Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Int J Toxicol ; 43(1_suppl): 50S-63S, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38146080

RESUMO

The Expert Panel for Cosmetic Ingredient Safety (Panel) assessed the safety of Adenosine, Adenosine Phosphate, Adenosine Triphosphate, Disodium Adenosine Phosphate, and Disodium Adenosine Triphosphate. These ingredients are reported to function in cosmetics as skin-conditioning agents - miscellaneous. The Panel considered the available data and concluded that the five adenosine ingredients reviewed in this report are safe in cosmetics in the present practices of use and concentration described in this safety assessment.


Assuntos
Adenosina , Cosméticos , Adenosina/toxicidade , Qualidade de Produtos para o Consumidor , Cosméticos/toxicidade , Trifosfato de Adenosina , Medição de Risco
2.
Bioorg Med Chem ; 46: 116364, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34450570

RESUMO

The nucleoside metabolite of remdesivir, GS-441524 displays potent anti-SARS-CoV-2 efficacy, and is being evaluated in clinical as an oral antiviral therapeutic for COVID-19. However, this nucleoside has a poor oral bioavailability in non-human primates, which may affect its therapeutic efficacy. Herein, we reported a variety of GS-441524 analogs with modifications on the base or the sugar moiety, as well as some prodrug forms, including five isobutyryl esters, two l-valine esters, and one carbamate. Among the new nucleosides, only the 7-fluoro analog 3c had moderate anti-SARS-CoV-2 activity, and its phosphoramidate prodrug 7 exhibited reduced activity in Vero E6 cells. As for the prodrugs, the 3'-isobutyryl ester 5a, the 5'-isobutyryl ester 5c, and the tri-isobutyryl ester 5g hydrobromide showed excellent oral bioavailabilities (F = 71.6%, 86.6% and 98.7%, respectively) in mice, which provided good insight into the pharmacokinetic optimization of GS-441524.


Assuntos
Adenosina/análogos & derivados , Antivirais/farmacologia , SARS-CoV-2/efeitos dos fármacos , Adenosina/farmacocinética , Adenosina/farmacologia , Adenosina/toxicidade , Animais , Antivirais/síntese química , Antivirais/farmacocinética , Antivirais/toxicidade , Chlorocebus aethiops , Masculino , Camundongos Endogâmicos ICR , Testes de Sensibilidade Microbiana , Pró-Fármacos/síntese química , Pró-Fármacos/farmacocinética , Pró-Fármacos/farmacologia , Pró-Fármacos/toxicidade , Células Vero
3.
Toxicol Sci ; 181(1): 13-22, 2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33616673

RESUMO

Epitranscriptomics, the study of chemically modified RNAs, is a burgeoning field being explored in a variety of scientific disciplines. Of the currently known epitranscriptomic modifications, N6-methyladenosine (m6A) methylation is the most abundant. The m6A modification is predominantly regulated by 3 tiers of protein modulators classified as writers, erasers, and readers. Depending upon cellular needs, these proteins function to deposit, remove, or read the methyl modifications on cognate mRNAs. Many environmental chemicals including heavy metals, pesticides, and other toxic pollutants, are all known to perturb transcription and translation machinery to exert their toxic responses. As such, we herein review how the m6A modification may be affected under different toxicological paradigms. Furthermore, we discuss how toxicants can affect the 3 tiers of regulation directly, and how these effects influence the m6A-modified mRNAs. Lastly, we highlight the disparities between published findings and theories, especially those concerning the m6A reader tier of regulation. In the far-reaching field of toxicology, m6A epitranscriptomics provides another enticing avenue to explore new mechanisms and therapies for a diverse range of environmentally linked disorders and diseases.


Assuntos
Adenosina , RNA , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/toxicidade , Humanos , Metilação , RNA Mensageiro/metabolismo
4.
Stem Cells ; 39(2): 170-182, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33159685

RESUMO

Heart transplantation is a life-saving therapy for end-stage organ failure. Organ deterioration during transportation limits storage to 4 hours, limiting hearts available. Approaches ameliorating organ damage could increase the number of hearts acceptable for transplantation. Prior studies show that adipose-derived stem/stromal cell secretome (ASC-S) rescues tissues from postischemic damage in vivo. This study tested whether ASC-S preserved the function of mouse hearts and human induced pluripotent stem cell-derived cardiomyocytes (iCM) exposed to organ transportation and transplantation conditions. Hearts were subjected to cold University of Wisconsin (UW) cardioplegic solution ± ASC-S for 6 hours followed by analysis using the Langendorff technique. In parallel, the effects of ASC-S on the recovery of iCM from UW solution were examined when provided either during or after cold cardioplegia. Exposure of hearts and iCM to UW deteriorated contractile activity and caused cell apoptosis, worsening in iCM as a function of exposure time; these were ameliorated by augmenting with ASC-S. Silencing of superoxide dismutase 3 and catalase expression prior to secretome generation compromised the ASC-S cardiomyocyte-protective effects. In this study, a novel in vitro iCM model was developed to complement a rodent heart model in assessing efficacy of approaches to improve cardiac preservation. ASC-S displays strong cardioprotective activity on iCM either with or following cold cardioplegia. This effect is associated with ASC-S-mediated cellular clearance of reactive oxygen species. The effect of ASC-S on the temporal recovery of iCM function supports the possibility of lengthening heart storage by augmenting cardioplegic transport solution with ASC-S, expanding the pool of hearts for transplantation.


Assuntos
Soluções Cardioplégicas/toxicidade , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Miócitos Cardíacos/metabolismo , Soluções para Preservação de Órgãos/toxicidade , Recuperação de Função Fisiológica/fisiologia , Adenosina/toxicidade , Alopurinol/toxicidade , Animais , Glutationa/toxicidade , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Insulina/toxicidade , Preparação de Coração Isolado/métodos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Rafinose/toxicidade , Recuperação de Função Fisiológica/efeitos dos fármacos
5.
Eur J Med Chem ; 201: 112408, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32574901

RESUMO

Lipid metabolism in Mycobacterium tuberculosis (Mtb) relies on 34 fatty acid adenylating enzymes (FadDs) that can be grouped into two classes: fatty acyl-CoA ligases (FACLs) involved in lipid and cholesterol catabolism and long chain fatty acyl-AMP ligases (FAALs) involved in biosynthesis of the numerous essential and virulence-conferring lipids found in Mtb. The precise biochemical roles of many FACLs remain poorly characterized while the functionally non-redundant FAALs are much better understood. Here we describe the systematic investigation of 5'-O-[N-(alkanoyl)sulfamoyl]adenosine (alkanoyl adenosine monosulfamate, alkanoyl-AMS) analogs as potential multitarget FadD inhibitors for their antitubercular activity and biochemical selectivity towards representative FAAL and FACL enzymes. We identified several potent compounds including 12-azidododecanoyl-AMS 28, 11-phenoxyundecanoyl-AMS 32, and nonyloxyacetyl-AMS 36 with minimum inhibitory concentrations (MICs) against M. tuberculosis ranging from 0.098 to 3.13 µM. Compound 32 was notable for its impressive biochemical selectivity against FAAL28 (apparent Ki = 0.7 µM) versus FACL19 (Ki > 100 µM), and uniform activity against a panel of multidrug and extensively drug-resistant TB strains with MICs ranging from 3.13 to 12.5 µM in minimal (GAST) and rich (7H9) media. The SAR analysis provided valuable insights for further optimization of 32 and also identified limitations to overcome.


Assuntos
Adenosina/análogos & derivados , Adenosina/farmacologia , Antituberculosos/farmacologia , Coenzima A Ligases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Adenosina/toxicidade , Animais , Antituberculosos/síntese química , Antituberculosos/toxicidade , Proteínas de Bactérias/antagonistas & inibidores , Chlorocebus aethiops , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/toxicidade , Testes de Sensibilidade Microbiana , Estrutura Molecular , Mycobacterium tuberculosis/enzimologia , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/farmacologia , Sulfonamidas/toxicidade , Células Vero
6.
J Antibiot (Tokyo) ; 72(12): 981-985, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31534199

RESUMO

Feline herpesvirus type 1 (FHV-1) causes a potentially fatal disease in cats. Through the use of virus inhibition and cytotoxicity assays, sinefungin, a nucleoside antibiotic, was assessed for its potential to inhibit the growth of FHV-1. Sinefungin inhibited in vitro growth of FHV-1 most significantly over other animal viruses, such as feline infectious peritonitis virus, equine herpesvirus, pseudorabies virus and feline calicivirus. Our results revealed that sinefungin specifically suppressed the replication of FHV-1 after its adsorption to the host feline kidney cells in a dose-dependent manner without obvious cytotoxicity to the host cells. This antibiotic can potentially offer a highly effective treatment for animals infected with FHV-1, providing alternative medication to currently available antiviral therapies.


Assuntos
Adenosina/análogos & derivados , Antivirais/farmacologia , Varicellovirus/efeitos dos fármacos , Adenosina/farmacologia , Adenosina/toxicidade , Animais , Antivirais/toxicidade , Calicivirus Felino/efeitos dos fármacos , Doenças do Gato/tratamento farmacológico , Gatos , Linhagem Celular , Coronavirus Felino/efeitos dos fármacos , Relação Dose-Resposta a Droga , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/efeitos dos fármacos , Herpesvirus Suídeo 1/efeitos dos fármacos , Cavalos , Rim/citologia , Rim/virologia , Testes de Toxicidade
7.
Acta Trop ; 190: 304-311, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30529445

RESUMO

Plant-based constituents have been proposed as eco-friendly alternatives to synthetic insecticides for control of mosquito vectors of malaria. In this study, we first screened the effects of methanolic leaf extracts of curry tree (Murraya koenigii) growing in tropical (Mombasa, Malindi) and semi-arid (Kibwezi, and Makindu) ecological zones of Kenya on third instar An. gambiae s.s. larvae. Extracts of the plant from the semi-arid region, and particularly from Kibwezi, led to high mortality of the larvae. Bioassay-guided fractionation of the methanolic extract of the leaves of the plants from Kibwezi was then undertaken and the most active fraction (20 fold more potent than the crude extract) was then analyzed by Liquid chromatography quadruple time of flight coupled with mass spectrometry (LC-QtoF-MS) and a number of constituents were identified, including a major alkaloid constituent, Neplanocin A (5). Exposure of the third instar larvae to a sub-lethal dose (4.43 ppm) of this fraction over 7-day periods induced gross morphogenetic abnormalities in the larvae, with reduced locomotion, and delayed pupation. Moreover, the few adults that emerged from some pupae failed to fly from the water surface, unlike in the untreated control group. These results demonstrate subtle growth-disrupting effects of the phytochemical blend from M. koenigii leaves on aquatic stages An. gambiae mosquito. The study lays down some useful groundwork for the downstream development of phytochemical blends that can be evaluated for integration into eco-friendly control of An. gambiae vector population targeting the often overlooked but important immature stages of the malaria vector.


Assuntos
Adenosina/análogos & derivados , Anopheles/efeitos dos fármacos , Larva/crescimento & desenvolvimento , Murraya , Extratos Vegetais/química , Extratos Vegetais/toxicidade , Adenosina/análise , Adenosina/toxicidade , Animais , Cromatografia Líquida , Feminino , Inseticidas/toxicidade , Quênia , Larva/efeitos dos fármacos , Larva/fisiologia , Locomoção/efeitos dos fármacos , Espectrometria de Massas , Folhas de Planta/química
8.
Sci Rep ; 8(1): 2717, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29426934

RESUMO

We examined whether orexin neurons might play a protective role against fasting- and adenosine-induced hypothermia. We first measured body temperature (BT) in orexin neuron-ablated (ORX-AB) mice and wild-type (WT) controls during 24 hours of fasting. As expected, the magnitude of BT drop and the length of time suffering from hypothermia were greater in ORX-AB mice than in WT mice. Orexin neurons were active just before onset of hypothermia and during the recovery period as revealed by calcium imaging in vivo using G-CaMP. We next examined adenosine-induced hypothermia via an intracerebroventricular administration of an adenosine A1 receptor agonist, N6-cyclohexyladenosine (CHA), which induced hypothermia in both ORX-AB and WT mice. The dose of CHA required to initiate a hypothermic response in ORX-AB mice was more than 10 times larger than the dose for WT mice. Once hypothermia was established, the recovery was seemingly slower in ORX-AB mice. Activation of orexin neurons during the recovery phase was confirmed by immunohistochemistry for c-Fos. We propose that orexin neurons play dual roles (enhancer in the induction phase and compensator during the recovery phase) in adenosine-induced hypothermia and a protective/compensatory role in fasting-induced hypothermia.


Assuntos
Adenosina/análogos & derivados , Regulação da Temperatura Corporal , Jejum , Hipotermia/prevenção & controle , Neurônios/efeitos dos fármacos , Orexinas/fisiologia , Adenosina/toxicidade , Agonistas do Receptor A1 de Adenosina/toxicidade , Animais , Hipotermia/induzido quimicamente , Masculino , Camundongos , Camundongos Knockout , Neurônios/patologia , Receptor A1 de Adenosina/química
9.
Toxicol Appl Pharmacol ; 338: 9-19, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29122671

RESUMO

AZD8848 is a TLR7 agonist antedrug developed for administration by inhalation dosing for the treatment of allergic diseases, such as asthma. Allergic asthma is associated with increased levels of Th2 cytokines which are suppressed for extended periods by TLR7 agonists in a number of preclinical models of allergic airway inflammation. However, TLRs form a central part of innate immunity and their activation often results in proinflammatory responses. Whilst AZD8848's antedrug mechanism is designed to restrict its pharmacological action beyond the lung, the effect of chronic, supramaximal dosing to the target tissue has yet to be defined. To support clinical development of this potentially disease modifying approach the nonclinical safety and pharmacodynamics of AZD8848 were evaluated in cynomolgus monkeys in studies examining single or multiple weekly inhaled doses. Here we show that following a single dose nearly all responses returned to baseline within a week. During multiple dosing serum biomarkers were quantified over the dosing period and indicated a limited systemic response. The dose at which maximal interferon responses were seen was dependent on dose. Thorough histopathological examination revealed a dose related increase of size and cells of lymphoid tissues in the lung and nose. Local lymphoid responses were recovered after the treatment free period. These studies are the first to evaluate safety of an inhaled TLR7 agonist and demonstrate AZD8848 is safe with a no observed adverse effect level at 26µg/kg allowing progression to man with weekly inhalation dosing.


Assuntos
Adenosina/análogos & derivados , Citocinas/sangue , Tecido Linfoide/efeitos dos fármacos , Fenilacetatos/toxicidade , Receptor 7 Toll-Like/agonistas , Adenosina/administração & dosagem , Adenosina/farmacocinética , Adenosina/toxicidade , Administração por Inalação , Animais , Citocinas/genética , Feminino , Tecido Linfoide/imunologia , Macaca fascicularis , Masculino , Fenilacetatos/administração & dosagem , Fenilacetatos/farmacocinética
10.
Neuroimmunomodulation ; 24(1): 54-59, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28793299

RESUMO

OBJECTIVE: It has been suggested that the adenosinergic system and cytokines play a role in the pathogenesis of epilepsy. Although the role of the adenosinergic system in the modulation of seizure activity is well known, the mechanism of this modulation needs to be described in detail. We performed this study to determine the contribution of the proinflammatory cytokines to the generalized seizure activity during adenosine and caffeine treatment. METHODS: We induced generalized tonic-clonic seizures with the administration of 60 mg/kg pentylenetetrazole (PTZ) in male Wistar Albino rats. Adenosine (500 mg/kg) or caffeine (5 mg/kg) was administered before PTZ injection. We monitored seizure activity and then determined the TNF-α, IL-1ß, and IL-6 levels in the cortical and thalamic brain regions of rats by ELISA. RESULTS: Adenosine pretreatment significantly extended seizure latency (p < 0.05), but did not affect seizure duration and entry time to stage 4 seizure. Caffeine pretreatment did not change seizure latency and seizure duration. PTZ treatment did not change brain cytokine levels significantly (p > 0.05) compared to the control group. Whereas adenosine pretreatment decreased brain TNF-α, IL-1ß, and IL-6 levels significantly (p < 0.05), caffeine pretreatment reduced brain cytokine levels slightly but nonsignificantly (p > 0.05). CONCLUSION: Our results show that there is a clear relation between adenosinergic system and brain tissue cytokine levels. Our findings indicated that TNF-α, IL-1ß, and IL-6 participate in the pathogenesis of generalized seizures, and the inhibition of TNF-α, IL-1ß, and IL-6 with adenosinergic modulation may decrease seizure severity.


Assuntos
Encéfalo/metabolismo , Citocinas/metabolismo , Convulsões/imunologia , Adenosina/toxicidade , Animais , Encéfalo/efeitos dos fármacos , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Convulsivantes/toxicidade , Modelos Animais de Doenças , Interações Medicamentosas , Masculino , Pentilenotetrazol/toxicidade , Agonistas do Receptor Purinérgico P1/toxicidade , Ratos , Ratos Wistar , Convulsões/induzido quimicamente
11.
Bioorg Med Chem ; 25(17): 4579-4594, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28739157

RESUMO

A series of cycloalkyl substituted analogues of the natural product sinefungin lacking the amino-acid moiety was designed and synthesized. Two stereoisomers (6-R and 6-S) were separated and their bioactivities examined against EHMT1/2. Of which, compound 14d showed an inhibitory activity against EHMT1/2 (88.9%, IC50=21.8µM for EHMT1 and 77.6%, IC50=39.6µM for EHMT2, respectively) similar to that of sinefungin (100.0%, IC50=28.4µM for EHMT1 and 79.5%, IC50=30.1µM for EHMT2, respectively). Further studies against other methyltransferases such as PRMT1 showed no activity except that 12d displayed about 20% inhibition.


Assuntos
Adenosina/análogos & derivados , Inibidores Enzimáticos/química , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Adenosina/química , Adenosina/metabolismo , Adenosina/toxicidade , Alcanos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/toxicidade , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Concentração Inibidora 50 , Relação Estrutura-Atividade
12.
Thromb Haemost ; 117(8): 1651-1659, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28569920

RESUMO

The efficiency of current dual antiplatelet therapy might be further improved by its combination with a glycoprotein (GP) VI-targeting strategy without increasing bleeding. GPVI-Fc, a recombinant dimeric fusion protein binding to plaque collagen and concealing binding sites for platelet GPVI, acts as a lesion-focused antiplatelet drug, and does not increase bleeding in vivo. We investigated, whether GPVI-Fc added in vitro on top of acetylsalicylic acid (ASA), the P2Y12 antagonist ticagrelor, and the fibrinogen receptor antagonist abciximab alone or in combination would increase inhibition of platelet activation by atherosclerotic plaque. Under static conditions, GPVI-Fc inhibited plaque-induced platelet aggregation by 53 %, and increased platelet inhibition by ASA (51 %) and ticagrelor (64 %) to 66 % and 80 %, respectively. Under arterial flow, GPVI-Fc inhibited plaque-induced platelet aggregation by 57 %, and significantly increased platelet inhibition by ASA (28 %) and ticagrelor (47 %) to about 81 % each. The triple combination of GPVI-Fc, ASA and ticagrelor achieved almost complete inhibition of plaque-induced platelet aggregation (93 %). GPVI-Fc alone or in combination with ASA or ticagrelor did not increase closure time measured by the platelet function analyzer (PFA)-200. GPVI-Fc added on top of abciximab, a clinically used anti-fibrinogen receptor antibody which blocks platelet aggregation, strongly inhibited total (81 %) and stable (89 %) platelet adhesion. We conclude that GPVI-Fc added on top of single or dual antiplatelet therapy with ASA and/or a P2Y12 antagonist is likely to improve anti-atherothrombotic protection without increasing bleeding risk. In contrast, the strong inhibition of platelet adhesion by GPVI-Fc in combination with GPIIb/IIIa inhibitors could be harmful.


Assuntos
Adenosina/análogos & derivados , Anticorpos Monoclonais/farmacologia , Aspirina/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Glicoproteínas/farmacologia , Fragmentos Fab das Imunoglobulinas/farmacologia , Fragmentos Fc das Imunoglobulinas/farmacologia , Placa Aterosclerótica , Inibidores da Agregação Plaquetária/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Trombose/prevenção & controle , Abciximab , Adenosina/farmacologia , Adenosina/toxicidade , Anticorpos Monoclonais/toxicidade , Aspirina/toxicidade , Plaquetas/metabolismo , Quimioterapia Combinada , Glicoproteínas/toxicidade , Hemorragia/induzido quimicamente , Humanos , Fragmentos Fab das Imunoglobulinas/toxicidade , Fragmentos Fc das Imunoglobulinas/toxicidade , Adesividade Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/toxicidade , Antagonistas do Receptor Purinérgico P2Y/toxicidade , Trombose/sangue , Trombose/patologia , Ticagrelor , Fatores de Tempo
13.
Cell Prolif ; 50(4)2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28370734

RESUMO

OBJECTIVES: Breast cancer stem cells (CSCs) are a small population of tumour cells with the ability of self-renewal and resistance to chemotherapy. Targeting CSCs is a promising strategy for treatment of cancer. A recent study demonstrated that adenosine receptor agonists inhibit glioblastoma CSCs proliferation. At present, the effect of adenosine on breast CSCs has not been reported. Therefore, this study was designed to evaluate the effect of adenosine and its signalling pathways in breast CSCs. MATERIALS AND METHODS: Anti-proliferative effect of adenosine on breast CSCs was evaluated by mammosphere formation and MTS assay. The effect of adenosine on cell cycle progression was examined using flow cytometry. Detection of apoptosis was conducted by Annexin V-FITC. The expression levels of cell cycle and apoptosis regulatory proteins as well as ERK1/2, and GLI-1 were measured by Western blot. RESULTS: Adenosine reduced CSCs population and mammosphere formation in breast CSCs. Adenosine induced G1 cell cycle arrest in breast CSCs in conjunction with a marked down-regulation of cyclin D1 and CDK4. Adenosine also induced apoptosis by regulation of Bax/Bcl-2 ratio, mitochondrial membrane potential depletion and activation of caspase-6. Moreover, adenosine inhibited ERK1/2 phosphorylation and GLI-1 protein expression. CONCLUSIONS: These findings indicated that adenosine induces cell cycle arrest and apoptosis through inhibition of GLI-1 and ERK1/2 pathways in breast CSCs.


Assuntos
Adenosina/toxicidade , Apoptose/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Proteína GLI1 em Dedos de Zinco/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Antígeno CD24/metabolismo , Caspase 6/metabolismo , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Receptores de Hialuronatos/metabolismo , Sistema de Sinalização das MAP Quinases , Células MCF-7 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Células-Tronco Neoplásicas/citologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
14.
J Clin Pathol ; 70(9): 733-739, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28159767

RESUMO

AIM: Management of ticagrelor-induced bleeding is challenging as platelet transfusion is ineffective. An effective strategy is needed. This study aimed to investigate in vitro the efficacy of four haemostatic drugs (HDs), namely recombinant activated factor VII (rFVIIa), fibrinogen concentrate (Fib), tranexamic acid (TXA) and factor XIII concentrate (FXIII) to improve the haemostatic capacity in the presence of ticagrelor. METHODS: Blood was spiked with ticagrelor then supplemented by either HD or control. Several assays were performed: ADP-induced platelet aggregation measured by impedance aggregometry, light transmission and two global assays, thrombolastography with the platelet mapping device (TEG-PM) and a platelet-dependent thrombin generation assay (TGA). RESULTS: Ticagrelor inhibited ADP-induced platelet aggregation and decreased the clot strength maximum amplitude (MA) in TEG-PMADP. None of the HDs corrected these parameters. However, rFVIIa shortened the coagulation time R using TEG-PMthrombin and the time to peak prolonged by ticagrelor in TGA. Fib increased MAthrombin and FXIII decreased LY30. TXA had no effects. CONCLUSIONS: Whereas none of the HDs corrected ticagrelor-induced platelet inhibition, rFVIIa shortened coagulation times, Fib increased clot firmness and FXIII decreased fibrinolysis. Consequently, they may bypass ticagrelor effects by acting on fibrin formation or fibrinolysis. Further studies are needed to confirm these data in vivo.


Assuntos
Adenosina/análogos & derivados , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Hemorragia/prevenção & controle , Hemostáticos/farmacologia , Inibidores da Agregação Plaquetária/toxicidade , Adenosina/toxicidade , Plaquetas/metabolismo , Relação Dose-Resposta a Droga , Fator VIIa/farmacologia , Fibrinogênio/farmacologia , Fibrinolisina/farmacologia , Hemorragia/sangue , Hemorragia/induzido quimicamente , Humanos , Agregação Plaquetária/efeitos dos fármacos , Testes de Função Plaquetária , Proteínas Recombinantes/farmacologia , Tromboelastografia , Ticagrelor , Fatores de Tempo , Ácido Tranexâmico/farmacologia
15.
J Pain ; 18(5): 574-582, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28089711

RESUMO

Hyperalgesic priming, a sexually dimorphic model of transition to chronic pain, is expressed as prolongation of prostaglandin E2-induced hyperalgesia by the activation of an additional pathway including an autocrine mechanism at the plasma membrane. The autocrine mechanism involves the transport of cyclic adenosine monophosphate (AMP) to the extracellular space, and its conversion to AMP and adenosine, by ecto-5'phosphodiesterase and ecto-5'nucleotidase, respectively. The end product, adenosine, activates A1 receptors, producing delayed onset prolongation of prostaglandin E2 hyperalgesia. We tested the hypothesis that the previously reported, estrogen-dependent, sexual dimorphism observed in the induction of priming is present in the mechanisms involved in its expression, as a regulatory effect on ecto-5'nucleotidase by estrogen receptor α (EsRα), in female rats. In the primed paw AMP hyperalgesia was dependent on conversion to adenosine, being prevented by ecto-5'nucleotidase inhibitor α,ß-methyleneadenosine 5'-diphosphate sodium salt and A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine. To investigate an interaction between EsRα and ecto-5'nucleotidase, we treated primed female rats with oligodeoxynucleotide antisense or mismatch against EsRα messenger RNA. Whereas in rats treated with antisense AMP-induced hyperalgesia was abolished, the A1 receptor agonist N6-cyclopentiladenosine still produced hyperalgesia. Thus, EsRα interacts with this autocrine pathway at the level of ecto-5'nucleotidase. These results demonstrate a sexually dimorphic mechanism for the expression of priming. PERSPECTIVE: This study presents evidence of an estrogen-dependent mechanism of expression of chronic pain in female rats, supporting the suggestion that differential targets must be considered when establishing protocols for the treatment of painful conditions in men and women.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica/fisiologia , 5'-Nucleotidase/metabolismo , Adenosina/análogos & derivados , Adenosina/toxicidade , Antagonistas do Receptor A1 de Adenosina/toxicidade , Monofosfato de Adenosina/toxicidade , Animais , Dor Crônica/induzido quimicamente , Dor Crônica/fisiopatologia , DNA Antissenso/uso terapêutico , Dinoprostona/toxicidade , Modelos Animais de Doenças , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia/tratamento farmacológico , Masculino , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Rianodina/toxicidade , Fatores Sexuais , Fatores de Tempo , Xantinas/toxicidade
16.
Circulation ; 134(6): 486-98, 2016 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-27462069

RESUMO

BACKGROUND: Adenosine provokes atrial fibrillation (AF) with a higher activation frequency in right atria (RA) versus left atria (LA) in patients, but the underlying molecular and functional substrates are unclear. We tested the hypothesis that adenosine-induced AF is driven by localized reentry in RA areas with highest expression of adenosine A1 receptor and its downstream GIRK (G protein-coupled inwardly rectifying potassium channels) channels (IK,Ado). METHODS: We applied biatrial optical mapping and immunoblot mapping of various atrial regions to reveal the mechanism of adenosine-induced AF in explanted failing and nonfailing human hearts (n=37). RESULTS: Optical mapping of coronary-perfused atria (n=24) revealed that adenosine perfusion (10-100 µmol/L) produced more significant shortening of action potential durations in RA (from 290±45 to 239±41 ms, 17.3±10.4%; P<0.01) than LA (from 307±24 to 286±23 ms, 6.7±6.6%; P<0.01). In 10 hearts, adenosine induced AF (317±116 s) that, when sustained (≥2 minutes), was primarily maintained by 1 to 2 localized reentrant drivers in lateral RA. Tertiapin (10-100 nmol/L), a selective GIRK channel blocker, counteracted adenosine-induced action potential duration shortening and prevented AF induction. Immunoblotting showed that the superior/middle lateral RA had significantly higher adenosine A1 receptor (2.7±1.7-fold; P<0.01) and GIRK4 (1.7±0.8-fold; P<0.05) protein expression than lateral/posterior LA. CONCLUSIONS: This study revealed a 3-fold RA-to-LA adenosine A1 receptor protein expression gradient in the human heart, leading to significantly greater RA versus LA repolarization sensitivity in response to adenosine. Sustained adenosine-induced AF is maintained by reentrant drivers localized in lateral RA regions with the highest adenosine A1 receptor/GIRK4 expression. Selective atrial GIRK channel blockade may effectively treat AF during conditions with increased endogenous adenosine.


Assuntos
Adenosina/toxicidade , Fibrilação Atrial/induzido quimicamente , Fibrilação Atrial/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/biossíntese , Átrios do Coração/metabolismo , Receptor A1 de Adenosina/biossíntese , Adulto , Idoso , Feminino , Regulação da Expressão Gênica , Coração/diagnóstico por imagem , Coração/efeitos dos fármacos , Átrios do Coração/diagnóstico por imagem , Átrios do Coração/efeitos dos fármacos , Sistema de Condução Cardíaco/diagnóstico por imagem , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada
17.
Eur J Anaesthesiol ; 33(5): 361-7, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26351831

RESUMO

BACKGROUND: Management of ticagrelor-induced bleeding is challenging, as no antidote is currently available. Platelet transfusion, usually proposed to reverse antiplatelet drugs, has been suggested to be ineffective but few data are available. OBJECTIVE: To assess the efficacy of platelet supplementation to restore platelet aggregation inhibited by ticagrelor. DESIGN: In vitro study. SETTING: Blood samples were obtained from the French Blood Bank Institute. PARTICIPANTS: Healthy blood donors. INTERVENTIONS: Whole blood from healthy donors was spiked with ticagrelor or aspirin (used as a positive control). MAIN OUTCOME MEASURES: Platelet aggregation was investigated with impedance aggregometry on whole blood [expressed in ohms (V)] and light transmission aggregometry (expressed in %) on platelet-rich plasma using ADP or arachidonic acid as agonists for ticagrelor or aspirin, respectively. Platelet supplementation was defined as the addition of washed platelet suspension increasing at least 60% of whole blood platelet count. RESULTS: Ticagrelor (3.25 mM) inhibited ADP-induced platelet aggregation compared with control either in whole blood (2 vs. 13 V, P < 0.05) or in platelet-rich plasma (15 vs. 75% P < 0.05). Aspirin (25 mM) inhibited arachidonic acid-induced aggregation (1 vs. 7.5 V, P < 0.05 in whole blood and 5 vs. 77.5%, P = 0.01 in platelet-rich plasma). Platelet supplementation completely restored arachidonic acid-induced platelet aggregation in whole blood (10 vs. 1 V, P = 0.008) and platelet-rich plasma (73 vs. 5%, P < 0.01) in aspirin-treated samples, whereas it failed to correct ADP-induced aggregation (2 vs. 2 V in whole blood and 13.5 vs. 15% in platelet-rich plasma, P > 0.05) in ticagrelor-treated samples. We also report a case of a ticagrelor-treated patient in whom platelet transfusion failed to restore ADP-induced platelet aggregation. CONCLUSION: Platelet supplementation restored platelet aggregation in aspirin-spiked but not in ticagrelor-spiked samples. These results do not support the use of platelet transfusion to reverse the effects of ticagrelor.


Assuntos
Adenosina/análogos & derivados , Inibidores da Agregação Plaquetária/toxicidade , Agregação Plaquetária/efeitos dos fármacos , Transfusão de Plaquetas , Adenosina/toxicidade , Difosfato de Adenosina/farmacologia , Ácido Araquidônico/farmacologia , Aspirina/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Testes de Função Plaquetária , Ticagrelor
18.
Pain ; 157(3): 698-709, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26588695

RESUMO

We have recently shown that repeated exposure of the peripheral terminal of the primary afferent nociceptor to the mu-opioid receptor (MOR) agonist DAMGO ([D-Ala, N-Me-Phe, Gly-ol]-enkephalin acetate salt) induces a model of transition to chronic pain that we have termed type II hyperalgesic priming. Similar to type I hyperalgesic priming, there is a markedly prolonged response to subsequent administration of proalgesic cytokines, prototypically prostaglandin E2 (PGE2). However, type II hyperalgesic priming differs from type I in being rapidly induced, protein kinase A (PKA), rather than PKCε dependent, not reversed by a protein translation inhibitor, occurring in female as well as in male rats, and isolectin B4-negative neuron dependent. We report that, as with the repeated injection of a MOR agonist, the repeated administration of an agonist at the A1-adenosine receptor, also a Gi-protein coupled receptor, N-cyclopentyladenosine (CPA), also produces priming similar to DAMGO-induced type II hyperalgesic priming. In this study, we demonstrate that priming induced by repeated exposure to this A1-adenosine receptor agonist shares the same mechanisms, as MOR-agonist induced priming. However, the prolongation of PGE2 hyperalgesia induced by repeated administration of CPA depends on G-protein αi subunit activation, differently from DAMGO-induced type II priming, in which it depends on the ß/γ subunit. These data implicate a novel form of Gi-protein signaling pathway in the type II hyperalgesic priming induced by repeated administration of an agonist at A1-adenosine receptor to the peripheral terminal of the nociceptor.


Assuntos
Agonistas do Receptor A1 de Adenosina/toxicidade , Adenosina/análogos & derivados , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Receptor A1 de Adenosina/metabolismo , Adenosina/toxicidade , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/toxicidade , Feminino , Hiperalgesia/patologia , Masculino , Ratos , Ratos Sprague-Dawley
19.
Purinergic Signal ; 11(3): 371-87, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26111639

RESUMO

We reported that 2-(3,4-difluorophenylethynyl)-N (6)-3-chlorobenzyl (N)-methanocarba adenosine derivative 1 (MRS5698) binds selectively to human and mouse A3 adenosine receptors (A3ARs, K i 3 nM). It is becoming an important pharmacological tool for defining A3AR effects and is orally active in a chronic neuropathic pain model. Here, we introduce a new synthetic route for MRS5698 from D-ribose, suitable for a scale-up on a multi-gram scale, and we measure in vitro and in vivo ADME-Tox parameters. MRS5698 was very stable in vitro, failed to inhibit CYPs at <10 µM, and was largely bound to plasma proteins. It was well tolerated in the rat at doses of ≤200 mg/kg i.p. A 1 mg/kg i.p. dose in the mouse displayed t 1/2 of 1.09 h and plasma Cmax of 204 nM at 1 h with an AUC of 213 ng × h/mL. CACO-2 bidirectional transport studies suggested intestinal efflux of MRS5698 (efflux ratio 86). Although the oral %F is only 5 %, the beneficial effect to reverse pain lasted for at least 2 h in the CCI model in rats, using the same vehicle for oral administration of a high dose. The stability, low toxicity, lack of CYP interaction, pharmacokinetic half-life, and in vivo efficacy suggest that MRS5698 is a preferred compound for further consideration as a treatment for neuropathic pain.


Assuntos
Agonistas do Receptor A3 de Adenosina/uso terapêutico , Adenosina/análogos & derivados , Neuralgia/tratamento farmacológico , Adenosina/farmacocinética , Adenosina/uso terapêutico , Adenosina/toxicidade , Agonistas do Receptor A3 de Adenosina/farmacocinética , Agonistas do Receptor A3 de Adenosina/toxicidade , Animais , Transporte Biológico Ativo , Proteínas Sanguíneas/metabolismo , Células CACO-2 , Doença Crônica , Constrição Patológica/complicações , Meia-Vida , Humanos , Técnicas In Vitro , Masculino , Microssomos Hepáticos , Testes de Mutagenicidade , Ligação Proteica , Ratos , Ratos Sprague-Dawley
20.
ChemMedChem ; 10(1): 173-82, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25319940

RESUMO

3-Deazaneplanocin A (DzNep) is a potential epigenetic drug for the treatment of various cancers. DzNep has been reported to deplete histone methylations, including oncogenic EZH2 complex, giving rise to epigenetic modifications that reactivate many silenced tumor suppressors in cancer cells. Despite its promise as an anticancer drug, little is known about the structure-activity relationships of DzNep in the context of epigenetic modifications and apoptosis induction. In this study, a number of analogues of DzNep were examined for DzNep-like ability to induce synergistic apoptosis in cancer cells in combination with trichostatin A, a known histone deacetylase (HDAC) inhibitor. The structure-activity relationship data thus obtained provide valuable information on the structural requirements for biological activity. The studies identified three compounds that show similar activities to DzNep. Two of these compounds show good pharmacokinetics and safety profiles. Attempts to correlate the observed synergistic apoptotic activities with measured S-adenosylhomocysteine hydrolase (SAHH) inhibitory activities suggest that the apoptotic activity of DzNep might not be directly due to its inhibition of SAHH.


Assuntos
Adenosina/análogos & derivados , Adenosina/química , Adenosina/uso terapêutico , Adenosina/toxicidade , Adenosil-Homocisteinase/antagonistas & inibidores , Adenosil-Homocisteinase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/toxicidade , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/tratamento farmacológico , Relação Estrutura-Atividade , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA