Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Angiogenesis ; 27(1): 91-103, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37733132

RESUMEN

Extracranial arteriovenous malformations (AVMs) are regarded as rare diseases and are prone to complications such as pain, bleeding, relentless growth, and high volume of shunted blood. Due to the high vascular pressure endothelial cells of AVMs are exposed to mechanical stress. To control symptoms and lesion growth pharmacological treatment strategies are urgently needed in addition to surgery and interventional radiology. AVM cells were isolated from three patients and exposed to cyclic mechanical stretching for 24 h. Thalidomide and bevacizumab, both VEGF inhibitors, were tested for their ability to prevent the formation of circular networks and proliferation of CD31+ endothelial AVM cells. Furthermore, the effect of thalidomide and bevacizumab on stretched endothelial AVM cells was evaluated. In response to mechanical stress, VEGF gene and protein expression increased in patient AVM endothelial cells. Thalidomide and bevacizumab reduced endothelial AVM cell proliferation. Bevacizumab inhibited circular network formation of endothelial AVM cells and lowered VEGF gene and protein expression, even though the cells were exposed to mechanical stress. With promising in vitro results, bevacizumab was used to treat three patients with unresectable AVMs or to prevent regrowth after incomplete resection. Bevacizumab controlled bleeding, pulsation, and pain over the follow up of eight months with no patient-reported side effects. Overall, mechanical stress increases VEGF expression in the microenvironment of AVM cells. The monoclonal VEGF antibody bevacizumab alleviates this effect, prevents circular network formation and proliferation of AVM endothelial cells in vitro. The clinical application of bevacizumab in AVM treatment demonstrates effective symptom control with no side effects.


Asunto(s)
Malformaciones Arteriovenosas , Células Endoteliales , Humanos , Células Endoteliales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Bevacizumab/farmacología , Bevacizumab/uso terapéutico , Bevacizumab/metabolismo , Talidomida/metabolismo , Malformaciones Arteriovenosas/genética , Dolor/metabolismo
2.
J Biochem ; 175(5): 507-519, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38140952

RESUMEN

Recently, the development of protein degraders (protein-degrading compounds) has prominently progressed. There are two remarkable classes of protein degraders: proteolysis-targeting chimeras (PROTACs) and molecular glue degraders (MGDs). Almost 70 years have passed since thalidomide was initially developed as a sedative-hypnotic drug, which is currently recognized as one of the most well-known MGDs. During the last two decades, a myriad of PROTACs and MGDs have been developed, and the molecular mechanism of action (MOA) of thalidomide was basically elucidated, including identifying its molecular target cereblon (CRBN). CRBN forms a Cullin Ring Ligase 4 with Cul4 and DDB1, whose substrate specificity is controlled by its binding ligands. Thalidomide, lenalidomide and pomalidomide, three CRBN-binding MGDs, were clinically approved to treat several intractable diseases (including multiple myeloma). Several other MGDs and CRBN-based PROTACs (ARV-110 and AVR-471) are undergoing clinical trials. In addition, several new related technologies regarding PROTACs and MGDs have also been developed, and achievements of protein degraders impact not only therapeutic fields but also basic biological science. In this article, I introduce the history of protein degraders, from the development of thalidomide to the latest PROTACs and related technologies.


Asunto(s)
Proteolisis , Talidomida , Talidomida/análogos & derivados , Ubiquitina-Proteína Ligasas , Talidomida/farmacología , Talidomida/química , Talidomida/metabolismo , Humanos , Proteolisis/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Quimera Dirigida a la Proteólisis
3.
Microb Pathog ; 181: 106168, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37224982

RESUMEN

Macrophage-derived inflammatory cytokines are critical for host defense against Talaromyces marneffei (T. marneffei) infection among HIV/AIDS patients, and excessive inflammatory cytokines are associated with poor outcomes of AIDS-associated talaromycosis. However, the underlying mechanisms of macrophage-caused pyroptosis and cytokine storm are poorly understood. Here, in the T. marneffei-infected mice and macrophages, we show that T. marneffei induced pyroptosis in macrophages through the NLRP3/caspase-1 pathway. The immunomodulatory drug thalidomide could promote the pyroptosis of macrophages infected T. marneffei. In T. marneffei-infected mice, the splenic macrophages underwent increasing pyroptosis as talaromycosis deteriorated. Thalidomide ameliorated inflammation of mice, while amphotericin B (AmB) in combination with thalidomide did not improve overall survival compared with AmB alone. Taken together, our findings suggest that thalidomide promotes NLRP3/caspase-1-mediated pyroptosis of macrophages in T. marneffei infection.


Asunto(s)
Talaromyces , Talidomida , Animales , Ratones , Talidomida/farmacología , Talidomida/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Caspasa 1/metabolismo , Piroptosis , Macrófagos/metabolismo , Anfotericina B , Citocinas/metabolismo
4.
Inflammopharmacology ; 31(3): 1167-1182, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36966238

RESUMEN

The "Thalidomide tragedy" is a landmark in the history of the pharmaceutical industry. Despite limited clinical trials, there is a continuous effort to investigate thalidomide as a drug for cancer and inflammatory diseases such as rheumatoid arthritis, lepromatous leprosy, and COVID-19. This review focuses on the possibilities of targeting inflammation by repurposing thalidomide for the treatment of idiopathic pulmonary fibrosis (IPF). Articles were searched from the Scopus database, sorted, and selected articles were reviewed. The content includes the proven mechanisms of action of thalidomide relevant to IPF. Inflammation, oxidative stress, and epigenetic mechanisms are major pathogenic factors in IPF. Transforming growth factor-ß (TGF-ß) is the major biomarker of IPF. Thalidomide is an effective anti-inflammatory drug in inhibiting TGF-ß, interleukins (IL-6 and IL-1ß), and tumour necrosis factor-α (TNF-α). Thalidomide binds cereblon, a process that is involved in the proposed mechanism in specific cancers such as breast cancer, colon cancer, multiple myeloma, and lung cancer. Cereblon is involved in activating AMP-activated protein kinase (AMPK)-TGF-ß/Smad signalling, thereby attenuating fibrosis. The past few years have witnessed an improvement in the identification of biomarkers and diagnostic technologies in respiratory diseases, partly because of the COVID-19 pandemic. Hence, investment in clinical trials with a systematic plan can help repurpose thalidomide for pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , Inmunosupresores , Talidomida , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Pulmón/metabolismo , Talidomida/uso terapéutico , Talidomida/metabolismo , Talidomida/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Inmunosupresores/farmacocinética , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico
5.
Drug Metab Dispos ; 51(1): 123-129, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35772770

RESUMEN

Many drug oxygenations are mainly mediated by polymorphic cytochromes P450 (P450s) and also by flavin-containing monooxygenases (FMOs). More than 50 years of research on P450/FMO-mediated drug oxygenations have clarified their catalytic roles. The natural product coumarin causes hepatotoxicity in rats via the reactive coumarin 3,4-epoxide, a reaction catalyzed by P450 1A2; however, coumarin undergoes rapid 7-hydroxylation by polymorphic P450 2A6 in humans. The primary oxidation product of the teratogen thalidomide in rats is deactivated 5'-hydroxythalidomide plus sulfate and glucuronide conjugates; however, similar 5'-hydroxythalidomide and 5-hydroxythalidomide are formed in rabbits in vivo. Thalidomide causes human P450 3A enzyme induction in liver (and placenta) and is also activated in vitro and in vivo by P450 3A through the primary human metabolite 5-hydroxythalidomide (leading to conjugation with glutathione/nonspecific proteins). Species differences exist in terms of drug metabolism in rodents and humans, and such differences can be very important when determining the contributions of individual enzymes. The approaches used for investigating the roles of human P450 and FMO enzymes in understanding drug oxidations and clinical therapy have not yet reached maturity and still require further development. SIGNIFICANCE STATEMENT: Drug oxidations in animals and humans mediated by P450s and FMOs are important for understanding the pharmacological properties of drugs, such as the species-dependent teratogenicity of the reactive metabolites of thalidomide and the metabolism of food-derived odorous trimethylamine to non-odorous (but proatherogenic) trimethylamine N-oxide. Recognized differences exist in terms of drug metabolism between rodents, non-human primates, and humans, and such differences are important when determining individual liver enzyme contributions with substrates in in vitro and in vivo systems.


Asunto(s)
Sistema Enzimático del Citocromo P-450 , Talidomida , Embarazo , Femenino , Humanos , Ratas , Conejos , Animales , Talidomida/metabolismo , Especificidad de la Especie , Oxidación-Reducción , Sistema Enzimático del Citocromo P-450/metabolismo , Microsomas Hepáticos/metabolismo , Cumarinas/metabolismo
6.
ACS Chem Biol ; 17(11): 3229-3237, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36325969

RESUMEN

Lenalidomide is a ligand of the E3 ligase substrate adapter cereblon (CRBN) that achieves its clinical effects in part by the promotion of substrate recruitment and degradation. In contrast to prior targets, eIF3i is recruited but not degraded upon complex formation with lenalidomide and CRBN, although the structural details and mechanistic outcomes of this interaction are unresolved. Here, we characterize the structural basis and mechanistic outcomes of lenalidomide-induced sequestration of eIF3i from the eIF3 complex. Identification of the binding interface on eIF3i by a covalent lenalidomide probe and mass spectrometry rationalizes the sequestration event. We further connect eIF3i and CRBN to lenalidomide-driven effects on angiogenic markers, Akt1 phosphorylation, and associated antiangiogenesis phenotypes. Finally, we find that eIF3i sequestration is observed in MM.1S and MOLM13 cells after the degradation of other substrates, such as IKZF1. The defined binding interface elucidated by chemical proteomics and the observation of eIF3i sequestration as a lenalidomide function open future directions in designing new chemical adapters for protein sequestration as a strategy to selectively control protein functions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Talidomida , Lenalidomida , Talidomida/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Factores de Transcripción/metabolismo , Péptido Hidrolasas/metabolismo
7.
Nucleic Acids Res ; 50(10): 5577-5598, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35640596

RESUMEN

A major pharmacological strategy toward HIV cure aims to reverse latency in infected cells as a first step leading to their elimination. While the unbiased identification of molecular targets physically associated with the latent HIV-1 provirus would be highly valuable to unravel the molecular determinants of HIV-1 transcriptional repression and latency reversal, due to technical limitations, this has been challenging. Here we use a dCas9 targeted chromatin and histone enrichment strategy coupled to mass spectrometry (Catchet-MS) to probe the differential protein composition of the latent and activated HIV-1 5'LTR. Catchet-MS identified known and novel latent 5'LTR-associated host factors. Among these, IKZF1 is a novel HIV-1 transcriptional repressor, required for Polycomb Repressive Complex 2 recruitment to the LTR. We find the clinically advanced thalidomide analogue iberdomide, and the FDA approved analogues lenalidomide and pomalidomide, to be novel LRAs. We demonstrate that, by targeting IKZF1 for degradation, these compounds reverse HIV-1 latency in CD4+ T-cells isolated from virally suppressed people living with HIV-1 and that they are able to synergize with other known LRAs.


Asunto(s)
Infecciones por VIH , VIH-1 , Linfocitos T CD4-Positivos/metabolismo , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , VIH-1/genética , Humanos , Factor de Transcripción Ikaros/genética , Provirus/genética , Talidomida/metabolismo , Talidomida/farmacología , Factores de Transcripción/metabolismo , Activación Viral , Latencia del Virus
8.
Commun Biol ; 4(1): 1277, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34764413

RESUMEN

Pomalidomide and lenalidomide are immunomodulatory agents that were derived from thalidomide. Cereblon (CRBN) is a common direct target of thalidomide and related compounds and works as a Cullin Ring 4 E3 ubiquitin ligase (CRL4) with DDB1, CUL4, and ROC1. The substrate specificity of CRL4CRBN is modulated by thalidomide-related compounds. While lenalidomide is approved for the treatment of several diseases including multiple myeloma, 5q- syndrome, mantle cell lymphoma, and follicular lymphoma, pomalidomide is approved only for the treatment of lenalidomide-resistant multiple myeloma. Here we show that PLZF/ZBTB16 and its fusion proteins are pomalidomide-dependent neosubstrates of CRL4CRBN. PLZF joins to RARα or potentially other partner genes, and the translocation causes leukemias, such as acute promyelocytic leukemia and T-cell acute lymphoblastic leukemia. We demonstrate that pomalidomide treatment induces PLZF-RARα degradation, resulting in antiproliferation of leukemic cells expressing PLZF-RARα. This study highlights a potential therapeutic role of pomalidomide as a degrader of leukemogenic fusion proteins.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Talidomida/análogos & derivados , Ubiquitina-Proteína Ligasas/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Células HEK293 , Humanos , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Alineación de Secuencia , Talidomida/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
9.
J Toxicol Sci ; 46(7): 311-317, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34193768

RESUMEN

The approved drug thalidomide is teratogenic in humans, nonhuman primates, and rabbits but not in rodents. The extensive biotransformation of 5'-hydroxythalidomide after oral administration of thalidomide (250 mg/kg) in rats was investigated in detail using liquid chromatography-tandem mass spectrometry. Probable metabolites 5'-hydroxythalidomide sulfate and glucuronide were extensively formed, with approximately tenfold and onefold peak areas, respectively, to the primary 5'-hydroxythalidomide measured using authentic standards. As a minor metabolite, 5-hydroxythalidomide was also detected. The output of simplified physiologically based pharmacokinetic rat models was consistent with the observed in vivo data under a metabolic ratio of 0.05 for the hepatic intrinsic clearance of thalidomide to unconjugated 5'-hydroxythalidomide. The aggregate of unconjugated and sulfate/glucuronide conjugated 5'-hydroxythalidomide forms appear to be the predominant metabolites in rats. Two hours after oral administration of thalidomide (100 mg/kg) to chimeric mice humanized with four different batches of genotyped human hepatocytes, the plasma concentration ratios of 5-hydroxythalidomide to 5'-hydroxythalidomide were correlated with replacement indexes of human liver cells previously transplanted in immunodeficient mice. These results indicate that rodent livers mediate thalidomide primary oxidation, leading to extensive deactivation in vivo to unconjugated/conjugated 5'-hydroxythalidomide and suggest that thalidomide activation might be dependent on the humanized livers in mice transplanted with human hepatocytes.


Asunto(s)
Hepatocitos/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Teratógenos/farmacocinética , Teratógenos/toxicidad , Talidomida/farmacocinética , Talidomida/toxicidad , Animales , Humanos , Masculino , Redes y Vías Metabólicas , Ratones , Modelos Animales , Ratas , Especificidad de la Especie , Teratógenos/metabolismo , Talidomida/análogos & derivados , Talidomida/metabolismo
10.
Eur J Med Chem ; 223: 113645, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34217059

RESUMEN

Protein degradation is a promising strategy for drug development. Proteolysis-targeting chimeras (PROTACs) hijacking the E3 ligase cereblon (CRBN) exhibit enormous potential and universal degradation performance due to the small molecular weight of CRBN ligands. In this study, the CRBN-recruiting PROTACs were explored on the degradation of oncogenic fusion protein BCR-ABL, which drives the pathogenesis of chronic myeloid leukemia (CML). A series of novel PROTACs were synthesized by conjugating BCR-ABL inhibitor dasatinib to the CRBN ligand including pomalidomide and lenalidomide, and the extensive structure-activity relationship (SAR) studies were performed focusing on optimization of linker parameters. Therein, we uncovered that pomalidomide-based degrader 17 (SIAIS056), possessing sulfur-substituted carbon chain linker, exhibits the most potent degradative activity in vitro and favorable pharmacokinetics in vivo. Besides, degrader 17 also degrades a variety of clinically relevant resistance-conferring mutations of BCR-ABL. Furthermore, degrader 17 induces significant tumor regression against K562 xenograft tumors. Our study indicates that 17 as an efficacious BCR-ABL degrader warrants intensive investigation for the future treatment of BCR-ABL+ leukemia.


Asunto(s)
Diseño de Fármacos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Ubiquitina-Proteína Ligasas/química , Animales , Proliferación Celular/efectos de los fármacos , Dasatinib/farmacología , Proteínas de Fusión bcr-abl/metabolismo , Semivida , Humanos , Células K562 , Lenalidomida/química , Lenalidomida/metabolismo , Ligandos , Ratones , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteolisis , Relación Estructura-Actividad , Talidomida/análogos & derivados , Talidomida/química , Talidomida/metabolismo , Trasplante Heterólogo , Ubiquitina-Proteína Ligasas/metabolismo
11.
Bioorg Med Chem ; 43: 116271, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34171757

RESUMEN

Apremilast is an important active pharmaceutical ingredient that relies on a resolution to produce the key chiral amine intermediate. To provide a new catalytic and enzymatic process for Apremilast, we performed the directed evolution of the amine transaminase fromVibriofluvialis. Six rounds of evolution resulted in the VF-8M-E variant with > 400-fold increase specific activity over the wildtype enzyme. A homology model of VF-8M-E was built and a molecular docking study was performed to explain the increase in activity. The purified VF-8M-E was successfully applied to produce the key chiral amine intermediate in enantiopure form and 49% conversion via a kinetic resolution, representing a new enzymatic access towards Apremilast.


Asunto(s)
Aminas/metabolismo , Talidomida/análogos & derivados , Transaminasas/metabolismo , Aminas/química , Biocatálisis , Cinética , Estructura Molecular , Talidomida/química , Talidomida/metabolismo , Transaminasas/química , Vibrio/enzimología
12.
J Med Chem ; 64(11): 7296-7311, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34042448

RESUMEN

Whereas the PROTAC approach to target protein degradation greatly benefits from rational design, the discovery of small-molecule degraders relies mostly on phenotypic screening and retrospective target identification efforts. Here, we describe the design, synthesis, and screening of a large diverse library of thalidomide analogues against a panel of patient-derived leukemia and medulloblastoma cell lines. These efforts led to the discovery of potent and novel GSPT1/2 degraders displaying selectivity over classical IMiD neosubstrates, such as IKZF1/3, and high oral bioavailability in mice. Taken together, this study offers compound 6 (SJ6986) as a valuable chemical probe for studying the role of GSPT1/2 in vitro and in vivo, and it supports the utility of a diverse library of CRBN binders in the pursuit of targeting undruggable oncoproteins.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Factores de Terminación de Péptidos/metabolismo , Proteolisis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Ubiquitina-Proteína Ligasas/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Administración Oral , Animales , Sitios de Unión , Línea Celular Tumoral , Semivida , Humanos , Factor de Transcripción Ikaros/metabolismo , Ratones , Simulación de Dinámica Molecular , Estudios Retrospectivos , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Relación Estructura-Actividad , Talidomida/administración & dosificación , Talidomida/análogos & derivados , Talidomida/metabolismo , Talidomida/farmacología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
13.
Nat Chem Biol ; 16(11): 1208-1217, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32958952

RESUMEN

The immunomodulatory drug (IMiD) thalidomide and its derivatives lenalidomide and pomalidomide are therapeutic agents used in the treatment of multiple myeloma. Although pomalidomide offers considerable clinical benefits to patients with lenalidomide-resistant multiple myeloma, the molecular mechanisms underlying its superior efficacy remain unclear. Here we show that ARID2, a component of the polybromo-associated BAF (PBAF) chromatin-remodeling complex, is a pomalidomide-induced neosubstrate of CRL4CRBN. BRD7, another subunit of PBAF, is critical for pomalidomide-induced ARID2 degradation. ARID2 is involved in transcriptional regulation of pomalidomide target genes including MYC. Pomalidomide is more effective than lenalidomide in degrading ARID2 and is capable of inhibiting MYC expression and proliferation in lenalidomide-resistant cell lines. Notably, ARID2 expression is associated with a poor prognosis and is higher in chemoresistant minimal residual disease (MRD) populations, and in patients with relapsed/refractory multiple myeloma. These findings suggest that ARID2 is a promising target for overcoming lenalidomide resistance in patients with multiple myeloma.


Asunto(s)
Antineoplásicos/farmacología , Mieloma Múltiple/metabolismo , Talidomida/análogos & derivados , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Antineoplásicos/metabolismo , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Lenalidomida/farmacología , Mieloma Múltiple/tratamiento farmacológico , Mutación , Unión Proteica , Proteolisis/efectos de los fármacos , ARN Mensajero , ARN Interferente Pequeño , Talidomida/metabolismo , Talidomida/farmacología , Factores de Tiempo , Factores de Transcripción/genética , Ubiquitinación
14.
Commun Biol ; 3(1): 515, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32948804

RESUMEN

Regulating the amount of proteins in living cells is a powerful approach for understanding the functions of the proteins. Immunomodulatory drugs (IMiDs) induce the degradation of neosubstrates by interacting with celebron (CRBN) in the cullin E3 ubiquitin ligase complex (CRL4CRBN). Here, we developed the IMiD-dependent Sal-like protein 4 (SALL4) degron (S4D) system for chemical protein knockdown. In transient assays, an N- or C-terminal S4D tag induced the degradation of proteins localized to various subcellular compartments, including the plasma membrane. The activity of luciferase-S4D was reduced by 90% within 3 h of IMiD treatment. IMiD treatment reduced the expression of endogenous S4D-fused RelA and IκBα in knock-in (KI) experiments. Interestingly, the IκBα knockdown suggested that there may be another, unknown mechanism for RelA translocation to the nucleus. Furthermore, 5-hydroxythalidomide as a thalidomide metabolite specifically degradated S4D-tagged protein. These results indicate that the S4D system is a useful tool for cellular biology.


Asunto(s)
Factores Inmunológicos/genética , Proteolisis , Talidomida/metabolismo , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética , Membrana Celular/genética , Membrana Celular/inmunología , Técnicas de Silenciamiento del Gen/métodos , Células HeLa , Humanos , Factores Inmunológicos/inmunología , Factores Inmunológicos/farmacología , Especificidad por Sustrato , Talidomida/análogos & derivados , Talidomida/inmunología , Talidomida/farmacología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología , Factores de Transcripción/inmunología , Factores de Elongación Transcripcional/genética , Ubiquitina-Proteína Ligasas/inmunología
15.
ACS Chem Biol ; 15(10): 2722-2730, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-32865967

RESUMEN

Cereblon (CRBN) is an E3 ligase adapter protein that can be reprogrammed by imide-class compounds such as thalidomide, lenalidomide, and pomalidomide to induce the degradation of neo-substrate proteins. In order to identify additional small molecule CRBN modulators, we implemented a focused combinatorial library approach where we fused an imide-based CRBN-binding pharmacophore to a heterocyclic scaffold, which could be further elaborated. We screened the library for CRBN-dependent antiproliferative activity in the multiple myeloma cell line MM1.S and identified five hit compounds. Quantitative chemical proteomics of hit compounds revealed that they induced selective degradation of GSPT1, a translation termination factor that is currently being explored as a therapeutic target for the treatment of acute myeloid leukemia. Molecular docking studies with CRBN and GSPT1 followed by analogue synthesis identified a possible hydrogen bond interaction with the central pyrimidine ring as a molecular determinant of hit compounds' selectivity. This study demonstrates that a focused combinatorial library design, phenotypic screening, and chemical proteomics can provide a suitable workflow to efficiently identify novel CRBN modulators.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Terminación de Péptidos/metabolismo , Proteolisis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Talidomida/análogos & derivados , Talidomida/farmacología , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Enlace de Hidrógeno , Simulación del Acoplamiento Molecular , Factores de Terminación de Péptidos/química , Unión Proteica , Bibliotecas de Moléculas Pequeñas/metabolismo , Talidomida/metabolismo , Ubiquitina-Proteína Ligasas/química
16.
Ann Clin Lab Sci ; 50(4): 474-480, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32826244

RESUMEN

OBJECTIVE: Epidermal growth factor receptor (EGFR) is one of the important targets in cancer treatment. However, EGFR inhibitors are reported to be ineffective in treating glioblastoma (GBM). In this study, we evaluated the potential mechanism of GBM resistance to EGFR inhibition. METHODS: EGFR, p38, extracellular signal-related kinase (ERK), and c-Jun N-terminal kinase (JNK) expression levels were detected by western blotting. Cell viability was evaluated by the MTT assay. Tumor necrosis factor (TNF) mRNA expression was assessed by qRT-PCR. TNF-α expression was detected by ELISA. The combined effect of EGFR inhibitor (afatinib) and TNF inhibitor (pomalidomide) was evaluated in xenograft athymic mouse model of GBM. RESULTS: Upon blocking TNF, GBM sensitivity to EGFR inhibitors was observed to recover. The combination of afatinib and pomalidomide was found to effectively inhibit cell growth of EG-FR-expressing GBM. In addition, the p38/JNK/ERK pathway was activated following EGFR inhibition. CONCLUSIONS: We demonstrated that GBM resistance to EGFR inhibition was mediated by the activation of TNF. The combination of EGFR inhibitor and TNF inhibitor may have potential clinical implication in treating patients with EGFR-positive GBM.


Asunto(s)
Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Talidomida/análogos & derivados , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , China , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Desnudos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Talidomida/metabolismo , Talidomida/farmacología , Inhibidores del Factor de Necrosis Tumoral/metabolismo , Inhibidores del Factor de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
17.
Chembiochem ; 21(20): 2916-2920, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32501609

RESUMEN

Novel chemical biology probes linking a serine hydrolase-directed fluorophosphonate warhead and cereblon-binding pomalidomide were assessed for the degradation of serine hydrolases. A quantitative proteomics approach to detect degraded proteins revealed that, despite the engagement of ∼40 serine hydrolases, degradation was achieved for only a single serine hydrolase, lysophospholipase II (LYPLA2).


Asunto(s)
Colorantes Fluorescentes/química , Hidrolasas/análisis , Fosfatos/química , Proteómica , Serina/análisis , Talidomida/análogos & derivados , Colorantes Fluorescentes/metabolismo , Hidrolasas/metabolismo , Estructura Molecular , Fosfatos/metabolismo , Serina/metabolismo , Talidomida/química , Talidomida/metabolismo
18.
Cell Chem Biol ; 27(7): 866-876.e8, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32413286

RESUMEN

Proteolysis-targeting chimeras (PROTACs) is a paradigm shift for small-molecule drug discovery. However, limited E3 ubiquitin ligase ligands with cellular activity are available. In vitro binding assays involve the expression and purification of a large amount of proteins and they often yield ligands that are inactive in cell-based assays due to poor cell permeability, stability, and other reasons. Herein, we report the development of a practical and efficient cell-based target engagement assay to evaluate the binding affinity of a small library of cereblon ligands to its E3 ligase in cells. Selected cell-permeable E3 ligase ligands derived from this assay are then used to construct HDAC6 degraders with cellular protein degradation activity. Because the assay does not involve any genetic engineering, it is relatively easy to transfer from one cell type to a different one.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Histona Desacetilasa 6/metabolismo , Ligandos , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Línea Celular , Histona Desacetilasa 6/antagonistas & inhibidores , Humanos , Lenalidomida/química , Lenalidomida/metabolismo , Unión Proteica , Proteolisis , Talidomida/análogos & derivados , Talidomida/química , Talidomida/metabolismo , Ubiquitina-Proteína Ligasas/química
19.
Sci Adv ; 6(8): eaay5154, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32128407

RESUMEN

By hijacking endogenous E3 ligase to degrade protein targets via the ubiquitin-proteasome system, PROTACs (PRoteolysis TArgeting Chimeras) provide a new strategy to inhibit protein targets that were regarded as undruggable before. However, the catalytic nature of PROTAC potentially leads to uncontrolled degradation that causes systemic toxicity issues, limiting the application of PROTAC in the clinic. Here, we introduce a light-inducible switch on PROTACs, thereafter termed as opto-PROTAC, to enable the degradation of protein targets in a spatiotemporal manner. By adding a photolabile caging group on pomalidomide as a parental compound and two additional PROTACs, dBET1 and dALK, we demonstrated light-inducible protein degradation. These opto-PROTACs display no activities in the dark, while the restricted degradation can be induced at a specific time and rate by ultraviolet A irradiation. Our approach provides a generalizable platform for the development of light-controlled PROTACs and enables PROTAC to be a precision medicine.


Asunto(s)
Luz , Proteolisis , Quinasa de Linfoma Anaplásico/metabolismo , Células HEK293 , Humanos , Factor de Transcripción Ikaros/metabolismo , Talidomida/análogos & derivados , Talidomida/metabolismo
20.
Sci Rep ; 10(1): 851, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31964914

RESUMEN

The Cereblon-CRL4 complex has been studied predominantly with regards to thalidomide treatment of multiple myeloma. Nevertheless, the role of Cereblon-CRL4 in Thalidomide Embryopathy (TE) is still not understood. Not all embryos exposed to thalidomide develop TE, hence here we evaluate the role of the CRL4-Cereblon complex in TE variability and susceptibility. We sequenced CRBN, DDB1, CUL4A, IKZF1, and IKZF3 in individuals with TE. To better interpret the variants, we suggested a score and a heatmap comprising their regulatory effect. Differential gene expression after thalidomide exposure and conservation of the CRL4-Cereblon protein complex were accessed from public repositories. Results suggest a summation effect of Cereblon variants on pre-axial longitudinal limb anomalies, and heatmap scores identify the CUL4A variant rs138961957 as potentially having an effect on TE susceptibility. CRL4-Cereblon gene expression after thalidomide exposure and CLR4-Cereblon protein conservation does not explain the difference in Thalidomide sensitivity between species. In conclusion, we suggest that CRL4-Cereblon variants act through several regulatory mechanisms, which may influence CRL4-Cereblon complex assembly and its ability to bind thalidomide. Human genetic variability must be addressed not only to further understand the susceptibility to TE, but as a crucial element in therapeutics, including in the development of pharmacogenomics strategies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Desarrollo Embrionario/genética , Enfermedades Fetales/inducido químicamente , Enfermedades Fetales/genética , Predisposición Genética a la Enfermedad/genética , Talidomida/efectos adversos , Ubiquitina-Proteína Ligasas/genética , Deformidades Congénitas de las Extremidades Superiores/inducido químicamente , Deformidades Congénitas de las Extremidades Superiores/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adolescente , Adulto , Niño , Embrión de Mamíferos , Femenino , Expresión Génica , Variación Genética , Humanos , Masculino , Persona de Mediana Edad , Unión Proteica , Talidomida/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...