RESUMEN
Acute myocardial infarction (AMI) is a prevalent cardiovascular disease with high morbidity and mortality rates worldwide. Pyroptosis is an inflammatory form of programmed cell death that has been linked to various pathological conditions. However, its exact contribution to the onset and progression of heart injury in AMI has not yet fully elucidated. Herein, we established mouse AMI model by ligating the left anterior descending artery and performed transcriptome analysis during the early phase of AMI. Mouse HL-1 and human AC-16 cardiomyocytes were subjected to hypoxia to simulate ischemic injury in vitro. Our results revealed a significant activation of the inflammatory response at 3 h post-ligation, as confirmed by RNA sequencing. We identified the occurrence of NLRP3 inflammasome-mediated pyroptosis in the cardiac tissues of human cases with AMI, as well as in mouse models of AMI and hypoxia-induced cardiomyocytes, using immunohistochemistry staining and Western blotting assays. Concurrently, pharmacological inhibition of NLRP3 inflammasome-mediated pyroptosis with MCC950 and VX-765 effectively decreased hypoxia-induced cardiomyocytes injury, while mitigating myocardial oxidative stress, apoptosis and inflammation caused by hypoxia. Moreover, the circulating levels of gasdermin D (GSDMD), the pyroptosis executor, were remarkably elevated in the plasma of mice with early AMI and in the supernatant of hypoxia-exposed cardiomyocytes in a time-dependent manner using ELISA and Western blotting. Furthermore, the change in circulating GSDMD positively correlated with Creatine Kinase-MB (CK-MB) in the plasma of early-stage AMI mouse. In summary, these findings indicated a critical role for NLRP3 inflammasome-mediated pyroptosis in the progression of AMI, the administration of MCC950 and VX-765 may be attractive candidate therapeutic approaches for cardiac injury caused by acute hypoxia or even AMI. Additionally, the circulating GSDMD exhibits potential as a newly diagnostic biomarker for AMI.
Asunto(s)
Apoptosis , Furanos , Inflamación , Ratones Endogámicos C57BL , Infarto del Miocardio , Miocitos Cardíacos , Estrés Oxidativo , Piroptosis , Sulfonamidas , Piroptosis/efectos de los fármacos , Animales , Ratones , Apoptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Sulfonamidas/farmacología , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/tratamiento farmacológico , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Masculino , Furanos/farmacología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/tratamiento farmacológico , Indenos/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , para-Aminobenzoatos/farmacología , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Modelos Animales de Enfermedad , Miocardio/metabolismo , Miocardio/patología , Hipoxia/metabolismo , Hipoxia/complicaciones , DipéptidosRESUMEN
Genetic hearing loss is a common health problem with no effective therapy currently available. DFNA15, caused by mutations of the transcription factor POU4F3, is one of the most common forms of autosomal dominant non-syndromic deafness. In this study, we established a novel mouse model of the human DFNA15 deafness, with a Pou4f3 gene mutation (Pou4f3Δ) identical to that found in a familial case of DFNA15. The Pou4f3(Δ/+) mice suffered progressive deafness in a similar manner to the DFNA15 patients. Hair cells in the Pou4f3(Δ/+) cochlea displayed significant stereociliary and mitochondrial pathologies, with apparent loss of outer hair cells. Progression of hearing and outer hair cell loss of the Pou4f3(Δ/+) mice was significantly modified by other genetic and environmental factors. Using Pou4f3(-/+) heterozygous knockout mice, we also showed that DFNA15 is likely caused by haploinsufficiency of the Pou4f3 gene. Importantly, inhibition of retinoic acid signaling by the aldehyde dehydrogenase (Aldh) and retinoic acid receptor inhibitors promoted Pou4f3 expression in the cochlear tissue and suppressed the progression of hearing loss in the mutant mice. These data demonstrate Pou4f3 haploinsufficiency as the main underlying cause of human DFNA15 deafness and highlight the therapeutic potential of Aldh inhibitors for treatment of progressive hearing loss.
Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Células Ciliadas Auditivas/patología , Pérdida Auditiva/tratamiento farmacológico , Pérdida Auditiva/etiología , Proteínas de Homeodominio/genética , Factor de Transcripción Brn-3C/genética , Animales , Benzaldehídos/farmacología , Modelos Animales de Enfermedad , Haploinsuficiencia/genética , Pérdida Auditiva/genética , Pérdida Auditiva/patología , Proteínas de Homeodominio/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ruido/efectos adversos , Quinolinas/farmacología , Factor de Transcripción Brn-3C/metabolismo , Tretinoina/farmacología , para-Aminobenzoatos/farmacologíaRESUMEN
Nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) is a critical mediator of vascular function. eNOS is tightly regulated at various levels, including transcription, co- and posttranslational modifications, and by various protein-protein interactions. Using stable isotope labeling with amino acids in cell culture (SILAC) and mass spectrometry (MS), we identified several eNOS interactors, including the protein plasminogen activator inhibitor-1 (PAI-1). In cultured human umbilical vein endothelial cells (HUVECs), PAI-1 and eNOS colocalize and proximity ligation assays demonstrate a protein-protein interaction between PAI-1 and eNOS. Knockdown of PAI-1 or eNOS eliminates the proximity ligation assay (PLA) signal in endothelial cells. Overexpression of eNOS and HA-tagged PAI-1 in COS7 cells confirmed the colocalization observations in HUVECs. Furthermore, the source of intracellular PAI-1 interacting with eNOS was shown to be endocytosis derived. The interaction between PAI-1 and eNOS is a direct interaction as supported in experiments with purified proteins. Moreover, PAI-1 directly inhibits eNOS activity, reducing NO synthesis, and the knockdown or antagonism of PAI-1 increases NO bioavailability. Taken together, these findings place PAI-1 as a negative regulator of eNOS and disruptions in eNOS-PAI-1 binding promote increases in NO production and enhance vasodilation in vivo.
Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Disponibilidad Biológica , Línea Celular , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Óxido Nítrico , Óxido Nítrico Sintasa de Tipo III/genética , Piperazinas/farmacología , Inhibidor 1 de Activador Plasminogénico/genética , Unión Proteica , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , para-Aminobenzoatos/farmacologíaRESUMEN
The sunscreen nanocapsules were successfully synthesized by the way of layer-by-layer self-assembly using charged droplets (prepared by emulsification of LAD-30, Tween-80 and EHA (2-Ethylhexyl-4-dimethylaminobenzoate)) as templates. Chitosan/sodium alginate/calcium chloride were selected as wall materials to wrap EHA. The emulsions with the ratio of Tween-80 to EHA (1:1) were stable. A stable NEI negative emulsion can be obtained when the ratio of Tween-80 and LAD-30 was 9:1. Chitosan solutions (50 kDa, 0.25 mg/mL) and sodium alginate solutions (0.5 mg/mL) were selected to prepare nanocapsules. The nanocapsules were characterized via some physico-chemical methods. Based on the synergistic effects of the electrostatic interaction between wall materials and emulsifiers, EHA was effectively encapsulated. DLS and TEM showed that the sunscreen nanocapsules were dispersed in a spherical shape with nano-size, with the increasing number of assembly layers, the size increased from 155 nm (NEI) to 189 nm (NEII) to 201 nm (NEIII) and 205 nm after solidification. The release studies in vitro showed sustained release behavior of the nanocapsules were observed with the increase of the number of deposition layers, implying a good coating effect. The sunscreen nanocapsules could control less than 50% the release of EHA after crosslinking of calcium chloride and sodium alginate, which also could effectively avoid the stimulation of the sun protection agent on the skin.
Asunto(s)
Alginatos/química , Cloruro de Calcio/química , Quitosano/química , Preparaciones de Acción Retardada/química , Protectores Solares/administración & dosificación , para-Aminobenzoatos/administración & dosificación , Animales , Liberación de Fármacos , Masculino , Ratones , Absorción Cutánea , Protectores Solares/farmacocinética , Protectores Solares/farmacología , para-Aminobenzoatos/farmacocinética , para-Aminobenzoatos/farmacologíaRESUMEN
The impairment of the cystic fibrosis transmembrane conductance regulator (CFTR) activity induces intracellular chloride (Cl- ) accumulation. The anion Cl- , acting as a second messenger, stimulates the secretion of interleukin-1ß (IL-1ß), which starts an autocrine positive feedback loop. Here, we show that NLR family pyrin domain containing 3 (NLRP3) and caspase 1 (CASP1) are indirectly modulated by the intracellular Cl- concentration, showing maximal expression and activity at 75 mM Cl- , in the presence of the ionophores nigericin and tributyltin. The expression of PYD and CARD domain containing (PYCARD/ASC) remained constant from 0 to 125 mM Cl- . The CASP1 inhibitor VX-765 and the NLRP3 inflammasome inhibitor MCC950 completely blocked the Cl- -stimulated IL-1ß mRNA expression and partially the IL-1ß secretion. DCF fluorescence (cellular reactive oxygen species, cROS) and MitoSOX fluorescence (mitochondrial ROS, mtROS) also showed maximal ROS levels at 75 mM Cl- , a response strongly inhibited by the ROS scavenger N-acetyl-L-cysteine (NAC) or the NADPH oxidase (NOX) inhibitor GKT137831. These inhibitors also affected CASP1 and NLRP3 mRNA and protein expression. More importantly, the serum/glucocorticoid regulated kinase 1 (SGK1) inhibitor GSK650394, or its shRNAs, completely abrogated the IL-1ß mRNA response to Cl- and the IL-1ß secretion, interrupting the autocrine IL-1ß loop. The results suggest that Cl- effects are mediated by SGK1, in which under Cl- modulation stimulates the secretion of mature IL-1ß, in turn, responsible for the upregulation of ROS, CASP1, NLRP3 and IL-1ß itself, through autocrine signalling.
Asunto(s)
Caspasa 1/metabolismo , Cloruros/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Interleucina-1beta/metabolismo , Espacio Intracelular/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Inhibidores de Caspasas/farmacología , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Dipéptidos/farmacología , Retroalimentación Fisiológica , Furanos/farmacología , Humanos , Proteínas Inmediatas-Precoces/genética , Indenos/farmacología , Interleucina-1beta/genética , Mutación/genética , Nigericina/farmacología , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sulfonamidas/farmacología , para-Aminobenzoatos/farmacologíaRESUMEN
BACKGROUND: Multiple studies have revealed that repeated or long-term exposure to ketamine causes neurodegeneration and cognitive dysfunction. Pyroptosis is an inflammatory form of programmed cell death that has been linked to various neurological diseases. However, the role of NLRP3/caspase-1 axis-related pyroptosis in ketamine-induced neurotoxicity and cognitive dysfunction remains uncertain. METHODS: To evaluate whether ketamine caused NLRP3/caspase1-dependent pyroptosis, flow cytometry analysis, western blotting, ELISA test, histopathological analysis, Morris water maze (MWM) test, cell viability assay, and lactate dehydrogenase release (LDH) assay were carried out on PC12 cells, HAPI cells, and 7-day-old rats. In addition, the NLRP3 inhibitor MCC950 or the caspase-1 inhibitor VX-765 was used to investigate the role of the NLRP3/caspase-1 axis in ketamine-induced neurotoxicity and cognitive dysfunction. RESULTS: Our findings demonstrated that ketamine exposure caused cell damage and increased the levels of pyroptosis in PC12 cells, HAPI cells, and the hippocampus of neonatal rats. After continuous exposure to ketamine, targeting NLRP3 and caspase-1 with MCC950 or VX765 improved pyroptosis, reduced neuropathological damages, and alleviated cognitive dysfunction. CONCLUSION: NLRP3/Caspase-1 axis-dependent pyroptosis is involved in ketamine-induced neuroinflammation and cognitive dysfunction, and it provides a promising strategy to treat ketamine-related neurotoxicity.
Asunto(s)
Caspasa 1/metabolismo , Disfunción Cognitiva/metabolismo , Hipocampo/metabolismo , Ketamina/toxicidad , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis/fisiología , Animales , Animales Recién Nacidos , Inhibidores de Caspasas/farmacología , Inhibidores de Caspasas/uso terapéutico , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/prevención & control , Dipéptidos/farmacología , Dipéptidos/uso terapéutico , Antagonistas de Aminoácidos Excitadores/toxicidad , Femenino , Furanos/farmacología , Furanos/uso terapéutico , Hipocampo/efectos de los fármacos , Indenos/farmacología , Indenos/uso terapéutico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Células PC12 , Piroptosis/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , para-Aminobenzoatos/farmacología , para-Aminobenzoatos/uso terapéuticoRESUMEN
Aim The oral MDM2 antagonist idasanutlin inhibits the p53-MDM2 interaction, enabling p53 activation, tumor growth inhibition, and increased survival in xenograft models. Methods We conducted a Phase I study of idasanutlin (microprecipitate bulk powder formulation) to determine the maximum tolerated dose (MTD), safety, pharmacokinetics, pharmacodynamics, food effect, and clinical activity in patients with advanced malignancies. Schedules investigated were once weekly for 3 weeks (QW × 3), once daily for 3 days (QD × 3), or QD × 5 every 28 days. We also analyzed p53 activation and the anti-proliferative effects of idasanutlin. Results The dose-escalation phase included 85 patients (QW × 3, n = 36; QD × 3, n = 15; QD × 5, n = 34). Daily MTD was 3200 mg (QW × 3), 1000 mg (QD × 3), and 500 mg (QD × 5). Most common adverse events were diarrhea, nausea/vomiting, decreased appetite, and thrombocytopenia. Dose-limiting toxicities were nausea/vomiting and myelosuppression; myelosuppression was more frequent with QD dosing and associated with pharmacokinetic exposure. Idasanutlin exposure was approximately dose proportional at low doses, but less than dose proportional at > 600 mg. Although inter-patient variability in exposure was high with all regimens, cumulative idasanutlin exposure over the whole 28-day cycle was greatest with a QD × 5 regimen. No major food effect on pharmacokinetic exposure occurred. MIC-1 levels were higher with QD dosing, increasing in an exposure-dependent manner. Best response was stable disease in 30.6% of patients, prolonged (> 600 days) in 2 patients with sarcoma. Conclusions Idasanutlin demonstrated dose- and schedule-dependent p53 activation with durable disease stabilization in some patients. Based on these findings, the QD × 5 schedule was selected for further development. TRIAL REGISTRATION: NCT01462175 (ClinicalTrials.gov), October 31, 2011.
Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Pirrolidinas/farmacología , Pirrolidinas/uso terapéutico , para-Aminobenzoatos/farmacología , para-Aminobenzoatos/uso terapéutico , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/patología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Pirrolidinas/efectos adversos , Pirrolidinas/farmacocinética , para-Aminobenzoatos/efectos adversos , para-Aminobenzoatos/farmacocinéticaRESUMEN
RG7388 (Idasanutlin) is a potent inhibitor of oncoprotein murine double minute 2 (MDM2). Herein we investigated the feasibility of developing 18F-labeled RG7388 as a radiotracer for imaging MDM2 expression in tumors with positron emission tomography (PET). Two fluorinated analogues of RG7388, 6 and 7, were synthesized by attaching a fluoronicotinyl moiety to RG7388 via a polyethylene glycol (PEG3) or a propyl linker. The inhibitory potency (IC50) of 6 and 7 against MDM2 was determined by a fluorescence polarization (FP)-based assay. Next, compound 6 was labeled with 18F using a trimethylammonium triflate precursor to obtain [18F]FN-PEG3-RG7388 ([18F]6), and its properties were evaluated in MDM2 expressing wild-type p53 tumor cell lines (SJSA-1 and HepG2) in vitro and in tumor xenografts in vivo. The FP assays revealed an IC50 against MDM2 of 119 nM and 160 nM for 6 and 7, respectively. 18F-labeling of 6 was achieved in 50.3 ± 7.5% radiochemical yield. [18F]6 exhibited a high uptake (â¼70% of input dose) and specificity in SJSA-1 and HepG2 cell lines. Saturation binding assays revealed a binding affinity (Kd) of 128 nM for [18F]6 on SJSA-1 cells. In mice, [18F]6 showed fast clearance from blood with a maximum tumor uptake of 3.80 ± 0.85% injected dose per gram (ID/g) in HepG2 xenografts at 30 min postinjection (p.i.) and 1.32 ± 0.32% ID/g in SJSA-1 xenografts at 1 h p.i. Specificity of [18F]6 uptake in tumors was demonstrated by pretreatment of mice with SJSA-xenografts with a blocking dose of RG7388 (35 mg/kg body weight, i.p.). In vivo stability studies in mice using HPLC showed â¼60% and â¼30% intact [18F]6 remaining in plasma at 30 min and 1 h p.i., respectively, with the remaining activity attributed to polar peaks. Our results suggest that RG7388 is a promising molecular scaffold for 18F-labeled probe development for MDM2. Additional labeling strategies and functionalizing locations on RG7388 are under development to improve binding affinity and in vivo stability of the 18F-labeled compound to make it more amenable for PET imaging of MDM2 in vivo.
Asunto(s)
Antineoplásicos/farmacología , Radioisótopos de Flúor , Tomografía de Emisión de Positrones/métodos , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Pirrolidinas/farmacología , para-Aminobenzoatos/farmacología , Animales , Antineoplásicos/uso terapéutico , Unión Competitiva , Células Hep G2/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Pirrolidinas/uso terapéutico , para-Aminobenzoatos/uso terapéuticoRESUMEN
Multiple sclerosis (MS) is a progressive inflammatory demyelinating disease of the CNS of unknown cause that remains incurable. Inflammasome-associated caspases mediate the maturation and release of the proinflammatory cytokines IL-1ß and IL-18 and activate the pore-forming protein gasdermin D (GSDMD). Inflammatory programmed cell death, pyroptosis, was recently shown to be mediated by GSDMD. Here, we report molecular evidence for GSDMD-mediated inflammasome activation and pyroptosis in both myeloid cells (macrophages/microglia) and, unexpectedly, in myelin-forming oligodendrocytes (ODCs) in the CNS of patients with MS and in the MS animal model, experimental autoimmune encephalomyelitis (EAE). We observed inflammasome activation and pyroptosis in human microglia and ODCs in vitro after exposure to inflammatory stimuli and demonstrate caspase-1 inhibition by the small-molecule inhibitor VX-765 in both cell types. GSDMD inhibition by siRNA transduction suppressed pyroptosis in human microglia. VX-765 treatment of EAE animals reduced the expression of inflammasome- and pyroptosis-associated proteins in the CNS, prevented axonal injury, and improved neurobehavioral performance. Thus, GSDMD-mediated pyroptosis in select glia cells is a previously unrecognized mechanism of inflammatory demyelination and represents a unique therapeutic opportunity for mitigating the disease process in MS and other CNS inflammatory diseases.
Asunto(s)
Caspasa 1/metabolismo , Inhibidores de Caspasas/farmacología , Dipéptidos/farmacología , Modelos Biológicos , Esclerosis Múltiple/enzimología , Oligodendroglía/enzimología , Piroptosis/efectos de los fármacos , para-Aminobenzoatos/farmacología , Células Cultivadas , Humanos , Esclerosis Múltiple/patología , Oligodendroglía/patologíaRESUMEN
Exposure to UV radiation damages the skin and increases the risk of skin cancer. Sunscreen is used to protect the skin from the harmful effects of UV radiation. However, the chemical UV filters used in sunscreen can show toxicity and cause allergic reactions. A safe sunscreen that includes a lower content of chemical UV filters and exerts an excellent effect on UV protection needs to be developed. The objective of this study was to investigate whether the addition of afzelin to sunscreen could improve the sun protection factor (SPF). A synergistic effect between afzelin and organic sunscreen agents including padimate O and oxybenzone was confirmed. Interestingly, 100% in vitro SPF-boosting was observed when afzelin (0.05%) was applied with a standard SPF formulation containing organic sunscreens while afzelin alone had no contribution to the SPF. In vivo SPF analysis of the standard SPF formulation showed an SPF value of 13.3 that increased to 20.1 when supplemented with afzelin (0.05%). Additionally, afzelin showed no skin irritation in a human trial. These results suggest that afzelin is useful as a natural additive in sunscreen formulations and provides an SPF-boosting effect. Afzelin supplementation to the formulation showed the potential to reduce the use of synthetic photoprotectors, which could minimize the risk of synthetic agent toxicity.
Asunto(s)
Cosméticos/química , Manósidos/química , Proantocianidinas/química , Factor de Protección Solar/métodos , Protectores Solares/química , Adolescente , Adulto , Benzofenonas/farmacología , Ensayos Clínicos como Asunto , Cosméticos/farmacología , Composición de Medicamentos , Femenino , Humanos , Masculino , Manósidos/farmacología , Persona de Mediana Edad , Proantocianidinas/farmacología , Piel , Protectores Solares/farmacología , Rayos Ultravioleta , para-Aminobenzoatos/farmacologíaRESUMEN
Ovarian cancer is a gynecological cancer that has the highest mortality rate and is often resistant to conventional treatments. Therefore, development of new therapies is essential. Metformin (MET), which is the priority drug for treatment of type 2 diabetes, has received increasing attention because of its anti-tumor effects. Here, we examined combined anti-tumor effects of MET and RG7388, the only MDM2 (mouse double minute 2 homolog) antagonist that has entered phase III clinical trials, on ovarian cancer cell lines. We examined effects on proliferation by Cell Counting Kit-8 (CCK-8) and colony formation assays, and effects on apoptosis by flow cytometric analysis and Hoechst staining. Western blotting was used to measure protein expression in cells and tissues treated with MET and/or RG7388. Flow cytometry was used to measure reactive oxygen species (ROS). We also examined the effects of MET and/or RG7388 on inhibition of A2780 cell growth in vivo. The combination of MET and RG7388 significantly increased growth inhibition, apoptosis, and ROS of A2780 and SKOV3 cells compared with either agent alone. Additionally, in vitro and in vivo results showed that MET and/or RG7388 inhibited the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway and their combination had a stronger effect. Our findings suggest that the combination of MET and RG7388 enhances growth inhibition and apoptosis induction of ovarian cancer cells through the PI3K/AKT/mTOR pathway and accumulation of intracellular ROS.
Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metformina/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Pirrolidinas/farmacología , para-Aminobenzoatos/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/genética , Ratones Desnudos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de XenoinjertoRESUMEN
BACKGROUND: Excessive inflammation might activate and injure the blood-brain barrier (BBB), a common feature of many central nervous system (CNS) disorders. We previously developed an in vitro BBB injury model in which the organophosphate paraoxon (PX) affects the BBB endothelium by attenuating junctional protein expression leading to weakened barrier integrity. The objective of this study was to investigate the inflammatory cellular response at the BBB to elucidate critical pathways that might lead to effective treatment in CNS pathologies in which the BBB is compromised. We hypothesized that caspase-1, a core component of the inflammasome complex, might have important role in BBB function since accumulating evidence indicates its involvement in brain inflammation and pathophysiology. METHODS: An in vitro human BBB model was employed to investigate BBB functions related to inflammation, primarily adhesion and transmigration of peripheral blood mononuclear cells (PBMCs). Caspase-1 pathway was studied by measurements of its activation state and its role in PBMCs adhesion, transmigration, and BBB permeability were investigated using the specific caspase-1 inhibitor, VX-765. Expression level of adhesion and junctional molecules and the secretion of pro-inflammatory cytokines were measured in vitro and in vivo at the BBB endothelium after exposure to PX. The potential repair effect of blocking caspase-1 and downstream molecules was evaluated by immunocytochemistry, ELISA, and Nanostring technology. RESULTS: PX affected the BBB in vitro by elevating the expression of the adhesion molecules E-selectin and ICAM-1 leading to increased adhesion of PBMCs to endothelial monolayer, followed by elevated transendothelial-migration which was ICAM-1 and LFA-1 dependent. Blocking caspase-8 and 9 rescued the viability of the endothelial cells but not the elevated transmigration of PBMCs. Inhibition of caspase-1, on the other hand, robustly restored all of barrier insults tested including PBMCs adhesion and transmigration, permeability, and VE-cadherin protein levels. The in vitro inflammatory response induced by PX and the role of caspase-1 in BBB injury were corroborated in vivo in isolated blood vessels from hippocampi of mice exposed to PX and treated with VX-765. CONCLUSIONS: These results shed light on the important role of caspase-1 in BBB insult in general and specifically in the inflamed endothelium, and suggest therapeutic potential for various CNS disorders, by targeting caspase-1 in the injured BBB.
Asunto(s)
Barrera Hematoencefálica/metabolismo , Caspasa 1/metabolismo , Células Endoteliales/metabolismo , Inflamasomas/metabolismo , Pericitos/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/lesiones , Muerte Celular/fisiología , Movimiento Celular/fisiología , Técnicas de Cocultivo , Dipéptidos/farmacología , Humanos , Interleucina-8/metabolismo , Masculino , Ratones , para-Aminobenzoatos/farmacologíaRESUMEN
Psoriasis is a skin disease characterized by abnormal keratinocyte proliferation and inflammation. Currently, there are no cures for this disease, so the goal of treatment is to decrease inflammation and slow down the associated rapid cell growth and shedding. Recent advances have led to the usage of phosphodiesterase 4 (PDE4) inhibitors for treatment of this condition. For example, apremilast is an oral, selective PDE4 inhibitor that is able to reduce skin inflammation and is Food and Drug Administration (FDA)-approved to treat adults with moderate to severe psoriasis and/or psoriatic arthritis. However, common target-related adverse events, including diarrhea, nausea, headache, and insomnia limit the usage of this drug. To circumvent these effects, the usage of PDE4 inhibitors specifically designed for topical treatment, such as CHF6001, may combine local anti-inflammatory activity with limited systemic exposure, improving tolerability. In this study, we showed that CHF6001, currently undergoing clinical development for COPD, suppresses human keratinocyte proliferation as assessed via BrdU incorporation. We also observed decreased re-epithelialization in a scratch-wound model after CHF6001 treatment. At the molecular level, CHF6001 inhibited translocation of phosphorylated NF-κB subunit p65, promoting loss of nuclear cyclin D1 accumulation and an increase of cell cycle inhibitor p21. Furthermore, CHF6001 decreased oxidative stress, measured by assessing lipid peroxidation (4-HNE adduct formation), through the inactivation of the NADPH oxidase. These results suggest that CHF6001 has the potential to treat skin disorders associated with hyperproliferative keratinocytes, such as psoriasis by targeting oxidative stress, abnormal re-epithelization, and inflammation.
Asunto(s)
Proliferación Celular/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Fosfodiesterasa 4/farmacología , Sulfonamidas/farmacología , para-Aminobenzoatos/farmacología , Aldehídos/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclina D1/metabolismo , Humanos , NADPH Oxidasas/metabolismo , Inhibidores de Fosfodiesterasa 4/toxicidad , Psoriasis/tratamiento farmacológico , Sulfonamidas/toxicidad , Factor de Transcripción ReIA/metabolismo , para-Aminobenzoatos/toxicidadRESUMEN
Inhibition of immune and inflammatory reaction induced by the expose of nucleus pulposus (NP) could effectively ameliorate neuropathic pain in the lumbar disc herniation. Maresin1 (MaR1), as a macrophage-derived mediator of inflammation resolution, displayed potent anti-inflammatory action. In the present study, we attempted to elucidate the impact of MaR1 on radicular pain and the interaction with NLRP3 inflammasome. We established a rat model of non-compressive lumbar disc herniation and different administration (MaR1 or Caspase-1 inhibitor) was given to them. The paw withdrawal latency (PWL) and paw withdrawal thresholds (PWTs) were observed to assess pain behaviors. The spinal cord horns were collected and the levels of IL-1ß and IL-18 were measured by ELISA. The mRNA and protein expression levels of NLRP3 inflammasome components were tested by RT-PCR, western blot and immunohistochemistry. The endogenous MaR1 levels of the spinal cord were analyzed using LC-MS/MS. The application of NP in the models lead to mechanical and thermal hypersensitivity, increased IL-1ß and IL-18 levels and expressions of NLRP3 inflammasome components, which were reversed markedly by administration of MaR1. Caspase-1 inhibition also improved mechanical hypersensitivity, decreased the expressions of inflammatory cytokines and restrained the activation of inflammasome. Meanwhile, Caspase-1 inhibitor promoted the endogenous MaR1 synthesis, which was hindered in the pain models. Altogether, our study indicated that the negative interaction between MaR1 and NLRP3 inflammasome mediated the inflammatory response in spinal dorsal horn, which involved in the pathogenesis of radicular pain.
Asunto(s)
Analgésicos/uso terapéutico , Antiinflamatorios/uso terapéutico , Ácidos Docosahexaenoicos/uso terapéutico , Inflamasomas/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuralgia/tratamiento farmacológico , Animales , Caspasa 1/metabolismo , Inhibidores de Caspasas/farmacología , Dipéptidos/farmacología , Inflamación/tratamiento farmacológico , Inflamación/epidemiología , Desplazamiento del Disco Intervertebral/complicaciones , Dolor de la Región Lumbar/tratamiento farmacológico , Dolor de la Región Lumbar/epidemiología , Vértebras Lumbares/efectos de los fármacos , Masculino , Neuralgia/epidemiología , Ratas Sprague-Dawley , para-Aminobenzoatos/farmacologíaRESUMEN
Pyroptosis is a newly identified lytic form of programmed cell death which is characterized by plasma membrane blebbing and rupture. Pyroptosis occurs in cerebral ischemia injury, and contributes to the activation and secretion of the inflammatory cytokines interleukin (IL)-1ß, IL-18, and IL-6. Previous reports have found that Dendrobium alkaloids (DNLA) can exert neuroprotective effects against oxygen-glucose deprivation/reperfusion (OGD/R) damage in vitro, but the mechanisms underlying these effects remain elusive. In this study, we investigated whether DNLA exerted therapeutic benefits against cerebral ischemia-reperfusion (CIR) damage via ameliorating pyroptosis and inflammation. OGD/R damage was induced in HT22 cells pretreated with DNLA (0.03, 0.3, or 3 mg/ml, 24 h prior to OGD/R), MCC950 (10 ng/ml, 1 h prior), and VX765 (10 ng/ml, 1 h prior). Neuronal apoptosis, necrosis, pyroptosis, and pathological changes were analyzed 24 h following OGD/R. Further to this, male C57/BL mice pretreated with different concentrations of DNLA (0.5 or 5 mg/kg, ip.) for 24 h and VX765 (50 mg/kg, ip., 1 h before CIR) underwent transient middle cerebral artery occlusion and reperfusion. We found that DNLA pretreatment resulted in a lower neurologic deficit score, a reduced infarct volume, fewer pyroptotic cells, and reduced levels of inflammatory factors 24 h after CIR. Furthermore, DNLA administration also reduced the levels of the pyroptosis-associated proteins Caspase-1 and gasdermin-D, particularly in the hippocampal CA1 region. Similar decreases were observed in the levels of the inflammatory factors IL-1ß, IL-6, and IL-18. OGD/R-associated ultrastructural damage was seen to improve following DNLA administration, likely due to the regulation of the tight junction protein Pannexin-1 by DNLA. Overall, these findings demonstrate that DNLA can protect against CIR damage through inhibiting pyroptosis-induced neuronal death, providing new therapeutic insights for CIR injury.
Asunto(s)
Alcaloides/uso terapéutico , Infarto de la Arteria Cerebral Media/prevención & control , Necrosis/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Piroptosis/efectos de los fármacos , Daño por Reperfusión/prevención & control , Animales , Caspasa 1/metabolismo , Inhibidores de Caspasas/farmacología , Línea Celular , Dipéptidos/farmacología , Hipocampo/metabolismo , Hipocampo/patología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones Endogámicos C57BL , Necrosis/metabolismo , Necrosis/patología , Proteínas de Unión a Fosfato/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , para-Aminobenzoatos/farmacologíaRESUMEN
BACKGROUND: Alcohol use disorders affect millions of people worldwide, and there is growing evidence that excessive alcohol intake causes severe damage to the brain of both humans and animals. Numerous studies on chronic alcohol exposure in animal models have identified that many functional impairments are associated with the hippocampus, which is a structure exhibiting substantial vulnerability to alcohol exposure. However, the precise mechanisms that lead to structural and functional impairments of the hippocampus are poorly understood. Herein, we report a novel cell death type, namely pyroptosis, which accounts for alcohol neurotoxicity in mice. METHODS: For this study, we used an in vivo model to induce alcohol-related neurotoxicity in the hippocampus. Adult male C57BL/6 mice were treated with 95% alcohol vapor either alone or in combination with selective cannabinoid receptor antagonists or agonists, and VX765 (Belnacasan), which is a selective caspase-1 inhibitor. RESULTS: Alcohol-induced in vivo pyroptosis occurs because of an increase in the levels of pyroptotic proteins such as nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3), caspase-1, gasdermin D (GSDMD), and amplified inflammatory response. Our results indicated that VX765 suppressed the expression of caspase-1 and inhibited the maturation of the proinflammatory cytokines interleukin-1ß (IL-1ß) and IL-18. Additionally, chronic alcohol intake created an imbalance in the endocannabinoid system and regulated 2 cannabinoid receptors (CB1R and CB2R) in the hippocampus. Specific antagonists of CB1R (AM251 and AM281) significantly ameliorated alcohol-induced pyroptosis signaling and inactivated the inflammatory response. CONCLUSIONS: Alcohol induces hippocampal pyroptosis, which leads to neurotoxicity, thereby indicating that pyroptosis may be an essential pathway involved in chronic alcohol-induced hippocampal neurotoxicity. Furthermore, cannabinoid receptors are regulated during this process, which suggests promising therapeutic strategies against alcohol-induced neurotoxicity through pharmacologic inhibition of CB1R.
Asunto(s)
Trastornos del Sistema Nervioso Inducidos por Alcohol/metabolismo , Antagonistas de Receptores de Cannabinoides/farmacología , Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Hipocampo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Piroptosis/efectos de los fármacos , Receptor Cannabinoide CB1/antagonistas & inhibidores , Animales , Agonistas de Receptores de Cannabinoides/farmacología , Caspasa 1/efectos de los fármacos , Caspasa 1/metabolismo , Inhibidores de Caspasas/farmacología , Dipéptidos/farmacología , Inflamación , Interleucina-18/metabolismo , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Morfolinas/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Síndromes de Neurotoxicidad , Proteínas de Unión a Fosfato/efectos de los fármacos , Proteínas de Unión a Fosfato/metabolismo , Piperidinas/farmacología , Pirazoles/farmacología , para-Aminobenzoatos/farmacologíaRESUMEN
The discovery of environmentally friendly and inexpensive plant growth regulators (PGRs) for agronomically important crops is a necessity and must be considered a priority worldwide. This study provides the synthesis, structure determination and the biological evaluation of two binary organic salts as potential PGRs. New compounds have dual biological activity and are based on natural metabolite p-aminobenzoic acid (pABAH) and different alkanolamines. Studied compounds exhibit hydrogen-bonded 3D supramolecular architectures with different crystal packing due to the formation of one homosynthon and various heterosynthons. The biological profile of new compounds was investigated in laboratory and greenhouse on Solanum lycopersicum L., revealing the efficiency in promoting plant rooting and plant productivity. The results may have a positive impact on agricultural economics, developing new sustainable PGRs for tomatoes.
Asunto(s)
Reguladores del Crecimiento de las Plantas/síntesis química , Solanum lycopersicum/crecimiento & desarrollo , para-Aminobenzoatos/síntesis química , Productos Agrícolas/efectos de los fármacos , Productos Agrícolas/crecimiento & desarrollo , Tecnología Química Verde , Solanum lycopersicum/efectos de los fármacos , Conformación Molecular , Reguladores del Crecimiento de las Plantas/química , Reguladores del Crecimiento de las Plantas/farmacología , Raíces de Plantas/efectos de los fármacos , Raíces de Plantas/crecimiento & desarrollo , para-Aminobenzoatos/química , para-Aminobenzoatos/farmacologíaRESUMEN
High-risk neuroblastoma, a predominantly TP53 wild-type (wt) tumour, is incurable in >50% patients supporting the use of MDM2 antagonists as novel therapeutics. Idasanutlin (RG7388) shows in vitro synergy with chemotherapies used to treat neuroblastoma. This is the first study to evaluate the in vivo efficacy of the intravenous idasanutlin prodrug, RO6839921 (RG7775), both alone and in combination with temozolomide in TP53 wt orthotopic neuroblastoma models. Detection of active idasanutlin using liquid chromatography-mass spectrometry and p53 pathway activation by ELISA assays and Western analysis showed peak plasma levels 1 h post-treatment with maximal p53 pathway activation 3-6 h post-treatment. RO6839921 and temozolomide, alone or in combination in mice implanted with TP53 wt SHSY5Y-Luc and NB1691-Luc cells showed that combined RO6839921 and temozolomide led to greater tumour growth inhibition and increase in survival compared to vehicle control. Overall, RO6839921 had a favourable pharmacokinetic profile consistent with intermittent dosing and was well tolerated alone and in combination. These preclinical studies support the further development of idasanutlin in combination with temozolomide in neuroblastoma in early phase clinical trials.
Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neuroblastoma/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Pirrolidinas/farmacología , Temozolomida/farmacología , para-Aminobenzoatos/farmacología , Animales , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Ratones , Neuroblastoma/genética , Neuroblastoma/metabolismo , Profármacos/farmacocinética , Profármacos/farmacología , Pirrolidinas/farmacocinética , Distribución Aleatoria , Temozolomida/administración & dosificación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , para-Aminobenzoatos/farmacocinéticaRESUMEN
BACKGROUND: We compared the anti-inflammatory effects of phosphodiesterase type 4 (PDE4) inhibitor roflumilast with CHF6001, a novel PDE4 inhibitor designed for inhaled administration, using human alveolar macrophages (AM) and lung tissue explants models. METHODS: AM from 13 chronic obstructive pulmonary disease (COPD) patients and 10 smoking controls and lung tissue from 7 COPD patients were stimulated with LPS following preincubation with roflumilast (0.000001-10⯵M), CHF6001 (0.000001-0.1⯵M), or vehicle. After 24â¯h, supernatants were analysed for cytokines by ELISA. The effects of both compounds on the phosphorylation and cellular localisation of cAMP response element binding protein (CREB) were assessed by immunofluorescence and Western blot analysis. Extracted RNA was used for quantitative PCR analysis of PDE4 A, B and D mRNA. RESULTS: PDE4 A, B and D expression were increased in alveolar macrophages and lung tissue of COPD patients compared to controls. Roflumilast and CHF6001 significantly reduced TNF-α production in AM and lung tissue. CHF6001 was more potent than roflumilast with lower EC50s of 0.02, 0.01 and 0.31â¯nM compared to 0.87, 0.47 and 10.8â¯nM in respective samples. PDE4 inhibition also inhibited secretion of the chemokines CCL2 and CCL4 from macrophages. Both compounds increased nuclear levels of phosphorylated CREB. CONCLUSION: PDE4 inhibitors caused a robust anti-inflammatory effect on TNF-α production from COPD AM, with inhibition of selective chemokines also observed. CHF6001 caused more potent inhibition of TNF-α production from COPD AM and lung tissue compared to roflumilast.
Asunto(s)
Aminopiridinas/farmacología , Benzamidas/farmacología , Macrófagos Alveolares/inmunología , Inhibidores de Fosfodiesterasa 4/farmacología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Sulfonamidas/farmacología , para-Aminobenzoatos/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Quimiocina CCL2/inmunología , Quimiocina CCL4/inmunología , Ciclopropanos/farmacología , Femenino , Humanos , Macrófagos Alveolares/patología , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/patología , Factor de Necrosis Tumoral alfa/inmunologíaRESUMEN
Chronic lymphocytic leukemia (CLL) is a clinically heterogeneous hematologic malignancy. In approximately 90% of cases the TP53 gene is in its wildtype state at diagnosis of this malignancy. As mouse double-minute-2 homolog (MDM2) is a primary repressor of p53, targeting this protein is an attractive therapeutic approach for non-genotoxic reactivation of p53. Since the discovery of the first MDM2 inhibitor, Nutlin-3a, newer potent and bioavailable compounds have been developed. In this study we tested the second-generation MDM2 inhibitor, RG7388, in patient-derived CLL cells and normal cells, examining its effect on the induction of p53-transcriptional targets. RG7388 potently decreased viability in p53-functional CLL cells, whereas p53-non-functional samples were more resistant to the drug. RG7388 induced a pro-apoptotic gene expression signature with upregulation of p53-target genes involved in the intrinsic (PUMA, BAX) and extrinsic (TNFRSF10B, FAS) pathways of apoptosis, as well as MDM2 Only a slight induction of CDKN1A was observed and upregulation of pro-apoptotic genes dominated, indicating that CLL cells are primed for p53-dependent apoptosis. Consequently, RG7388 led to a concentration-dependent increase in caspase-3/7 activity and cleaved poly (ADP-ribose) polymerase. Importantly, we observed a preferential pro-apoptotic signature in CLL cells but not in normal blood and bone marrow cells, including CD34+ hematopoietic cells. These data support the further evaluation of MDM2 inhibitors as a novel additional treatment option for patients with p53-functional CLL.