Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Biol Pharm Bull ; 47(3): 669-679, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38508765

RESUMEN

Osteoporosis is caused by imbalance between osteogenesis and bone resorption, thus, osteogenic drugs and resorption inhibitors are used for treatment of osteoporosis. The present study examined the effects of (R)-4-(1-hydroxyethyl)-3-{4-[2-(tetrahydropyran-4-yloxy)ethoxy]phenoxy}benzamide (KY-273), a diphenyl ether derivative, on CDK8/19 activity, osteoblast differentiation and femoral bone using micro-computed tomography in female rats. KY-273 potently inhibited CDK8/19 activity, promoted osteoblast differentiation with an increase in alkaline phosphatase (ALP) activity, and gene expression of type I collagen, ALP and BMP-4 in mesenchymal stem cells (ST2 cells). In female rat femur, ovariectomy decreased metaphyseal trabecular bone volume (Tb.BV), mineral content (Tb.BMC), yet had no effect on metaphyseal and diaphyseal cortical bone volume (Ct.BV), mineral content (Ct.BMC) and strength parameters (BSPs). In ovaries-intact and ovariectomized rats, oral administration of KY-273 (10 mg/kg/d) for 6 weeks increased metaphyseal and diaphyseal Ct.BV, Ct.BMC, and BSPs without affecting medullary volume (Med.V), but did not affect Tb.BV and Tb.BMC. In ovariectomized rats, alendronate (3 mg/kg/d) caused marked restoration of Tb.BV, Tb.BMC and structural parameters after ovariectomy, and increased metaphyseal but not diaphyseal Ct.BV, Ct.BMC, and BSPs. In ovaries-intact and ovariectomized rats, by the last week, KY-273 increased bone formation rate/bone surface at the periosteal but not the endocortical side. These findings indicate that KY-273 causes osteogenesis in cortical bone at the periosteal side without reducing Med.V. In conclusion, KY-273 has cortical-bone-selective osteogenic effects by osteoblastogenesis via CDK8/19 inhibition in ovaries-intact and ovariectomized rats, and is an orally active drug candidate for bone diseases such as osteoporosis in monotherapy and combination therapy.


Asunto(s)
Células Madre Mesenquimatosas , Osteoporosis , Humanos , Ratas , Femenino , Animales , Osteogénesis , Densidad Ósea , Ratas Sprague-Dawley , Microtomografía por Rayos X , Osteoporosis/tratamiento farmacológico , Ovariectomía , Minerales/farmacología , Quinasa 8 Dependiente de Ciclina
2.
Biol Pharm Bull ; 46(10): 1435-1443, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37779045

RESUMEN

Osteoporosis is treated with oral and parenteral bone resorption inhibitors such as bisphosphonates, and parenteral osteogenic drugs including parathyroid hormone (PTH) analogues and anti-sclerostin antibodies. In the present study, we synthesized KY-054, a 4,6-substituted coumarin derivative, and found that it potently promoted osteoblast differentiation with an increase in alkaline phosphatase (ALP) activity at 0.01-1 µM in mouse-derived mesenchymal stem cells (ST2 cells) and rat bone marrow-derived mesenchymal stem cells (BMSCs). In the ovariectomized (OVX) rats, KY-054 (10 mg/kg/d, 8 weeks) increased plasma bone-type ALP activity, suggesting in vivo promoting effects on osteoblast differentiation and/or activation. In dual-energy X-ray absorption (DEXA) scanning, KY-054 significantly increased the distal and diaphyseal femurs areal bone mineral density (aBMD) that was decreased by ovariectomy, indicating its beneficial effects on bone mineral contents (BMC) and/or bone volume (BV). In micro-computed tomography (micro-CT) scanning, KY-054 had no effect on metaphysis trabecular bone loss and microarchitecture parameters weakened by ovariectomy, but instead increased metaphysis and diaphysis cortical bone volume (Ct.BV) and cortical BMC (Ct.BMC) without reducing medullary volume (Med.V), resulting in increased bone strength parameters. It is concluded that KY-054 preferentially promotes metaphysis and diaphysis cortical bone osteogenesis with little effect on metaphysis trabecular bone resorption, and is a potential orally active osteogenic anti-osteoporosis drug candidate.


Asunto(s)
Osteogénesis , Osteoporosis , Ratas , Femenino , Animales , Ratones , Humanos , Microtomografía por Rayos X , Huesos , Densidad Ósea , Fémur , Osteoporosis/tratamiento farmacológico , Hueso Cortical , Ovariectomía
3.
Chem Pharm Bull (Tokyo) ; 71(12): 859-878, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38044139

RESUMEN

Hurler syndrome, a type of Mucopolysaccharidosis type I, is an inherited disorder caused by the accumulation of glycosaminoglycans (GAG) due to a deficiency in lysosomal α-L-iduronidase (IDUA), resulting in multiorgan dysfunction. In many patients with Hurler syndrome, IDUA proteins are not produced due to nonsense mutations in their genes; therefore, readthrough-inducing compounds, such as gentamycin, are expected to restore IDUA proteins by skipping the premature termination codon. In the present study, we synthesized a series of chromenopyridine derivatives to identify novel readthrough-inducing compounds. The readthrough-inducing activities of synthesized compounds were examined by measuring cellular IDUA activities and GAG concentrations in Hurler syndrome patient-derived cells. Compounds with a difluorophenyl group at the 2-position of chromenopyridine, a cyclobutyl group at the 3-position, and a basic side chain or basic fused ring exhibited excellent readthrough-inducing activities. KY-640, a chromenopyridine derivative with a tetrahydroisoquinoline sub-structure, increased the cellular IDUA activities of patient-derived cells by 3.2-fold at 0.3 µM and significantly reduced GAG concentrations, and also significantly increased enzyme activity in mouse models, suggesting its therapeutic potential in patients with Hurler syndrome.


Asunto(s)
Mucopolisacaridosis I , Ratones , Animales , Humanos , Mucopolisacaridosis I/tratamiento farmacológico , Mucopolisacaridosis I/genética , Codón sin Sentido
4.
Chem Pharm Bull (Tokyo) ; 71(9): 701-716, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37661376

RESUMEN

The readthrough mechanism, which skips the premature termination codon and restores the biosynthesis of the defective enzyme, is an emerging therapeutic tactic for nonsense mutation-related diseases, such as Hurler syndrome, a type of mucopolysaccharidosis. In the present study, novel triaryl derivatives were synthesized and their readthrough-inducing activities were evaluated by a luciferase reporter assay with a partial α-L-iduronidase (IDUA) DNA sequence containing the Q70X nonsense mutation found in Hurler syndrome and by measuring the enzyme activity of IDUA knockout cells transfected with the mutant IDUA gene. KY-516, a representative compound in which the meta position carboxyl group of the left ring of the clinically used ataluren was converted to the para position sulfamoylamino group, the central ring to triazole, and the right ring to cyanobenzene, exhibited the most potent readthrough-inducing activity in the Q70X/luciferase reporter assay. In Q70X mutant IDUA transgenic cells, KY-516 significantly increased enzyme activity at 0.1 µM. After the oral administration of KY-516 (10 mg/kg), the highest plasma concentration of KY-516 was above 5 µM in rats. These results indicate that KY-516, a novel triaryl derivative, exhibits potent readthrough-inducing activity and has potential as a therapeutic agent for Hurler syndrome.


Asunto(s)
Mucopolisacaridosis I , Animales , Ratas , Mucopolisacaridosis I/tratamiento farmacológico , Mucopolisacaridosis I/genética , Codón sin Sentido , Administración Oral , Bioensayo , Triazoles
5.
Biol Pharm Bull ; 44(5): 659-668, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33952822

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARγ) modulators are expected to exert anti-diabetic effects without PPARγ-related adverse effects, such as fluid retention, weight gain, and bone loss. The present study showed that the novel tetrazole derivative KY-903 exerted similar selective PPARγ partial agonist properties to INT-131, a known PPARγ modulator, in transactivation assays, and decreased plasma glucose and triglyceride levels with increases in adiponectin levels in diabetic KK-Ay mice. These effects were similar to those of pioglitazone. Pioglitazone, but not KY-903, increased adipose tissue and heart weights. In pre-adipocytes (3T3-L1), KY-903, in contrast to pioglitazone, increased adiponectin mRNA levels without adipocyte differentiation, indicating anti-diabetic effects via adiponectin without adipogenesis. In ovariectomized rats fed a high-fat diet (OVX/HFD), KY-903 and pioglitazone decreased plasma triglyceride and non-esterified fatty acid levels and increased adiponectin levels, indicating insulin sensitization via adiponectin. KY-903 reduced body weight gain and adipose tissue weight, while pioglitazone increased heart weight and markedly reduced bone mineral density. In mesenchymal stem cell-like ST2 cells, KY-903 slightly reduced osteoblast differentiation without adipocyte differentiation, while pioglitazone markedly reduced it with adipocyte differentiation. In conclusion, KY-903 is a novel PPARγ modulator that exerts anti-diabetic effects without body weight gain or cardiac hypertrophy in diabetic mice and anti-obesity effects with minor bone loss in OVX/HFD, possibly due to increases in adiponectin levels without adipogenesis.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Obesidad/tratamiento farmacológico , PPAR gamma/agonistas , Células 3T3-L1 , Adipogénesis/efectos de los fármacos , Adiponectina/análisis , Adiponectina/metabolismo , Animales , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/genética , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Hipoglucemiantes/uso terapéutico , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Obesidad/sangre , Obesidad/etiología , PPAR gamma/metabolismo , Pioglitazona/farmacología , Pioglitazona/uso terapéutico , Ratas , Tetrazoles/química , Triglicéridos/sangre , Triglicéridos/metabolismo , Aumento de Peso/efectos de los fármacos
6.
Chem Pharm Bull (Tokyo) ; 69(4): 333-351, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790079

RESUMEN

A novel series of 7-substituted-2-[3-(2-furyl)acryloyl]-6-tetrazolyl-1,2,3,4-tetrahydroisoquinoline derivatives were synthesized to clarify structure-activity relationships for peroxisome proliferator-activated receptor γ (PPARγ) partial agonist activity and identify more efficacious PPARγ partial agonists with minor adverse effects. Among the derivatives synthesized, compound 26v with a 2-(2,5-dihydropyrrol-1-yl)-5-methyloxazol-4-ylmethoxy group at the 7-position of the tetrahydroisoquinoline structure exhibited stronger PPARγ agonist and antagonist activities (EC50 = 6 nM and IC50 = 101 nM) than previously reported values for compound 1 (EC50 = 13 nM and IC50 = 512 nM). Compound 26v had very weak protein tyrosine phosphatase 1B (PTP1B) inhibitory activity and showed higher oral absorption (Cmax = 11.4 µg/mL and area under the curve (AUC) = 134.7 µg·h/mL) than compound 1 (Cmax = 7.0 µg/mL and AUC = 63.9 µg·h/mL) in male Sprague-Dawley (SD) rats. A computational docking calculation revealed that 26v bound to PPARγ in a similar manner to that of compound 1. In male Zucker fatty rats, 26v and pioglitazone at 10 and 30 mg/kg for 4 weeks similarly reduced plasma triglyceride levels, increased plasma adiponectin levels, and attenuated increases in plasma glucose levels in the oral glucose tolerance test, while only pioglitazone decreased hematocrit values. In conclusion, 6-tetrazolyl-1,2,3,4-tetrahydroisoquinoline derivatives provide a novel scaffold for selective PPARγ partial agonists and 26v attenuates insulin resistance possibly by adiponectin enhancements with minor adverse effects.


Asunto(s)
PPAR gamma/agonistas , Tetrahidroisoquinolinas/química , Tetrahidroisoquinolinas/farmacología , Animales , Técnicas de Química Sintética , Descubrimiento de Drogas , Humanos , Masculino , Modelos Moleculares , PPAR gamma/metabolismo , Ratas Sprague-Dawley , Ratas Zucker , Tetrahidroisoquinolinas/síntesis química
7.
Chem Pharm Bull (Tokyo) ; 67(11): 1211-1224, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31685749

RESUMEN

A novel series of 2,6,7-substituted 3-unsubstituted 1,2,3,4-tetrahydroisoquinoline derivatives were synthesized to find a peroxisome proliferator-activated receptor γ (PPARγ) partial agonist. Among the derivatives, (E)-7-[2-(cyclopent-3-eny)-5-methyloxazol-4-ylmethoxy]-2-[3-(2-furyl)acryloyl]-6-(1H-tetrazol-5-yl)-1,2,3,4-tetrahydroisoquinoline (20g) exhibited potent partial agonist activity (EC50 = 13 nM, maximal response 30%) and very weak protein tyrosine phosphatase 1B (PTP1B) inhibition (IC50 = 1100 nM), indicating a selective PPARγ partial agonist. A computational docking calculation revealed that 20g bound to PPARγ in a similar manner to that of known partial agonists. In male and female KK-Ay mice with insulin resistance and hyperglycemia, 20g at 30 mg/kg for 7 d significantly reduced plasma glucose levels, but not triglyceride levels. The effects of 20g were similar to those of pioglitazone at 10 mg/kg. In conclusion, the 2,6,7-substituted 1,2,3,4-tetrahydroisoquinoline with an acidic group at the 6-position provides a novel scaffold for selective PPARγ partial agonists and 20g exerted anti-diabetic effects via the partial activation of PPARγ.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Hiperglucemia/tratamiento farmacológico , Hipoglucemiantes/farmacología , PPAR gamma/agonistas , Tetrahidroisoquinolinas/farmacología , Administración Oral , Animales , Glucemia/efectos de los fármacos , Cristalografía por Rayos X , Femenino , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/química , Resistencia a la Insulina , Masculino , Ratones , Ratones Transgénicos , Simulación del Acoplamiento Molecular , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Tetrahidroisoquinolinas/administración & dosificación , Tetrahidroisoquinolinas/química
8.
J Pharmacol Sci ; 137(1): 38-46, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29731242

RESUMEN

The anti-diabetic and anti-obesity effects of the allosteric protein tyrosine phosphatase 1B (PTP1B) inhibitor 4-(biphenyl-4-ylmethylsulfanylmethyl)-N-(hexane-1-sulfonyl)benzoylamide (KY-226) were pharmacologically evaluated. KY-226 inhibited human PTP1B activity (IC50 = 0.28 µM), but did not exhibit peroxisome proliferator-activated receptor γ (PPARγ) agonist activity. In rodent preadipocytes (3T3-L1), KY-226 up to 10 µM had no effects on adipocyte differentiation, whereas pioglitazone, a PPARγ agonist, markedly promoted it. In human hepatoma-derived cells (HepG2), KY-226 (0.3-10 µM) increased the phosphorylated insulin receptor (pIR) produced by insulin. In db/db mice, the oral administration of KY-226 (10 and 30 mg/kg/day, 4 weeks) significantly reduced plasma glucose and triglyceride levels as well as hemoglobin A1c values without increasing body weight gain, while pioglitazone exerted similar effects with increases in body weight gain. KY-226 attenuated plasma glucose elevations in the oral glucose tolerance test. KY-226 also increased pIR and phosphorylated Akt in the liver and femoral muscle. In high-fat diet-induced obese mice, the oral administration of KY-226 (30 and 60 mg/kg/day, 4 weeks) decreased body weight gain, food consumption, and fat volume gain with increases in phosphorylated STAT3 in the hypothalamus. In conclusion, KY-226 exerted anti-diabetic and anti-obesity effects by enhancing insulin and leptin signaling, respectively.


Asunto(s)
Benzamidas/farmacología , Compuestos de Bifenilo/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Insulina/metabolismo , Leptina/metabolismo , Obesidad/tratamiento farmacológico , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Células 3T3 , Adipocitos/citología , Animales , Benzamidas/uso terapéutico , Compuestos de Bifenilo/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/genética , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/uso terapéutico , Células Hep G2 , Humanos , Insulina/fisiología , Leptina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Terapia Molecular Dirigida , Obesidad/genética , Fosforilación , Factor de Transcripción STAT3/metabolismo
9.
Chem Pharm Bull (Tokyo) ; 66(12): 1131-1152, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30504630

RESUMEN

A novel series of 2-acyl-3-carboxyl-tetrahydroisoquinoline derivatives were synthesized and biologically evaluated. Among them, (S)-2-{(E)-3-furan-2-ylacryloyl}-7-[(2E,4E)-5-(2,4,6-trifluorophenyl)penta-2,4-dienyloxy]-1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (compound 17u) was identified as a potent protein tyrosine phosphatase 1B (PTP1B) inhibitor without peroxisome proliferator-activated receptor (PPAR) γ activation: PTP1B inhibition IC50=0.19 µM and PPARγ ΕC50>10 µM. Compound 17u exhibited mixed-type inhibition for PTP1B, and this mode of inhibition was rationalized by computational ligand docking into the catalytic and allosteric sites of PTP1B. Compound 17u also showed high oral absorption at 10 mg/kg (per os (p.o.), Cmax=4.67 µM) in rats, significantly reduced non-fasting plasma glucose and triglyceride levels with no side effects at 30 mg/kg/d (p.o.) for 4 weeks, and attenuated elevations in plasma glucose levels in the oral glucose tolerance test performed 24 h after its final administration in db/db mice. In conclusion, the substituted 2-acyl-3-carboxyl-tetrahydroisoquinoline is a novel scaffold of mixed-type PTP1B inhibitors without PPARγ activation, and compound 17u has potential as an efficacious and safe anti-diabetic drug as well as a useful tool for investigations on the physiological and pathophysiological effects of mixed-type PTP1B inhibition.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hipoglucemiantes/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Tetrahidroisoquinolinas/farmacología , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/química , Humanos , Hipoglucemiantes/sangre , Hipoglucemiantes/química , Masculino , Ratones , Ratones Endogámicos , Simulación del Acoplamiento Molecular , Estructura Molecular , PPAR gamma/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Tetrahidroisoquinolinas/sangre , Tetrahidroisoquinolinas/química
10.
Chem Pharm Bull (Tokyo) ; 65(12): 1144-1160, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29199219

RESUMEN

A novel series of benzoylsulfonamide derivatives were synthesized and biologically evaluated. Among them, 4-(biphenyl-4-ylmethylsulfanylmethyl)-N-(hexane-1-sulfonyl)benzamide (compound 18K) was identified as a protein tyrosine phosphatase 1B (PTP1B) inhibitor with potent and selective inhibitory activity against PTP1B (IC50=0.25 µM). Compound 18K functioned as a non-competitive inhibitor and bound to the allosteric site of PTP1B. It also showed high oral absorption in mice (the maximum drug concentration (Cmax)=45.5 µM at 30 mg/kg), rats (Cmax=53.6 µM at 30 mg/kg), and beagles (Cmax=37.8 µM at 10 mg/kg), and significantly reduced plasma glucose levels at 30 mg/kg/d (per os (p.o.)) for one week with no side effects in db/db mice. In conclusion, the substituted benzoylsulfonamide was shown to be a novel scaffold of a non-competitive and allosteric PTP1B inhibitor, and compound 18K has potential as an efficacious and safe anti-diabetic drug as well as a useful tool for investigations of the physiological and pathophysiological effects of allosteric PTP1B inhibition.


Asunto(s)
Inhibidores Enzimáticos/química , Hipoglucemiantes/química , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Sulfonamidas/química , Administración Oral , Regulación Alostérica/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Glucemia/análisis , Perros , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Concentración 50 Inhibidora , Masculino , Ratones , Ratones Obesos , Simulación de Dinámica Molecular , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Ratas , Ratas Sprague-Dawley , Alineación de Secuencia , Relación Estructura-Actividad , Sulfonamidas/metabolismo , Sulfonamidas/farmacología
11.
Chem Pharm Bull (Tokyo) ; 63(12): 998-1014, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26633022

RESUMEN

A novel series of 2,7-substituted 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid derivatives were synthesized and biologically evaluated. (S)-2-(2-Furylacryloyl)-7-[2-(2-methylindane-2-yl)-5-methyloxazol-4-yl]methoxy-1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid tert-butylamine salt (13jE) was identified as a potent human peroxisome proliferator-activated receptor γ (PPARγ)-selective agonist (EC50=85 nM) and human protein-tyrosine phosphatase 1B (PTP-1B) inhibitor (IC50=1.0 µM). Compound 13jE partially activated PPARγ, but not PPARα or PPARδ, and antagonized farglitazar, a full PPARγ agonist. Cmax after the oral administration of 13jE at 10 mg/kg was 28.6 µg/mL (53 µM) in male Sprague-Dawley (SD) rats. Repeated administration of 13jE and rosiglitazone for 14 d at 10 mg/kg/d decreased plasma glucose and triglyceride levels significantly in male KK-A(y) mice. Rosiglitazone, but not 13jE, significantly increased the plasma volume and liver weight. In conclusion, 13jE showed stronger hypoglycemic and hypolipidemic effects and weaker hemodilution and hepatotoxic effects than rosiglitazone, suggesting that its safer efficacy may be due to its partial PPARγ agonism and PTP-1B inhibition.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Hipoglucemiantes/farmacología , PPAR gamma/agonistas , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Tetrahidroisoquinolinas/farmacología , Animales , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/química , Humanos , Hipoglucemiantes/sangre , Hipoglucemiantes/química , Masculino , Ratones , Ratones Endogámicos , Estructura Molecular , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad , Tetrahidroisoquinolinas/sangre , Tetrahidroisoquinolinas/química
12.
J Pharmacol Sci ; 124(2): 276-85, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24553405

RESUMEN

The pharmacological profile of (S)-7-(2-{2-[(E)-2-cyclopentylvinyl]-5-methyloxazol-4-yl}-ethoxy)-2-[(2E,4E)-hexadienoyl]-1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (KY-201), a peroxisome proliferator-activated receptor (PPAR) γ agonist, was compared with that of rosiglitazone in ovariectomized rats. The serum triglyceride and non-esterified fatty acid reducing effects of KY-201 at 3 and 10 mg/kg per day for 6 weeks were similar to those of rosiglitazone despite its weaker PPARγ agonistic activity. KY-201 had no effects on body weight gain, blood volume, or heart and adipose weights, while rosiglitazone at 10 mg/kg per day increased them. KY-201 had few effects on bone mineral density (BMD) or fat in marrow (FM), whereas rosiglitazone strongly decreased BMD and increased FM. The PPARγ agonistic activity of KY-201 was weaker than that of rosiglitazone in ST-2 cells, and KY-201 reduced osteoblast differentiation and increased adipocyte differentiation less potently than rosiglitazone in rat bone marrow-derived mesenchymal stem cells. KY-201, but not rosiglitazone inhibited protein tyrosine phosphatase 1B (PTP1B) and increased phosphorylation of the insulin receptor in HepG2 cells. These results suggest that the hypolipidemic effects of KY-201 are similar to those of rosiglitazone, but with less adverse effects, due to the combination of PPARγ partial activation and PTP1B inhibition. KY-201 would be useful for treatments of diabetic patients at high risk of osteoporosis, cardiovascular disease, and/or obesity.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Isoquinolinas/farmacología , Osteoporosis Posmenopáusica/etiología , Oxazoles/farmacología , PPAR gamma/agonistas , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Receptor de Insulina/metabolismo , Adipocitos/citología , Animales , Densidad Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Tipo 2/complicaciones , Modelos Animales de Enfermedad , Ácidos Grasos no Esterificados/sangre , Femenino , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/efectos adversos , Hipolipemiantes , Isoquinolinas/administración & dosificación , Isoquinolinas/efectos adversos , Osteoblastos/citología , Ovariectomía , Oxazoles/administración & dosificación , Oxazoles/efectos adversos , Ratas , Ratas Endogámicas F344 , Riesgo , Rosiglitazona , Tiazolidinedionas/administración & dosificación , Tiazolidinedionas/efectos adversos , Tiazolidinedionas/farmacología , Triglicéridos/sangre
13.
Stem Cell Reports ; 17(7): 1576-1588, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35777359

RESUMEN

Bone marrow mesenchymal stem cells (MSCs) are critical regulators of postnatal bone homeostasis. Osteoporosis is characterized by bone volume and strength deterioration, partly due to MSC dysfunction. Cyclin-dependent kinase 8 (CDK8) belongs to the transcription-related CDK family. Here, CDK8 in MSCs was identified as important for bone homeostasis. CDK8 level was increased in aged MSCs along with the association with aging-related signals. Mouse genetic studies revealed that CDK8 in MSCs plays a crucial role in bone resorption and homeostasis. Mechanistically, CDK8 in MSCs extrinsically controls osteoclastogenesis through the signal transducer and transcription 1 (STAT1)-receptor activator of the nuclear factor κ Β ligand (RANKL) axis. Moreover, aged MSCs have high osteoclastogenesis-supporting activity, partly through a CDK8-dependent manner. Finally, pharmacological inhibition of CDK8 effectively repressed MSC-dependent osteoclastogenesis and prevented ovariectomy-induced osteoclastic activation and bone loss. These findings highlight that the CDK8-STAT1-RANKL axis in MSCs could play a crucial role in bone resorption and homeostasis.


Asunto(s)
Resorción Ósea , Quinasa 8 Dependiente de Ciclina/metabolismo , Células Madre Mesenquimatosas , Animales , Resorción Ósea/genética , Diferenciación Celular , Quinasa 8 Dependiente de Ciclina/genética , Femenino , Homeostasis , Células Madre Mesenquimatosas/metabolismo , Ratones , FN-kappa B/metabolismo , Osteoclastos , Osteogénesis/genética , Ligando RANK/metabolismo , Ligando RANK/farmacología
14.
Oncogene ; 40(15): 2803-2815, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33727660

RESUMEN

Glioblastoma (GBM) is the most malignant form of glioma. Glioma stem cells (GSCs) contribute to the initiation, progression, and recurrence of GBM as a result of their self-renewal potential and tumorigenicity. Cyclin-dependent kinase 8 (CDK8) belongs to the transcription-related CDK family. Although CDK8 has been shown to be implicated in the malignancy of several types of cancer, its functional role and mechanism in gliomagenesis remain largely unknown. Here, we demonstrate how CDK8 plays an essential role in maintaining stemness and tumorigenicity in GSCs. The genetic inhibition of CDK8 by shRNA or CRISPR interference resulted in an abrogation of the self-renewal potential and tumorigenicity of patient-derived GSCs, which could be significantly rescued by the ectopic expression of c-MYC, a stem cell transcription factor. Moreover, we demonstrated that the pharmacological inhibition of CDK8 significantly attenuated the self-renewal potential and tumorigenicity of GSCs. CDK8 expression was significantly higher in human GBM tissues than in normal brain tissues, and its expression was positively correlated with stem cell markers including c-MYC and SOX2 in human GBM specimens. Additionally, CDK8 expression is associated with poor survival in GBM patients. Collectively, these findings highlight the importance of the CDK8-c-MYC axis in maintaining stemness and tumorigenicity in GSCs; these findings also identify the CDK8-c-MYC axis as a potential target for GSC-directed therapy.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Quinasa 8 Dependiente de Ciclina/metabolismo , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Quinasa 8 Dependiente de Ciclina/genética , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal
15.
Yakugaku Zasshi ; 139(1): 19-25, 2019.
Artículo en Japonés | MEDLINE | ID: mdl-30606923

RESUMEN

Osteoporosis increases the risk of bone fractures (e.g., the femur), reduces a person's activities of daily living (ADL) and increases the likelihood of being bedridden. Therapeutic drugs for osteoporosis include oral bisphosphonates and intravenous receptor activator of nuclear factor-κB ligand (RANKL) antibodies, both of which suppress osteoclast activity, as well as the subcutaneously administered recombinant human parathyroid hormone (PTH), which activates osteoblasts. However, there is currently no oral osteogenesis-promoting drug. In the present study, we found a low-molecular-weight compound, KY-273, with osteogenesis promoting effects. KY-273 induced osteoblast differentiation in ST2 cells and in rat bone marrow-derived mesenchymal stem cells at a dose of 0.1 µM. On the other hand, KY-273 did not clearly exert differentiation effects in osteoclasts, chondrocytes, adipocytes, or myoblasts. In ovariectomized rats, KY-273 clearly increased serum bone alkaline phosphatase (ALP) by at a dose of 3 mg/kg for 8 weeks, and increased both the cortical bone volume and medullary volume of the diaphyseal and epiphyseal regions of femoral bone, but did not affect trabecular bone. Although alendronate (used to decrease bone loss) increased trabecular bone, it did not have any significant effects on cortical bone. PTH increased epiphysis cortical and trabecular bone volume, and reduced medullary volume. KY-273 also displayed good oral absorption in rats. In conclusion, KY-273 is a promising candidate for use as an oral anti-osteoporosis drug with osteogenesis promoting effects.


Asunto(s)
Conservadores de la Densidad Ósea/administración & dosificación , Conservadores de la Densidad Ósea/farmacología , Diferenciación Celular/efectos de los fármacos , Descubrimiento de Drogas , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Osteogénesis/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Administración Oral , Fosfatasa Alcalina/sangre , Animales , Densidad Ósea/efectos de los fármacos , Remodelación Ósea/efectos de los fármacos , Huesos/metabolismo , Células Cultivadas , Quinasa 8 Dependiente de Ciclina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Terapia Molecular Dirigida , Osteoporosis/genética , Ratas , Estimulación Química
16.
Neuropharmacology ; 55(5): 654-60, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18573265

RESUMEN

T-817MA (1-{3-[2-(1-benzothiophen-5-yl)ethoxy]propyl} azetidin-3-ol maleate) is a candidate therapeutic agent for Alzheimer's disease that inhibits oxidative stress and nitric oxide-induced neurotoxicity and acts as a neurotrophic factor. The present study examines the effect of T-817MA on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic neurotoxicity in C57BL/6J mice. MPTP treatment (10mg/kg, s.c.x4 at 2-h intervals) impaired rotarod performance, and T-817MA improved this deficit. MPTP treatment also decreased dopamine levels and tyrosine hydroxylase immunostaining in the substantia nigra (SNc) and striatum. Pretreatment with T-817MA (10 and 30mg/kg as T-817, p.o.) attenuated these decreases in dopamine levels and tyrosine hydroxylase immunoreactivity, but did not affect brain levels of 1-methyl-4-phenylpyridinium ion, an active metabolite of MPTP. The protective effect was almost complete in the SNc, but only partial in the striatum. MPTP increased levels of the lipid peroxidation product, thiobarbituric acid reactive substance, only in the midbrain, which could be blocked by T-817MA. MPTP caused microglial activation both in the SNc and striatum, but T-817MA did not affect the activation of microglia. These results suggest that T-817MA protects against MPTP-induced neurotoxicity by blocking lipid peroxidation in the SNc, and imply that this compound may be useful for treating neurodegenerative disorders related to oxidative stress, such as Parkinson's disease.


Asunto(s)
Intoxicación por MPTP/prevención & control , Maleatos/farmacología , Fármacos Neuroprotectores/farmacología , Tiofenos/farmacología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/metabolismo , Análisis de Varianza , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Peroxidación de Lípido/efectos de los fármacos , Intoxicación por MPTP/inducido químicamente , Intoxicación por MPTP/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Prueba de Desempeño de Rotación con Aceleración Constante
17.
Neurochem Int ; 57(1): 58-66, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20447431

RESUMEN

Nitric oxide (NO) is involved in many pathological conditions including neurodegenerative disorders. We have previously found that sodium nitroprusside (SNP), an NO donor, stimulates mitogen-activated protein kinases (MAPKs) such as extracellular signal-regulating kinase (ERK), c-jun N-terminal protein kinase (JNK) and p38 MAPK, leading to caspase-independent apoptosis in cultured astrocytes. In view of the previous observation that NO stimulates the activity of the Na(+)/Ca(2+) exchanger (NCX), this study examines the involvement of NCX in cytotoxicity. The specific NCX inhibitor SEA0400 blocked SNP-induced phosphorylation of ERK, JNK and p38 MAPK, and decrease in cell viability. SNP-induced phosphorylation of ERK, JNK and p38 MAPK was blocked by removal of external Ca(2+), and SNP treatment caused an increase in (45)Ca(2+) influx. This increase in (45)Ca(2+) influx was blocked by SEA0400, but not the Ca(2+) channel blocker nifedipine. In addition, SNP-induced (45)Ca(2+) influx and cytotoxicity were reduced in NCX1-deficient cells which were transfected with NCX1 siRNA. Inhibitors of intracellular Ca(2+)-dependent proteins such as calpain and calmodulin blocked SNP-induced ERK phosphorylation and decrease in cell viability. Furthermore, the guanylate cyclase inhibitor LY83583 and the cGMP-dependent protein kinase inhibitor KT5823 blocked SNP-induced cytotoxicity. These findings suggest that NCX-mediated Ca(2+) influx triggers SNP-induced apoptosis in astrocytes, which may be mediated by a cGMP-dependent pathway.


Asunto(s)
Astrocitos/metabolismo , Calcio/metabolismo , Calcio/toxicidad , Donantes de Óxido Nítrico/toxicidad , Intercambiador de Sodio-Calcio/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/fisiología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , GMP Cíclico/fisiología , Nitroprusiato/toxicidad , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Intercambiador de Sodio-Calcio/genética
18.
Glia ; 55(13): 1325-33, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17626263

RESUMEN

Nitric oxide induces apoptosis-like cell death in cultured astrocytes, but the exact mechanism is not known. This study further characterized the mechanism of nitric oxide-induced cytotoxicity, and examined the effect of edaravone, a radical scavenger, on cytotoxicity. Treatment of cultured rat astrocytes with sodium nitroprusside (SNP), a nitric oxide donor, for 72 h, decreased cell viability by causing apoptosis-like cell death. The injury was accompanied by increases in the production of reactive oxygen species and in the level of nuclear apoptosis-inducing factor, but not in caspase activity. SNP-induced cytotoxicity was blocked by the c-jun N-terminal protein kinase (JNK) inhibitor SP600125 (20 microM), the p38 mitogen-activated protein (MAP) kinase inhibitor SB203580 (20 microM), and the extracellular signal-regulating kinase (ERK) inhibitor U0126 (10 microM), and the nitric oxide donor stimulated the phosphorylation of p38 MAP kinase, JNK, and ERK. Edaravone (10 microM) protected astrocytes against SNP-induced cell injury and it inhibited SNP-induced phosphorylation of p38 MAP kinase, JNK, and ERK, and the production of reactive oxygen species. Edaravone also attenuated SNP-induced increase in nuclear apoptosis-inducing factor levels. These results suggest that MAP kinase pathways play a key role in nitric oxide-induced apoptosis and that edaravone protects against nitric oxide-induced cytotoxicity by inhibiting nitric oxide-induced MAP kinase activation in astrocytes.


Asunto(s)
Antipirina/análogos & derivados , Apoptosis , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Depuradores de Radicales Libres/farmacología , Óxido Nítrico/farmacología , Animales , Antipirina/farmacología , Factor Inductor de la Apoptosis/antagonistas & inhibidores , Factor Inductor de la Apoptosis/metabolismo , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Edaravona , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
J Immunol ; 176(8): 4675-81, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16585560

RESUMEN

We investigated the cytoprotective effect of NO on H2O2-induced cell death in mouse macrophage-like cell line RAW264. H2O2-treated cells showed apoptotic features, such as activation of caspase-9 and caspase-3, nuclear fragmentation, and DNA fragmentation. These apoptotic features were significantly inhibited by pretreatment for 24 h with NO donors, sodium nitroprusside and 1-hydroxy-2-oxo-3,3-bis-(2-aminoethyl)-1-triazene, at a low nontoxic concentration. The cytoprotective effect of NO was abrogated by the catalase inhibitor 3-amino-1,2,4-triazole but was not affected by a glutathione synthesis inhibitor, L-buthionine-(S,R)-sulfoximine. NO donors increased the level of catalase and its activity in a concentration-dependent manner. Cycloheximide, a protein synthesis inhibitor, inhibited both the NO-induced increase in the catalase level and the cytoprotective effect of NO. These results indicate that NO at a low concentration protects macrophages from H2O2-induced apoptosis by inducing the production of catalase.


Asunto(s)
Apoptosis/efectos de los fármacos , Catalasa/biosíntesis , Peróxido de Hidrógeno/toxicidad , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Animales , Línea Celular , GMP Cíclico/metabolismo , Macrófagos/citología , Ratones , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Compuestos Nitrosos/farmacología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA