Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Angiogenesis ; 27(1): 91-103, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37733132

RESUMO

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.


Assuntos
Malformações Arteriovenosas , Células Endoteliais , Humanos , Células Endoteliais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Bevacizumab/farmacologia , Bevacizumab/uso terapêutico , Bevacizumab/metabolismo , Talidomida/metabolismo , Malformações Arteriovenosas/genética , Dor/metabolismo
2.
J Biochem ; 175(5): 507-519, 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38140952

RESUMO

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.


Assuntos
Proteólise , Talidomida , Talidomida/análogos & derivados , Ubiquitina-Proteína Ligases , Talidomida/farmacologia , Talidomida/química , Talidomida/metabolismo , Humanos , Proteólise/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Quimera de Direcionamento de Proteólise
3.
Microb Pathog ; 181: 106168, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37224982

RESUMO

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.


Assuntos
Talaromyces , Talidomida , Animais , Camundongos , Talidomida/farmacologia , Talidomida/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Caspase 1/metabolismo , Piroptose , Macrófagos/metabolismo , Anfotericina B , Citocinas/metabolismo
4.
Inflammopharmacology ; 31(3): 1167-1182, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36966238

RESUMO

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.


Assuntos
Fibrose Pulmonar Idiopática , Imunossupressores , Talidomida , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Pulmão/metabolismo , Talidomida/uso terapêutico , Talidomida/metabolismo , Talidomida/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Imunossupressores/farmacocinética , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico
5.
Commun Biol ; 4(1): 1277, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34764413

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteína com Dedos de Zinco da Leucemia Promielocítica/genética , Talidomida/análogos & derivados , Ubiquitina-Proteína Ligases/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Células HEK293 , Humanos , Proteína com Dedos de Zinco da Leucemia Promielocítica/metabolismo , Alinhamento de Sequência , Talidomida/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
6.
Eur J Med Chem ; 223: 113645, 2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34217059

RESUMO

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.


Assuntos
Desenho de Fármacos , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Ubiquitina-Proteína Ligases/química , Animais , Proliferação de Células/efeitos dos fármacos , Dasatinibe/farmacologia , Proteínas de Fusão bcr-abl/metabolismo , Meia-Vida , Humanos , Células K562 , Lenalidomida/química , Lenalidomida/metabolismo , Ligantes , Camundongos , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteólise , Relação Estrutura-Atividade , Talidomida/análogos & derivados , Talidomida/química , Talidomida/metabolismo , Transplante Heterólogo , Ubiquitina-Proteína Ligases/metabolismo
7.
J Med Chem ; 64(11): 7296-7311, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34042448

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Fatores de Terminação de Peptídeos/metabolismo , Proteólise/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Ubiquitina-Proteína Ligases/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Administração Oral , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Meia-Vida , Humanos , Fator de Transcrição Ikaros/metabolismo , Camundongos , Simulação de Dinâmica Molecular , Estudos Retrospectivos , Bibliotecas de Moléculas Pequenas/administração & dosagem , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Relação Estrutura-Atividade , Talidomida/administração & dosagem , Talidomida/análogos & derivados , Talidomida/metabolismo , Talidomida/farmacologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
8.
ACS Chem Biol ; 15(10): 2722-2730, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-32865967

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , Proteólise/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Talidomida/análogos & derivados , Talidomida/farmacologia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Ligação de Hidrogênio , Simulação de Acoplamento Molecular , Fatores de Terminação de Peptídeos/química , Ligação Proteica , Bibliotecas de Moléculas Pequenas/metabolismo , Talidomida/metabolismo , Ubiquitina-Proteína Ligases/química
9.
Nat Chem Biol ; 16(11): 1208-1217, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32958952

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Mieloma Múltiplo/metabolismo , Talidomida/análogos & derivados , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Lenalidomida/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Mutação , Ligação Proteica , Proteólise/efeitos dos fármacos , RNA Mensageiro , RNA Interferente Pequeno , Talidomida/metabolismo , Talidomida/farmacologia , Fatores de Tempo , Fatores de Transcrição/genética , Ubiquitinação
10.
Commun Biol ; 3(1): 515, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32948804

RESUMO

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.


Assuntos
Fatores Imunológicos/genética , Proteólise , Talidomida/metabolismo , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/genética , Membrana Celular/genética , Membrana Celular/imunologia , Técnicas de Silenciamento de Genes/métodos , Células HeLa , Humanos , Fatores Imunológicos/imunologia , Fatores Imunológicos/farmacologia , Especificidade por Substrato , Talidomida/análogos & derivados , Talidomida/imunologia , Talidomida/farmacologia , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/imunologia , Fatores de Transcrição/imunologia , Fatores de Elongação da Transcrição/genética , Ubiquitina-Proteína Ligases/imunologia
11.
Ann Clin Lab Sci ; 50(4): 474-480, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32826244

RESUMO

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.


Assuntos
Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Talidomida/análogos & derivados , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , China , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Nus , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Talidomida/metabolismo , Talidomida/farmacologia , Inibidores do Fator de Necrose Tumoral/metabolismo , Inibidores do Fator de Necrose Tumoral/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
12.
Sci Adv ; 6(8): eaay5154, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32128407

RESUMO

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.


Assuntos
Luz , Proteólise , Quinase do Linfoma Anaplásico/metabolismo , Células HEK293 , Humanos , Fator de Transcrição Ikaros/metabolismo , Talidomida/análogos & derivados , Talidomida/metabolismo
13.
Sci Rep ; 10(1): 851, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31964914

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Desenvolvimento Embrionário/genética , Doenças Fetais/induzido quimicamente , Doenças Fetais/genética , Predisposição Genética para Doença/genética , Talidomida/efeitos adversos , Ubiquitina-Proteína Ligases/genética , Deformidades Congênitas das Extremidades Superiores/induzido quimicamente , Deformidades Congênitas das Extremidades Superiores/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adolescente , Adulto , Criança , Embrião de Mamíferos , Feminino , Expressão Gênica , Variação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Ligação Proteica , Talidomida/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Adulto Jovem
14.
Chem Commun (Camb) ; 55(98): 14765-14768, 2019 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-31754664

RESUMO

BCL-XL, an anti-apoptotic BCL-2 family protein, plays a key role in cancer cell survival. However, the potential of BCL-XL as an anti-cancer target has been hampered by the on-target platelet toxicity because platelets depend on BCL-XL to maintain their viability. Here we report the development of a PROTAC BCL-XL degrader, XZ424, which has increased selectivity for BCL-XL-dependent MOLT-4 cells over human platelets compared with conventional BCL-XL inhibitors. This proof-of-concept study demonstrates the potential of utilizing a PROTAC approach to achieve tissue selectivity.


Assuntos
Isoquinolinas/química , Talidomida/análogos & derivados , Proteína bcl-X/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/efeitos dos fármacos , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Linhagem Celular Tumoral , Humanos , Isoquinolinas/metabolismo , Isoquinolinas/farmacologia , Ligação Proteica , Talidomida/química , Talidomida/metabolismo , Talidomida/farmacologia , Ubiquitina-Proteína Ligases , Proteína bcl-X/antagonistas & inibidores
15.
Anal Chem ; 90(24): 14287-14293, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30431263

RESUMO

Protein-protein interactions (PPIs) mediate signal transduction in cells. Small molecules that regulate PPIs are important tools for biology and biomedicine. Dynamic imaging of small molecule induced PPIs characterizes and verifies these molecules in living cells. It is thus important to develop cellular assays for dynamic visualization of small molecule induced protein-protein association and dissociation in living cells. Here we have applied a fluorophore phase transition based principle and designed a PPI assay named SPPIER (separation of phases-based protein interaction reporter). SPPIER utilizes the green fluorescent protein (GFP) and is thus genetically encoded. Upon small molecule induced PPI, SPPIER rapidly forms highly fluorescent GFP droplets in living cells. SPPIER detects immunomodulatory drug (IMiD) induced PPI between cereblon and the transcription factor Ikaros. It also detects IMiD analogue (e.g., CC-885) induced PPI between cereblon and GSPT1. Furthermore, SPPIER can visualize bifunctional molecules (e.g. PROTAC)-induced PPI between an E3 ubiquitin ligase and a target protein. Lastly, SPPIER can be modified to image small molecule induced protein-protein dissociation, such as nutlin-induced dissociation between HDM2 and p53. The intense brightness and rapid kinetics of SPPIER enable robust and dynamic visualization of PPIs in living cells.


Assuntos
Fator de Transcrição Ikaros/metabolismo , Peptídeo Hidrolases/metabolismo , Mapas de Interação de Proteínas , Bibliotecas de Moléculas Pequenas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ciclo Celular , Transferência Ressonante de Energia de Fluorescência , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Fator de Transcrição Ikaros/química , Fatores Imunológicos/química , Fatores Imunológicos/metabolismo , Microscopia Confocal , Proteínas Nucleares/genética , Peptídeo Hidrolases/química , Peptídeo Hidrolases/genética , Compostos de Fenilureia/química , Compostos de Fenilureia/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , Bibliotecas de Moléculas Pequenas/química , Talidomida/análogos & derivados , Talidomida/química , Talidomida/metabolismo , Imagem com Lapso de Tempo , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases , Proteína Supressora de Tumor Von Hippel-Lindau/genética
16.
ACS Chem Biol ; 13(11): 3131-3141, 2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30335946

RESUMO

B-cell lymphoma 6 (BCL6) inhibition is a promising mechanism for treating hematological cancers but high quality chemical probes are necessary to evaluate its therapeutic potential. Here we report potent BCL6 inhibitors that demonstrate cellular target engagement and exhibit exquisite selectivity for BCL6 based on mass spectrometry analyses following chemical proteomic pull down. Importantly, a proteolysis-targeting chimera (PROTAC) was also developed and shown to significantly degrade BCL6 in a number of diffuse large B-cell lymphoma (DLBCL) cell lines, but neither BCL6 inhibition nor degradation selectively induced marked phenotypic response. To investigate, we monitored PROTAC directed BCL6 degradation in DLBCL OCI-Ly1 cells by immunofluorescence and discovered a residual BCL6 population. Analysis of subcellular fractions also showed incomplete BCL6 degradation in all fractions despite having measurable PROTAC concentrations, together providing a rationale for the weak antiproliferative response seen with both BCL6 inhibitor and degrader. In summary, we have developed potent and selective BCL6 inhibitors and a BCL6 PROTAC that effectively degraded BCL6, but both modalities failed to induce a significant phenotypic response in DLBCL despite achieving cellular concentrations.


Assuntos
Antineoplásicos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-6/antagonistas & inibidores , Quinolonas/farmacologia , Talidomida/análogos & derivados , Talidomida/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Células HEK293 , Humanos , Ligantes , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Peptídeo Hidrolases/metabolismo , Ligação Proteica , Proteólise , Proteínas Proto-Oncogênicas c-bcl-6/química , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Quinolonas/síntese química , Quinolonas/metabolismo , Talidomida/síntese química , Talidomida/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
17.
J Biol Chem ; 293(16): 6187-6200, 2018 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-29449372

RESUMO

Upon binding to thalidomide and other immunomodulatory drugs, the E3 ligase substrate receptor cereblon (CRBN) promotes proteosomal destruction by engaging the DDB1-CUL4A-Roc1-RBX1 E3 ubiquitin ligase in human cells but not in mouse cells, suggesting that sequence variations in CRBN may cause its inactivation. Therapeutically, CRBN engagers have the potential for broad applications in cancer and immune therapy by specifically reducing protein expression through targeted ubiquitin-mediated degradation. To examine the effects of defined sequence changes on CRBN's activity, we performed a comprehensive study using complementary theoretical, biophysical, and biological assays aimed at understanding CRBN's nonprimate sequence variations. With a series of recombinant thalidomide-binding domain (TBD) proteins, we show that CRBN sequence variants retain their drug-binding properties to both classical immunomodulatory drugs and dBET1, a chemical compound and targeting ligand designed to degrade bromodomain-containing 4 (BRD4) via a CRBN-dependent mechanism. We further show that dBET1 stimulates CRBN's E3 ubiquitin-conjugating function and degrades BRD4 in both mouse and human cells. This insight paves the way for studies of CRBN-dependent proteasome-targeting molecules in nonprimate models and provides a new understanding of CRBN's substrate-recruiting function.


Assuntos
Proteínas Culina/metabolismo , Peptídeo Hidrolases/química , Peptídeo Hidrolases/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Azepinas/farmacologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Sequência Conservada , Humanos , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Lenalidomida/farmacologia , Ligantes , Camundongos , Sondas Moleculares , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Linfócitos T/metabolismo , Talidomida/análogos & derivados , Talidomida/metabolismo , Talidomida/farmacologia , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Triazóis/farmacologia , Ubiquitina/metabolismo
18.
Chem Res Toxicol ; 30(8): 1622-1628, 2017 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-28745489

RESUMO

Thalidomide [α-(N-phthalimido)glutarimide] (1) is a sedative and antiemetic drug originally introduced into the clinic in the 1950s for the treatment of morning sickness. Although marketed as entirely safe, more than 10 000 babies were born with severe birth defects. Thalidomide was banned and subsequently approved for the treatment of multiple myeloma and complications associated with leprosy. Although known for more than 5 decades, the mechanism of teratogenicity remains to be conclusively understood. Various theories have been proposed in the literature including DNA damage and ROS and inhibition of angiogenesis and cereblon. All of the theories have their merits and limitations. Although the recently proposed cereblon theory has gained wide acceptance, it fails to explain the metabolism and low-dose requirement reported by a number of groups. Recently, we have provided convincing structural evidence in support of the presence of arene oxide and the quinone-reactive intermediates. However, the ability of these reactive intermediates to impart toxicity/teratogenicity needs investigation. Herein we report that the oxidative metabolite of thalidomide, dihydroxythalidomide, is responsible for generating ROS and causing DNA damage. We show, using cell lines, the formation of comet (DNA damage) and ROS. Using DNA-cleavage assays, we also show that catalase, radical scavengers, and desferal are capable of inhibiting DNA damage. A mechanism of teratogenicity is proposed that not only explains the DNA-damaging property but also the metabolism, low concentration, and species-specificity requirements of thalidomide.


Assuntos
Dano ao DNA/efeitos dos fármacos , Talidomida/toxicidade , Catalase/metabolismo , Clivagem do DNA , Sequestradores de Radicais Livres/química , Células HEK293 , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana , Humanos , Microscopia de Fluorescência , Plasmídeos/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo , Teratogênicos/química , Teratogênicos/metabolismo , Teratogênicos/toxicidade , Talidomida/química , Talidomida/metabolismo
19.
Math Med Biol ; 34(1): 1-13, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-26443812

RESUMO

278: The Cytochrome P450 (CYP) system is involved in 90% of the human body's interactions with xenobiotics and due to this, it has become an area of avid research including the creation of transgenic mice. This paper proposes a three-compartment model which is used to explain the drug metabolism in the Hepatic Reductase Null (HRN) mouse developed by the University of Dundee (Henderson, C. J., Otto, D. M. E., Carrie, D., Magnuson, M. A., McLaren, A. W., Rosewell, I. and Wolf, C. R. (2003) Inactivation of the hepatic cytochrome p450 system by conditional deletion of hepatic cytochrome p450 reductase. J. Biol. Chem. , 13480-13486). The model is compared with a two-compartment model using experimental data from studies using wild-type and HRN mice. This comparison allowed for metabolic differences between the two types of mice to be isolated. The three sets of drug data (Gefitinib, Midazolam and Thalidomide) showed that the transgenic mouse has a decreased rate of metabolism.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Fígado/enzimologia , Modelos Biológicos , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Inibidores da Angiogênese/metabolismo , Animais , Modelos Animais de Doenças , Moduladores GABAérgicos/metabolismo , Gefitinibe , Camundongos , Camundongos Transgênicos/metabolismo , Midazolam/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Quinazolinas/metabolismo , Talidomida/metabolismo
20.
Clin Exp Med ; 17(2): 193-207, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-26961690

RESUMO

We have previously found that ex vivo expanded human CD4+CD25+Treg cells suppress proliferation of lymphoma B-cell lines. Here we demonstrate that the immunomodulatory drug lenalidomide potentiates suppression of lymphoma B-cell proliferation by freshly isolated CD4+CD25+Tregs, as well as suppression by Tregs expanded polyclonally in the presence of rapamycin from CD4+CD25+T cells or CD4+CD25+CD127loT cells. The regulation of lymphoma cell proliferation by Tregs pre-expanded with "third-party" allogeneic MoDCs in the presence of rapamycin was also potentiated by lenalidomide. Lenalidomide contributed to the suppression exerted by Tregs despite concomitant downregulation of Treg proliferation. Lenalidomide did not reduce the suppression of conventional T cells by expanded Tregs. The exposure of polyclonally expanded Tregs to lenalidomide did not significantly alter their phenotype. There was no uniform pattern of lenalidomide effect on Treg-mediated regulation of lymphoma B cells freshly isolated from patients. Freshly isolated lymphoma cells activated with multimeric CD40L and IL-4 to support their survival in vitro varied in their sensitivity to lenalidomide, and the regulatory effect of Tregs on such lymphoma cells ranged from suppression to help in individual patients. Lenalidomide potentiated or attenuated Treg effects on the survival of freshly isolated lymphoma cells. A combination of lenalidomide treatment with adoptive transfer of CD4+CD25+Tregs or CD4+CD25+CD127loTregs expanded ex vivo could be used to suppress proliferation of residual lymphoma in select patients with lymphoma responsive to the regulation by Tregs and sensitive to lenalidomide.


Assuntos
Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Proliferação de Células , Fatores Imunológicos/metabolismo , Linfoma/imunologia , Linfócitos T Reguladores/imunologia , Talidomida/análogos & derivados , Adulto , Idoso , Linfócitos B/fisiologia , Doadores de Sangue , Antígenos CD4/análise , Células Cultivadas , Feminino , Humanos , Subunidade alfa de Receptor de Interleucina-2/análise , Lenalidomida , Linfoma/patologia , Masculino , Pessoa de Meia-Idade , Sirolimo/metabolismo , Linfócitos T Reguladores/química , Talidomida/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA