Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 289
Filtrar
1.
Bioorg Med Chem Lett ; 105: 129760, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38641151

RESUMEN

The naturally occurring bile acid lithocholic acid (LCA) has been a crucial core structure for many non-sugar-containing sialyltranferase (ST) inhibitors documented in literature. With the aim of elucidating the impact of the terminal carboxyl acid substituent of LCA on its ST inhibition, in this present study, we report the (bio)isosteric replacement-based design and synthesis of sulfonate and sulfate analogues of LCA. Among these compounds, the sulfate analogue SPP-002 was found to selectively inhibit N-glycan sialylation by at least an order of magnitude, indicating a substantial improvement in both potency and selectivity when compared to the unmodified parent bile acid. Molecular docking analysis supported the stronger binding of the synthetic analogue in the enzyme active site. Treatment with SPP-002 also hampered the migration, adhesion, and invasion of MDA-MB-231 cells in vitro by suppressing the expression of signaling proteins involved in the cancer metastasis-associated integrin/FAK/paxillin pathway. In totality, these findings offer not only a novel structural scaffold but also valuable insights for the future development of more potent and selective ST inhibitors with potential therapeutic effects against tumor cancer metastasis.


Asunto(s)
Ácido Litocólico , Simulación del Acoplamiento Molecular , Sialiltransferasas , Ácido Litocólico/farmacología , Ácido Litocólico/química , Ácido Litocólico/síntesis química , Ácido Litocólico/análogos & derivados , Humanos , Sialiltransferasas/antagonistas & inhibidores , Sialiltransferasas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/síntesis química , Relación Estructura-Actividad , Sulfatos/química , Sulfatos/farmacología , Sulfatos/síntesis química , Metástasis de la Neoplasia , Ácidos Sulfónicos/farmacología , Ácidos Sulfónicos/química , Ácidos Sulfónicos/síntesis química , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Estructura Molecular , Adhesión Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Paxillin/metabolismo , Paxillin/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/metabolismo , Descubrimiento de Drogas
2.
Infect Immun ; 92(5): e0008024, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38534100

RESUMEN

Traditional folk treatments for the prevention and management of urinary tract infections (UTIs) and other infectious diseases often include plants and plant extracts that are rich in phenolic compounds. These have been ascribed a variety of activities, including inhibition of bacterial interactions with host cells. Here, we tested a panel of four well-studied phenolic compounds-caffeic acid phenethyl ester (CAPE), resveratrol, catechin, and epigallocatechin gallate-for the effects on host cell adherence and invasion by uropathogenic Escherichia coli (UPEC). These bacteria, which are the leading cause of UTIs, can bind and subsequently invade bladder epithelial cells via an actin-dependent process. Intracellular UPEC reservoirs within the bladder are often protected from antibiotics and host defenses and likely contribute to the development of chronic and recurrent infections. In cell culture-based assays, only resveratrol had a notable negative effect on UPEC adherence to bladder cells. However, both CAPE and resveratrol significantly inhibited UPEC entry into the host cells, coordinate with attenuated phosphorylation of the host actin regulator Focal Adhesion Kinase (FAK or PTK2) and marked increases in the numbers of focal adhesion structures. We further show that the intravesical delivery of resveratrol inhibits UPEC infiltration of the bladder mucosa in a murine UTI model and that resveratrol and CAPE can disrupt the ability of other invasive pathogens to enter host cells. Together, these results highlight the therapeutic potential of molecules like CAPE and resveratrol, which could be used to augment antibiotic treatments by restricting pathogen access to protective intracellular niches.IMPORTANCEUrinary tract infections (UTIs) are exceptionally common and increasingly difficult to treat due to the ongoing rise and spread of antibiotic-resistant pathogens. Furthermore, the primary cause of UTIs, uropathogenic Escherichia coli (UPEC), can avoid antibiotic exposure and many host defenses by invading the epithelial cells that line the bladder surface. Here, we identified two plant-derived phenolic compounds that disrupt activation of the host machinery needed for UPEC entry into bladder cells. One of these compounds, resveratrol, effectively inhibited UPEC invasion of the bladder mucosa in a mouse UTI model, and both phenolic compounds significantly reduced host cell entry by other invasive pathogens. These findings suggest that select phenolic compounds could be used to supplement existing antibacterial therapeutics by denying uropathogens shelter within host cells and tissues and help explain some of the benefits attributed to traditional plant-based medicines.


Asunto(s)
Adhesión Bacteriana , Catequina/análogos & derivados , Infecciones por Escherichia coli , Fenoles , Alcohol Feniletílico/análogos & derivados , Infecciones Urinarias , Escherichia coli Uropatógena , Escherichia coli Uropatógena/efectos de los fármacos , Animales , Ratones , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Infecciones Urinarias/microbiología , Infecciones Urinarias/tratamiento farmacológico , Fenoles/farmacología , Humanos , Adhesión Bacteriana/efectos de los fármacos , Resveratrol/farmacología , Células Epiteliales/microbiología , Células Epiteliales/efectos de los fármacos , Vejiga Urinaria/microbiología , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/patología , Extractos Vegetales/farmacología , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Línea Celular , Catequina/farmacología , Ácidos Cafeicos/farmacología
3.
Mol Cancer Ther ; 23(1): 56-67, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37703580

RESUMEN

Triple-negative breast cancer (TNBC) represents the most lethal and treatment-resistant breast cancer subtype with limited treatment options. We previously identified a protein complex unique to TNBC composed of the gap junction protein connexin 26 (Cx26), the pluripotency transcription factor NANOG, and focal adhesion kinase (FAK). We sought to determine whether a peptide mimetic of the interaction region of Cx26 attenuated tumor growth in preclinical models. We designed peptides based on Cx26 juxtamembrane domains and performed binding experiments with NANOG and FAK using surface plasmon resonance. Binding studies revealed that the Cx26 C-terminal tail and intracellular loop bound to NANOG and FAK with submicromolar-to-micromolar affinity and that a 5-amino acid sequence in the C-terminal tail of Cx26 (RYCSG) was sufficient for binding. Peptides with high affinity were engineered with a cell-penetrating antennapedia sequence and assessed in functional assays including cell proliferation, tumorsphere formation, and in vivo tumor growth, and downstream signaling changes were measured. The cell-penetrating Cx26 peptide (aCx26-pep) disrupted self-renewal while reducing nuclear FAK and NANOG and inhibiting NANOG target gene expression in TNBC cells but not luminal mammary epithelial cells. In vivo, aCx26-pep reduced tumor growth and proliferation and induced cell death. Here, we provide proof-of-concept that a Cx26 peptide-based strategy inhibits growth and alters NANOG activity specifically in TNBC, indicating the therapeutic potential of this targeting approach.


Asunto(s)
Péptidos de Penetración Celular , Conexina 26 , Quinasa 1 de Adhesión Focal , Proteína Homeótica Nanog , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/terapia , Proteína Homeótica Nanog/antagonistas & inhibidores , Humanos , Animales , Ratones , Línea Celular Tumoral , Conexina 26/química , Conexina 26/uso terapéutico , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/uso terapéutico
4.
Chem Biodivers ; 20(4): e202300146, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36919922

RESUMEN

Febrifugine is a kind of quinazolinone compound with high biological activity from a Chinese herb called Chang Shan (Dichroa febrifuga). Febrifugine and its derivatives possess extensive biological activities, some of which exhibited anti-tumor activities as FAK inhibitors. However, they are not very effective at inhibiting tumor metastasis, perhaps because tumors gain energy through compensatory activation of other signaling pathways that promote cell migration and invasion. Therefore, seventeen novel febrifugine derivatives with quinazolinone skeleton were designed, synthesized and acted as potential FAK/PLK1 dual inhibitors. These compounds were determined by 1 H-NMR, 13 C-NMR and MS. Most of the compounds exhibited good inhibitory activity against cancer cell lines by computer-assisted screening, antitumor activity test and FAK/PLK1 inhibitory activity test, wherein compound 3b was screened as a high-efficiency lead compound.


Asunto(s)
Antineoplásicos , Inhibidores de Proteínas Quinasas , Quinazolinonas , Antineoplásicos/química , Línea Celular , Proliferación Celular , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Espectroscopía de Resonancia Magnética , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinonas/química , Quinazolinonas/farmacología , Esqueleto , Relación Estructura-Actividad , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa Tipo Polo 1
5.
Cell Mol Life Sci ; 79(3): 156, 2022 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-35218437

RESUMEN

Signaling through adhesion-related molecules is important for cancer growth and metastasis and cancer cells are resistant to anoikis, a form of cell death ensued by cell detachment from the extracellular matrix. Herein, we report that detached carcinoma cells and immortalized fibroblasts display defects in TNF and CD40 ligand (CD40L)-induced MEK-ERK signaling. Cell detachment results in reduced basal levels of the MEK kinase TPL2, compromises TPL2 activation and sensitizes carcinoma cells to death-inducing receptor ligands, mimicking the synthetic lethal interactions between TPL2 inactivation and TNF or CD40L stimulation. Focal Adhesion Kinase (FAK), which is activated in focal adhesions and mediates anchorage-dependent survival signaling, was found to sustain steady state TPL2 protein levels and to be required for TNF-induced TPL2 signal transduction. We show that when FAK levels are reduced, as seen in certain types of malignancy or malignant cell populations, the formation of cIAP2:RIPK1 complexes increases, leading to reduced TPL2 expression levels by a dual mechanism: first, by the reduction in the levels of NF-κΒ1 which is required for TPL2 stability; second, by the engagement of an RelA NF-κΒ pathway that elevates interleukin-6 production, leading to activation of STAT3 and its transcriptional target SKP2 which functions as a TPL2 E3 ubiquitin ligase. These data underscore a new mode of regulation of TNF family signal transduction on the TPL2-MEK-ERK branch by adhesion-related molecules that may have important ramifications for cancer therapy.


Asunto(s)
Adhesión Celular , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Animales , Ligando de CD40/genética , Ligando de CD40/metabolismo , Ligando de CD40/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteínas de Complejo Poro Nuclear/antagonistas & inhibidores , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
6.
Front Immunol ; 13: 837180, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35178052

RESUMEN

Sepsis and septic shock are associated with high mortality and are considered one of the major public health concerns. The onset of sepsis is known as a hyper-inflammatory state that contributes to organ failure and mortality. Recent findings suggest a potential role of two non-receptor protein tyrosine kinases, namely Focal adhesion kinase (FAK) and Proline-rich tyrosine kinase 2 (Pyk2), in the inflammation associated with endometriosis, cancer, atherosclerosis and asthma. Here we investigate the role of FAK-Pyk2 in the pathogenesis of sepsis and the potential beneficial effects of the pharmacological modulation of this pathway by administering the potent reversible dual inhibitor of FAK and Pyk2, PF562271 (PF271) in a murine model of cecal ligation and puncture (CLP)-induced sepsis. Five-month-old male C57BL/6 mice underwent CLP or Sham surgery and one hour after the surgical procedure, mice were randomly assigned to receive PF271 (25 mg/kg, s.c.) or vehicle. Twenty-four hours after surgery, organs and plasma were collected for analyses. In another group of mice, survival rate was assessed every 12 h over the subsequent 5 days. Experimental sepsis led to a systemic cytokine storm resulting in the formation of excessive amounts of both pro-inflammatory cytokines (TNF-α, IL-1ß, IL-17 and IL-6) and the anti-inflammatory cytokine IL-10. The systemic inflammatory response was accompanied by high plasma levels of ALT, AST (liver injury), creatinine, (renal dysfunction) and lactate, as well as a high, clinical severity score. All parameters were attenuated following PF271 administration. Experimental sepsis induced an overactivation of FAK and Pyk2 in liver and kidney, which was associated to p38 MAPK activation, leading to increased expression/activation of several pro-inflammatory markers, including the NLRP3 inflammasome complex, the adhesion molecules ICAM-1, VCAM-1 and E-selectin and the enzyme NOS-2 and myeloperoxidase. Treatment with PF271 inhibited FAK-Pyk2 activation, thus blunting the inflammatory abnormalities orchestrated by sepsis. Finally, PF271 significantly prolonged the survival of mice subjected to CLP-sepsis. Taken together, our data show for the first time that the FAK-Pyk2 pathway contributes to sepsis-induced inflammation and organ injury/dysfunction and that the pharmacological modulation of this pathway may represents a new strategy for the treatment of sepsis.


Asunto(s)
Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/fisiopatología , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL , Insuficiencia Multiorgánica/tratamiento farmacológico , Insuficiencia Multiorgánica/fisiopatología , Distribución Aleatoria , Sepsis , Tasa de Supervivencia
7.
J Pathol ; 256(2): 235-247, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34743335

RESUMEN

A common limitation of cancer treatments is chemotherapy resistance. We have previously identified that endothelial cell (EC)-specific deletion of focal adhesion kinase (FAK) sensitises tumour cells to DNA-damaging therapies, reducing tumour growth in mice. The present study addressed the kinase activity dependency of EC FAK sensitisation to the DNA-damaging chemotherapeutic drug, doxorubicin. FAK is recognised as a therapeutic target in tumour cells, leading to the development of a range of inhibitors, the majority being ATP competitive kinase inhibitors. We demonstrate that inactivation of EC FAK kinase domain (kinase dead; EC FAK-KD) in established subcutaneous B16F0 tumours improves melanoma cell sensitisation to doxorubicin. Doxorubicin treatment in EC FAK-KD mice reduced the percentage change in exponential B16F0 tumour growth further than in wild-type mice. There was no difference in tumour blood vessel numbers, vessel perfusion or doxorubicin delivery between genotypes, suggesting a possible angiocrine effect on the regulation of tumour growth. Doxorubicin reduced perivascular malignant cell proliferation, while enhancing perivascular tumour cell apoptosis and DNA damage in tumours grown in EC FAK-KD mice 48 h after doxorubicin injection. Human pulmonary microvascular ECs treated with the pharmacological FAK kinase inhibitors defactinib, PF-562,271 or PF-573,228 in combination with doxorubicin also reduced cytokine expression levels. Together, these data suggest that targeting EC FAK kinase activity may alter angiocrine signals that correlate with improved acute tumour cell chemosensitisation. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Células Endoteliales/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , Melanoma Experimental/enzimología , Neovascularización Fisiológica , Neoplasias Cutáneas/enzimología , Inhibidores de la Angiogénesis/farmacología , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Citocinas/metabolismo , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Femenino , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/genética , Humanos , Masculino , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Carga Tumoral
8.
Eur J Med Chem ; 228: 113978, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34810020

RESUMEN

Focal adhesion kinase (FAK) promotes tumor progression by intracellular signal transduction and regulation of gene expression and protein turnover, which is a compelling therapeutic target for various cancer types, including ovarian cancer. However, the clinical responses of FAK inhibitors remain unsatisfactory. Here, we describe the discovery of FAK inhibitors using a scaffold hopping strategy. Structure-activity relationship (SAR) exploration identified 36 as a potent FAK inhibitor, which exhibited inhibitory activities against FAK signaling in vitro. Treatment with 36 not only decreased migration and invasion of PA-1 cells, but also reduced expression of MMP-2 and MMP-9. Moreover, 36 inhibited tumor growth and metastasis, and no obvious adverse effects were observed during the in vivo study. These results revealed the potential of FAK inhibitor 36 for treatment of ovarian cancer.


Asunto(s)
Antineoplásicos/farmacología , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Indanos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Indanos/síntesis química , Indanos/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Pirimidinas/síntesis química , Pirimidinas/química , Pirroles/síntesis química , Pirroles/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
9.
JCI Insight ; 7(3)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34914633

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) is an extremely metastatic and lethal disease. Here, in both murine and human PDA, we demonstrate that extracellular matrix architecture regulates cell extrusion and subsequent invasion from intact ductal structures through tumor-associated collagen signatures (TACS). This results in early dissemination from histologically premalignant lesions and continual invasion from well-differentiated disease, and it suggests TACS as a biomarker to aid in the pathologic assessment of early disease. Furthermore, we show that pancreatitis results in invasion-conducive architectures, thus priming the stroma prior to malignant disease. Analysis in potentially novel microfluidic-derived microtissues and in vivo demonstrates decreased extrusion and invasion following focal adhesion kinase (FAK) inhibition, consistent with decreased metastasis. Thus, data suggest that targeting FAK or strategies to reengineer and normalize tumor microenvironments may have roles not only in very early disease, but also for limiting continued dissemination from unresectable disease. Likewise, it may be beneficial to employ stroma-targeting strategies to resolve precursor diseases such as pancreatitis in order to remove stromal architectures that increase risk for early dissemination.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Experimentales , Neoplasias Pancreáticas/genética , ARN Interferente Pequeño/genética , Microambiente Tumoral/genética , Animales , Apoptosis , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/terapia , Línea Celular Tumoral , Movimiento Celular , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/biosíntesis , Humanos , Ratones , Ratones Transgénicos , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia
10.
J Enzyme Inhib Med Chem ; 37(1): 349-372, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34923887

RESUMEN

In this study, different assortments of 2-arylquinolines and 2,6-diarylquinolines have been developed. Recently, we have developed a new series of 6,7-dimethoxy-4-alkoxy-2-arylquinolines as Topoisomerase I (TOP1) inhibitors with potent anticancer activity. Utilising the SAR outputs from this study, we tried to enhance anticancer and TOP1 inhibitory activities. Though target quinolines demonstrated potent antiproliferative effect, specifically against colorectal cancer DLD-1 and HCT-116, they showed weak TOP1 inhibition which may be attributable to their non-coplanarity. Thereafter, screening against kinase panel revealed their dual inhibitory activity against EGFR and FAK. Quinolines 6f, 6h, 6i, and 20f were the most potent EGFR inhibitors (IC50s = 25.39, 20.15, 22.36, and 24.81 nM, respectively). Meanwhile, quinolines 6f, 6h, 6i, 16d, and 20f exerted the best FAK inhibition (IC50s = 22.68, 14.25, 18.36, 17.36, and 15.36 nM, respectively). Finally, molecular modelling was employed to justify the promising EGFR/FAK inhibition. The study outcomes afforded the first reported quinolines with potent EGFR/FAK dual inhibition.


Asunto(s)
Antineoplásicos/farmacología , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Quinolinas/síntesis química , Quinolinas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
11.
Sci Rep ; 11(1): 23285, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857846

RESUMEN

Machine learning approaches have shown great promise in biology and medicine discovering hidden information to further understand complex biological and pathological processes. In this study, we developed a deep learning-based machine learning algorithm to meaningfully process image data and facilitate studies in vascular biology and pathology. Vascular injury and atherosclerosis are characterized by neointima formation caused by the aberrant accumulation and proliferation of vascular smooth muscle cells (VSMCs) within the vessel wall. Understanding how to control VSMC behaviors would promote the development of therapeutic targets to treat vascular diseases. However, the response to drug treatments among VSMCs with the same diseased vascular condition is often heterogeneous. Here, to identify the heterogeneous responses of drug treatments, we created an in vitro experimental model system using VSMC spheroids and developed a machine learning-based computational method called HETEROID (heterogeneous spheroid). First, we established a VSMC spheroid model that mimics neointima-like formation and the structure of arteries. Then, to identify the morphological subpopulations of drug-treated VSMC spheroids, we used a machine learning framework that combines deep learning-based spheroid segmentation and morphological clustering analysis. Our machine learning approach successfully showed that FAK, Rac, Rho, and Cdc42 inhibitors differentially affect spheroid morphology, suggesting that multiple drug responses of VSMC spheroid formation exist. Overall, our HETEROID pipeline enables detailed quantitative drug characterization of morphological changes in neointima formation, that occurs in vivo, by single-spheroid analysis.


Asunto(s)
Aprendizaje Automático , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Aterosclerosis/patología , Células Cultivadas , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/fisiología , Humanos , Neointima/patología , Esferoides Celulares/fisiología , Lesiones del Sistema Vascular/patología , Proteína de Unión al GTP cdc42/antagonistas & inhibidores , Proteína de Unión al GTP cdc42/fisiología , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Proteínas de Unión al GTP rac/fisiología
12.
Bioorg Med Chem Lett ; 54: 128433, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34757216

RESUMEN

Forty-one new focal adhesion kinase (FAK) covalent inhibitors were designed and synthesized based on FAK inhibitor TAE226. Compound 11w displayed the highest inhibition of FAK with an IC50 value of 35 nM and exhibited potent anticancer activity against Hela, HCT116 and MDA-MB-231 cell lines with IC50 values of 0.41, 0.01 and 0.11 µM respectively, compared to TAE226 (2.68, 0.64 and 4.19 µM respectively). 11w also inhibited the clone formation and migration of HCT-116 cells and stimulated cell cycle arrest in the G2/M phase, inducing tumor cell apoptosis. Compound 11w formed a covalent bond with the Cys427 residue of FAK in a docking model, inhibiting the autophosphorylation of FAK and downstream proteins in a dose-dependent manner. Moreover, 11w showed adequate oral bioavailability of 21.02%. A 74.20% inhibition of tumor growth in the HCT116 xenograft model was also observed. These data indicate that 11w is a promising covalent inhibitor of FAK.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
13.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638565

RESUMEN

Malignant pleural mesothelioma (MPM) is an aggressive malignancy with limited effective treatment options. Focal adhesion kinase (FAK) inhibitors have been shown to efficiently suppress MPM cell growth initially, with limited utility in the current clinical setting. In this study, we utilised a large collection of MPM cell lines and MPM tissue samples to study the role of E-cadherin (CDH1) and microRNA on the efficacy of FAK inhibitors in MPM. The immunohistochemistry (IHC) results showed that the majority of MPM FFPE samples exhibited either the absence of, or very low, E-cadherin protein expression in MPM tissue. We showed that MPM cells with high CDH1 mRNA levels exhibited resistance to the FAK inhibitor PND-1186. In summary, MPM cells that did not express CDH1 mRNA were sensitive to PND-1186, and MPM cells that retained CDH1 mRNA were resistant. A cell cycle analysis showed that PND-1186 induced cell cycle disruption by inducing the G2/M arrest of MPM cells. A protein-protein interaction study showed that EGFR is linked to the FAK pathway, and a target scan of the microRNAs revealed that microRNAs (miR-17, miR221, miR-222, miR137, and miR148) interact with EGFR 3'UTR. Transfection of MPM cells with these microRNAs sensitised the CHD1-expressing FAK-inhibitor-resistant MPM cells to the FAK inhibitor.


Asunto(s)
Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Mesotelioma Maligno/tratamiento farmacológico , Mesotelioma Maligno/genética , MicroARNs/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Aminopiridinas/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Receptores ErbB/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Mapas de Interacción de Proteínas
14.
Nat Commun ; 12(1): 5256, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34489407

RESUMEN

Tissue repair and healing remain among the most complicated processes that occur during postnatal life. Humans and other large organisms heal by forming fibrotic scar tissue with diminished function, while smaller organisms respond with scarless tissue regeneration and functional restoration. Well-established scaling principles reveal that organism size exponentially correlates with peak tissue forces during movement, and evolutionary responses have compensated by strengthening organ-level mechanical properties. How these adaptations may affect tissue injury has not been previously examined in large animals and humans. Here, we show that blocking mechanotransduction signaling through the focal adhesion kinase pathway in large animals significantly accelerates wound healing and enhances regeneration of skin with secondary structures such as hair follicles. In human cells, we demonstrate that mechanical forces shift fibroblasts toward pro-fibrotic phenotypes driven by ERK-YAP activation, leading to myofibroblast differentiation and excessive collagen production. Disruption of mechanical signaling specifically abrogates these responses and instead promotes regenerative fibroblast clusters characterized by AKT-EGR1.


Asunto(s)
Indoles/farmacología , Mecanotransducción Celular/fisiología , Piel/lesiones , Sulfonamidas/farmacología , Cicatrización de Heridas/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Colágeno/metabolismo , Femenino , Fibroblastos , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/metabolismo , Regeneración Tisular Dirigida , Humanos , Indoles/sangre , Mecanotransducción Celular/efectos de los fármacos , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Piel/efectos de los fármacos , Piel/patología , Fenómenos Fisiológicos de la Piel , Estrés Mecánico , Sulfonamidas/sangre , Porcinos , Cicatrización de Heridas/efectos de los fármacos
15.
J Mater Chem B ; 9(37): 7848-7865, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34586154

RESUMEN

Vanadium is an important trace element in bone and is involved in bone metabolism, bone formation, and bone growth, but the roles of various vanadium ions, especially of pentavalent vanadium, in bone tissue regenerative repair have been underestimated and even misinterpreted for a long time. The main purposes of this study are to investigate the release profile of Si, Ca, P, and V ions from vanadium doped mesoporous bioactive glass (V-MBG) particles and to explore the effect of pentavalent vanadium ions on proliferation and osteogenic differentiation of BMSCs as well as the corresponding osteogenic signaling pathway. On the basis of preparations of V-MBG particles with different pentavalent vanadium contents, the ion release behavior from V-MBG in distilled water and simulated body fluid was systemically investigated. Furthermore, the cytocompatibility and osteogenic effect of V-MBG extracts were studied in rBMSCs, and the related molecular mechanisms were preliminarily discussed. The results of dissolution experiments showed that the V ionic concentration exhibited a burst increase and then a sustained slow increase in the two media. The resultant V ions from 1.0V-MBG, 4.0V-MBG and 10.0V-MBG at 21 days were about 1.1, 5.8, and 12.5 mg L-1 in water, respectively, and 1.6, 4.8 and 12.8 mg L-1 in SBF, respectively. The release behaviors of Si, Ca, P, and V ions were evidently affected by high contents of incorporated vanadium. The cellular results indicated that compared to the control and MBG groups, the V(V) ions in V-MBG extracts at about 19.4 µM markedly promoted the proliferation, the gene and protein expression of BMP-2 and COL-I, and the ALP activity of rBMSCs in non-osteoinductive media, but insignificantly stimulated the OCN protein synthesis. More deeply, V(V) ions at about 19.4 µM significantly upregulated the gene and protein expressions of Itga 2b, FAK, and pERK1/2, demonstrating that V(V) ions could regulate osteogenic differentiation of rBMSCs through the activation of the Itga 2b-FAK-MAPK (pERK1/2) signaling pathway. The in vivo results further confirmed that V-MBG induced and promoted new bone formation in the defect area compared to the PGC and PGC/V-M0 groups. These results would contribute to modify the perception about the biocompatibility and osteogenic promotion of pentavalent vanadium at an appropriate concentration.


Asunto(s)
Materiales Biocompatibles/química , Diferenciación Celular , Vidrio/química , Osteogénesis , Transducción de Señal , Vanadio/química , Animales , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/uso terapéutico , Enfermedades Óseas/patología , Enfermedades Óseas/terapia , Enfermedades Óseas/veterinaria , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/metabolismo , Expresión Génica/efectos de los fármacos , Iones/química , Iones/metabolismo , Iones/farmacología , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogénesis/efectos de los fármacos , Polímeros/química , Porosidad , Prótesis e Implantes , Ratas , Transducción de Señal/efectos de los fármacos
16.
Biochem Pharmacol ; 192: 114722, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34384759

RESUMEN

Mast cells (MCs) initiate and maintain allergic inflammation. Upon being stimulated with immunoglobulin (Ig)E and antigen (Ag), MCs exhibit FcεRI (high-affinity IgE) receptor-mediated degranulation, cytokine secretion, and increased focal adhesion kinase (FAK) activity. The aims of this study were to examine mechanisms of FAK regulation in IgE-mediated MC activation and the effects of FAK inhibition on MC-mediated allergic responses. FAK activity was manipulated with short hairpin RNA (shRNA) knockdown, FAK overexpression, and the FAK inhibitor PF-431396 (PF). Gene expression and kinase activation were analyzed with quantitative molecular biology assays. PF effects were tested in the passive cutaneous anaphylaxis (PCA), active systemic anaphylaxis (ASA), and allergic conjunctivitis (AC) mouse models. Our results showed that FAK overexpression increased IgE-mediated degranulation and reduced the dexamethasone inhibitory effect on MCs activation. The FAK inhibitor PF diminished MC release of ß-hexosaminidase (ß-hex), histamine, and inflammatory cytokines, via a mechanism that involves MAPK and NF-κB signaling pathways. CaMKII was identified as a robust FAK-associating protein. Inhibition of CaMKII activation by KN-93 suppressed FAK activity and its downstream pathway. PF attenuated inflammatory responses in our PCA and ASA models, and relieved signs of allergic disease in AC model mice. In conclusions, MC degranulation and production of inflammatory mediators in allergic disease may be consequent to FcεRI crosslinking inducing CaMKII-mediated activation of FAK activity. FAK inhibition may represent a new MC-suppressing treatment strategy for the treatment of allergic diseases.


Asunto(s)
Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/metabolismo , Hipersensibilidad/metabolismo , Inmunoglobulina E/toxicidad , Mastocitos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Quinasa 1 de Adhesión Focal/inmunología , Hipersensibilidad/tratamiento farmacológico , Hipersensibilidad/inmunología , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Inhibidores de Proteínas Quinasas/uso terapéutico
17.
Future Med Chem ; 13(18): 1559-1590, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34340532

RESUMEN

FAK, a nonreceptor tyrosine kinase, has been recognized as a novel target class for the development of targeted anticancer agents. Overexpression of FAK is a common occurrence in several solid tumors, in which the kinase has been implicated in promoting metastases. Consequently, designing and developing potent FAK inhibitors is becoming an attractive goal, and FAK inhibitors are being recognized as a promising tool in our armamentarium for treating diverse cancers. This review comprehensively summarizes the different classes of synthetically derived compounds that have been reported as potent FAK inhibitors in the last three decades. Finally, the future of FAK-targeting smart drugs that are designed to slow down the emergence of drug resistance is discussed.


Asunto(s)
Antineoplásicos/química , Biomarcadores de Tumor/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Secuencia de Aminoácidos , Antineoplásicos/farmacología , Sitios de Unión , Diseño de Fármacos , Compuestos Heterocíclicos/química , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Relación Estructura-Actividad
18.
Angew Chem Int Ed Engl ; 60(43): 23327-23334, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34416073

RESUMEN

Focal adhesion kinase (FAK) is a key mediator of tumour progression and metastasis. To date, clinical trials of FAK inhibitors have reported disappointing efficacy for oncology indications. We report the design and characterisation of GSK215, a potent, selective, FAK-degrading Proteolysis Targeting Chimera (PROTAC) based on a binder for the VHL E3 ligase and the known FAK inhibitor VS-4718. X-ray crystallography revealed the molecular basis of the highly cooperative FAK-GSK215-VHL ternary complex, and GSK215 showed differentiated in-vitro pharmacology compared to VS-4718. In mice, a single dose of GSK215 induced rapid and prolonged FAK degradation, giving a long-lasting effect on FAK levels (≈96 h) and a marked PK/PD disconnect. This tool PROTAC molecule is expected to be useful for the study of FAK-degradation biology in vivo, and our results indicate that FAK degradation may be a differentiated clinical strategy versus FAK inhibition for the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Proteolisis/efectos de los fármacos , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Benzamidas/química , Benzamidas/farmacocinética , Benzamidas/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Dipéptidos/química , Dipéptidos/farmacocinética , Dipéptidos/farmacología , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Ratones , Estructura Molecular , Ubiquitina-Proteína Ligasas/metabolismo
19.
Cancer Res ; 81(21): 5438-5450, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34417202

RESUMEN

Despite the approval of several multikinase inhibitors that target SRC and the overwhelming evidence of the role of SRC in the progression and resistance mechanisms of many solid malignancies, inhibition of its kinase activity has thus far failed to improve patient outcomes. Here we report the small molecule eCF506 locks SRC in its native inactive conformation, thereby inhibiting both enzymatic and scaffolding functions that prevent phosphorylation and complex formation with its partner FAK. This mechanism of action resulted in highly potent and selective pathway inhibition in culture and in vivo. Treatment with eCF506 resulted in increased antitumor efficacy and tolerability in syngeneic murine cancer models, demonstrating significant therapeutic advantages over existing SRC/ABL inhibitors. Therefore, this mode of inhibiting SRC could lead to improved treatment of SRC-associated disorders. SIGNIFICANCE: Small molecule-mediated inhibition of SRC impairing both catalytic and scaffolding functions confers increased anticancer properties and tolerability compared with other SRC/ABL inhibitors.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Animales , Apoptosis , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Conformación Proteica , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/química , Familia-src Quinasas/metabolismo
20.
J Med Chem ; 64(16): 11934-11957, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34324343

RESUMEN

Focal adhesion kinase (FAK) is overexpressed in highly invasive and metastatic cancers. To identify novel FAK inhibitors, we designed and synthesized various thieno[3,2-d]pyrimidine derivatives. An intensive structure-activity relationship (SAR) study led to the identification of 26 as a lead. Moreover, 26, a multitargeted kinase inhibitor, possesses excellent potencies against FLT3 mutants as well as FAK. Gratifyingly, 26 remarkably inhibits recalcitrant FLT3 mutants, including F691L, that cause drug resistance. Importantly, 26 is superior to PF-562271 in terms of apoptosis induction, anchorage-independent growth inhibition, and tumor burden reduction in the MDA-MB-231 xenograft mouse model. Also, 26 causes regression of tumor growth in the MV4-11 xenograft mouse model, indicating that it could be effective against acute myeloid leukemia (AML). Finally, in an orthotopic mouse model using MDA-MB-231, 26 remarkably prevents metastasis of orthotopic tumors to lymph nodes. Taken together, the results indicate that 26 possesses potential therapeutic value against highly invasive cancers and relapsed AML.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Tiofenos/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Simulación del Acoplamiento Molecular , Estructura Molecular , Metástasis de la Neoplasia/prevención & control , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Pirimidinas/farmacología , Relación Estructura-Actividad , Tiofenos/síntesis química , Tiofenos/metabolismo , Tiofenos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...