Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Pept Sci ; 28(7): e3389, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34937123

RESUMEN

Aberrant activation of the Wnt signaling pathway has been identified in numerous types of cancer. One common feature of oncogenic Wnt regulation involves an increase in the cellular levels of ß-catenin due to interference with its constitutive ubiquitin-dependent degradation. Targeting ß-catenin has therefore emerged as an appealing approach for the treatment of Wnt-dependent cancers. Here, we report a strategy that employs multifunctional stapled peptides to recruit an E3 ubiquitin ligase to ß-catenin, thereby rescuing ß-catenin degradation by hijacking the endogenous ubiquitin-proteasome pathway. Specifically, we designed, synthesized, and evaluated a panel of multifunctional stapled peptides that have a ß-catenin binding moiety (StAx-35) covalently linked to a second stapled peptide moiety (SAH-p53-8), which is capable to interact with the E3 ubiquitin ligase MDM2. We found that in vitro these multifunctional peptides can recruit the MDM2 protein to ß-catenin and induce poly-ubiquitination on ß-catenin. In cellulo, treatment of the human colorectal cancer cell line SW480 with the multifunctional stapled peptides showed dose-dependent degradation of endogenous ß-catenin levels. In addition, a luciferase reporter assay showed that the multifunctional stapled peptides can suppress ß-catenin-mediated gene expression via the Wnt signaling pathway. Therefore, these multifunctional stapled peptides provide a unique research tool for examining the Wnt signaling pathway by targeted knockdown of ß-catenin at the protein level, and may serve as leads for potential drug candidates in the treatment of Wnt-dependent cancers.


Asunto(s)
Vía de Señalización Wnt , beta Catenina , Humanos , Péptidos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Ubiquitinas/genética , Ubiquitinas/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
2.
Invest New Drugs ; 39(1): 131-141, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32915418

RESUMEN

Today, pancreatic cancer (PC) is a major health problem in the United States. It remains a challenge to develop efficacious clinically useful PC therapies. New avenues, based on translational approaches and innovative validated biomarkers could be a preclinical option to evaluate PC drug candidates or drug combinations before clinical trials. Herein, we describe evaluation of combination therapies by incorporating a novel pathway modulator, p53-Activator Wnt Inhibitor-2 (PAWI-2) with other FDA-approved cancer drugs that have been used in PC clinical trials. PAWI-2 is a potent inhibitor of drug-resistant PC cells that has been shown to selectively ameliorate human pancreatic cancer stem cells (i.e., hPCSCs, FGß3 cells). In the present study, we showed PAWI-2 produced therapeutic synergism with certain types of anti-cancer drugs. These drugs themselves oftentimes do not ameliorate PC cells (especially PCSCs) due to high levels of drug-resistance. PAWI-2 has the ability to rescue the potency of drugs (i.e., erlotinib, trametinib) and inhibit PC cell growth. Key molecular regulators of PAWI-2 could be used to predict synergistic/antagonistic effects between PAWI-2 and other anti-cancer drugs. Anti-cancer results showed potency could be quite accurately correlated to phosphorylation of optineurin (OPTN) in PC cells. Synergism/antagonism was also associated with inhibition of PCSC marker SOX2 that was observed in FGß3 cells. Synergism broadens the potential use of PAWI-2 as an adjunct chemotherapy in patients with PC that have developed resistance to first-line targeted therapies or chemotherapies.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Pancreáticas/patología , Quinoxalinas/farmacología , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Proteínas de Transporte de Membrana/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Quinoxalinas/administración & dosificación , Factores de Transcripción SOXB1/efectos de los fármacos
3.
J Pharmacol Exp Ther ; 371(3): 703-712, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31582422

RESUMEN

Prostate cancer (PCa) is the second leading cause of cancer-related death for men in the United States. Approximately 35% of PCa recurs and is often transformed to castration-resistant prostate cancer (CRPCa), the most deadly and aggressive form of PCa. However, the CRPCa standard-of-care treatment (enzalutamide with abiraterone) usually has limited efficacy. Herein, we report a novel molecule (PAWI-2) that inhibits cellular proliferation of androgen-sensitive and androgen-insensitive cells (LNCaP and PC-3, respectively). In vivo studies in a PC-3 xenograft model showed that PAWI-2 (20 mg/kg per day i.p., 21 days) inhibited tumor growth by 49% compared with vehicle-treated mice. PAWI-2 synergized currently clinically used enzalutamide in in vitro inhibition of PCa cell viability and resensitized inhibition of in vivo PC-3 tumor growth. Compared with vehicle-treated mice, PC-3 xenograft studies also showed that PAWI-2 (20 mg/kg per day i.p., 21 days) and enzalutamide (5 mg/kg per day i.p., 21 days) inhibited tumor growth by 63%. Synergism was mainly controlled by the imbalance of prosurvival factors (e.g., Bcl-2, Bcl-xL, Mcl-1) and antisurvival factors (e.g., Bax, Bak) induced by affecting mitochondrial membrane potential/mitochondria dynamics. Thus, PAWI-2 utilizes a distinct mechanism of action to inhibit PCa growth independently of androgen receptor signaling and overcomes enzalutamide-resistant CRPCa. SIGNIFICANCE STATEMENT: Castration-resistant prostate cancer (CRPCa) is the most aggressive human prostate cancer (PCa) but standard chemotherapies for CRPCa are largely ineffective. PAWI-2 potently inhibits PCa proliferation in vitro and in vivo regardless of androgen receptor status and uses a distinct mechanism of action. PAWI-2 has greater utility in treating CRPCa than standard-of-care therapy. PAWI-2 possesses promising therapeutic potency in low-dose combination therapy with a clinically used drug (e.g., enzalutamide). This study describes a new approach to address the overarching challenge in clinical treatment of CRPCa.


Asunto(s)
Antineoplásicos/farmacología , Feniltiohidantoína/análogos & derivados , Piperazinas/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Quinoxalinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Aspartato Aminotransferasas/sangre , Benzamidas , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Masculino , Ratones , Nitrilos , Feniltiohidantoína/farmacología , Neoplasias de la Próstata/patología , Proteína p53 Supresora de Tumor/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Bioorg Med Chem Lett ; 28(20): 3363-3367, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30201292

RESUMEN

The Notch signaling pathway is involved in cell proliferation and differentiation, and has been recognized as an active pathway in regenerating tissue and cancerous cells. Notch signaling inhibition is considered a viable approach to the treatment of a variety of conditions including colorectal cancer, pancreatic cancer, breast cancer and metastatic melanoma. The discovery that the b-annulated dihydropyridine FLI-06 (1) is an inhibitor of the Notch pathway with an EC50 ≈ 2.5 µM prompted us to screen a library of related analogs. After structure activity studies were conducted, racemic compound 7 was identified with an EC50 = 0.36 µM. Synthesis of individual enantiomers provided (+)-7 enantiomer with an EC50 = 0.13 µM, or about 20-fold the potency of 1.


Asunto(s)
Antineoplásicos/farmacología , Dihidropiridinas/farmacología , Receptor Notch1/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Dihidropiridinas/síntesis química , Dihidropiridinas/química , Células HCT116 , Humanos , Estructura Molecular , Quinolinas/síntesis química , Quinolinas/química , Quinolinas/farmacología , Estereoisomerismo
5.
Bioorg Med Chem ; 26(15): 4441-4451, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30075999

RESUMEN

For adult women in the United States, breast cancer is the most prevalent form of cancer. Compounds that target dysregulated signal transduction can be efficacious anti-cancer therapies. A prominent signaling pathway frequently dysregulated in breast cancer cells is the Wingless-related integration site (Wnt) pathway. The purpose of the work was to optimize a "hit" from a screening campaign. 76,000 compounds were tested in a Wnt transcription assay and revealed potent and reproducible "hit," compound 1. Medicinal chemistry optimization of 1 led to more potent and drug-like molecules, 19, 24 and 25 (i.e., Wnt pathway IC50 values = 11, 18 and 7 nM, respectively). The principal results showed compounds 19, 24 and 25 were potent anti-proliferative agents in breast cancer cell lines, MCF-7 (i.e., IC50 values = 10, 7 and 4 nM, respectively) and MDA-MB 231 (i.e., IC50 values = 13, 13 and 16 nM, respectively). Compound 19 synergized anti-proliferation with chemotherapeutic Doxorubicin in vitro. A major conclusion was that compound 19 enhanced anti-proliferation of Doxorubicin in vitro and in a xenograft animal model of breast cancer.


Asunto(s)
Antineoplásicos/química , Sulfonamidas/química , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Doxorrubicina/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones Desnudos , Relación Estructura-Actividad , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Transcripción Genética/efectos de los fármacos , Trasplante Heterólogo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
6.
J Biol Chem ; 288(21): 14863-73, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23576432

RESUMEN

Peripheral membrane proteins can be targeted to specific organelles or the plasma membrane by differential recognition of phospholipid headgroups. Although molecular determinants of specificity for several headgroups, including phosphatidylserine and phosphoinositides are well defined, specific recognition of the headgroup of the zwitterionic phosphatidylcholine (PC) is less well understood. In cytosolic proteins the cation-π box provides a suitable receptor for choline recognition and binding through the trimethylammonium moiety. In PC, this moiety might provide a sufficient handle to bind to peripheral proteins via a cation-π cage, where the π systems of two or more aromatic residues are within 4-5 Å of the quaternary amine. We prove this hypothesis by engineering the cation-π box into secreted phosphatidylinositol-specific phospholipase C from Staphylococcus aureus, which lacks specific PC recognition. The N254Y/H258Y variant selectively binds PC-enriched vesicles, and x-ray crystallography reveals N254Y/H258Y binds choline and dibutyroylphosphatidylcholine within the cation-π motif. Such simple PC recognition motifs could be engineered into a wide variety of secondary structures providing a generally applicable method for specific recognition of PC.


Asunto(s)
Proteínas Bacterianas/química , Membrana Celular/química , Fosfatidilcolinas/química , Receptores de Superficie Celular/química , Staphylococcus aureus/química , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cationes/química , Cationes/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Mutación Missense , Fosfatidilcolinas/genética , Fosfatidilcolinas/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
7.
Chem Commun (Camb) ; 60(17): 2369-2372, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38318781

RESUMEN

Conventional methods for nitrile synthesis bring inherent environmental risks due to their reliance on oxidants and harsh reaction conditions. Meanwhile, direct electrooxidation of amines to nitriles suffers from low current density. In this study, we propose an innovative indirect electrooxidation strategy for nitrile formation, mediated by Br-/Br2, utilizing a highly efficient CoS2/CoS@Graphite Felt (GF) electrode. Notably, the anodic nitrile generation can be synergistically coupled with the cathodic hydrogen evolution reaction (HER). Through meticulous optimization of reaction parameters, we achieve an impressive 98% selectivity for octanenitrile at a current density of 60 mA cm-2 with a remarkable faradaic efficiency (FE) of 87%. Furthermore, our approach demonstrates excellent versatility, as we successfully evaluate both aliphatic and aromatic primary amines, highlighting its promising potential for practical applications in the field.

8.
Biophys J ; 104(1): 185-95, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23332071

RESUMEN

The enzymatic activity of secreted phosphatidylinositol-specific phospholipase C (PI-PLC) enzymes is associated with bacterial virulence. Although the PI-PLC active site has no obvious lid, molecular-dynamics simulations suggest that correlated loop motions may limit access to the active site, and two Pro residues, Pro(245) and Pro(254), are associated with these correlated motions. Whereas the region containing both Pro residues is quite variable among PI-PLCs, it shows high conservation in virulence-associated, secreted PI-PLCs that bind to the surface of cells. These regions of the protein are also associated with phosphatidylcholine binding, which enhances PI-PLC activity. In silico mutagenesis of Pro(245) disrupts correlated motions between the two halves of Bacillus thuringiensis PI-PLC, and Pro(245) variants show significantly reduced enzymatic activity in all assay systems. PC still enhanced activity, but not to the level of wild-type enzyme. Mutagenesis of Pro(254) appears to stiffen the PI-PLC structure, but experimental mutations had minor effects on activity and membrane binding. With the exception of P245Y, reduced activity was not associated with reduced membrane affinity. This combination of simulations and experiments suggests that correlated motions between the two halves of PI-PLC may be more important for enzymatic activity than for vesicle binding.


Asunto(s)
Bacillus thuringiensis/citología , Bacillus thuringiensis/enzimología , Membrana Celular/enzimología , Fosfoinositido Fosfolipasa C/metabolismo , Secuencia de Aminoácidos , Bacillus cereus/enzimología , Biocatálisis , Secuencia Conservada , Micelas , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Movimiento (Física) , Proteínas Mutantes/metabolismo , Fosfoinositido Fosfolipasa C/química , Hidrolasas Diéster Fosfóricas/metabolismo , Fosfotransferasas/metabolismo , Prolina/química , Unión Proteica , Estructura Secundaria de Proteína , Liposomas Unilamelares/química
9.
J Biol Chem ; 287(48): 40317-27, 2012 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-23038258

RESUMEN

BACKGROUND: Bacterial phosphatidylinositol-specific phospholipase C targets PI and glycosylphosphatidylinositol-linked proteins of eukaryotic cells. RESULTS: Functional relevance of a homodimeric S. aureus PI-PLC crystal structure is supported by enzyme kinetics and mutagenesis. Nonsubstrate phosphatidylcholine increases activity by facilitating enzyme dimerization. CONCLUSION: Activating transient dimerization is antagonized by anions binding to a discrete site. SIGNIFICANCE: Interplay of protein oligomerization and anion binding controls enzyme activity. Staphylococcus aureus phosphatidylinositol-specific phospholipase C (PI-PLC) is a secreted virulence factor for this pathogenic bacterium. A novel crystal structure shows that this PI-PLC can form a dimer via helix B, a structural feature present in all secreted, bacterial PI-PLCs that is important for membrane binding. Despite the small size of this interface, it is critical for optimal enzyme activity. Kinetic evidence, increased enzyme specific activity with increasing enzyme concentration, supports a mechanism where the PI-PLC dimerization is enhanced in membranes containing phosphatidylcholine (PC). Mutagenesis of key residues confirm that the zwitterionic phospholipid acts not by specific binding to the protein, but rather by reducing anionic lipid interactions with a cationic pocket on the surface of the S. aureus enzyme that stabilizes monomeric protein. Despite its structural and sequence similarity to PI-PLCs from other Gram-positive pathogenic bacteria, S. aureus PI-PLC appears to have a unique mechanism where enzyme activity is modulated by competition between binding of soluble anions or anionic lipids to the cationic sensor and transient dimerization on the membrane.


Asunto(s)
Aniones/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Fosfoinositido Fosfolipasa C/química , Fosfoinositido Fosfolipasa C/metabolismo , Staphylococcus aureus/enzimología , Proteínas Bacterianas/genética , Sitios de Unión , Dimerización , Cinética , Fosfoinositido Fosfolipasa C/genética , Unión Proteica , Staphylococcus aureus/química , Staphylococcus aureus/genética , Especificidad por Sustrato
10.
J Med Chem ; 66(1): 95-106, 2023 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-36580278

RESUMEN

Therapeutic peptides have revolutionized treatment for a number of human diseases. In particular, the past two decades have witnessed rapid progress of stapled helical peptides in drug discovery. Stapled helical peptides are chemically modified and constrained in their bioactive α-helical conformation. Compared to unstabilized linear peptides, stapled helical peptides exhibit superior binding affinity and selectivity, enhanced membrane permeability, and improved metabolic stability, presenting exciting promise for targeting otherwise challenging protein-protein interfaces. In this Perspective, we summarize recent applications of high-throughput screening technologies for identification of potent stapled helical peptides with optimized binding properties. We expect to provide a broad reference to accelerate the development of stapled helical peptides as the next generation of therapeutic peptides for various human diseases.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Péptidos , Humanos , Estructura Secundaria de Proteína , Péptidos/farmacología , Péptidos/química , Descubrimiento de Drogas , Conformación Proteica en Hélice alfa
11.
RSC Med Chem ; 14(12): 2496-2508, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38107173

RESUMEN

Protein-protein interactions (PPIs) control many essential biological pathways which are often misregulated in disease. As such, selective PPI modulators are desirable to unravel complex functions of PPIs and thus expand the repertoire of therapeutic targets. However, the large size and relative flatness of PPI interfaces make them challenging molecular targets for conventional drug modalities, rendering most PPIs "undruggable". Therefore, there is a growing need to discover innovative molecules that are able to modulate crucial PPIs. Peptides are ideal candidates to deliver such therapeutics attributed to their ability to closely mimic structural features of protein interfaces. However, their inherently poor proteolysis resistance and cell permeability inevitably hamper their biomedical applications. The introduction of a constraint (i.e., peptide cyclization) to stabilize peptides' secondary structure is a promising strategy to address this problem as witnessed by the rapid development of cyclic peptide drugs in the past two decades. Here, we comprehensively review the recent progress on stabilized cyclic peptides in targeting challenging PPIs. Technological advancements and emerging chemical approaches for stabilizing active peptide conformations are categorized in terms of α-helix stapling, ß-hairpin mimetics and macrocyclization. To discover potent and selective ligands, cyclic peptide library technologies were updated based on genetic, biochemical or synthetic methodologies. Moreover, several advances to improve the permeability and oral bioavailability of biologically active cyclic peptides enable the de novo development of cyclic peptide ligands with pharmacological properties. In summary, the development of cyclic peptide-based PPI modulators carries tremendous promise for the next generation of therapeutic agents to target historically "intractable" PPI systems.

12.
Biochemistry ; 51(12): 2579-87, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22390775

RESUMEN

Staphylococcus aureus secretes a phosphatidylinositol-specific phospholipase C (PI-PLC) as a virulence factor that is unusual in exhibiting higher activity at acidic pH values than other enzymes in this class. We have determined the crystal structure of this enzyme at pH 4.6 and pH 7.5. Under slightly basic conditions, the S. aureus PI-PLC structure closely follows the conformation of other bacterial PI-PLCs. However, when crystallized under acidic conditions, a large section of mobile loop at the αß-barrel rim in the vicinity of the active site shows ~10 Å shift. This loop displacement at acidic pH is the result of a titratable intramolecular π-cation interaction between His258 and Phe249. This was verified by a structure of the mutant protein H258Y crystallized at pH 4.6, which does not exhibit the large loop shift. The intramolecular π-cation interaction for S. aureus PI-PLC provides an explanation for the activity of the enzyme at acid pH and also suggests how phosphatidylcholine, as a competitor for Phe249, may kinetically activate this enzyme.


Asunto(s)
Dominio Catalítico , Fosfoinositido Fosfolipasa C/química , Fosfoinositido Fosfolipasa C/metabolismo , Staphylococcus aureus/enzimología , Concentración de Iones de Hidrógeno , Fosfatos de Inositol/química , Fosfatos de Inositol/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Fosfoinositido Fosfolipasa C/genética , Solubilidad , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/patogenicidad , Especificidad por Sustrato , Agua/química
13.
Am J Cancer Res ; 12(5): 2226-2248, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35693088

RESUMEN

Effective biomarkers that guide therapeutics with limited adverse effects, have emerged as attractive research topics in cancer diagnosis and treatment. Cancer-derived exosomes, a type of extracellular vesicles representing molecular signatures of cells of origin, could serve as stable reservoirs for potential biomarkers (i.e., proteins, nucleic acids) in non-invasive cancer diagnosis and prognosis. In this review, the physiological and pathological roles of exosomes and their protein components in facilitating tumorigenesis are highlighted. Exosomes carrying proteins can participate in tumor development and progression through multiple signaling pathways, including EMT, invasion and metastasis. Meanwhile, the practical applications of exosomal proteins in detecting and monitoring several solid-tumor cancers (including lung, breast, pancreatic, colorectal and prostate cancers) were also summarized. More clinically relevant, exosomal proteins play pivotal roles in transmitting oncogenic potential or resistance to therapies in recipient cells, which might further support therapeutic strategy determinations.

14.
Cell Chem Biol ; 28(5): 625-635.e5, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-33503403

RESUMEN

Wnt signaling plays a central role in tissue maintenance and cancer. Wnt activates downstream genes through ß-catenin, which interacts with TCF/LEF transcription factors. A major question is how this signaling is coordinated relative to tissue organization and renewal. We used a recently described class of small molecules that binds tubulin to reveal a molecular cascade linking stress signaling through ATM, HIPK2, and p53 to the regulation of TCF/LEF transcriptional activity. These data suggest a mechanism by which mitotic and genotoxic stress can indirectly modulate Wnt responsiveness to exert coherent control over cell shape and renewal. These findings have implications for understanding tissue morphogenesis and small-molecule anticancer therapeutics.


Asunto(s)
Sondas Moleculares/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción TCF/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Animales , Células Cultivadas , Humanos , Masculino , Sondas Moleculares/química , Bibliotecas de Moléculas Pequeñas/química , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Xenopus , Pez Cebra , beta Catenina/genética , beta Catenina/metabolismo
16.
Sci Rep ; 10(1): 9162, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32514015

RESUMEN

Today, pancreatic cancer (PC) remains a major health problem in the US. The fact that cancer stem cells (CSCs) become enriched in humans following anti-cancer therapy implicates CSCs as key contributors to tumor dormancy, metastasis, and relapse in PC. A highly validated CSC model (FGß3 cells) was used to test a novel compound (PAWI-2) to eradicate CSCs. Compared to parental bulk FG cells, PAWI-2 showed greater potency to inhibit cell viability and self-renewal capacity of FGß3 cells. For FGß3 cells, dysregulated integrin ß3-KRAS signaling drives tumor progression. PAWI-2 inhibited ß3-KRAS signaling independent of KRAS. This is clinically relevant. PAWI-2 targeted the downstream TBK1 phosphorylation cascade that was negatively regulated by optineurin phosphorylation via a feedback mechanism. This was confirmed by TBK1 genetic knockdown or co-treatment with TBK1-specific inhibitor (MRT67307). PAWI-2 also overcame erlotinib (an EGFR inhibitor) resistance in FGß3 cells more potently than bortezomib. In the proposed working model, optineurin acts as a key regulator to link inhibition of KRAS signaling and cell cycle arrest (G2/M). The findings show PAWI-2 is a new approach to reverse tumor stemness that resensitizes CSC tumors to drug inhibition.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Integrina beta3/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Quinoxalinas/farmacología , Antineoplásicos , Humanos , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células Tumorales Cultivadas
17.
Tissue Eng Part C Methods ; 26(11): 577-589, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33086948

RESUMEN

Despite considerable research effort, there is a significant need for safe agents that stimulate bone formation. Treatment of large or complex bone defects remains a challenge. Implantation of small molecule-induced human bone marrow-derived mesenchymal stromal cells (hBMSCs) on an appropriate tricalcium phosphate (TCP) scaffold offers a robust system for noninvasive therapy for spinal fusion. To show the efficacy of this approach, we identified a small molecule curcuminoid that when combined with TCP ceramic in the presence of hBMSCs selectively induced growth of bone cells: after 8- or 25-day incubations, alkaline phosphatase was elevated. Treatment of hBMSCs with curcuminoid 1 and TCP ceramic increased osteogenic target gene expression (i.e., Runx2, BMP2, Osteopontin, and Osteocalcin) over time. In the presence of curcuminoid 1 and TCP ceramic, osteogenesis of hBMSCs, including proliferation, differentiation, and mineralization, was observed. No evidence of chondrogenic or adipogenic potential using this protocol was observed. Transplantation of curcuminoid 1-treated hBMSC/TCP mixtures into the spine of immunodeficient rats showed that it achieved spinal fusion and provided greater stability of the spinal column than untreated hBMSC-TCP implants or TCP alone implants. On the basis of histological analysis, greater bone formation was associated with curcuminoid 1-treated hBMSC implants manifested as contiguous growth plates with extensive hematopoietic territories. Stimulation of hBMSCs by administration of small molecule curcuminoid 1 in the presence of TCP ceramic afforded an effective noninvasive strategy that increased spinal fusion repair and provided greater stability of the spinal column after 8 weeks in immunodeficient rats. Impact statement Bone defects only slowly regenerate themselves in humans. Current procedures to restore spinal defects are not always effective. Some have side effects. In this article, a new method to produce bone growth within 8 weeks in rats is presented. In the presence of tricalcium phosphate ceramic, curcuminoid-1 small molecule-stimulated human bone marrow-derived mesenchymal stromal cells showed robust bone cell growth in vitro. Transplantation of this mixture into the spine showed efficient spinal fusion in rats. The approach presented herein provides an efficient biocompatible scaffold for delivery of a potentially clinically useful system that could be applicable in patients.


Asunto(s)
Fosfatos de Calcio/farmacología , Cerámica/farmacología , Diarilheptanoides/farmacología , Fusión Vertebral , Fosfatasa Alcalina/metabolismo , Animales , Calcio/metabolismo , Fosfatos de Calcio/química , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno/farmacología , Diarilheptanoides/química , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Implantes Experimentales , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/enzimología , Minerales/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Ratas Desnudas , Andamios del Tejido/química , Proteínas Wnt/metabolismo
18.
Analyst ; 134(7): 1392-5, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19562207

RESUMEN

This paper reports the electrochemical behavior of lactate dehydrogenase (LDH) immobilized in the silica sol-gel film on gold electrode after adding nanometre-sized tridecameric aluminium polycation (nano-Al13, also called nanopolynuclear Al13) as a promoter. A pair of surface controlled quasi-reversible cyclic voltammetry peaks with the formal potential (E0') of 154 mV (vs. SCE) was found in the presence of nano-Al13. A potential application of the nano-Al13-LDH electrode for the determination of resorcinol and p-xylene was also investigated. The experimental results showed that both resorcinol and p-xylene inhibited LDH activity, and the calibration ranges were 5.0 x 10(-6)-3.0 x 10(-4) mol L(-1) for resorcinol and 1.0 x 10(-6)-1.0 x 10(-5) mol L(-1) for p-xylene, respectively. The nano-Al13-LDH electrode can be anticipated to be applied to environmental toxic assessments.


Asunto(s)
Aluminio/química , Enzimas Inmovilizadas/química , Oro/química , L-Lactato Deshidrogenasa/química , Nanopartículas del Metal/química , Dióxido de Silicio/química , Animales , Técnicas Biosensibles , Bovinos , Electroquímica , Electrodos , Contaminantes Ambientales/análisis , Enzimas Inmovilizadas/metabolismo , Geles , L-Lactato Deshidrogenasa/metabolismo
19.
Am J Cancer Res ; 9(2): 390-405, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30906636

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC), constitutes >90% of pancreatic cancers (PC) and is one of the most aggressive human tumors. Standard chemotherapies for PDAC (e.g., gemcitabine, FOLFIRINOX, etc.) has proven to be largely ineffective. Herein, we report a novel molecule (i.e., compound 1) that potently inhibits proliferation and induces apoptosis of PDAC cells. As we observed in other cancer types (i.e., colorectal, breast cancer), the effect of 1 against PDAC cells is also related to microtubule destabilization and DNA damage checkpoint activation. However, in PDAC cells, the inhibitory effect of 1 was mainly controlled by mitochondrial p53-dependent apoptosis. Compound 1 worked with cells of different p53 mutant status and affected p53 activation/phosphorylation not simply by stabilizing p53 protein but through antagonizing anti-apoptotic effects of Bcl-xL and restoring p53 to activate mitochondrial-apoptotic pathways (i.e., cytochrome c release, caspase activation and PARP cleavage). Compound 1 was more efficient than a typical PDAC combination therapy (i.e., gemcitabine with paclitaxel) and showed synergism in inhibiting PDAC cell proliferation with gemcitabine (or gemcitabine with paclitaxel). This synergism varied between different types of PDAC cells and was partially controlled by the phosphorylation of p53 on Serine15 (phospho-Ser15-p53). In vivo studies in an orthotopic syngeneic murine model showed that 1 (20 mg/kg/day, 28 days, i.p.) inhibited tumor growth by 65% compared to vehicle-treated mice. No apparent acute or chronic toxicity was observed. Thus, compound 1 utilizes a distinct mechanism of action to inhibit PC growth in vitro and in vivo and is a novel anti-PDAC compound.

20.
Artículo en Inglés | MEDLINE | ID: mdl-17434334

RESUMEN

We present a novel method for the study of the aggregation of protein induced by metal ion aluminum(III) using resonance Rayleigh scattering (RRS) technique. In neutral Tris-HCl medium, the effect of this aggregation of protein results in the enhancement of RRS intensity and the relationship between the enhancement of the RRS signal and the Al concentration is nonlinear. On this basis, we established a new method for the determination of the critical induced-aggregation concentrations (C(CIAC)) of metal ion Al(III) inducing the protein aggregation. Our results show that many factors, such as, pH value, anions, salts, temperature and solvents have obvious effects. We also studied the extent of aggregation and structural changes using ultra-violet spectrometry, protein intrinsic fluorescence and circular dichroism to further understand the exact mechanisms of the aggregation characteristics of proteins induced by metal ion Al(III) at the molecular level, to help us to develop effective methods to investigate the toxicity of metal ion Al, and to provide theoretical and quantitative evidences for the development of appropriate treatments for neurodementia such as Parkinson's disease, Alzheimer's disease and dementia related to dialysis.


Asunto(s)
Aluminio/química , Técnicas de Química Analítica/métodos , Albúmina Sérica Bovina/química , Albúmina Sérica/química , Animales , Bovinos , Dicroismo Circular , Humanos , Concentración de Iones de Hidrógeno , Estructura Cuaternaria de Proteína , Solventes , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA