Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 596
Filtrar
1.
J Feline Med Surg ; 26(3): 1098612X241230941, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38511293

RESUMEN

OBJECTIVES: The main objective of this study was to compare the postoperative analgesic effects of grapiprant with those of robenacoxib in cats undergoing ovariohysterectomy (OVH). METHODS: In total, 37 female cats (age range 4 months-10 years, weighing ⩾2.5 kg) were enrolled in a prospective, randomized, masked, non-inferiority (NI) clinical trial. Cats received oral robenacoxib (1 mg/kg) or grapiprant (2 mg/kg) 2 h before OVH. Analgesia was assessed via the Feline Grimace Scale (FGS), the Glasgow Composite Measure Pain Scale-Feline (CMPS-F), von Frey monofilaments (vFFs) and pressure algometry (ALG) 2 h before treatment administration, at extubation, and 2, 4, 6, 8, 18 and 24 hours after extubation. Hydromorphone (<8 h postoperatively) or buprenorphine (>18 h postoperatively) were administered to cats with scores of ⩾5/20 on CMPS-F and/or ⩾4/10 on FGS. NI margins for CMPS-F and vFFs were set at 3 and -0.2, respectively. A mixed-effect ANOVA was used for FGS scores (P <0.05). Data are reported as mean ± SEM. RESULTS: The data from 33 cats were analyzed. The upper limit of the 95% confidence interval (CI) (0.35) was less than the NI margin of 3 for CMPS-F, and the lower limit of the 95% CI (0.055) was greater than the NI margin of -0.2 for vFFs, indicating NI of grapiprant. The FGS scores were greater than baseline at extubation for both treatments (1.65 ± 0.63; P = 0.001); however, there was no difference between treatments. There was no difference between treatments, nor treatment by time interaction, for vFFs (P <0.001). The CMPS-F scores for both treatments were higher at extubation but returned to baseline after 4 h (P <0.001). For ALG, there was no difference in treatment or treatment by time interaction. The robenacoxib group had lower pressure readings at extubation and 6 h compared with baseline. CONCLUSIONS AND RELEVANCE: These results indicate that grapiprant was non-inferior to robenacoxib for mitigating postsurgical pain in cats after OVH performed via ventral celiotomy. The impact of grapiprant for analgesia in OVH via the flank is unknown.


Asunto(s)
Analgésicos , Bencenosulfonamidas , Enfermedades de los Gatos , Difenilamina/análogos & derivados , Imidazoles , Fenilacetatos , Piridinas , Compuestos de Sulfonilurea , Gatos , Animales , Femenino , Ovariectomía/veterinaria , Estudios Prospectivos , Histerectomía/veterinaria , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/prevención & control , Dolor Postoperatorio/veterinaria , Enfermedades de los Gatos/tratamiento farmacológico , Enfermedades de los Gatos/cirugía
2.
Front Cell Infect Microbiol ; 14: 1275940, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38352056

RESUMEN

Chronic pulmonary bacterial infections and associated inflammation remain a cause of morbidity and mortality in people with cystic fibrosis (PwCF) despite new modulator therapies. Therapies targeting host factors that dampen detrimental inflammation without suppressing immune responses critical for controlling infections remain limited, while the development of lung infections caused by antimicrobial resistant bacteria is an increasing global problem, and a significant challenge in CF. Pharmacological compounds targeting the mammalian MAPK proteins MEK1 and MEK2, referred to as MEK1/2 inhibitor compounds, have potential combined anti-microbial and anti-inflammatory effects. Here we examined the immunomodulatory properties of MEK1/2 inhibitor compounds PD0325901, trametinib, and CI-1040 on CF innate immune cells. Human CF macrophage and neutrophil phagocytic functions were assessed by quantifying phagocytosis of serum opsonized pHrodo red E. coli, Staphylococcus aureus, and zymosan bioparticles. MEK1/2 inhibitor compounds reduced CF macrophage pro-inflammatory cytokine production without impairing CF macrophage or neutrophil phagocytic abilities. Wild-type C57BL6/J and Cftr tm1kth (F508del homozygous) mice were used to evaluate the in vivo therapeutic potential of PD0325901 compared to vehicle treatment in an intranasal methicillin-resistant Staphylococcus aureus (MRSA) infection with the community-acquired MRSA strain USA300. In both wild-type and CF mice, PD0325901 reduced inflammation associated body mass loss. Wild-type mice treated with PD0325901 had significant reduction in neutrophil-mediated inflammation compared to vehicle treatment groups, with preserved clearance of bacteria in lung, liver, or spleen 1 day after infection in either wild-type or CF mouse models. In summary, this study provides the first data evaluating the therapeutic potential of MEK1/2 inhibitor to modulate CF immune cells and demonstrates that MEK1/2 inhibitors diminish pro-inflammatory responses without impairing host defense mechanisms required for acute pathogen clearance.


Asunto(s)
Benzamidas , Fibrosis Quística , Difenilamina/análogos & derivados , Staphylococcus aureus Resistente a Meticilina , Humanos , Animales , Ratones , Fibrosis Quística/complicaciones , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/microbiología , Escherichia coli , Macrófagos , Inflamación/complicaciones , Gravedad del Paciente , Mamíferos
3.
J Vet Pharmacol Ther ; 47(3): 175-186, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38235901

RESUMEN

We evaluated the effect of administration timing of meloxicam and robenacoxib on renal function, platelet cyclo-oxygenase and perioperative analgesia in 60 cats undergoing ovariohysterectomy, in a prospective randomized blinded controlled study. Twelve cats were randomly allocated to one subcutaneous treatment group: meloxicam (0.2 mg/kg) or robenacoxib (2 mg/kg) at admission (MA, RA), at induction (MI, RI) and robenacoxib at the end of surgery (RE). All cats received the same anaesthesia protocol. Plasma renin activity (PRA), plasma creatinine, drug concentrations and serum thromboxane (TxB2) were measured sequentially. Anaesthesia significantly increased PRA, as activity at end of the surgery was higher than 2 h later (mean ± SD: 26.6 ± 2.8 versus 10.0 ± 3.9 ng/mL/h). PRA remained higher at 2 h post-surgery in admission groups compared to induction groups (p = .01). Serum TxB2 was lower with meloxicam than robenacoxib (p = .001), and was lower in the MA than each robenacoxib group at catheter placement. Admission groups (16/24 from RA and MA groups) received earlier rescue analgesia than other groups (p = .033). In conclusion, the renin-angiotensin system was activated during anaesthesia despite cyclo-oxygenase inhibition, possibly due to hypotension or surgical stimulation. There was no effect of drug or timing on the markers of renal function but one cat receiving meloxicam at induction had suspected IRIS grade II acute kidney injury.


Asunto(s)
Difenilamina , Difenilamina/análogos & derivados , Histerectomía , Meloxicam , Ovariectomía , Dolor Postoperatorio , Fenilacetatos , Animales , Meloxicam/administración & dosificación , Meloxicam/farmacología , Meloxicam/uso terapéutico , Femenino , Gatos , Histerectomía/veterinaria , Dolor Postoperatorio/veterinaria , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/prevención & control , Difenilamina/farmacología , Difenilamina/administración & dosificación , Ovariectomía/veterinaria , Fenilacetatos/administración & dosificación , Fenilacetatos/farmacología , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Riñón/efectos de los fármacos , Analgesia/veterinaria , Analgesia/métodos
4.
Sci Rep ; 14(1): 1871, 2024 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-38253821

RESUMEN

Better agents are needed to improve glaucoma filtration surgery outcomes compared to current ones. The purpose of this study is to determine whether mitogen-activated protein kinase kinase (MEK) inhibitors can effectively arrest the cell cycle of human conjunctival fibroblasts (HCFs) and inhibit the formation of fibrosis and scarring following glaucoma filtration surgery. A cell counting kit­8 assay revealed that the MEK inhibitor PD0325901 exhibited concentration-dependent growth inhibition of HCFs. Quantitative PCR, immunocytochemistry, and western blotting demonstrated decreased expression of proliferating cell nuclear antigen (PCNA) and cyclin D1 and increased expression of p27 in HCFs treated with PD0325901. Flow cytometry indicated that PD0325901 arrested the cell cycle of HCFs in the G0/1 phase. The cell-migration assay showed that HCF migration rate was significantly suppressed by PD0325901 exposure. Rabbits were divided into PD0325901-treatment and control groups, and glaucoma filtration surgery was performed. Although intraocular pressure did not differ between PD0325901-treatment and control groups, bleb height was greater in the treatment group. Histopathological evaluation revealed that fibrotic changes were significantly attenuated in the PD0325901-treatment group compared to the control group. In conclusion, the MEK inhibitor impedes HCF proliferation via cell-cycle arrest and may be beneficial for glaucoma filtration surgery by reducing bleb scarring.


Asunto(s)
Benzamidas , Difenilamina/análogos & derivados , Cirugía Filtrante , Glaucoma , Animales , Humanos , Conejos , Cicatriz/tratamiento farmacológico , Cicatriz/prevención & control , Ciclo Celular , Glaucoma/cirugía , Inhibidores de Proteínas Quinasas/farmacología
5.
Cell Death Dis ; 15(1): 27, 2024 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-38199990

RESUMEN

Intestinal epithelial renewal, which depends on the proliferation and differentiation of intestinal stem cells (ISCs), is essential for epithelial homoeostasis. Understanding the mechanism controlling ISC activity is important. We found that death receptor 5 (DR5) gene deletion (DR5-/-) mice had impaired epithelial absorption and barrier function, resulting in delayed weight gain, which might be related to the general reduction of differentiated epithelial cells. In DR5-/- mice, the expression of ISC marker genes, the number of Olfm4+ ISCs, and the number of Ki67+ and BrdU+ cells in crypt were reduced. Furthermore, DR5 deletion inhibited the expression of lineage differentiation genes driving ISC differentiation into enterocytes, goblet cells, enteroendocrine cells, and Paneth cells. Therefore, DR5 gene loss may inhibit the intestinal epithelial renewal by dampening ISC activity. The ability of crypts from DR5-/- mice to form organoids decreased, and selective DR5 activation by Bioymifi promoted organoid growth and the expression of ISC and intestinal epithelial cell marker genes. Silencing of endogenous DR5 ligand TRAIL in organoids down-regulated the expression of ISC and intestinal epithelial cell marker genes. So, DR5 expressed in intestinal crypts was involved in the regulation of ISC activity. DR5 deletion in vivo or activation in organoids inhibited or enhanced the activity of Wnt, Notch, and BMP signalling through regulating the production of Paneth cell-derived ISC niche factors. DR5 gene deletion caused apoptosis and DNA damage in transit amplifying cells by inhibiting ERK1/2 activity in intestinal crypts. Inhibition of ERK1/2 with PD0325901 dampened the ISC activity and epithelial regeneration. In organoids, when Bioymifi's effect in activating ERK1/2 activity was completely blocked by PD0325901, its role in stimulating ISC activity and promoting epithelial regeneration was also eliminated. In summary, DR5 in intestinal crypts is essential for ISC activity during epithelial renewal under homoeostasis.


Asunto(s)
Benzamidas , Difenilamina , Ftalimidas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Células Madre , Tiazolidinas , Animales , Ratones , Difenilamina/análogos & derivados , Homeostasis
6.
Biochem Pharmacol ; 204: 115233, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36041543

RESUMEN

Antiestrogen resistance of breast cancer has been related to enhanced growth factor receptor expression and activation. We have previously shown that ectopic expression and subsequent activation of the insulin-like growth factor-1 receptor (IGF1R) or the epidermal growth factor receptor (EGFR) in MCF7 or T47D breast cancer cells results in antiestrogen resistance. In order to identify novel therapeutic targets to prevent this antiestrogen resistance, we performed kinase inhibitor screens with 273 different inhibitors in MCF7 cells overexpressing IGF1R or EGFR. Kinase inhibitors that antagonized antiestrogen resistance but are not directly involved in IGF1R or EGFR signaling were prioritized for further analyses. Various ALK (anaplastic lymphoma receptor tyrosine kinase) inhibitors inhibited cell proliferation in IGF1R expressing cells under normal and antiestrogen resistance conditions by preventing IGF1R activation and subsequent downstream signaling; the ALK inhibitors did not affect EGFR signaling. On the other hand, MEK (mitogen-activated protein kinase kinase)1/2 inhibitors, including PD0325901, selumetinib, trametinib and TAK-733, selectively antagonized IGF1R signaling-mediated antiestrogen resistance but did not affect cell proliferation under normal growth conditions. RNAseq analysis revealed that MEK inhibitors PD0325901 and selumetinib drastically altered cell cycle progression and cell migration networks under IGF1R signaling-mediated antiestrogen resistance. In a group of 219 patients with metastasized ER + breast cancer, strong pMEK staining showed a significant correlation with no clinical benefit of first-line tamoxifen treatment. We propose a critical role for MEK activation in IGF1R signaling-mediated antiestrogen resistance and anticipate that dual-targeted therapy with a MEK inhibitor and antiestrogen could improve treatment outcome.


Asunto(s)
Neoplasias de la Mama , Moduladores de los Receptores de Estrógeno , Quinasa de Linfoma Anaplásico , Benzamidas , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Difenilamina/análogos & derivados , Resistencia a Antineoplásicos , Receptores ErbB , Antagonistas de Estrógenos/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Moduladores de los Receptores de Estrógeno/uso terapéutico , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina , Quinasas de Proteína Quinasa Activadas por Mitógenos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor IGF Tipo 1 , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico
7.
J Biol Chem ; 298(8): 102226, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35787369

RESUMEN

Increased MAPK signaling is a hallmark of various cancers and is a central regulator of cell survival. Direct ERK1/2 inhibition is considered a promising approach to avoid ERK1/2 reactivation caused by upstream kinases BRAF, MEK1/2, and KRAS, as well as by receptor tyrosine kinase inhibitors, but the dynamics and selectivity of ERK1/2 inhibitors are much less studied compared with BRAF or MEK inhibitors. Using ERK1/2 and downstream kinase ELK1 reporter cell lines of lung cancer (H1299; NRASQ61K), colon cancer (HCT-116; KRASG13D), neuroblastoma (SH-SY5Y), and leukemia (U937), we examined the relationship between ERK inhibition and drug-induced toxicity for five ERK inhibitors: SCH772984, ravoxertinib, LY3214996, ulixertinib, and VX-11e, as well as one MEK inhibitor, PD0325901. Comparing cell viability and ERK inhibition revealed different ERK dependencies for these cell lines. We identify several drugs, such as SCH772984 and VX-11e, which induce excessive toxicity not directly related to ERK1/2 inhibition in specific cell lines. We also show that PD0325901, LY3214996, and ulixertinib are prone to ERK1/2 reactivation over time. We distinguished two types of ERK1/2 reactivation: the first could be reversed by adding a fresh dose of inhibitors, while the second persists even after additional treatments. We also showed that cells that became resistant to the MEK1/2 inhibitor PD0325901 due to ERK1/2 reactivation remained sensitive to ERK1/2 inhibitor ulixertinib. Our data indicate that correlation of ERK inhibition with drug-induced toxicity in multiple cell lines may help to find more selective and effective ERK1/2 inhibitors.


Asunto(s)
Antineoplásicos , Quinasas de Proteína Quinasa Activadas por Mitógenos , Neuroblastoma , Inhibidores de Proteínas Quinasas , Aminopiridinas , Antineoplásicos/farmacología , Benzamidas , Línea Celular Tumoral , Supervivencia Celular , Difenilamina/análogos & derivados , Humanos , Indazoles , Sistema de Señalización de MAP Quinasas , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neuroblastoma/tratamiento farmacológico , Piperazinas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Pirazoles , Piridonas , Pirimidinas , Pirroles
8.
J Vet Pharmacol Ther ; 45(4): 325-351, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35460083

RESUMEN

Robenacoxib is a veterinary-approved non-steroidal anti-inflammatory drug (NSAID) of the coxib group. It possesses anti-hyperalgesic, anti-inflammatory and anti-pyretic properties. Robenacoxib inhibits the cyclooxygenase (COX)-2 isoform of COX selectively (in vitro IC50 ratios COX-1:COX-2, 129:1 in dogs, 32:1 in cats). At registered dosages (2 mg/kg subcutaneously in dogs and cats, 1-4 mg/kg orally in dogs and 1-2.4 mg/kg orally in cats), robenacoxib produces significant inhibition of COX-2 whilst sparing COX-1. The pharmacokinetic (PK) profile of robenacoxib is characterized by a high degree of binding to plasma proteins (>98%) and moderate volume of distribution (at steady state, 240 ml/kg in dogs and 190 ml/kg in cats). In consequence, the terminal half-life in blood (<2 h) is short, despite moderate body clearance (0.81 L/kg/h) in dogs and low clearance (0.44 L/kg/h) in cats. Excretion is principally in the bile (65% in dogs and 72% in cats). Robenacoxib concentrates in inflamed tissues, and clinical efficacy is achieved with once-daily dosing, despite the short blood terminal half-life. In dogs, no relevant breed differences in robenacoxib PK have been detected. Robenacoxib has a wide safety margin; in healthy laboratory animals daily oral doses 20-fold (dog, 1 month), eight-fold (cat, 6 weeks) and five-fold (dog, 6 months) higher than recommended clinical doses were well tolerated. Clinical efficacy and safety have been demonstrated in orthopaedic and soft tissue surgery, and in musculoskeletal disorders in dogs and cats.


Asunto(s)
Enfermedades de los Gatos , Enfermedades de los Perros , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Enfermedades de los Gatos/inducido químicamente , Enfermedades de los Gatos/tratamiento farmacológico , Gatos , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/efectos adversos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Difenilamina/uso terapéutico , Enfermedades de los Perros/tratamiento farmacológico , Perros , Fenilacetatos/farmacología , Fenilacetatos/uso terapéutico
9.
Can J Vet Res ; 86(1): 40-47, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34975221

RESUMEN

The aim of this study was to evaluate the perioperative effects of robenacoxib on serum C-reactive protein (CRP) and iron concentrations in dogs undergoing gonadectomy. In a prospective, blinded, controlled clinical trial, 60 healthy dogs were randomly assigned to receive preoperative subcutaneous injection of either robenacoxib [2 mg/kg body weight (BW)], meloxicam (0.2 mg/kg BW), or saline (0.04 mL/kg BW), followed by oral administration over 72 h (robenacoxib: 2 to 4 mg/kg BW; meloxicam: 0.1 mg/kg BW; saline: gelatin capsules). Blood samples were taken before surgery and 12, 24, 48, 72 h, and 7 d after surgery. Pain scores were assessed via the short-form Glasgow Composite Pain Scale over 72 h postoperatively. C-reactive protein (CRP) and iron serum levels increased and decreased (P < 0.01, both), respectively, after surgery and returned to baseline within 1 wk. No differences were observed among treatments (P > 0.05) or based on surgery/gender (P > 0.05). Pain assessment revealed a higher incidence of treatment failure in saline (6 females versus 2 and 1 female in robenacoxib and meloxicam, respectively). In conclusion, robenacoxib and meloxicam had no influence on postoperative CRP or iron in dogs, which suggests that these nonsteroidal anti-inflammatory drugs (NSAIDs) do not have a relevant effect on these biomarkers.


Le but de cette étude était d'évaluer les effets périopératoires du robenacoxib sur les concentrations sériques de protéine C réactive (CRP) et de fer chez des chiens subissant une gonadectomie. Dans un essai clinique prospectif, en aveugle et contrôlé, 60 chiens en bonne santé ont été randomisés pour recevoir une injection sous-cutanée préopératoire de robenacoxib [2 mg/kg de poids corporel (PC)], de méloxicam (0,2 mg/kg de poids corporel) ou de solution saline (0,04 mL/kg de poids corporel), suivie d'une administration orale pendant 72 h (robenacoxib : 2 à 4 mg/kg de poids corporel; méloxicam : 0,1 mg/kg de poids corporel; saline : gélules). Des échantillons de sang ont été prélevés avant la chirurgie et 12, 24, 48, 72 h et 7 jours après la chirurgie. Les pointages de douleur ont été évalués via l'échelle abrégée Glasgow Composite Pain Scale sur 72 h après l'opération. Les taux sériques de CRP et de fer ont augmenté et diminué (P < 0,01, les deux), respectivement, après la chirurgie et sont revenus à la valeur de base en 1 semaine. Aucune différence n'a été observée entre les traitements (P > 0,05) ou en fonction de la chirurgie/du sexe (P > 0,05). L'évaluation de la douleur a révélé une incidence plus élevée d'échec du traitement avec la saline (6 femelles contre 2 et 1 femelles pour le robenacoxib et le méloxicam, respectivement). En conclusion, le robenacoxib et le méloxicam n'ont eu aucune influence sur la CRP ou le fer postopératoire chez le chien, ce qui suggère que ces anti-inflammatoires non stéroïdiens (AINS) n'ont pas d'effet pertinent sur ces biomarqueurs.(Traduit par Docteur Serge Messier).


Asunto(s)
Proteína C-Reactiva , Castración , Difenilamina/análogos & derivados , Hierro , Fenilacetatos , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Biomarcadores/sangre , Proteína C-Reactiva/análisis , Proteína C-Reactiva/efectos de los fármacos , Castración/veterinaria , Difenilamina/administración & dosificación , Difenilamina/farmacología , Perros , Femenino , Hierro/sangre , Meloxicam/administración & dosificación , Meloxicam/farmacología , Dolor Postoperatorio/prevención & control , Dolor Postoperatorio/veterinaria , Atención Perioperativa/veterinaria , Fenilacetatos/administración & dosificación , Fenilacetatos/farmacología , Estudios Prospectivos
10.
Cell Rep ; 36(11): 109705, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525377

RESUMEN

Mouse embryonic stem cells (mESCs) can be maintained in vitro in defined N2B27 medium supplemented with two chemical inhibitors for GSK3 and MEK (2i) and the cytokine leukemia inhibitory factor (LIF), which act synergistically to promote self-renewal and pluripotency. Here, we find that genetic deletion of the four genes encoding the TCF/LEF transcription factors confers mESCs with the ability to self-renew in N2B27 medium alone. TCF/LEF quadruple knockout (qKO) mESCs display dysregulation of several genes, including Aire, Dnmt3l, and IcosL, located adjacent to each other within a topologically associated domain (TAD). Aire, Dnmt3l, and IcosL appear to be regulated by TCF/LEF in a ß-catenin independent manner. Moreover, downregulation of Aire and Dnmt3l in wild-type mESCs mimics the loss of TCF/LEF and increases mESC survival in the absence of 2iL. Hence, this study identifies TCF/LEF effectors that mediate exit from the pluripotent state.


Asunto(s)
Autorrenovación de las Células , Factor Nuclear 1-alfa del Hepatocito/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Proteína 1 Similar al Factor de Transcripción 7/genética , Proteína 2 Similar al Factor de Transcripción 7/genética , Animales , Benzamidas/farmacología , Autorrenovación de las Células/efectos de los fármacos , Medios de Cultivo/química , Medios de Cultivo/farmacología , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Difenilamina/análogos & derivados , Difenilamina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Edición Génica , Factor Nuclear 1-alfa del Hepatocito/deficiencia , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Ligando Coestimulador de Linfocitos T Inducibles/antagonistas & inhibidores , Ligando Coestimulador de Linfocitos T Inducibles/genética , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Factor de Unión 1 al Potenciador Linfoide/deficiencia , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína 1 Similar al Factor de Transcripción 7/deficiencia , Proteína 1 Similar al Factor de Transcripción 7/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/deficiencia , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , beta Catenina/deficiencia , beta Catenina/genética , Proteína AIRE
11.
Stem Cell Res ; 56: 102537, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34562798

RESUMEN

As an important mechanical cue in the extracellular microenvironment, osmotic stress directly affects the proliferation, migration, and differentiation of cells. In this paper, we focused on the influence of hypertonic pressure on the colony morphology, stemness, and self-renew of mouse embryonic stem cells (mESCs). Our results showed that culture media with hypertonic pressure are more conducive to the maintenance of 3D colony morphology and pluripotency of mESCs after withdrawing the glycogen synthase kinase 3ß (GSK3ß) inhibitor CHIR99021 and the mitogen-activated protein kinase (MEK) inhibitor PD0325901 (hereinafter referred to as 2i) for 48 h. Furthermore, we revealed the microscopic mechanisms of the this finding: hypertonic pressure resulted in the depolymerization of F-actin cytoskeleton and limits Yes-associated protein (hereinafter referred to as YAP) transmission into the nucleus which play a vital role in the regulation of cell proliferation, and resulting in cell-cycle arrest at last.


Asunto(s)
Células Madre Embrionarias de Ratones , Presión Osmótica , Animales , Benzamidas , Diferenciación Celular , Proliferación Celular , Difenilamina/análogos & derivados , Ratones , Proteínas Quinasas Activadas por Mitógenos , Células Madre Embrionarias de Ratones/fisiología
12.
Int J Mol Sci ; 22(13)2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34201614

RESUMEN

The use of MEK inhibitors in the therapy of uveal melanoma (UM) has been investigated widely but has failed to show benefits in clinical trials due to fast acquisition of resistance. In this study, we investigated a variety of therapeutic compounds in primary-derived uveal melanoma cell lines and found monosomy of chromosome 3 (M3) and mutations in BAP1 to be associated with higher resistance to MEK inhibition. However, reconstitution of BAP1 in a BAP1-deficient UM cell line was unable to restore sensitivity to MEK inhibition. We then compared UM tumors from The Cancer Genome Atlas (TCGA) with mutations in BAP1 with tumors with wild-type BAP1. Principal component analysis (PCA) clearly differentiated both groups of tumors, which displayed disparate overall and progression-free survival data. Further analysis provided insight into differential expression of genes involved in signaling pathways, suggesting that the downregulation of the eukaryotic translation initiation factor 2A (EIF2A) observed in UM tumors with BAP1 mutations and M3 UM cell lines might lead to a decrease in ribosome biogenesis while inducing an adaptive response to stress. Taken together, our study links loss of chromosome 3 with decreased sensitivity to MEK inhibition and gives insight into possible related mechanisms, whose understanding is fundamental to overcome resistance in this aggressive tumor.


Asunto(s)
Cromosomas Humanos Par 3/genética , Resistencia a Antineoplásicos/genética , Melanoma/genética , Monosomía , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Úvea/genética , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN , Difenilamina/análogos & derivados , Difenilamina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Melanoma/tratamiento farmacológico , Melanoma/mortalidad , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Piridonas/farmacología , Pirimidinonas/farmacología , Sulfonamidas/farmacología , Análisis de Supervivencia , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Neoplasias de la Úvea/tratamiento farmacológico , Neoplasias de la Úvea/mortalidad
13.
J Vet Intern Med ; 35(5): 2384-2394, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34196973

RESUMEN

BACKGROUND: Evaluate the clinical safety of robenacoxib in cats with chronic musculoskeletal disease (CMSD). ANIMALS: Four hundred forty-nine client-owned cats with CMSD. METHODS: Pooled analysis of safety variables from 4 prospective randomized blinded clinical trials of robenacoxib (n = 222) versus placebo (n = 227), administered orally once daily for 4 to 12 weeks. Safety was evaluated from reported adverse events (AEs) and abnormalities detected on hematology and serum and urine chemistry analyses. RESULTS: The number of cats with at least 1 AE was not significantly different (P = .15) with robenacoxib (n = 106, 47.8%) compared to placebo (n = 93, 41.0%). The relative risk of at least 1 AE (incidence robenacoxib/placebo) was 1.15 (95% confidence interval 0.93-1.43). There was no significant difference between groups in the number of clinical signs (range, 0-9) per cat (P = .23). Serum creatinine concentrations were higher during robenacoxib administration compared to placebo (+4.36 µmol/L, 95% confidence interval 0.21-8.50), but no related adverse clinical effects were detected. In the subgroup of 126 cats with evidence of chronic kidney disease, the relative risk of at least 1 AE (robenacoxib/placebo) was 1.09 (95% confidence interval 0.78-1.52, P = .61). CONCLUSIONS AND CLINICAL IMPORTANCE: Robenacoxib was not associated with increased risk of AEs compared to placebo when administered for 4 to 12 weeks to cats with CMSD. The generalizability of the results to general practice is limited by the fact that cases with severe and uncontrolled concomitant diseases were not included.


Asunto(s)
Enfermedades de los Gatos , Enfermedades Musculoesqueléticas , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Enfermedades de los Gatos/tratamiento farmacológico , Gatos , Difenilamina/efectos adversos , Difenilamina/análogos & derivados , Enfermedades Musculoesqueléticas/tratamiento farmacológico , Enfermedades Musculoesqueléticas/veterinaria , Fenilacetatos , Estudios Prospectivos
14.
J Zoo Wildl Med ; 52(2): 529-537, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34130395

RESUMEN

Postoperative antinociception control in fish is currently suboptimal, as commonly used antiinflammatory drugs last for only a few hours at tested temperatures. Therefore, long-acting anti-inflammatory drugs, such as robenacoxib, could improve the welfare of fish. The pharmacokinetics, duration of antinociceptive action, and potential adverse effects of robenacoxib were evaluated through two prospective randomized blinded trials in rainbow trout (Oncorhynchus mykiss). Six healthy rainbow trout received a single IM administration of robenacoxib (2 mg/kg), and two control fish received the same volume of saline IM. Blood samples were collected at predetermined time points for 5 d. Plasma robenacoxib concentrations were measured using high-performance liquid chromatography-high-resolution hybrid orbitrap mass spectrometry and noncompartmental pharmacokinetic analysis. Ten additional rainbow trout received an intralabial injection of 0.05 ml of 2% acetic acid following a previously validated nociceptive model. The treated group (n = 6) received 2 mg/kg of robenacoxib IM and the control group (n = 4) received an equivalent volume of saline IM. The behavior, appetite, and opercular rate of the fish were evaluated every hour for 5 h, then once daily for 3 d. All 12 treated trout and 6 controls underwent histopathologic evaluation. Average maximum plasma concentration (Cmax) was 329.9 ± 137.3 ng/ml observed at 2.1 ± 0.7 h (Tmax) and terminal half-life was 12.6 ± 2.27 h. Plasma concentrations described as antinociceptive in domestic carnivores were measured for 3-4 d. This dose was associated with a significant decrease in rocking behavior (P = 0.017). No adverse effects were detected clinically nor on histopathology. Robenacoxib administered IM at 2 mg/kg appears to be safe and may provide an antinociceptive effect in rainbow trout. This study presents a new therapeutic option to provide long-lasting antinociception in rainbow trout.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/farmacocinética , Difenilamina/análogos & derivados , Oncorhynchus mykiss/sangre , Fenilacetatos/farmacocinética , Animales , Área Bajo la Curva , Inhibidores de la Ciclooxigenasa 2/efectos adversos , Difenilamina/efectos adversos , Difenilamina/farmacocinética , Femenino , Semivida , Masculino , Fenilacetatos/efectos adversos
15.
FEBS Lett ; 595(14): 1949-1961, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34056710

RESUMEN

In embryonic stem (ES) cells, oxidative stress control is crucial for genomic stability, self-renewal, and cell differentiation. Heme oxygenase-1 (HO-1) is a key player of the antioxidant system and is also involved in stem cell differentiation and pluripotency acquisition. We found that the HO-1 gene is expressed in ES cells and induced after promoting differentiation. Moreover, downregulation of the pluripotency transcription factor (TF) OCT4 increased HO-1 mRNA levels in ES cells, and analysis of ChIP-seq public data revealed that this TF binds to the HO-1 gene locus in pluripotent cells. Finally, ectopic expression of OCT4 in heterologous systems repressed a reporter carrying the HO-1 gene promoter and the endogenous gene. Hence, this work highlights the connection between pluripotency and redox homeostasis.


Asunto(s)
Regulación de la Expresión Génica , Hemo-Oxigenasa 1/genética , Proteínas de la Membrana/genética , Células Madre Embrionarias de Ratones/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/metabolismo , ARN Mensajero/genética , Animales , Benzamidas/farmacología , Diferenciación Celular/efectos de los fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Embrión de Mamíferos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hemo-Oxigenasa 1/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células 3T3 NIH , Proteína Homeótica Nanog/antagonistas & inhibidores , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/antagonistas & inhibidores , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Regiones Promotoras Genéticas , Piridinas/farmacología , Pirimidinas/farmacología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factores de Transcripción SOXB1/antagonistas & inhibidores , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Transcripción Genética
16.
J Vet Pharmacol Ther ; 44(3): 411-416, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33881783

RESUMEN

The objective of the studies was to determine the route of excretion, faecal or urinary, of the nonsteroidal anti-inflammatory drug (NSAID) robenacoxib (Onsior™) in cats and dogs. The studies employed a two-part crossover design in 4 beagle dogs (2 female and 2 male, age 36-41 months and body weight 9.0-10.3 kg) and a parallel group comparison of two groups each of 3 domestic short-hair cats (2 female and 4 castrated male, age 35-73 months and body weight 3.0-5.7 kg). Animals were administered single doses of 1 (dog) or 2 (cat) mg/kg of [14 C]-robenacoxib by intravenous (IV) and oral routes. Venous blood samples were taken and analysed for robenacoxib concentration. Faeces and urine were collected for 4 (cats) or 7 (dogs) days and analysed for radioactivity. Robenacoxib was eliminated rapidly from blood (≤ 8 hr). In dogs, expressed as the percentage of the administered dose and adjusted so that faecal plus urine recovery was 100%, the mean (SD) excretion in faeces and urine was, respectively, 64.6% (4.30) and 35.4% (4.3) after IV and 66.7% (6.9) and 33.3% (6.9) after oral administration. The respective values in cats, in faeces and urine, were 72.5% (4.6) and 27.5% (4.6) after IV and 78.5% (2.6) and 21.5% (2.6) after oral administration. In conclusion, excretion of systemically available robenacoxib in cats and dogs was mixed via both faeces and urine, but predominately faecal (~64.6% in dogs and ~72.5% in cats) and assumed to be via biliary excretion.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Gatos/metabolismo , Difenilamina , Perros/metabolismo , Fenilacetatos , Animales , Difenilamina/análogos & derivados , Femenino , Masculino , Proyectos Piloto
17.
Sci Rep ; 11(1): 7721, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33833276

RESUMEN

The main objective of this pilot clinical trial was to evaluate outcome measures for the assessment of the nonsteroidal anti-inflammatory drug (NSAID) robenacoxib in cats with degenerative joint disease-associated pain (DJD-pain). Otherwise healthy cats (n = 109) with DJD-pain entered a parallel group, randomized, blinded clinical trial. Cats received placebo (P) or robenacoxib (R) for two consecutive 3-week periods. Treatment groups were PP, RR, and RP. Actimetry and owner-assessment data were collected. Data were analyzed using mixed-effects and generalized mixed-effects linear models. Activity data showed high within-cat and between-cat variability, and 82.4% of the values were zero. Compared to placebo, mean total activity was higher (5.7%) in robenacoxib-treated cats (p = 0.24); for the 80th percentile of activity, more robenacoxib-treated cats had a > 10% increase in activity after 3 (p = 0.046) and 6 weeks (p = 0.026). Robenacoxib treatment significantly decreased owner-assessed disability, (p = 0.01; 49% reduction in disability; effect size ~ 0.3), and improved temperament (p = 0.0039) and happiness (p = 0.021) after 6 weeks. More robenacoxib-treated cats were successes at 6 weeks (p = 0.018; NNT: 3.8). Adverse effect frequencies were similar across groups. Results identified suitable endpoints for confirmatory studies, while also indicating efficacy of robenacoxib in cats with DJD-pain.


Asunto(s)
Enfermedades de los Gatos/tratamiento farmacológico , Difenilamina/análogos & derivados , Artropatías/veterinaria , Osteoartritis/veterinaria , Manejo del Dolor/veterinaria , Dolor/veterinaria , Fenilacetatos/uso terapéutico , Animales , Gatos , Difenilamina/uso terapéutico , Femenino , Artropatías/complicaciones , Masculino , Osteoartritis/complicaciones , Dolor/complicaciones , Proyectos Piloto , Placebos
18.
Biochem Biophys Res Commun ; 553: 58-64, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33756346

RESUMEN

Human embryonic stem cells (hESCs) have the unique feature of unlimited self-renewal and differentiation into derivatives of all three germ layers in human body, providing a powerful in vitro model for studying cell differentiation. FGF2, BMP4 and TGF-ß signaling have been shown to play crucial roles in mesendodermal differentiation of hESCs. However, their underlying molecular mechanisms and other signaling pathways potentially involved in mesendodermal differentiation of hESCs remain to be further investigated. In this study, we uncover that VEGF signaling pathway plays a critical role in the mesendodermal induction of hESCs. Treating hESCs with Lenvatinib, a pan-inhibitor of VEGF receptors (VEGFRs), impedes their mesendodermal induction. Conversely, overexpression of VEGFA165, a major human VEGF isoform, promotes the mesendodermal differentiation. Similar to the VEGFR inhibitor, MEK inhibitor PD0325901 hinders mesendodermal induction of hESCs. In contrast, overexpression of ERK2GOF, an intrinsically active ERK2 mutant, markedly reduces the inhibitory effect of the VEGFR inhibitor. Thus, the MEK-ERK cascade plays an important role for the function of VEGF signaling pathway in the mesendodermal induction of hESCs. All together, this study identifies the critical role of VEGF signaling pathway as well as potential crosstalk of VEGF signaling pathway with other known signaling pathways in mesendodermal differentiation of hESCs.


Asunto(s)
Endodermo/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Sistema de Señalización de MAP Quinasas , Mesodermo/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Benzamidas/farmacología , Diferenciación Celular/efectos de los fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína Smad1/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Proteína Smad5/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
19.
Nat Commun ; 12(1): 1863, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33767186

RESUMEN

Embryonic stem cells (ESCs) can be maintained in the naïve state through inhibition of Mek1/2 and Gsk3 (2i). A relevant effect of 2i is the inhibition of Cdk8/19, which are negative regulators of the Mediator complex, responsible for the activity of enhancers. Inhibition of Cdk8/19 (Cdk8/19i) stimulates enhancers and, similar to 2i, stabilizes ESCs in the naïve state. Here, we use mass spectrometry to describe the molecular events (phosphoproteome, proteome, and metabolome) triggered by 2i and Cdk8/19i on ESCs. Our data reveal widespread commonalities between these two treatments, suggesting overlapping processes. We find that post-transcriptional de-repression by both 2i and Cdk8/19i might support the mitochondrial capacity of naive cells. However, proteome reprogramming in each treatment is achieved by different mechanisms. Cdk8/19i acts directly on the transcriptional machinery, activating key identity genes to promote the naïve program. In contrast, 2i stabilizes the naïve circuitry through, in part, de-phosphorylation of downstream transcriptional effectors.


Asunto(s)
Quinasa 8 Dependiente de Ciclina/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Células Madre Embrionarias de Ratones/citología , Células Madre Pluripotentes/citología , Animales , Benzamidas/farmacología , Línea Celular , Difenilamina/análogos & derivados , Difenilamina/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Fosforilación/fisiología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
20.
Development ; 148(6)2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33782043

RESUMEN

Rostro-caudal patterning of vertebrates depends on the temporally progressive activation of HOX genes within axial stem cells that fuel axial embryo elongation. Whether the pace of sequential activation of HOX genes, the 'HOX clock', is controlled by intrinsic chromatin-based timing mechanisms or by temporal changes in extrinsic cues remains unclear. Here, we studied HOX clock pacing in human pluripotent stem cell-derived axial progenitors differentiating into diverse spinal cord motor neuron subtypes. We show that the progressive activation of caudal HOX genes is controlled by a dynamic increase in FGF signaling. Blocking the FGF pathway stalled induction of HOX genes, while a precocious increase of FGF, alone or with GDF11 ligand, accelerated the HOX clock. Cells differentiated under accelerated HOX induction generated appropriate posterior motor neuron subtypes found along the human embryonic spinal cord. The pacing of the HOX clock is thus dynamically regulated by exposure to secreted cues. Its manipulation by extrinsic factors provides synchronized access to multiple human neuronal subtypes of distinct rostro-caudal identities for basic and translational applications.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Relojes Circadianos , Proteínas de Homeodominio/metabolismo , Neuronas Motoras/metabolismo , Células Madre Pluripotentes/metabolismo , Benzamidas/farmacología , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular , Relojes Circadianos/efectos de los fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Regulación del Desarrollo de la Expresión Génica , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Factores de Diferenciación de Crecimiento/farmacología , Proteínas de Homeodominio/genética , Humanos , Neuronas Motoras/citología , Células Madre Pluripotentes/citología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Médula Espinal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...