. Interestingly, autophagy markers were down regulated in MCF7 cells upon treatment with nicotinamide, an inhibitor of SIRT1 activity and dorsomorphin, a phospho-AMPK inhibitor when treated separately under nitrosative stress. To further confirm the role of SIRT1 in the induction of autophagy associated cell death, it was knocked down using si-RNA and nitrosative stress was applied. SIRT1 knock down led to increase in MCF7 cell viability along with down regulation of autophagic markers and phospho-AMPK as well as accumulation of acetylated p53. The increase in p53 controlled DRAM1 mRNA expression in MCF7 cells under nitrosative stress further confirmed a complex interplay between p53, SIRT1 and AMPK under nitrosative stress in MCF7 cells. Altogether our work for the first time suggests a complex inter-twined partnership between AMPK, SIRT1 and p53 in regulating autophagy in response to nitrosative stress in MCF7 cells.


Assuntos
Autofagia/fisiologia , Estresse Nitrosativo/fisiologia , Proteínas Quinases/metabolismo , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Autofagia/efeitos dos fármacos , Humanos , Células MCF-7 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , NAD/metabolismo , Compostos Nitrosos/farmacologia , Oxirredução
17.
Artigo em Inglês | MEDLINE | ID: mdl-31285226

RESUMO

Tuberculosis (TB) is a global health concern, and this situation has further worsened due to the emergence of drug-resistant strains and the failure of BCG vaccine to impart protection. There is an imperative need to develop highly sensitive, specific diagnostic tools, novel therapeutics, and vaccines for the eradication of TB. In the present study, a chemical screen of a pharmacologically active compound library was performed to identify antimycobacterial compounds. The phenotypic screen identified a few novel small-molecule inhibitors, including NU-6027, a known CDK-2 inhibitor. We demonstrate that NU-6027 inhibits Mycobacterium bovis BCG growth in vitro and also displayed cross-reactivity with Mycobacterium tuberculosis protein kinase D (PknD) and protein kinase G (PknG). Comparative structural and sequence analysis along with docking simulation suggest that the unique binding site stereochemistry of PknG and PknD accommodates NU-6027 more favorably than other M. tuberculosis Ser/Thr protein kinases. Further, we also show that NU-6027 treatment induces the expression of proapoptotic genes in macrophages. Finally, we demonstrate that NU-6027 inhibits M. tuberculosis growth in both macrophage and mouse tissues. Taken together, these results indicate that NU-6027 can be optimized further for the development of antimycobacterial agents.


Assuntos
Antituberculosos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Mycobacterium bovis/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Compostos Nitrosos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirimidinas/farmacologia , Antituberculosos/química , Proteínas Reguladoras de Apoptose/agonistas , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Regulação da Expressão Gênica , Ensaios de Triagem em Larga Escala , Interações Hospedeiro-Patógeno , Macrófagos/metabolismo , Macrófagos/microbiologia , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Mycobacterium bovis/enzimologia , Mycobacterium bovis/genética , Mycobacterium bovis/crescimento & desenvolvimento , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crescimento & desenvolvimento , Compostos Nitrosos/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteína Quinase C/química , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Secundária de Proteína , Pirimidinas/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
18.
Theranostics ; 9(13): 3918-3939, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281522

RESUMO

Carbon monoxide and nitric oxide are two of the most important vasoprotective mediators. Their downregulation observed during vascular dysfunction, which is associated with cancer progression, leads to uncontrolled platelet activation. Therefore, the aim of our studies was to improve vasoprotection and to decrease platelet activation during progression of mouse mammary gland cancer by concurrent use of CO and NO donors (CORM-A1 and DETA/NO, respectively). Methods: Mice injected intravenously with 4T1-luc2-tdTomato or orthotopically with 4T1 mouse mammary gland cancer cells were treated with CORM-A1 and DETA/NO. Ex vivo aggregation and activation of platelets were assessed in the blood of healthy donors and breast cancer patients. Moreover, we analyzed the compounds' direct effect on 4T1 mouse and MDA-MB-231 human breast cancer cells proliferation, adhesion and migration in vitro. Results: We have observed antimetastatic effect of combination therapy, which was only transient in orthotopic model. During early stages of tumor progression concurrent use of CORM-A1 and DETA/NO demonstrated vasoprotective ability (decreased endothelin-1, sICAM and sE-selectin plasma level) and downregulated platelets activation (decreased bound of fibrinogen and vWf to platelets) as well as inhibited EMT process. Combined treatment with CO and NO donors diminished adhesion and migration of breast cancer cells in vitro and inhibited aggregation as well as TGF-ß release from breast cancer patients' platelets ex vivo. However, antimetastatic effect was not observed at a later stage of tumor progression which was accompanied by increased platelets activation and endothelial dysfunction related to a decrease of VASP level. Conclusion: The therapy was shown to have antimetastatic action and resulted in normalization of endothelial metabolism, diminution of platelet activation and inhibition of EMT process. The effect was more prominent during early stages of tumor dissemination. Such treatment could be applied to inhibit metastasis during the first stages of this process.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Boranos/farmacologia , Carbonatos/farmacologia , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/patologia , Óxido Nítrico/metabolismo , Compostos Nitrosos/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Boranos/uso terapêutico , Carbonatos/uso terapêutico , Bovinos , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Progressão da Doença , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Endotélio/efeitos dos fármacos , Endotélio/fisiopatologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Hidrazinas/farmacologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Animais/irrigação sanguínea , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos/metabolismo , Metástase Neoplásica , Óxido Nítrico/farmacologia , Compostos Nitrosos/uso terapêutico , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Fatores de Tempo
19.
Biomaterials ; 212: 28-38, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31102854

RESUMO

Metallic stents cause vascular wall damage with subsequent smooth muscle cell (SMC) proliferation, neointimal hyperplasia, and treatment failure. To combat in-stent restenosis, drug-eluting stents (DES) delivering mTOR inhibitors such as sirolimus or everolimus have become standard for coronary stenting. However, the relatively non-specific action of mTOR inhibitors prevents efficient endothelium recovery and mandates dual antiplatelet therapy to prevent thrombosis. Unfortunately, long-term dual antiplatelet therapy leads to increased risk of bleeding/stroke and, paradoxically, myocardial infarction. Here, we took advantage of the fact that nitric oxide (NO) increases Fas receptors on the SMC surface. Fas forms a death-inducing complex upon binding to Fas ligand (FasL), while endothelial cells (ECs) are relatively resistant to this pathway. Selected doses of FasL and NO donor synergistically increased SMC apoptosis and inhibited SMC growth more potently than did everolimus or sirolimus, while having no significant effect on EC viability and proliferation. This differential effect was corroborated in ex vivo pig coronaries, where the neointimal formation was inhibited by the drug combination, but endothelial viability was retained. We also deployed FasL-NO donor-releasing ethylene-vinyl acetate copolymer (EVAc)-coated stents into pig coronary arteries, and cultured them in perfusion bioreactors for one week. FasL and NO donor, released from the stent coating, killed SMCs close to the stent struts, even in the presence of flow rates mimicking those of native arteries. Thus, the FasL-NO donor-combination has a potential to prevent intimal hyperplasia and in-stent restenosis, without harming endothelial restoration, and hence may be a superior drug delivery strategy for DES.


Assuntos
Células Endoteliais/citologia , Proteína Ligante Fas/farmacologia , Miócitos de Músculo Liso/citologia , Óxido Nítrico/farmacologia , Sirolimo/farmacologia , Animais , Reatores Biológicos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Vasos Coronários/citologia , Células Endoteliais/efeitos dos fármacos , Everolimo/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Miócitos de Músculo Liso/efeitos dos fármacos , Compostos Nitrosos/farmacologia , Polímeros/química , Suínos
20.
Pediatr Blood Cancer ; 66(8): e27785, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31044544

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) is a heterogeneous disease regarding morphology, immunophenotyping, genetic abnormalities, and clinical behavior. The overall survival rate of pediatric AML is 60% to 70%, and has not significantly improved over the past two decades. Children with Down syndrome (DS) are at risk of developing acute megakaryoblastic leukemia (AMKL), which can be preceded by a transient myeloproliferative disorder during the neonatal period. Intensification of current treatment protocols is not feasible due to already high treatment-related morbidity and mortality. Instead, more targeted therapies with less severe side effects are highly needed. PROCEDURE: To identify potential novel therapeutic targets for myeloid disorders in children, including DS-AMKL and non-DS-AML, we performed an unbiased compound screen of 80 small molecules targeting epigenetic regulators in three pediatric AML cell lines that are representative for different subtypes of pediatric AML. Three candidate compounds were validated and further evaluated in normal myeloid precursor cells during neutrophil differentiation and in (pre-)leukemic pediatric patient cells. RESULTS: Candidate drugs LMK235, NSC3852, and bromosporine were effective in all tested pediatric AML cell lines with antiproliferative, proapoptotic, and differentiation effects. Out of these three compounds, the pan-histone deacetylase inhibitor NSC3852 specifically induced growth arrest and apoptosis in pediatric AML cells, without disrupting normal neutrophil differentiation. CONCLUSION: NSC3852 is a potential candidate drug for further preclinical testing in pediatric AML and DS-AMKL.


Assuntos
Ensaios de Seleção de Medicamentos Antitumorais/métodos , Epigênese Genética , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/química , Hidroxiquinolinas/farmacologia , Leucemia Mieloide Aguda/patologia , Compostos Nitrosos/farmacologia , Apoptose , Proliferação de Células , Criança , Síndrome de Down/tratamento farmacológico , Síndrome de Down/genética , Síndrome de Down/patologia , Ensaios de Triagem em Larga Escala , Histona Desacetilases/genética , Humanos , Leucemia Megacarioblástica Aguda/tratamento farmacológico , Leucemia Megacarioblástica Aguda/genética , Leucemia Megacarioblástica Aguda/patologia , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Reação Leucemoide/tratamento farmacológico , Reação Leucemoide/genética , Reação Leucemoide/patologia , Prognóstico , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...