Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Molecules ; 29(7)2024 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-38611729

RESUMO

Royal jelly (RJ) is recognized as beneficial to mammalian health. Multilineage differentiation potential is an important property of mesenchymal stem cells (MSCs). C2C12 cells have an innate ability to differentiate into myogenic cells. Like MSCs, C2C12 cells can also differentiate into osteoblast- and adipocyte-lineage cells. We recently reported that RJ enhances the myogenic differentiation of C2C12 cells. However, the effect of RJ on osteoblast or adipocyte differentiation is still unknown. Here in this study, we have examined the effect of RJ on the osteoblast and adipocyte differentiation of C2C12 cells. Protease-treated RJ was used to reduce the adverse effects caused by RJ supplementation. To induce osteoblast or adipocyte differentiation, cells were treated with bone morphogenetic proteins (BMP) or peroxisome proliferator-activated receptor γ (PPARγ) agonist, respectively. RNA-seq was used to analyze the effect of RJ on gene expression. We found that RJ stimulates osteoblast and adipocyte differentiation. RJ regulated 279 genes. RJ treatment upregulated glutathione-related genes. Glutathione, the most abundant antioxidative factor in cells, has been shown to promote osteoblast differentiation in MSC and MSC-like cells. Therefore, RJ may promote osteogenesis, at least in part, through the antioxidant effects of glutathione. RJ enhances the differentiation ability of C2C12 cells into multiple lineages, including myoblasts, osteoblasts, and adipocytes.


Assuntos
Antioxidantes , Ácidos Graxos , Animais , Diferenciação Celular , Glutationa , Mioblastos , Mamíferos
2.
Ann Rheum Dis ; 82(9): 1153-1161, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37400117

RESUMO

OBJECTIVES: Recent studies demonstrate that extracellular-released aminoacyl-tRNA synthetases (aaRSs) play unique roles in immune responses and diseases. This study aimed to understand the role of extracellular aaRSs in the pathogenesis of rheumatoid arthritis (RA). METHODS: Primary macrophages and fibroblast-like synoviocytes were cultured with aaRSs. aaRS-induced cytokine production including IL-6 and TNF-α was detected by ELISA. Transcriptomic features of aaRS-stimulated macrophages were examined using RNA-sequencing. Serum and synovial fluid (SF) aaRS levels in patients with RA were assessed using ELISA. Peptidyl arginine deiminase (PAD) 4 release from macrophages stimulated with aaRSs was detected by ELISA. Citrullination of aaRSs by themselves was examined by immunoprecipitation and western blotting. Furthermore, aaRS inhibitory peptides were used for inhibition of arthritis in two mouse RA models, collagen-induced arthritis and collagen antibody-induced arthritis. RESULTS: All 20 aaRSs functioned as alarmin; they induced pro-inflammatory cytokines through the CD14-MD2-TLR4 axis. Stimulation of macrophages with aaRSs displayed persistent innate inflammatory responses. Serum and SF levels of many aaRSs increased in patients with RA compared with control subjects. Furthermore, aaRSs released PAD4 from living macrophages, leading to their citrullination. We demonstrate that aaRS inhibitory peptides suppress cytokine production and PAD4 release by aaRSs and alleviate arthritic symptoms in a mouse RA model. CONCLUSIONS: Our findings uncovered the significant role of aaRSs as a novel alarmin in RA pathogenesis, indicating that their blocking agents are potent antirheumatic drugs.


Assuntos
Artrite Experimental , Artrite Reumatoide , Animais , Camundongos , Alarminas , Células Cultivadas , Citocinas , Modelos Animais de Doenças , Fibroblastos/patologia , Inflamação , Líquido Sinovial , Humanos
3.
Genomics ; 114(5): 110477, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36058475

RESUMO

Fish exhibit different muscle structures and growth characteristics compared with mammals. We used a spatial transcriptomics approach and examined myotomal muscle sections from zebrafish. Adult muscles were divided into eight regions according to spatial gene expression characteristics. Slow muscle was located in the wedge-shaped region near the lateral line and at the base of the dorsal fin, intermediate muscle was located in a ribbon-shaped region adjacent to slow muscle, and fast muscle was located in the deep region of the trunk, surrounded by intermediate muscle; the interior of fast muscle was further divided into 6 parts by their transcriptomic features. Combined analysis of adult and larval data revealed that adult muscles contain specific regions similar to larval muscles. These regions showed active myogenesis and a high expression of genes associated with muscle hyperplasia. This is the first study to apply spatial transcriptomics to fish myotomal muscle structure and growth.


Assuntos
Transcriptoma , Peixe-Zebra , Animais , Larva , Mamíferos , Desenvolvimento Muscular/genética , Músculos , Peixe-Zebra/genética
4.
Chem Soc Rev ; 51(15): 6234-6250, 2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35796627

RESUMO

Progress in strategies aimed at breaking down therapeutic target proteins has led to a paradigm shift in drug discovery. Thalidomide and its derivatives are the only protein degraders currently used in clinical practice. Our understanding of the molecular mechanism of action of thalidomide and its derivatives has advanced dramatically since the identification of cereblon (CRBN) as their direct target. The binding of thalidomide derivatives to CRBN, a substrate recognition receptor for Cullin 4 RING E3 ubiquitin ligase (CRL4), induces the recruitment of non-native substrates to CRL4CRBN and their subsequent degradation. This discovery was a breakthrough in the current rapid development of protein-degrading agents because clarification of the mechanism of action of thalidomide derivatives has demonstrated the clinical value of these compounds. This review provides an overview of the mechanism of action of thalidomide and its derivatives and describes perspectives for protein degraders.


Assuntos
Peptídeo Hidrolases , Talidomida , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Peptídeo Hidrolases/metabolismo , Talidomida/química , Talidomida/farmacologia , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
5.
Blood ; 135(24): 2146-2158, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32219443

RESUMO

Immunomodulatory drugs (IMiDs) are key agents for the treatment of multiple myeloma and myelodysplastic syndrome with chromosome 5q deletion. IMiDs exert their pleiotropic effects through the recruitment of neosubstrates to cereblon, a substrate receptor of the E3 ubiquitin ligase complex; therefore, identification of cell-specific neosubstrates is important to understand the effects of IMiDs. In clinical practice, IMiDs induce thrombocytopenia, which frequently results in the discontinuation of IMiD treatment. In the current study, we sought to identify the molecular mechanism underlying thrombocytopenia induced by IMiD treatment. We found that IMiDs strongly impaired proplatelet formation, a critical step in functional platelet production, through the inhibition of autocrine estradiol signaling in human megakaryocytes. Furthermore, we identified aromatase, an indispensable enzyme for estradiol biosynthesis, as a novel neosubstrate of cereblon. IMiDs promoted the recruitment of aromatase to cereblon, resulting in the degradation of aromatase in a proteasome-dependent manner. Finally, aromatase was significantly degraded in the bone marrow of patients with multiple myeloma who developed thrombocytopenia with IMiD treatment. These data suggest that aromatase is a neosubstrate of cereblon that is responsible for IMiD-induced thrombocytopenia.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aromatase/metabolismo , Fatores Imunológicos/efeitos adversos , Trombocitopenia/induzido quimicamente , Trombocitopenia/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Idoso , Idoso de 80 Anos ou mais , Feminino , Células HEK293 , Humanos , Células K562 , Masculino , Megacariócitos/efeitos dos fármacos , Megacariócitos/patologia , Pessoa de Meia-Idade , Mieloma Múltiplo/tratamento farmacológico , Síndromes Mielodisplásicas/tratamento farmacológico , Mielopoese/efeitos dos fármacos , Mielopoese/fisiologia , Especificidade por Substrato , Trombocitopenia/patologia
6.
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
7.
Nature ; 535(7611): 252-7, 2016 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-27338790

RESUMO

Immunomodulatory drugs bind to cereblon (CRBN) to confer differentiated substrate specificity on the CRL4(CRBN) E3 ubiquitin ligase. Here we report the identification of a new cereblon modulator, CC-885, with potent anti-tumour activity. The anti-tumour activity of CC-885 is mediated through the cereblon-dependent ubiquitination and degradation of the translation termination factor GSPT1. Patient-derived acute myeloid leukaemia tumour cells exhibit high sensitivity to CC-885, indicating the clinical potential of this mechanism. Crystallographic studies of the CRBN-DDB1-CC-885-GSPT1 complex reveal that GSPT1 binds to cereblon through a surface turn containing a glycine residue at a key position, interacting with both CC-885 and a 'hotspot' on the cereblon surface. Although GSPT1 possesses no obvious structural, sequence or functional homology to previously known cereblon substrates, mutational analysis and modelling indicate that the cereblon substrate Ikaros uses a similar structural feature to bind cereblon, suggesting a common motif for substrate recruitment. These findings define a structural degron underlying cereblon 'neosubstrate' selectivity, and identify an anti-tumour target rendered druggable by cereblon modulation.


Assuntos
Antineoplásicos/farmacologia , Peptídeo Hidrolases/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , Compostos de Fenilureia/farmacologia , Talidomida/análogos & derivados , Proteínas Adaptadoras de Transdução de Sinal , Motivos de Aminoácidos , Antineoplásicos/química , Sítios de Ligação , Cristalografia por Raios X , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Humanos , Fator de Transcrição Ikaros/química , Fator de Transcrição Ikaros/metabolismo , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Peptídeo Hidrolases/química , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/deficiência , Compostos de Fenilureia/química , Ligação Proteica , Proteólise/efeitos dos fármacos , Especificidade por Substrato , Talidomida/química , Talidomida/farmacologia , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
8.
Rinsho Ketsueki ; 63(6): 573-579, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-35831190

RESUMO

Thalidomide was developed as a sedative drug during the 1950s. Unfortunately, it has serious teratogenic properties. When pregnant women ingested thalidomide, their infants developed serious malformations such as short limbs. However, thalidomide is now recognized as a clinically useful drug, with several countries approving it as an anti-myeloma treatment. Although the direct target of thalidomide was largely debated until recently, our groups discovered cereblon (CRBN), a substrate receptor of an E3 ubiquitin ligase as a primary target of thalidomide in 2010. CRBN binds not only to thalidomide, but also to various thalidomide derivatives such as lenalidomide and pomalidomide, as well as compounds containing a thalidomide moiety. These compounds are known as cereblon modulators, which induced specific neosubstrates of CRBN E3 ubiquitin ligase such as Ikaros and Aiolos. Several groups have now joined the CRBN research and have reported the basic mechanism of CRBN and its binding compounds. In this review, we present our findings as well as recent advances in this subject area.


Assuntos
Peptídeo Hidrolases , Talidomida , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Feminino , Humanos , Lenalidomida/uso terapêutico , Peptídeo Hidrolases/metabolismo , Gravidez , Talidomida/farmacologia , Talidomida/uso terapêutico , Ubiquitina-Proteína Ligases
9.
Nat Chem Biol ; 15(11): 1077-1084, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31591562

RESUMO

Cereblon (CRBN) is a primary target of thalidomide and mediates its multiple pharmacological activities, including teratogenic and antimyeloma activities. CRBN functions as a substrate receptor of the E3 ubiquitin ligase CRL4, whose substrate specificity is modulated by thalidomide and its analogs. Although a number of CRL4CRBN substrates have recently been identified, the substrate involved in thalidomide teratogenicity is unclear. Here we show that p63 isoforms are thalidomide-dependent CRL4CRBN neosubstrates that are responsible, at least in part, for its teratogenic effects. The p53 family member p63 is associated with multiple developmental processes. ∆Np63α is essential for limb development, while TAp63α is important for cochlea development and hearing. Using a zebrafish model, we demonstrate that thalidomide exerts its teratogenic effects on pectoral fins and otic vesicles by inducing the degradation of ∆Np63α and TAp63α, respectively. These results may contribute to the invention of new thalidomide analogs lacking teratogenic activity.


Assuntos
Proteínas de Membrana/metabolismo , Teratogênicos/toxicidade , Talidomida/toxicidade , Células HEK293 , Humanos , Especificidade por Substrato
10.
J Pharmacol Sci ; 147(2): 192-199, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34384567

RESUMO

In a previous study, we suggested that the Hsp90 inhibitor 17-AAG prevents cardiac dysfunction in the failing heart following myocardial infarction in rats. Although it is assumed that the RIP1/RIP3/MLKL necroptotic pathway, which comprises client proteins for Hsp90, is involved; however, the relationship between the cardioprotective effects of 17-AAG and the activity of the cardiac RIP1/RIP3/MLKL necrosome-associated proteins in the failing heart following myocardial infarction remained unclear. Therefore, the levels of phosphorylated MLKL after myocardial infarction with or without Hsp90 inhibitor treatment were measured. Myocardial infarction was induced by ligation of the coronary artery (CAL) in Wistar rats. 17-AAG was injected from the 2nd to the 8th week after myocardial infarction. The administration of 17-AAG attenuated the cardiac dysfunction, hypertrophy, and fibrosis at the 8th week after CAL, simultaneously lessening the increases in the expression and phosphorylation levels of RIP1, RIP3, and MLKL in the area of the left ventricular muscle without infarct. These results indicate that the activation of the RIP1/RIP3/MLKL pathway is a common event in the development of chronic heart failure. Furthermore, our findings suggest that the effects of 17-AAG treatment on the improvement of cardiac function in rats after myocardial infarction is related to the attenuation of this RIP1/RIP3/MLKL pathway.


Assuntos
Benzoquinonas/farmacologia , Cardiotônicos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/prevenção & controle , Lactamas Macrocíclicas/farmacologia , Infarto do Miocárdio/complicações , Proteínas Quinases/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Benzoquinonas/administração & dosagem , Insuficiência Cardíaca/etiologia , Injeções , Lactamas Macrocíclicas/administração & dosagem , Masculino , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Transdução de Sinais/genética
11.
Proc Jpn Acad Ser B Phys Biol Sci ; 96(6): 189-203, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32522938

RESUMO

Thalidomide, originally developed as a sedative drug, causes multiple defects due to severe teratogenicity, but it has been re-purposed for treating multiple myeloma, and derivatives such as lenalidomide and pomalidomide have been developed for treating blood cancers. Although the molecular mechanisms of thalidomide and its derivatives remained poorly understood until recently, we identified cereblon (CRBN), a primary direct target of thalidomide, using ferrite glycidyl methacrylate (FG) beads. CRBN is a ligand-dependent substrate receptor of the E3 ubiquitin ligase complex cullin-RING ligase 4 (CRL4CRBN). When a ligand such as thalidomide binds to CRBN, it recognizes various 'neosubstrates' depending on the shape of the ligand. CRL4CRBN binds many neosubstrates in the presence of various ligands. CRBN has been utilized in a novel protein knockdown technology named proteolysis targeting chimeras (PROTACs). Heterobifunctional molecules such as dBET1 are being developed to specifically degrade proteins of interest. Herein, we review recent advances in CRBN research.


Assuntos
Talidomida/química , Animais , Reposicionamento de Medicamentos , Humanos , Terapia de Alvo Molecular , Talidomida/efeitos adversos , Talidomida/farmacologia
12.
Rinsho Ketsueki ; 60(9): 1013-1019, 2019.
Artigo em Japonês | MEDLINE | ID: mdl-31597822

RESUMO

Various derivatives of thalidomide, a drug that is well-known for its teratogenicity, have recently been developed; among them, lenalidomide and pomalidomide, known as immunomodulatory drugs (IMiDs), have potent anticancer activity. These drugs have been approved by Food and Drug Administration for the treatment of several diseases, including multiple myeloma, under strict control. The primary direct target protein of thalidomide and IMiDs is cereblon (CRBN), a substrate receptor of Cullin-RING ligase 4 (CRL4). CRL4CRBN is a unique E3 ubiquitin ligase because its substrate selectivity is altered by ligands such as IMiDs. Each IMiD induces degradation of neosubstrates, such as Ikaros or CK1a. Because the activity of new CRBN-binding compounds is not limited to immunomodulation, the designation of "cereblon modulators" has been proposed for these small CRBN-binding compounds. Currently, researchers are investigating CC-122 (avadomide) and CC-220 (iberdomide) for the treatment of diffuse large B-cell lymphoma and systemic lupus erythematosus, respectively. Other recent studies have been investigating heterobifunctional molecules called proteolysis-targeting chimeras (PROTACs) for protein of interest degradation. Moreover, several proteins, such as BRD4, CDK9, or Tau, have already been successfully degraded by CRBN-based PROTACs. In this review, recent advances in CRBN and its binding compounds have been discussed.


Assuntos
Imunomodulação , Peptídeo Hidrolases/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Morfolinas , Ftalimidas , Piperidonas , Inibidores de Proteases , Especificidade por Substrato , Ubiquitina-Proteína Ligases/antagonistas & inibidores
13.
Rinsho Ketsueki ; 58(10): 2067-2073, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-28978850

RESUMO

Immunomodulatory drugs (IMiDs) are a new class of anticancer compounds that are derived from thalidomide. Lenalidomide and pomalidomide are well-known IMiDs, and they have already been approved by FDA for the treatment of several diseases, including multiple myeloma. Cereblon (CRBN) is a common primary target for IMiDs. It works as a substrate receptor of CRL4. Accumulating evidence has shown that the substrate specificity of CRL4CRBN is altered by ligands such as IMiDs. Recently, novel CRBN-binding compounds have been developed and are called "cereblon modulators". Among these, CC-122 and CC-220 are currently under clinical development for the treatment of diffuse large B-cell lymphoma and systemic lupus erythematosus, respectively. Another new cereblon modulator CC-885 is shown to induce degradation of the translation termination factor GSPT1, resulting in an antiproliferative effect in acute myeloid leukemia. Structural analyses have revealed that CC-885 provides an interaction hotspot between CRBN and GSPT1. On the other hand, several groups have been investigating linker-based approaches for targeted protein degradation via CRBN. Several proteins, such as BRD4 and BCR-ABL, have been successfully degraded by CRL4CRBN using these technologies. In this review, we introduce recent topics in CRBN and its binding compounds.


Assuntos
Fatores Imunológicos/uso terapêutico , Terapia de Alvo Molecular , Peptídeo Hidrolases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sítios de Ligação , Desenho de Fármacos , Humanos , Fatores Imunológicos/química , Ligantes , Peptídeo Hidrolases/química , Especificidade por Substrato , Ubiquitina-Proteína Ligases
15.
Rinsho Ketsueki ; 57(5): 556-62, 2016 05.
Artigo em Japonês | MEDLINE | ID: mdl-27263779

RESUMO

Half a century ago, the sedative thalidomide caused a serious drug disaster because of its teratogenicity and was withdrawn from the market. However, thalidomide, which has returned to the market, is now used for the treatment of leprosy and multiple myeloma (MM) under strict control. The mechanism of thalidomide action had been a long-standing question. We developed a new affinity bead technology and identified cereblon (CRBN) as a thalidomide-binding protein. We found that CRBN functions as a substrate receptor of an E3 cullin-Ring ligase complex 4 (CRL4) and is a primary target of thalidomide teratogenicity. Recently, new thalidomide derivatives, called immunomodulatory drugs (IMiDs), have been developed by Celgene. Among them, lenalidomide (Len) and pomalidomide (Pom) were shown to exert strong therapeutic effects against MM. It was found that Len and Pom both bind CRBN-CRL4 and recruit neomorphic substrates (Ikaros and Aiolos). More recently it was reported that casein kinase 1a (Ck1a) was identified as a substrate for CRBN-CRL4 in the presence of Len, but not Pom. Ck1a breakdown explains why Len is specifically effective for myelodysplastic syndrome with 5q deletion. It is now proposed that binding of IMiDs to CRBN appears to alter the substrate specificity of CRBN-CRL4. In this review, we introduce recent findings on IMiDs.


Assuntos
Imunossupressores/uso terapêutico , Descoberta de Drogas , Humanos , Imunossupressores/química , Terapia de Alvo Molecular , Mieloma Múltiplo/tratamento farmacológico , Especificidade por Substrato , Ubiquitina-Proteína Ligases/metabolismo
16.
Nihon Rinsho ; 73(1): 143-8, 2015 Jan.
Artigo em Japonês | MEDLINE | ID: mdl-25626320

RESUMO

Half a century ago, thalidomide was developed as a sedative drug and was wildly used over 40 countries. However the drug has serious birth defects such as amelia and phocomelia. Now thalidomide is regarded as a clinically effective drug and used for the treatment of multiple myeloma under strict controls. The direct target of thalidomide had been a long-standing question. We identified cereblon as a primary direct target protein for thalidomide teratogenicity using new affinity bead technology in 2010. In this review, we introduce an overview of thalidomide teratogenicity, a story about how we identified cereblon, and recent advances in cereblon studies.


Assuntos
Mieloma Múltiplo/tratamento farmacológico , Teratogênese , Talidomida/uso terapêutico , Proteínas Adaptadoras de Transdução de Sinal , Animais , Humanos , Terapia de Alvo Molecular , Peptídeo Hidrolases/metabolismo , Talidomida/efeitos adversos , Talidomida/química , Ubiquitina-Proteína Ligases
17.
Br J Haematol ; 164(6): 811-21, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24328678

RESUMO

Cereblon (CRBN), the molecular target of lenalidomide and pomalidomide, is a substrate receptor of the cullin ring E3 ubiquitin ligase complex, CRL4(CRBN) . T cell co-stimulation by lenalidomide or pomalidomide is cereblon dependent: however, the CRL4(CRBN) substrates responsible for T cell co-stimulation have yet to be identified. Here we demonstrate that interaction of the transcription factors Ikaros (IKZF1, encoded by the IKZF1 gene) and Aiolos (IKZF3, encoded by the IKZF3 gene) with CRL4(CRBN) is induced by lenalidomide or pomalidomide. Each agent promotes Aiolos and Ikaros binding to CRL4(CRBN) with enhanced ubiquitination leading to cereblon-dependent proteosomal degradation in T lymphocytes. We confirm that Aiolos and Ikaros are transcriptional repressors of interleukin-2 expression. The findings link lenalidomide- or pomalidomide-induced degradation of these transcriptional suppressors to well documented T cell activation. Importantly, Aiolos could serve as a proximal pharmacodynamic marker for lenalidomide and pomalidomide, as healthy human subjects administered lenalidomide demonstrated Aiolos degradation in their peripheral T cells. In conclusion, we present a molecular model in which drug binding to cereblon results in the interaction of Ikaros and Aiolos to CRL4(CRBN) , leading to their ubiquitination, subsequent proteasomal degradation and T cell activation.


Assuntos
Fator de Transcrição Ikaros/metabolismo , Peptídeo Hidrolases/metabolismo , Linfócitos T/efeitos dos fármacos , Talidomida/análogos & derivados , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Inibidores da Angiogênese/farmacologia , Humanos , Fator de Transcrição Ikaros/genética , Fatores Imunológicos/farmacologia , Lenalidomida , Peptídeo Hidrolases/genética , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Linfócitos T/metabolismo , Talidomida/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
18.
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
19.
Blood Adv ; 8(3): 785-796, 2024 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-38163319

RESUMO

ABSTRACT: Immunomodulatory drugs (IMiDs) are key drugs for treating multiple myeloma and myelodysplastic syndrome with chromosome 5q deletion. IMiDs exert their pleiotropic effects through the interaction between cell-specific substrates and cereblon, a substrate receptor of the E3 ubiquitin ligase complex. Thus, identification of cell-specific substrates is important for understanding the effects of IMiDs. IMiDs increase the risk of thromboembolism, which sometimes results in fatal clinical outcomes. In this study, we sought to clarify the molecular mechanisms underlying IMiDs-induced thrombosis. We investigated cereblon substrates in human megakaryocytes using liquid chromatography-mass spectrometry and found that thrombospondin-1 (THBS-1), which is an inhibitor of a disintegrin-like and metalloproteinase with thrombospondin type 1 motifs 13, functions as an endogenous substrate in human megakaryocytes. IMiDs inhibited the proteasomal degradation of THBS-1 by impairing the recruitment of cereblon to THBS-1, leading to aberrant accumulation of THBS-1. We observed a significant increase in THBS-1 in peripheral blood mononuclear cells as well as larger von Willebrand factor multimers in the plasma of patients with myeloma, who were treated with IMiDs. These results collectively suggest that THBS-1 represents an endogenous substrate of cereblon. This pairing is disrupted by IMiDs, and the aberrant accumulation of THBS-1 plays an important role in the pathogenesis of IMiDs-induced thromboembolism.


Assuntos
Mieloma Múltiplo , Tromboembolia , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Agentes de Imunomodulação , Leucócitos Mononucleares/metabolismo , Mieloma Múltiplo/genética , Tromboembolia/etiologia , Trombospondinas/metabolismo , Trombospondinas/uso terapêutico
20.
Mol Pharmacol ; 83(5): 930-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23393163

RESUMO

Vesnarinone is a synthetic quinolinone derivative used in the treatment of cardiac failure and cancer. It is also known to cause agranulocytosis as a side effect, which restricts its use, although the mechanism underlying agranulocytosis is not well understood. Here, we show that vesnarinone binds to valosin-containing protein (VCP), which interacts with polyubiquitinated proteins and is essential for the degradation of IκBα to activate nuclear factor (NF)κB. We show that vesnarinone impairs the degradation of IκBα, and that the impairment of the degradation of IκBα is the result of the inhibition of the interaction between VCP and the 26S proteasome by vesnarinone. These results suggest that vesnarinone suppresses NFκB activation by inhibiting the VCP-dependent degradation of polyubiquitinated IκBα, resulting in the suppression of tumor necrosis factor-α mRNA expression.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Proteínas de Ciclo Celular/antagonistas & inibidores , Quinolinas/farmacologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Fator de Necrose Tumoral alfa/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Células HEK293 , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/genética , NF-kappa B/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Pirazinas , RNA Mensageiro/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteína com Valosina
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa