Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(4)2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35216371

RESUMEN

Chronic cardiac muscle inflammation and subsequent fibrotic tissue deposition are key features in Duchenne Muscular Dystrophy (DMD). The treatment of choice for delaying DMD progression both in skeletal and cardiac muscle are corticosteroids, supporting the notion that chronic inflammation in the heart plays a pivotal role in fibrosis deposition and subsequent cardiac dysfunction. Nevertheless, considering the adverse effects associated with long-term corticosteroid treatments, there is a need for novel anti-inflammatory therapies. In this study, we used our recently described exercised mdx (ex mdx) mouse model characterised by accelerated heart pathology, and the specific PKCθ inhibitor Compound 20 (C20), to show that inhibition of this kinase leads to a significant reduction in the number of immune cells infiltrating the heart, as well as necrosis and fibrosis. Functionally, C20 treatment also prevented the reduction in left ventricle fractional shortening, which was typically observed in the vehicle-treated ex mdx mice. Based on these findings, we propose that PKCθ pharmacological inhibition could be an attractive therapeutic approach to treating dystrophic cardiomyopathy.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Corazón/efectos de los fármacos , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Proteína Quinasa C-theta/antagonistas & inhibidores , Animales , Cardiomiopatías/metabolismo , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Distrofina/metabolismo , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Miocardio/metabolismo , Fenotipo
2.
J Med Chem ; 64(16): 11886-11903, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34355886

RESUMEN

The PKC-θ isoform of protein kinase C is selectively expressed in T lymphocytes and plays an important role in the T cell antigen receptor (TCR)-triggered activation of mature T cells, T cell proliferation, and the subsequent release of cytokines such as interleukin-2 (IL-2). Herein, we report the synthesis and structure-activity relationship (SAR) of a novel series of PKC-θ inhibitors. Through a combination of structure-guided design and exploratory SAR, suitable replacements for the basic C4 amine of the original lead (3) were identified. Property-guided design enabled the identification of appropriately substituted C2 groups to afford potent analogs with metabolic stability and permeability to support in vivo testing. With exquisite general kinase selectivity, cellular inhibition of T cell activation as assessed by IL-2 expression, a favorable safety profile, and demonstrated in vivo efficacy in models of acute and chronic T cell activation with oral dosing, CC-90005 (57) was selected for clinical development.


Asunto(s)
Ciclohexanoles/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico , Proteína Quinasa C-theta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Animales , Células CACO-2 , Proliferación Celular/efectos de los fármacos , Ciclohexanoles/síntesis química , Ciclohexanoles/metabolismo , Humanos , Factores Inmunológicos/síntesis química , Factores Inmunológicos/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-theta/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/metabolismo , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos
3.
J Med Chem ; 63(21): 12978-12991, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33100009

RESUMEN

Protein kinases C (PKCs) are a family of serine/threonine kinases involved in various cellular processes, including proliferation, differentiation, cell survival, and apoptosis. Here, we report the identification, structure-activity relationship (SAR), and 3D-QSAR studies of 69 natural indolocarbazoles, including 15 new compounds, from marine streptomyces strains. Interestingly, we found that the chair conformational isomer of 7-oxo-staurosporine (compound 15) inhibited PKCθ more potently than the corresponding boat isomer. An evaluation of kinase selectivity and antitumor efficacy revealed that 15 was a potent dual PKCθ/δ inhibitor and that it could efficiently inhibit tumor growth in pancreatic cancer (PC) by inducing cellular apoptosis and suppressing the NF-κB/p-P65 pathway. In addition, we demonstrated that overexpression of p-PKCδ and p-P65 was associated with poor survival rates in patients with PC, and p-PKCθ expression also showed significant positive correlations with p-PKCδ and p-P65 levels. Finally, the PC patient-derived xenograft model further confirmed the potential anti-PC efficacy of 15.


Asunto(s)
Carbazoles/química , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-theta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión , Carbazoles/metabolismo , Carbazoles/farmacología , Carbazoles/uso terapéutico , Línea Celular Tumoral , Cristalografía por Rayos X , Humanos , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , FN-kappa B/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-theta/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Agua de Mar/microbiología , Transducción de Señal/efectos de los fármacos , Streptomyces/química , Streptomyces/metabolismo , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Breast Cancer Res ; 22(1): 72, 2020 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-32600444

RESUMEN

BACKGROUND: Protein kinase C theta, (PRKCQ/PKCθ) is a serine/threonine kinase that is highly expressed in a subset of triple-negative breast cancers (TNBC) and promotes their growth, anoikis resistance, epithelial-mesenchymal transition (EMT), and invasion. Here, we show that PRKCQ regulates the sensitivity of TNBC cells to apoptosis triggered by standard-of-care chemotherapy by regulating levels of pro-apoptotic Bim. METHODS: To determine the effects of PRKCQ expression on chemotherapy-induced apoptosis, shRNA and cDNA vectors were used to modulate the PRKCQ expression in MCF-10A breast epithelial cells or triple-negative breast cancer cells (MDA-MB231Luc, HCC1806). A novel PRKCQ small-molecule inhibitor, 17k, was used to inhibit kinase activity. Viability and apoptosis of cells treated with PRKCQ cDNA/shRNA/inhibitor +/-chemotherapy were measured. Expression levels of Bcl2 family members were assessed. RESULTS: Enhanced expression of PRKCQ is sufficient to suppress apoptosis triggered by paclitaxel or doxorubicin treatment. Downregulation of PRKCQ also enhanced the apoptosis of chemotherapy-treated TNBC cells. Regulation of chemotherapy sensitivity by PRKCQ mechanistically occurs via regulation of levels of Bim, a pro-apoptotic Bcl2 family member; suppression of Bim prevents the enhanced apoptosis observed with combined PRKCQ downregulation and chemotherapy treatment. Regulation of Bim and chemotherapy sensitivity is significantly dependent on PRKCQ kinase activity; overexpression of a catalytically inactive PRKCQ does not suppress Bim or chemotherapy-associated apoptosis. Furthermore, PRKCQ kinase inhibitor treatment suppressed growth, increased anoikis and Bim expression, and enhanced apoptosis of chemotherapy-treated TNBC cells, phenocopying the effects of PRKCQ downregulation. CONCLUSIONS: These studies support PRKCQ inhibition as an attractive therapeutic strategy and complement to chemotherapy to inhibit the growth and survival of TNBC cells.


Asunto(s)
Proteína 11 Similar a Bcl2/metabolismo , Doxorrubicina/farmacología , Paclitaxel/farmacología , Proteína Quinasa C-theta/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Invasividad Neoplásica , Proteína Quinasa C-theta/genética , Proteína Quinasa C-theta/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Int Immunopharmacol ; 80: 106133, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31931364

RESUMEN

BACKGROUND: T help 2 (Th2) cell differentiation by morphine has been verified. However, the underlying mechanism of morphine induces Th2 cell differentiation remains elusive. The aim of the present study was to explore the possible basis of morphine induced Th2 cell differentiation. METHODS: Flow cytometry analysis was used to detect the content of T help 1(Th1) cell and Th2 cell. Enzyme linked immunosorbent assay (ELISA) was performed to determine the levels of IL-4 and IFN-γ. Real-time quantitative polymerase chain reaction, electrophoretic mobility shift assay and Western blotting was conducted in this study. RESULTS: Th2 cell subset and IL-4 level were elevated in morphine induced naïve T cells. Pathway determining found the protein phosphorylation level of PKC-θ and the expression and activity of the transcription factor GATA3 was also enhanced in the naïve T cells challenged by morphine. Moreover, inhibitor of morphine(naltrexone) or PKC-θ(AEB071) can reverse morphine-induced Th2 cell differentiation. CONCLUSION: These results suggested that morphine induce naïve T cell differentiation to Th2 cells via the PKC-θ/GATA3 signal pathway.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Factor de Transcripción GATA3/genética , Interferón gamma/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Masculino , Ratones Endogámicos C57BL , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Proteína Quinasa C-theta/antagonistas & inhibidores , Pirroles/farmacología , Quinazolinas/farmacología , Linfocitos T Colaboradores-Inductores/citología
6.
Inflammation ; 42(6): 1980-1989, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31297750

RESUMEN

Acute lung injury (ALI)/acute respiratory distress syndrome is characterized by increased pulmonary inflammation, where T helper 17 (Th17) cells play an important regulatory role. Notch signaling critically regulates Th17 differentiation and is known to be linked with proximal T cell by protein kinase C theta (PKCθ). We hypothesized that PKCθ inhibition could attenuate ALI by suppressing Th17 response via the Notch signaling pathway. Male C57BL/6 mice were treated with phosphate-buffered saline (PBS), lipopolysaccharide (LPS), LPS and N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT, a Notch signaling inhibitor), or LPS and PKCθ inhibitor (PI), and the bronchoalveolar lavage fluid (BALF), blood, and lung tissues were harvested at 48 h after the LPS challenge. CD4+ T cells were treated with DAPT or PI and harvested after 72 h. PKCθ inhibition markedly attenuated pathological changes and decreased the wet to dry weight ratio of the mouse lungs. The total cell and neutrophil counts, tumor necrosis factor-α (TNF- α) in BALF, myeloperoxidase activity in lung tissue, and the leukocyte count in whole blood were markedly reduced by PKCθ inhibition. The concentration of interleukin (IL)-17 and IL-22 in BALF, and the percentage of CD4+IL-17A+ T cells in the lungs were significantly downregulated by PKCθ inhibition. A similar trend was observed for the expression of retinoic acid-related orphan receptor gamma t and IL-23 receptor after PKCθ inhibition accompanied with inactivation of the Notch signaling pathway in vivo and in vitro. Collectively, these data demonstrated that PKCθ inhibition protects against LPS-induced ALI by suppressing the differentiation and pathogenicity of Th17, at least partially, through a Notch-dependent mechanism.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Proteína Quinasa C-theta/antagonistas & inhibidores , Receptores Notch/metabolismo , Transducción de Señal , Células Th17/efectos de los fármacos , Lesión Pulmonar Aguda/inducido químicamente , Animales , Diferenciación Celular , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Células Th17/inmunología , Células Th17/patología
7.
Bioorg Chem ; 82: 33-40, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30268972

RESUMEN

Six new (1-6) and nine known (7-15) staurosporine derivatives were isolated from the rice solid fermentation of the marine-derived Streptomyces sp. NB-A13. The structures of the new staurosporine derivatives were established by extensive spectroscopic data interpretation. The absolute configurations of 1 and 2 were assigned by quantum chemical calculations of the electronic circular dichroism (ECD) spectra. All of these compounds were screened for their cytotoxic activities against PC-3 and SW-620 cell lines. Compound 7 exhibited stronger inhibitory activity against SW-620 cell lines than the positive control staurosporine (25.10 nM), with IC50 values of 9.99 nM. Moreover, compounds 1-5, 8-13 and 15 also showed significant cytotoxicities with IC50 values ranging from 0.02 to 16.60 µM, while 6 exhibited no cytotoxic potency. Additionally, compounds 1-7 were also tested for enzyme inhibition activities of Protein kinase C theta (PKC-θ), and showed activity with IC50 values ranging from 0.06 to 9.43 µM except for compound 6, which has no inhibition activity.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Streptomyces/química , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Proteína Quinasa C-theta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/aislamiento & purificación , Estaurosporina/aislamiento & purificación , Estereoisomerismo , Relación Estructura-Actividad
8.
Bioorg Med Chem ; 26(20): 5499-5509, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30274941

RESUMEN

Protein kinase C theta (PKCθ) plays a critical role in T cell signaling and is an attractive target for the treatment of T cell-mediated diseases such as transplant rejection and autoimmune disease. To identify PKCθ inhibitors, we focused on the 2,6-diamino-3-carbamoyl-5-cyanopyrazine derivative 2, which exhibited moderate PKCθ inhibitory activity. Optimization of 2 identified the 2,4-diamino-5-cyanopyrimidine derivative 16c, which exhibited potent PKCθ inhibitory activity and showed good selectivity against other PKC isozymes. Compound 16c prolonged graft survival in an in vivo rat heterotopic cardiac transplant model.


Asunto(s)
Rechazo de Injerto/prevención & control , Trasplante de Corazón/métodos , Proteína Quinasa C-theta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/química , Pirimidinas/uso terapéutico , Animales , Femenino , Rechazo de Injerto/metabolismo , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Corazón/efectos adversos , Humanos , Simulación del Acoplamiento Molecular , Proteína Quinasa C-theta/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Ratas Sprague-Dawley , Trasplante Homólogo/efectos adversos , Trasplante Homólogo/métodos
9.
Bioorg Chem ; 78: 46-57, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29533214

RESUMEN

Recent developments in the literature have demonstrated that curcumin exhibit antioxidant properties supporting its anti-inflammatory, chemopreventive and antitumoral activities against aggressive and recurrent cancers. Despite the valuable findings of curcumin against different cancer cells, the clinical use of curcumin in cancer treatment is limited due to its extremely low aqueous solubility and instability, which lead to poor in vivo bioavailability and limited therapeutic effects. We therefore focused in the present study to evaluate the anti-tumor potential of curcumin analogues on the human breast carcinoma cell lines MDA-MB-231 and MCF-7, as well as their effects on non-tumorigenic normal breast epithelial cells (MCF-10). The IC50 values of curcumin analogue J1 in these cancer cell lines were determined to be 5 ng/ml and 10 ng/ml, in MDA-MB-231 and MCF-7 cells respectively. Interestingly, at these concentrations, the J1 did not affect the viability of non-tumorigenic normal breast epithelial cells MCF-10. Furthermore, we found that J1 strongly induced growth arrest of these cancer cells by modulating the mitochondrial membrane potentials without significant effect on normal MCF-10 cells using JC-1 staining and flow cytometry analysis. Using annexin-V/PI double staining assay followed by flow cytometry analysis, we found that J1 robustly enhanced the induction of apoptosis by increasing the activity of caspases in MDA-MB-231 and MCF-7 cancer cells. In addition, treatment of breast cancer cells with J1 revealed that, in contrast to the expression of cyclin B1, this curcumin analogue vigorously decreased the expression of cyclin A, CDK2 and cyclin E and subsequently sensitized tumor cells to cell cycle arrest. Most importantly, the phosphorylation of AKT, mTOR and PKC-theta in J1-treated cancer cells was markedly decreased and hence affecting the survival of these cancer cells. Most interestingly, J1-treated cancer cells exhibited a significant inhibition in the activation of RhoA followed by reduction in actin polymerization and cytoskeletal rearrangement in response to CXCL12. Our data reveal the therapeutic potential of the curcumin analogue J1 and the underlying mechanisms to fight breast cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Curcumina/análogos & derivados , Curcumina/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteína Quinasa C-theta/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Estructura Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C-theta/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas
10.
J Biomol Struct Dyn ; 36(15): 4029-4044, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29182053

RESUMEN

Protein kinases are ubiquitously expressed as Serine/Threonine kinases, and play a crucial role in cellular activities. Protein kinases have evolved through stringent regulation mechanisms. Protein kinases are also involved in tauopathy, thus are important targets for developing Anti-Alzheimer's disease compounds. Structures with an indole scaffold turned out to be potent new leads. With the aim of developing new inhibitors for human protein kinase C, here we report the generation of four point 3D geometric featured pharmacophore model. In order to identify novel and potent PKCθ inhibitors, the pharmacophore model was screened against 80,000,00 compounds from various chemical databases such as., ZINC, SPEC, ASINEX, which resulted in 127 compound hits, and were taken for molecular docking filters (HTVS, XP docking). After in-depth analysis of binding patterns, induced fit docking (flexible) was employed for six compounds along with the cocrystallized inhibitor. Molecular docking study reveals that compound 6F found to be tight binder at the active site of PKCθ as compared to the cocrystal and has occupancy of 90 percentile. MM-GBSA also confirmed the potency of the compound 6F as better than cocrystal. Molecular dynamics results suggest that compound 6F showed good binding stability of active sites residues similar to cocrystal 7G compound. Present study corroborates the pharmacophore-based virtual screening, and finds the compound 6F as a potent Inhibitor of PKC, having therapeutic potential for Alzheimer's disease. Worldwide, 46.8 million people are believed to be living with Alzheimer's disease. When elderly population increases rapidly and neurodegenerative burden also increases in parallel, we project the findings from this study will be useful for drug developing efforts targeting Alzheimer's disease.


Asunto(s)
Simulación de Dinámica Molecular , Nootrópicos/química , Proteína Quinasa C-theta/química , Inhibidores de Proteínas Quinasas/química , Pirroles/química , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/fisiopatología , Dominio Catalítico , Bases de Datos de Compuestos Químicos , Diseño de Fármacos , Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Humanos , Enlace de Hidrógeno , Cinética , Ligandos , Simulación del Acoplamiento Molecular , Nootrópicos/farmacología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteína Quinasa C-theta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/farmacología , Relación Estructura-Actividad Cuantitativa , Termodinámica
11.
J Pathol ; 244(3): 323-333, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29214629

RESUMEN

Chronic muscle inflammation is a critical feature of Duchenne muscular dystrophy and contributes to muscle fibre injury and disease progression. Although previous studies have implicated T cells in the development of muscle fibrosis, little is known about their role during the early stages of muscular dystrophy. Here, we show that T cells are among the first cells to infiltrate mdx mouse dystrophic muscle, prior to the onset of necrosis, suggesting an important role in early disease pathogenesis. Based on our comprehensive analysis of the kinetics of the immune response, we further identify the early pre-necrotic stage of muscular dystrophy as the relevant time frame for T-cell-based interventions. We focused on protein kinase C θ (PKCθ, encoded by Prkcq), a critical regulator of effector T-cell activation, as a potential target to inhibit T-cell activity in dystrophic muscle. Lack of PKCθ not only reduced the frequency and number of infiltrating T cells but also led to quantitative and qualitative changes in the innate immune cell infiltrate in mdx/Prkcq-/- muscle. These changes were due to the inhibition of T cells, since PKCθ was necessary for T-cell but not for myeloid cell infiltration of acutely injured muscle. Targeting T cells with a PKCθ inhibitor early in the disease process markedly diminished the size of the inflammatory cell infiltrate and resulted in reduced muscle damage. Moreover, diaphragm necrosis and fibrosis were also reduced following treatment. Overall, our findings identify the early T-cell infiltrate as a therapeutic target and highlight the potential of PKCθ inhibition as a therapeutic approach to muscular dystrophy. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Diafragma/efectos de los fármacos , Distrofia Muscular Animal/prevención & control , Proteína Quinasa C-theta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T/efectos de los fármacos , Animales , Diafragma/enzimología , Diafragma/inmunología , Diafragma/patología , Modelos Animales de Enfermedad , Fibrosis , Inmunidad Innata/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular Animal/enzimología , Distrofia Muscular Animal/inmunología , Distrofia Muscular Animal/patología , Necrosis , Proteína Quinasa C-theta/deficiencia , Proteína Quinasa C-theta/genética , Proteína Quinasa C-theta/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Linfocitos T/enzimología , Linfocitos T/inmunología , Factores de Tiempo
12.
Future Med Chem ; 9(11): 1141-1159, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28722471

RESUMEN

AIM: Ligand-based pharmacophore modeling requires long list of inhibitors, while pharmacophores based on single ligand-receptor crystallographic structure can be too restricted or promiscuous. METHODOLOGY: This prompted us to combine simulated annealing molecular dynamics (SAMD) with ligand-receptor contacts analysis as means to construct pharmacophore model(s) from single ligand-receptor complex. Ligand-receptor contacts that survive numerous heating-cooling SAMD cycles are considered critical and are used to guide pharmacophore development. RESULTS: This methodology was implemented to develop pharmacophores for acetylcholinesterase and protein kinase C-θ. The resulting models were validated by receiver-operating characteristic analysis and in vitro bioassay. Assay identified four new protein kinase C-θ inhibitors among captured hits, two of which exhibited nanomolar potencies. CONCLUSION: The results illustrate the ability of the new method to extract valid pharmacophores from single ligand-protein complex.


Asunto(s)
Acetilcolinesterasa/química , Inhibidores de la Colinesterasa/química , Simulación de Dinámica Molecular , Proteína Quinasa C-theta/antagonistas & inhibidores , Proteína Quinasa C-theta/química , Descubrimiento de Drogas , Activación Enzimática , Humanos , Ligandos , Modelos Moleculares , Estructura Molecular , Conformación Proteica , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA