Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 201
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nat Chem Biol ; 18(12): 1370-1379, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35970996

RESUMEN

Pyrvinium is a quinoline-derived cyanine dye and an approved anti-helminthic drug reported to inhibit WNT signaling and have anti-proliferative effects in various cancer cell lines. To further understand the mechanism by which pyrvinium is cytotoxic, we conducted a pooled genome-wide CRISPR loss-of-function screen in the human HAP1 cell model. The top drug-gene sensitizer interactions implicated the malate-aspartate and glycerol-3-phosphate shuttles as mediators of cytotoxicity to mitochondrial complex I inhibition including pyrvinium. By contrast, perturbation of the poorly characterized gene C1orf115/RDD1 resulted in strong resistance to the cytotoxic effects of pyrvinium through dysregulation of the major drug efflux pump ABCB1/MDR1. Interestingly, C1orf115/RDD1 was found to physically associate with ABCB1/MDR1 through proximity-labeling experiments and perturbation of C1orf115 led to mis-localization of ABCB1/MDR1. Our results are consistent with a model whereby C1orf115 modulates drug efflux through regulation of the major drug exporter ABCB1/MDR1.


Asunto(s)
Antineoplásicos , Compuestos de Pirvinio , Humanos , Compuestos de Pirvinio/farmacología , Vía de Señalización Wnt , Antineoplásicos/farmacología , Genómica
2.
J Biol Chem ; 298(8): 102227, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35780831

RESUMEN

The Cullin-RING ligase 4 E3 ubiquitin ligase component Cereblon (CRBN) is a well-established target for a class of small molecules termed immunomodulatory drugs (IMiDs). These drugs drive CRBN to modulate the degradation of a number of neosubstrates required for the growth of multiple cancers. Whereas the mechanism underlying the activation of CRBN by IMiDs is well described, the normal physiological regulation of CRBN is poorly understood. We recently showed that CRBN is activated following exposure to Wnt ligands and subsequently mediates the degradation of a subset of physiological substrates. Among the Wnt-dependent substrates of CRBN is Casein kinase 1α (CK1α), a known negative regulator of Wnt signaling. Wnt-mediated degradation of CK1α occurs via its association with CRBN at a known IMiD binding pocket. Herein, we demonstrate that a small-molecule CK1α agonist, pyrvinium, directly prevents the Wnt-dependent interaction of CRBN with CK1α, attenuating the consequent CK1α degradation. We further show that pyrvinium disrupts the ability of CRBN to interact with CK1α at the IMiD binding pocket within the CRBN-CK1α complex. Of note, this function of pyrvinium is independent of its previously reported ability to enhance CK1α kinase activity. Furthermore, we also demonstrate that pyrvinium attenuates CRBN-induced Wnt pathway activation in vivo. Collectively, these results reveal a novel dual mechanism through which pyrvinium inhibits Wnt signaling by both attenuating the CRBN-mediated destabilization of CK1α and activating CK1α kinase activity.


Asunto(s)
Caseína Quinasa Ialfa , Compuestos de Pirvinio , Caseína Quinasa Ialfa/metabolismo , Compuestos de Pirvinio/farmacología , Ubiquitina-Proteína Ligasas/metabolismo , Vía de Señalización Wnt
3.
J Biol Chem ; 298(10): 102479, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36096200

RESUMEN

The WNT-ß-catenin signaling pathway has a major role in regulating cell proliferation and differentiation. Aberrant activation of the pathway contributes to various human cancer types. Because casein kinase CK1α-initiated phosphorylation of ß-catenin is a key first step to restrain WNT signaling, effective restoration of CK1α activity represents an innovative strategy to combat WNT-driven cancer. A recent study in JBC reveals the anthelmintic pyrvinium directly binds to CK1α as an activator and also stabilizes CK1α protein, doubling against WNT-driven cancer activity.


Asunto(s)
Neoplasias , Compuestos de Pirvinio , Humanos , beta Catenina/genética , beta Catenina/metabolismo , Compuestos de Pirvinio/farmacología , Vía de Señalización Wnt , Neoplasias/tratamiento farmacológico , Neoplasias/genética
4.
Blood ; 137(17): 2403-2416, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33529322

RESUMEN

Chronic graft-versus-host disease (cGVHD) is a major life-threatening complication of allogeneic hematopoietic stem cell transplantation. The molecular mechanisms underlying cGVHD remain poorly understood, and targeted therapies for clinical use are not well established. Here, we examined the role of the canonical WNT pathway in sclerodermatous cGVHD (sclGVHD). WNT signaling was activated in human sclGVHD with increased nuclear accumulation of the transcription factor ß-catenin and a WNT-biased gene expression signature in lesional skin. Treatment with the highly selective tankryase inhibitor G007-LK, the CK1α agonist pyrvinium, or the LRP6 inhibitor salinomycin abrogated the activation of WNT signaling and protected against experimental cGVHD, without a significant impact on graft-versus-leukemia effect (GVL). Treatment with G007-LK, pyrvinium, or salinomycin almost completely prevented the development of clinical and histological features in the B10.D2 (H-2d) → BALB/c (H-2d) and LP/J (H-2b) → C57BL/6 (H-2b) models of sclGVHD. Inhibition of canonical WNT signaling reduced the release of extracellular matrix from fibroblasts and reduced leukocyte influx, suggesting that WNT signaling stimulates fibrotic tissue remodeling by direct effects on fibroblasts and by indirect inflammation-dependent effects in sclGVHD. Our findings may have direct translational potential, because pyrvinium is in clinical use, and tankyrase inhibitors are in clinical trials for other indications.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Piranos/farmacología , Compuestos de Pirvinio/farmacología , Esclerodermia Sistémica/prevención & control , Sulfonas/farmacología , Triazoles/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Animales , Antibacterianos/farmacología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Persona de Mediana Edad , Esclerodermia Sistémica/etiología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología
5.
J Biol Chem ; 294(29): 11259-11275, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31167790

RESUMEN

Transthyretin (TTR) is a major amyloidogenic protein associated with hereditary (ATTRm) and nonhereditary (ATTRwt) intractable systemic transthyretin amyloidosis. The pathological mechanisms of ATTR-associated amyloid fibril formation are incompletely understood, and there is a need for identifying compounds that target ATTR. C-terminal TTR fragments are often present in amyloid-laden tissues of most patients with ATTR amyloidosis, and on the basis of in vitro studies, these fragments have been proposed to play important roles in amyloid formation. Here, we found that experimentally-formed aggregates of full-length TTR are cleaved into C-terminal fragments, which were also identified in patients' amyloid-laden tissues and in SH-SY5Y neuronal and U87MG glial cells. We observed that a 5-kDa C-terminal fragment of TTR, TTR81-127, is highly amyloidogenic in vitro, even at neutral pH. This fragment formed amyloid deposits and induced apoptosis and inflammatory gene expression also in cultured cells. Using the highly amyloidogenic TTR81-127 fragment, we developed a cell-based high-throughput screening method to discover compounds that disrupt TTR amyloid fibrils. Screening a library of 1280 off-patent drugs, we identified two candidate repositioning drugs, pyrvinium pamoate and apomorphine hydrochloride. Both drugs disrupted patient-derived TTR amyloid fibrils ex vivo, and pyrvinium pamoate also stabilized the tetrameric structure of TTR ex vivo in patient plasma. We conclude that our TTR81-127-based screening method is very useful for discovering therapeutic drugs that directly disrupt amyloid fibrils. We propose that repositioning pyrvinium pamoate and apomorphine hydrochloride as TTR amyloid-disrupting agents may enable evaluation of their clinical utility for managing ATTR amyloidosis.


Asunto(s)
Amiloide/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Prealbúmina/metabolismo , Amiloide/efectos de los fármacos , Neuropatías Amiloides Familiares/metabolismo , Apomorfina/farmacología , Células Cultivadas , Reposicionamiento de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Inflamación/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Prealbúmina/química , Conformación Proteica , Proteolisis , Compuestos de Pirvinio/farmacología , Tripsina/metabolismo
6.
Pharm Res ; 37(3): 43, 2020 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-31989336

RESUMEN

PURPOSE: Pyrvinium pamoate (PP) is an anthelmintic drug that has been found to have anti-cancer activity in several cancer types. In the present study, we evaluated PP for potential anti-leukemic activity in B cell acute lymphoblastic leukemia (ALL) cell lines, in an effort to evaluate the repurposing potential of this drug in leukemia. METHODS: ALL cells were treated with PP at various concentrations to determine its effect on cell proliferation. Metabolic function was tested by evaluating Extracellular Acidification Rate (ECAR) and Oxygen Consumption Rate (OCR). Lastly, 3D spheroids were grown, and PP was reformulated into nanoparticles to evaluate distribution effectiveness. RESULTS: PP was found to inhibit ALL proliferation, with varied selectivity to different ALL cell subtypes. We also found that PP's cell death activity was specific for leukemic cells, as primary normal immune cells were resistant to PP-mediated cell death. Metabolic studies indicated that PP, in part, inhibits mitochondrial oxidative phosphorylation. To increase the targeting of PP to a hypoxic bone tumor microenvironment (BTME) niche, we successfully encapsulated PP in a nanoparticle drug delivery system and demonstrated that it retained its anti-leukemic activity in a hemosphere assay. CONCLUSION: We have demonstrated that PP is a novel therapeutic lead compound that counteracts the respiratory reprogramming found in refractory ALL cells and can be effectively formulated into a nanoparticle delivery system to target the BTME.


Asunto(s)
Antineoplásicos/farmacología , Huesos/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Compuestos de Pirvinio/farmacología , Microambiente Tumoral/efectos de los fármacos , Muerte Celular , Línea Celular Tumoral , Proliferación Celular , Composición de Medicamentos/métodos , Liberación de Fármacos , Humanos , Nanocápsulas/química , Fosforilación , Transducción de Señal
7.
Bioorg Med Chem Lett ; 30(8): 127037, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32081449

RESUMEN

Herein, we report the synthesis and evaluation of pyrvinium-based antimalarial and antitubercular compounds. Pyrvinium is an FDA approved drug for the treatment of pinworm infection, and it has been reported to have antiparasitic and antimicrobial activities. Pyrvinium contains quinoline core coupled with pyrrole. We replaced the pyrrole with various aryl or heteroaryl substituents to generate pyrvinium analogs. The profiling of these compounds against malaria parasite P. falciparum 3D7 revealed analogs with better antimalarial activity than pyrvinium pamoate. Compound 14 and 16 showed IC50 of 23 nM and 60 nM against P. falciparum 3D7, respectively. These compounds were also effective against drug-resistant malaria parasite P. falciparum Dd2 with IC50 of 53 nM and 97 nM, respectively. The cytotoxicity against CHO-K1, HEK and NRK-49F cells revealed better selectivity index for these new analogs compared to pyrvinium. Additionally, this series of compounds showed activity against M. tuberculosis H37Rv; particularly compounds 10, 13, 14 and 16 showed equipotent antitubercular activity to that of pyrvinium pamoate. The compounds 14 and 16 should be taken forward as leads for further optimization.


Asunto(s)
Antimaláricos/farmacología , Antituberculosos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Compuestos de Pirvinio/farmacología , Antimaláricos/síntesis química , Antimaláricos/química , Antituberculosos/síntesis química , Antituberculosos/química , Relación Dosis-Respuesta a Droga , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Pruebas de Sensibilidad Parasitaria , Compuestos de Pirvinio/síntesis química , Compuestos de Pirvinio/química , Relación Estructura-Actividad , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología
8.
Biochemistry ; 58(51): 5102-5106, 2019 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-31820934

RESUMEN

The serine/threonine protein kinase casein kinase 1α (CK1α) functions as a negative regulator of Wnt signaling, phosphorylating ß-catenin at serine 45 (P-S45) to initiate its eventual ubiquitin-mediated degradation. We previously showed that the repurposed, FDA-approved anthelminthic drug pyrvinium potently inhibits Wnt signaling in vitro and in vivo. Moreover, we proposed that pyrvinium's Wnt inhibitory activity was the result of its function as an activator of CK1α. An understanding of the mechanism by which pyrvinium activates CK1α is important because pyrvinium was given an orphan drug designation by the FDA to treat familial adenomatous polyposis, a precancerous condition driven by constitutive Wnt signaling. In the current study, we show that pyrvinium stimulates the phosphorylation of S45 ß-catenin, a known CK1α substrate, in a cell-based assay, and does so in a dose- and time-dependent manner. Alternative splicing of CK1α results in four forms of the protein with distinct biological properties. We evaluated these splice products and identified the CK1α splice variant, CK1αS, as the form that exhibits the most robust response to pyrvinium in cells. Kinetic studies indicate that pyrvinium also stimulates the kinase activity of purified, recombinant CK1αS in vitro, increasing its catalytic efficiency (kcat/Km) toward substrates. These studies provide strong and clear mechanistic evidence that pyrvinium enhances CK1α kinase activity.


Asunto(s)
Biocatálisis/efectos de los fármacos , Caseína Quinasa Ialfa/metabolismo , Compuestos de Pirvinio/farmacología , Activación Enzimática/efectos de los fármacos , Células HEK293 , Humanos , Cinética
9.
Blood ; 129(22): 2959-2970, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28348148

RESUMEN

There is accumulating evidence that functional alteration(s) of the bone marrow (BM) microenvironment contribute to the development of some myeloid disorders, such as myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). In addition to a cell-intrinsic role of WNT activation in leukemia stem cells, WNT activation in the BM niche is also thought to contribute to the pathogenesis of MDS and AML. We previously showed that the Apc-haploinsufficient mice (Apcdel/+ ) model MDS induced by an aberrant BM microenvironment. We sought to determine whether Apc, a multifunctional protein and key negative regulator of the canonical ß-catenin (Ctnnb1)/WNT-signaling pathway, mediates this disease through modulating WNT signaling, and whether inhibition of WNT signaling prevents the development of MDS in Apcdel/+ mice. Here, we demonstrate that loss of 1 copy of Ctnnb1 is sufficient to prevent the development of MDS in Apcdel/+ mice and that altered canonical WNT signaling in the microenvironment is responsible for the disease. Furthermore, the US Food and Drug Administration (FDA)-approved drug pyrvinium delays and/or inhibits disease in Apcdel/+ mice, even when it is administered after the presentation of anemia. Other groups have observed increased nuclear CTNNB1 in stromal cells from a high frequency of MDS/AML patients, a finding that together with our results highlights a potential new strategy for treating some myeloid disorders.


Asunto(s)
Genes APC , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/prevención & control , Nicho de Células Madre/genética , Vía de Señalización Wnt , Animales , Modelos Animales de Enfermedad , Haploinsuficiencia , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Síndromes Mielodisplásicos/patología , Compuestos de Pirvinio/farmacología , Vía de Señalización Wnt/genética , beta Catenina/genética
10.
Anticancer Drugs ; 30(7): e0747, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31305293

RESUMEN

Pyrvinium tosylate (PT) is an anthelminthic drug that has recently been shown to suppress various human cancers. However, whether PT is effective in nasopharyngeal carcinoma (NPC) has not been determined to date. In this work, we show the selective efficacy of PT in NPC while sparing normal nasopharyngeal epithelial cells, and its ability to increase chemosensitivity. We show that PT at 100 and 500 nmol/l significantly inhibits growth and induces apoptosis in several NPC cell lines without affecting normal nasopharyngeal epithelial cells. Using cell culture and xenograft mouse models, PT markedly enhances cisplatin's efficacy in NPC and the combination leads to almost complete tumor inhibition. Mechanism studies show that PT suppresses active, nuclear ß-catenin level and activity and increases Axin level in NPC cells. ß-Catenin overexpression completely reverses the inhibitory effects of PT, confirming that ß-catenin is the molecular mechanism of PT's action in NPC. In addition, the effects of PT on ß-catenin and Axin levels and on Wnt signaling in NPC cells are mediated by its activation of casine kinase 1α. Our work is the first to suggest that Wnt/ß-catenin is a selective target for NPC treatment, and provides the preclinical evidence on the translational potential of PT as a useful addition to the treatment armamentarium for NPC.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Carcinoma Nasofaríngeo/tratamiento farmacológico , Compuestos de Pirvinio/farmacología , Proteínas Wnt/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Apoptosis , Caseína Quinasa Ialfa/genética , Proliferación Celular , Cisplatino/farmacología , Quimioterapia Combinada , Humanos , Ratones , Ratones SCID , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Molecules ; 24(13)2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31261664

RESUMEN

Obesity is a serious health problem, while the current anti-obesity drugs are not very effective. The Connectivity Map (C-Map), an in-silico drug screening approach based on gene expression profiles, has recently been indicated as a promising strategy for drug repositioning. In this study, we performed mRNA expression profile analysis using microarray technology and identified 435 differentially expressed genes (DEG) during adipogenesis in both C3H10T1/2 and 3T3-L1 cells. Then, DEG signature was uploaded into C-Map, and using pattern-matching methods we discovered that pyrvinium, a classical anthelminthic, is a novel anti-adipogenic differentiation agent. Pyrvinium suppressed adipogenic differentiation in a dose-dependent manner, as evidenced by Oil Red O staining and the mRNA levels of adipogenic markers. Furthermore, we identified that the inhibitory effect of pyrvinium was resulted primarily from the early stage of adipogenesis. Molecular studies showed that pyrvinium downregulated the expression of key transcription factors C/EBPa and PPARγ. The mRNA levels of notch target genes Hes1 and Hey1 were obviously reduced after pyrvinium treatment. Taken together, this study identified many differentially expressed genes involved in adipogenesis and demonstrated for the first time that pyrvinium is a novel anti-adipogenic compound for obesity therapy. Meanwhile, we provided a new strategy to explore potential anti-obesity drugs.


Asunto(s)
Adipogénesis/efectos de los fármacos , Antihelmínticos/farmacología , Perfilación de la Expresión Génica/métodos , Compuestos de Pirvinio/farmacología , Células 3T3-L1 , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Reposicionamiento de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos
12.
J Cell Physiol ; 233(4): 2871-2881, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28500633

RESUMEN

Pyrvinium pamoate (PP) is a quinoline-derived cyanine dye which was officially approved by FDA for its anthelmintic properties and therapeutic function against animal-like protists such as Cryptosporidium parvum and Plasmodium falciparum in the 1950s. In the last 10 years, several studies have shown the novel activity of pyrvinium in tumor therapy. Some investigations have indicated that pyrvinium could delay or inhibit tumor cell proliferation in cancer models including colon, breast, lung and prostate cancer, and some hematological malignancies. In this review, we discuss multiple critical signaling pathways and mechanisms underlying the anticancer effects of PP. In details, pyrvinium acts through the following main mechanisms: (i) energy and autophagy depletion; and (ii) inhibition of Akt and Wnt-ß-catenin-dependent pathways. Interestingly, pyrvinium has also shown potent anti-cancer stem cell activity. The overwhelming insights into the mechanism of anticancer properties of PP can help establishing novel and future anti-tumor treatment strategies.


Asunto(s)
Neoplasias/tratamiento farmacológico , Compuestos de Pirvinio/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Humanos , Neoplasias/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Compuestos de Pirvinio/química , Compuestos de Pirvinio/farmacología , Transducción de Señal/efectos de los fármacos
13.
J Cell Physiol ; 233(9): 7391-7401, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29659015

RESUMEN

Malignant mesothelioma (MM) is a very aggressive asbestos-related cancer, whose incidence is increasing worldwide. Unfortunately, no effective therapies are currently available and the prognosis is extremely poor. Recently, the anti-helminthic drug pyrvinium pamoate has attracted a strong interest for its anti-cancer activity, which has been demonstrated in many cancer models. Considering the previously established inhibitory effect of pyrvinium pamoate on the Wnt/ß-catenin pathway and given the important role of this pathway in MM, we investigated the potential anti-tumor activity of this drug in MM cell lines. We observed that pyrvinium pamoate significantly impairs MM cell proliferation, cloning efficiency, migration, and tumor spheroid formation. At the molecular level, our data show that pyrvinium pamoate down-regulates the expression of ß-catenin and Wnt-regulates genes. Overall, our study suggests that the repurposing of pyrvinium pamoate for MM treatment could represent a new promising therapeutic approach.


Asunto(s)
Reposicionamiento de Medicamentos , Mesotelioma/tratamiento farmacológico , Compuestos de Pirvinio/uso terapéutico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Mesotelioma/genética , Mesotelioma/patología , Compuestos de Pirvinio/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Factores de Tiempo , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , beta Catenina/metabolismo
14.
Am J Pathol ; 187(8): 1660-1669, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28606794

RESUMEN

Wound healing after corneal injury typically involves fibrosis, with transforming growth factor ß1 (TGF-ß1) as one of its strongest mediators. A class of small molecules-peroxisome proliferator-activated receptor γ (PPARγ) ligands-exert potent antifibrotic effects in the cornea by blocking phosphorylation of p38 mitogen-activated protein kinase (MAPK). However, why this blocks fibrosis remains unknown. Herein, we show that PPARγ ligands (rosiglitazone, troglitazone, and 15-deoxy-Δ12,14-prostaglandin J2) decrease levels of ß-catenin. We also show that ß-catenin siRNA and the Wingless/integrated (Wnt) inhibitor pyrvinium block the ability of corneal fibroblasts to up-regulate synthesis of α-smooth muscle actin (α-SMA), collagen 1 (COL1), and fibronectin (FN) in response to TGF-ß1. Activation of TGF-ß receptors and p38 MAPK increased glycogen synthase kinase 3ß (GSK3ß) phosphorylation, whereas a chemical inhibitor of p38 MAPK (SB203580) reduced the phosphorylation of GSK3ß, decreasing active ß-catenin levels in both cytoplasmic and nuclear fractions. Finally, lithium chloride, a GSK3 inhibitor, also attenuated the TGF-ß1-induced increase in α-SMA, COL1, and FN expression. All in all, our results suggest that TGF-ß1 stimulation increases active ß-catenin concentration in cultured corneal fibroblasts through p38 MAPK regulation of canonical Wnt/ß-catenin signaling, increasing α-SMA, COL1, and FN synthesis. Thus, PPARγ ligands, by blocking TGF-ß1-induced p38 MAPK phosphorylation, prevent increases in both total and active ß-catenin through p38 MAPK-GSK3ß signaling.


Asunto(s)
Córnea/efectos de los fármacos , Fibroblastos/efectos de los fármacos , PPAR gamma/agonistas , beta Catenina/metabolismo , Actinas/metabolismo , Animales , Gatos , Cromanos/farmacología , Colágeno Tipo I/metabolismo , Córnea/metabolismo , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Fibrosis/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Cloruro de Litio/farmacología , Fosforilación/efectos de los fármacos , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacología , Compuestos de Pirvinio/farmacología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/farmacología , Factor de Crecimiento Transformador beta1/farmacología , Troglitazona , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Biol Pharm Bull ; 41(2): 220-228, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29386482

RESUMEN

Previously, we developed a method to evaluate states of cells treated with anticancer drugs via the comprehensive analysis of amino acids, termed amino acid metabolomics. In the present study, we evaluated the effects of the anticancer drugs, gemcitabine hydrochloride and pyrvinium pamoate, on the proliferation of a pancreatic cancer cell line (PANC-1) under hypoglycemic conditions using amino acid metabolomics. Intracellular and extracellular amino acid profiles of PANC-1 were determined by hydrophilic interaction chromatography-tandem mass spectrometry with simple pretreatment. Changes to the drugs' anticancer effects resulting from glucose starvation conditions were presented in score plots obtained from principal component analyses. In particular, the analysis of intracellular amino acids was found to be the superior approach because the results allowed a clearer assessment of the cell state. Further, orthogonal partial least squares discriminant analysis was performed to search for amino acid candidates that discriminate with anticancer drug-treated PANC-1 cells. We identified several amino acids that might be able to distinguish the drug-treated group from the control group. These results might provide a better understanding of the mechanisms underlying cell responses such as drug resistance or austerity. The present study is the first to evaluate the efficacy of anticancer drugs under glucose starvation based on the analysis of the variation of extracellular and intracellular amino acid profiles in vitro.


Asunto(s)
Aminoácidos/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Desoxicitidina/análogos & derivados , Líquido Extracelular/metabolismo , Líquido Intracelular/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Compuestos de Pirvinio/farmacología , Aminoácidos/química , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/metabolismo , Glucemia/análisis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Desoxicitidina/farmacología , Análisis Discriminante , Glucosa/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Hipoglucemia/sangre , Hipoglucemia/complicaciones , Hipoglucemia/metabolismo , Análisis de los Mínimos Cuadrados , Metabolómica/métodos , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/metabolismo , Análisis de Componente Principal , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem , Gemcitabina
16.
Macromol Rapid Commun ; 38(13)2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28117522

RESUMEN

Pyrylium salts combined with vinyl ethers are shown to act as new versatile dual-cure initiating systems for both photochemical and thermal initiation of oxirane monomers. The combination of both possibilities allows the curing of thick samples through photoinduced frontal polymerization. On the basis of quantum calculations and photochemical experiments, some clues are given about the reaction mechanisms involved. Interestingly, a sequential kick-starting effect is observed in the presence of vinyl ether enabling the curing of oxetane monomers. Thereby, this communication presents a short overview of potential of pyrylium salts in cationic polymerization of oxiranes.


Asunto(s)
Polimerizacion , Compuestos de Pirvinio/química , Compuestos de Vinilo/química , Compuestos Epoxi/química , Óxido de Etileno/química
17.
Med Sci Monit ; 23: 266-275, 2017 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-28090074

RESUMEN

BACKGROUND Aberrant activation of Wnt/ß-catenin has been shown to promote ovarian cancer proliferation and chemoresistance. Pyrvinium, an FDA-approved anthelmintic drug, has been identified as a potent Wnt inhibitor. Pyrvinium may sensitize ovarian cancer cells to chemotherapy. MATERIAL AND METHODS The effect of pyrvinium alone and its combination with paclitaxel in ovarian cancer was investigated using an in vitro culture system and in vivo xenograft models. The mechanisms of its action were also analyzed, focusing on the Wnt/ß-catenin pathway. RESULTS Pyrvinium inhibited growth and induced apoptosis of paclitaxel- and cisplatin-resistant epithelial ovarian cancer cell lines A2278/PTX and SK-OV-3. Its combination with paclitaxel was synergistic in targeting ovarian cancer cells in vitro. In 3 independent ovarian xenograft mouse models, pyrvinium alone inhibited tumor growth. More importantly, we observed significant inhibition of tumor growth throughout the treatment when using pyrvinium and paclitaxel combined. Mechanistically, pyrvinium increased the Wnt-negative regulator axin and decreased the b-catenin levels in ovarian cancer cells. In addition, pyrvinium suppressed Wnt/b-catenin-mediated transcription, as shown by the decreased mRNA levels of MYC, cyclin D, and BCL-9. In contrast, the inhibitory effects of pyrvinium were reversed by ß-catenin stabilization or overexpression, demonstrating that pyrvinium acted on ovarian cancer cells via targeting the Wnt/ß-catenin signaling pathway. CONCLUSIONS We demonstrated that the anthelmintic drug pyrvinium targets ovarian cancer cells through suppressing Wnt/ß-catenin signaling. Our work highlights the therapeutic value of inhibiting Wnt/ß-catenin in ovarian cancer.


Asunto(s)
Antihelmínticos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Compuestos de Pirvinio/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Humanos , Cloruro de Litio/farmacología , Ratones SCID , Paclitaxel/farmacología , beta Catenina/metabolismo
18.
Acta Biochim Biophys Sin (Shanghai) ; 49(10): 890-898, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28981601

RESUMEN

Uveal melanoma is the most common primary intraocular malignancy in adults. And there is an absence of targeted agents for patients with uveal melanoma. Pyrvinium pamoate is an old anthelminthic medicine approved by FDA for the treatment of enterobiasis in 1955, which recently re-attracts attention as an anti-cancer drug due to its inhibition of Wnt/ß-catenin pathway in some types of cancer. But the role of pyrvinium pamoate in uveal melanoma and the potential underlying mechanism remains unknown. In this study, we tested the anti-tumor effects of pyrvinium pamoate on four uveal melanoma cell lines (92.1, Mel270, Omm1, and Omm2.3) and evaluated the Wnt/ß-catenin signaling transduction, cell growth, cell death, cell migration, and invasion accordingly. The results revealed that pyrvinium pamoate treatment repressed the phosphorylation of GSK3ß at S9 which might be mediated by AKT, and decreased the protein levels of ß-catenin and its downstream targets (c-Myc, cyclin D1). Pyrvinium pamoate remarkably inhibited cell viability and colony formation ability. Treatment with pyrvinium pamoate induced intrinsic pathway-dependent apoptosis accompanied with a decline of anti-apoptotic XIAP and Survivin, and an overt increase of pro-apoptotic Bax. In addition, pyrvinium pamoate significantly inhibited the migration and invasion in vitro. Our studies suggest that pyrvinium pamoate may be a potential therapeutic agent for uveal melanoma.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Compuestos de Pirvinio/farmacología , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo , Antihelmínticos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Melanoma/metabolismo , Melanoma/patología , Fosforilación/efectos de los fármacos , Survivin , Neoplasias de la Úvea/metabolismo , Neoplasias de la Úvea/patología , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
19.
Microbiology (Reading) ; 162(7): 1059-1068, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27126388

RESUMEN

Microbial pathogens represent an increasing threat to human health. Although many infections can be successfully treated and cleared, drug resistance is a widespread problem. The existence of subpopulations of 'tolerant' cells (where a fraction of the population is able to grow above the population resistance level) may increase the rate of treatment failure; yet, existing methods to measure subpopulation effects are cumbersome. Here we describe diskImageR, a computational pipeline that analyses photographs of disk diffusion assays to determine the degree of drug susceptibility [the radius of inhibition, (RAD)], and two aspects of subpopulation growth [the fraction of growth (FoG) within the zone of inhibition, (ZOI), and the rate of change in growth from no drug to inhibitory drug concentrations, (SLOPE)]. diskImageR was used to examine the response of the human fungal pathogen Candida albicans to the antifungal drug fluconazole across different strain backgrounds and growth conditions. Disk diffusion assays performed under Clinical and Laboratory Standards Institute (CLSI) conditions led to more susceptibility and less tolerance than assays performed using rich medium conditions. We also used diskImageR to quantify the effects of three drugs in combination with fluconazole, finding that all three combinations affected tolerance, with the effect of one drug (doxycycline) being very strain dependent. The three drugs had different effects on susceptibility, with doxycycline generally having no effect, chloroquine generally increasing susceptibility and pyrvinium pamoate generally reducing susceptibility. The ability to simultaneously quantitate different aspects of microbial drug responses will facilitate the study of mechanisms of subpopulation responses in the presence of antimicrobial drugs.


Asunto(s)
Antifúngicos/farmacología , Candida albicans/efectos de los fármacos , Candida albicans/crecimiento & desarrollo , Pruebas Antimicrobianas de Difusión por Disco/métodos , Programas Informáticos , Candida albicans/aislamiento & purificación , Cloroquina/farmacología , Doxiciclina/farmacología , Farmacorresistencia Fúngica , Fluconazol/farmacología , Humanos , Compuestos de Pirvinio/farmacología
20.
Biochem Biophys Res Commun ; 469(3): 716-22, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26707639

RESUMEN

Targeting mitochondrial respiration has emerged as an attractive therapeutic strategy in blood cancer due to their unique metabolic dependencies. In this study, we show that pyrvinium, a FDA-approved anthelmintic drug, selectively targets lymphoma T-cells though inhibition of mitochondrial functions and JAK2/STAT5. Pyrvinium induces apoptosis of malignant T-cell line Jurkat and primary T-cells from lymphoma patients while sparing T-cells from healthy donors. Increased level of active caspase-3 and decreased levels of Bcl-2 and Mcl-1 were also observed in Jurkat and lymphoma T-cells but not normal T-cells treated with pyrvinium. In addition, pyrvinium impairs mitochondrial functions by inhibit mitochondrial respiration, suppressing mitochondrial respiratory complex I activity, increasing ROS and decreasing ATP levels. However, the effects of pyrvinium were abolished in mitochondrial respiration-deficient Jurkat ρ(0) cells, confirming that pyrvinium acts on lymphoma T-cells via targeting mitochondrial respiration. We further show that lymphoma T-cells derived from patients depend more on mitochondrial respiration than normal T-cells, and this explains the selective toxicity of pyrvinium in lymphoma versus normal T-cells. Finally, we demonstrate that pyrvinium also suppresses JAK2/STAT5 signaling pathway in Jurkat cells. Our study suggests that pyrvinium is a useful addition to T-cell lymphoma treatment, and emphasizes the potential therapeutic value of the differences in the mitochondrial characteristics between malignant and normal T-cells in blood cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Janus Quinasa 2/metabolismo , Linfoma/tratamiento farmacológico , Linfoma/metabolismo , Compuestos de Pirvinio/administración & dosificación , Factor de Transcripción STAT5/metabolismo , Antihelmínticos/administración & dosificación , Antineoplásicos/administración & dosificación , Respiración de la Célula/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Células Jurkat , Linfoma/patología , Mitocondrias , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA