Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Science ; 378(6619): 549-553, 2022 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-36378961

RESUMO

Cereblon (CRBN) is a ubiquitin ligase (E3) substrate receptor protein co-opted by CRBN E3 ligase modulatory drug (CELMoD) agents that target therapeutically relevant proteins for degradation. Prior crystallographic studies defined the drug-binding site within CRBN's thalidomide-binding domain (TBD), but the allostery of drug-induced neosubstrate binding remains unclear. We performed cryo-electron microscopy analyses of the DNA damage-binding protein 1 (DDB1)-CRBN apo complex and compared these structures with DDB1-CRBN in the presence of CELMoD compounds alone and complexed with neosubstrates. Association of CELMoD compounds to the TBD is necessary and sufficient for triggering CRBN allosteric rearrangement from an open conformation to the canonical closed conformation. The neosubstrate Ikaros only stably associates with the closed CRBN conformation, illustrating the importance of allostery for CELMoD compound efficacy and informing structure-guided design strategies to improve therapeutic efficacy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Ubiquitina-Proteína Ligases , Proteínas Adaptadoras de Transdução de Sinal/química , Microscopia Crioeletrônica , Talidomida/química , Ubiquitina-Proteína Ligases/química , Domínios Proteicos , Regulação Alostérica
2.
Nat Commun ; 12(1): 5218, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34471125

RESUMO

CD47 is the only 5-transmembrane (5-TM) spanning receptor of the immune system. Its extracellular domain (ECD) is a cell surface marker of self that binds SIRPα and inhibits macrophage phagocytosis, and cancer immuno-therapy approaches in clinical trials are focused on blocking CD47/SIRPα interaction. We present the crystal structure of full length CD47 bound to the function-blocking antibody B6H12. CD47 ECD is tethered to the TM domain via a six-residue peptide linker (114RVVSWF119) that forms an extended loop (SWF loop), with the fundamental role of inserting the side chains of W118 and F119 into the core of CD47 extracellular loop region (ECLR). Using hydrogen-deuterium exchange and molecular dynamics simulations we show that CD47's ECLR architecture, comprised of two extracellular loops and the SWF loop, creates a molecular environment stabilizing the ECD for presentation on the cell surface. These findings provide insights into CD47 immune recognition, signaling and therapeutic intervention.


Assuntos
Biomarcadores , Antígeno CD47/química , Antígeno CD47/metabolismo , Proteínas de Transporte/metabolismo , Receptores Imunológicos/metabolismo , Anticorpos Bloqueadores/química , Anticorpos Bloqueadores/farmacologia , Antígenos de Diferenciação/imunologia , Sítios de Ligação , Antígeno CD47/efeitos dos fármacos , Antígeno CD47/genética , Humanos , Macrófagos/metabolismo , Modelos Moleculares , Fagocitose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
3.
Methods Mol Biol ; 2365: 283-300, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34432250

RESUMO

Targeted protein degradation is garnering increased attention as a therapeutic modality due in part to its promise of modulating targets previously considered undruggable. Cereblon E3 Ligase Modulating Drugs (CELMoDs) are one of the most well-characterized therapeutics employing this modality. CELMoDs hijack Cereblon E3 ligase activity causing neosubstrates to be ubiquitinated and degraded in the proteasome. Here, we describe a suite of assays-cellular substrate degradation, confirmation of CELMoD mechanism of action, in vitro ubiquitination, and Cereblon binding-that can be used to characterize CELMoD-mediated degradation of Cereblon neosubstrates. While the assays presented herein can be run independently, when combined they provide a strong platform to support the discovery and optimization of CELMoDs and fuel validation of targets degraded by this drug modality.


Assuntos
Nanoestruturas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
4.
Blood Cancer Discov ; 2(3): 250-265, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34027417

RESUMO

Thalidomide analogs exert their therapeutic effects by binding to the CRL4CRBN E3 ubiquitin ligase, promoting ubiquitination and subsequent proteasomal degradation of specific protein substrates. Drug-induced degradation of IKZF1 and IKZF3 in B-cell malignancies demonstrates the clinical utility of targeting disease-relevant transcription factors for degradation. Here, we found that avadomide (CC-122) induces CRBN-dependent ubiquitination and proteasomal degradation of ZMYM2 (ZNF198), a transcription factor involved in balanced chromosomal rearrangements with FGFR1 and FLT3 in aggressive forms of hematologic malignancies. The minimal drug-responsive element of ZMYM2 is a zinc-chelating MYM domain and is contained in the N-terminal portion of ZMYM2 that is universally included in the derived fusion proteins. We demonstrate that avadomide has the ability to induce proteasomal degradation of ZMYM2-FGFR1 and ZMYM2-FLT3 chimeric oncoproteins, both in vitro and in vivo. Our findings suggest that patients with hematologic malignancies harboring these ZMYM2 fusion proteins may benefit from avadomide treatment.


Assuntos
Neoplasias Hematológicas , Talidomida , Proteínas de Ligação a DNA , Neoplasias Hematológicas/tratamento farmacológico , Humanos , Lenalidomida/farmacologia , Proteínas Oncogênicas , Fatores de Transcrição/metabolismo
5.
Blood ; 137(2): 232-237, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33443552

RESUMO

Emergence of drug resistance to all available therapies is the major challenge to improving survival in myeloma. Cereblon (CRBN) is the essential binding protein of the widely used immunomodulatory drugs (IMiDs) and novel CRBN E3 ligase modulator drugs (CELMoDs) in myeloma, as well as certain proteolysis targeting chimeras (PROTACs), in development for a range of diseases. Using whole-genome sequencing (WGS) data from 455 patients and RNA sequencing (RNASeq) data from 655 patients, including newly diagnosed (WGS, n = 198; RNASeq, n = 437), lenalidomide (LEN)-refractory (WGS, n = 203; RNASeq, n = 176), and pomalidomide (POM)-refractory cohorts (WGS, n = 54; RNASeq, n = 42), we found incremental increases in the frequency of 3 CRBN aberrations, namely point mutations, copy losses/structural variations, and a specific variant transcript (exon 10 spliced), with progressive IMiD exposure, until almost one-third of patients had CBRN alterations by the time they were POM refractory. We found all 3 CRBN aberrations were associated with inferior outcomes to POM in those already refractory to LEN, including those with gene copy losses and structural variations, a finding not previously described. This represents the first comprehensive analysis and largest data set of CBRN alterations in myeloma patients as they progress through therapy. It will help inform patient selection for sequential therapies with CRBN-targeting drugs.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Mieloma Múltiplo/tratamento farmacológico , Ubiquitina-Proteína Ligases/genética , Variação Genética , Humanos , Lenalidomida/uso terapêutico , Talidomida/análogos & derivados , Talidomida/uso terapêutico
6.
ACS Chem Biol ; 15(12): 3149-3158, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33206504

RESUMO

There is a growing interest in using targeted protein degradation as a therapeutic modality in view of its potential to expand the druggable proteome. One avenue to using this modality is via molecular glue based Cereblon E3 Ligase Modulating Drug compounds. Here, we report the identification of the transcription factor ZBTB16 as a Cereblon neosubstrate. We also report two new Cereblon modulators, CC-3060 and CC-647, that promote ZBTB16 degradation. Unexpectedly, CC-3060 and CC-647 target ZBTB16 for degradation by primarily engaging distinct structural degrons on different zinc finger domains. The reciprocal fusion proteins, ZBTB16-RARα and RARα-ZBTB16, which cause a rare acute promyelocytic leukemia, contain these same structural degrons and can be targeted for proteasomal degradation with Cereblon modulator treatment. Thus, a targeted protein degradation approach via Cereblon modulators may represent a novel therapeutic strategy in acute promyelocytic leukemia where ZBTB16/RARA rearrangements are critical disease drivers.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Humanos , Leucemia Promielocítica Aguda/metabolismo , Proteólise , Receptor alfa de Ácido Retinoico/metabolismo , Especificidade por Substrato
7.
Nat Struct Mol Biol ; 27(4): 319-322, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32251415

RESUMO

Thalidomide-dependent degradation of the embryonic transcription factor SALL4 by the CRL4CRBN E3 ubiquitin ligase is a plausible major driver of thalidomide teratogenicity. The structure of the second zinc finger of SALL4 in complex with pomalidomide, cereblon and DDB1 reveals the molecular details of recruitment. Sequence differences and a shifted binding position relative to Ikaros offer a path to the rational design of cereblon-binding drugs with reduced teratogenic risk.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas de Ligação a DNA/ultraestrutura , Complexos Multiproteicos/ultraestrutura , Fatores de Transcrição/ultraestrutura , Proteínas Adaptadoras de Transdução de Sinal/genética , Cristalografia por Raios X , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Ligação Proteica , Conformação Proteica , Proteólise/efeitos dos fármacos , Especificidade por Substrato , Talidomida/análogos & derivados , Talidomida/química , Talidomida/farmacologia , Fatores de Transcrição/química , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/ultraestrutura , Ubiquitinação/genética
8.
ACS Med Chem Lett ; 10(12): 1592-1602, 2019 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-31857833

RESUMO

Many cellular processes and pathways are mediated by the regulation of protein-protein complexes. For example, E3 ubiquitin ligases recruit substrate proteins and transfer a ubiquitin tag to target those proteins for destruction by the proteasome. It has now been shown that this cellular process for protein destruction can be redirected by small molecules in both laboratory and clinical settings. This presents a new paradigm in drug discovery, enabling the rapid removal of target proteins linked to disease. In this Innovations review, we will describe the work done on cereblon as a case study on the different strategies available for targeted protein degradation.

9.
Nat Chem Biol ; 15(10): 937-944, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31527835

RESUMO

Targeted protein degradation as a therapeutic modality has seen dramatic progress and massive investment in recent years because of the convergence of two key scientific breakthroughs: optimization of first-generation peptidic proteolysis-targeted chimeras (PROTACs) into more drug-like molecules able to support in vivo proof of concept and the discovery that clinical molecules function as degraders by binding and repurposing the proteins cereblon and DCAF15. This provided clinical validation for the general approach through the cereblon modulator class of drugs and provided highly drug-like and ligand-efficient E3 ligase binders upon which to tether target-binding moieties. Increasingly rational and systematic approaches including biophysical and structural studies on ternary complexes are being leveraged as the field advances. In this Perspective we summarize the discoveries that have laid the foundation for future degradation therapeutics, focusing on those classes of small molecules that redirect E3 ubiquitin ligases to non-native substrates.


Assuntos
Proteólise/efeitos dos fármacos , Sítios de Ligação , Humanos , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Bibliotecas de Moléculas Pequenas/química , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
10.
Drug Discov Today Technol ; 31: 29-34, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31200856

RESUMO

Targeted protein degradation has become an exciting new paradigm in drug discovery with the potential to target new protein families for therapeutic intervention. In 2010, Hiroshi Handa and colleagues discovered that the drug thalidomide binds to the protein cereblon, a component of the CRL4CRBN E3 ubiquitin ligase. In contrast to the heterobifunctional small molecule degraders reported in the literature, thalidomide is of very low molecular weight (∼258Da) with molecular properties (solubility, metabolic stability, permeability etc) that readily support pharmaceutical dosing. It was subsequently shown that thalidomide and the analogues lenalidomide and pomalidomide are able to degrade the transcription factors Ikaros and Aiolos. CK1α and GSPT1 were subsequently identified as substrates for specific ligands, indicating that this molecular class could be tuned for selective protein degradation. Structural studies showed that the thalidomide analogues bind to a shallow hydrophobic pocket on the surface of cereblon, and scaffold a protein-protein interaction with target proteins. Target proteins do not need any affinity for the cereblon modulators, and as such undruggable, or even unligandable, proteins can be targeted for degradation. A similar mechanism of action was subsequently identified for the clinical molecule indisulam, indicating that low molecular weight degraders are not unique to cereblon. The groundbreaking work on cereblon represents a case study for the discovery and characterization of low molecular weight protein degraders for other ligases.


Assuntos
Peptídeo Hidrolases/metabolismo , Proteólise , Proteínas Adaptadoras de Transdução de Sinal , Humanos , Peso Molecular , Ubiquitina-Proteína Ligases/metabolismo
11.
Cell Chem Biol ; 26(3): 307-308, 2019 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-30901555

RESUMO

Targeted protein degradation offers considerable promise for the discovery of new therapeutics. In Cell Chemical Biology, Brand et al. (2019) identify selective degraders of CDK6 derived from clinically approved CDK4/6 inhibitors. This approach offers the possibility of a differentiated therapeutic profile with potential to treat acute myeloid leukemia.


Assuntos
Leucemia Mieloide Aguda , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Humanos , Proteólise
12.
Nat Chem Biol ; 14(10): 981-987, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30190590

RESUMO

Targeted protein degradation via small-molecule modulation of cereblon offers vast potential for the development of new therapeutics. Cereblon-binding therapeutics carry the safety risks of thalidomide, which caused an epidemic of severe birth defects characterized by forelimb shortening or phocomelia. Here we show that thalidomide is not teratogenic in transgenic mice expressing human cereblon, indicating that binding to cereblon is not sufficient to cause birth defects. Instead, we identify SALL4 as a thalidomide-dependent cereblon neosubstrate. Human mutations in SALL4 cause Duane-radial ray, IVIC, and acro-renal-ocular syndromes with overlapping clinical presentations to thalidomide embryopathy, including phocomelia. SALL4 is degraded in rabbits but not in resistant organisms such as mice because of SALL4 sequence variations. This work expands the scope of cereblon neosubstrate activity within the formerly 'undruggable' C2H2 zinc finger family and offers a path toward safer therapeutics through an improved understanding of the molecular basis of thalidomide-induced teratogenicity.


Assuntos
Regulação da Expressão Gênica , Peptídeo Hidrolases/química , Teratogênicos/química , Talidomida/química , Fatores de Transcrição/química , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Homozigoto , Humanos , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas , Ligantes , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Peptídeo Hidrolases/genética , Proteólise , Coelhos , Testículo/metabolismo , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/metabolismo , Dedos de Zinco
13.
J Med Chem ; 61(2): 535-542, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-28425720

RESUMO

The drugs lenalidomide and pomalidomide bind to the protein cereblon, directing the CRL4-CRBN E3 ligase toward the transcription factors Ikaros and Aiolos to cause their ubiquitination and degradation. Here we describe CC-220 (compound 6), a cereblon modulator in clinical development for systemic lupus erythematosis and relapsed/refractory multiple myeloma. Compound 6 binds cereblon with a higher affinity than lenalidomide or pomalidomide. Consistent with this, the cellular degradation of Ikaros and Aiolos is more potent and the extent of substrate depletion is greater. The crystal structure of cereblon in complex with DDB1 and compound 6 reveals that the increase in potency correlates with increased contacts between compound 6 and cereblon away from the modeled binding site for Ikaros/Aiolos. These results describe a new cereblon modulator which achieves greater substrate degradation via tighter binding to the cereblon E3 ligase and provides an example of the effect of E3 ligase binding affinity with relevance to other drug discovery efforts in targeted protein degradation.


Assuntos
Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Fator de Transcrição Ikaros/metabolismo , Peptídeo Hidrolases/química , Peptídeo Hidrolases/metabolismo , Proteólise/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Linhagem Celular Tumoral , Cristalografia por Raios X , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Transferência Ressonante de Energia de Fluorescência , Compostos Heterocíclicos de 4 ou mais Anéis/química , Compostos Heterocíclicos de 4 ou mais Anéis/metabolismo , Humanos , Lenalidomida/química , Lenalidomida/metabolismo , Morfolinas , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Ftalimidas , Piperidonas , Ligação Proteica , Ubiquitina-Proteína Ligases
14.
J Med Chem ; 60(21): 8989-9002, 2017 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-28991472

RESUMO

Triple negative breast cancer (TNBC) remains a serious unmet medical need with discouragingly high relapse rates. We report here the synthesis and structure-activity relationship (SAR) of a novel series of 2,4,5-trisubstituted-7H-pyrrolo[2,3-d]pyrimidines with potent activity against TNBC tumor cell lines. These compounds were discovered from a TNBC phenotypic screen and possess a unique dual inhibition profile targeting TTK (mitotic exit) and CLK2 (mRNA splicing). Design and optimization, driven with a TNBC tumor cell assay, identified potent and selective compounds with favorable in vitro and in vivo activity profiles and good iv PK properties. This cell-based driven SAR produced compounds with strong single agent in vivo efficacy in multiple TNBC xenograft models without significant body weight loss. These data supported the nomination of CC-671 into IND-enabling studies as a single agent TNBC therapy.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/síntese química , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , Camundongos , Mitose/efeitos dos fármacos , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Splicing de RNA/efeitos dos fármacos , Relação Estrutura-Atividade , Neoplasias de Mama Triplo Negativas/enzimologia
15.
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
16.
Nature ; 523(7559): 183-188, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-26131937

RESUMO

Lenalidomide is a highly effective treatment for myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)). Here, we demonstrate that lenalidomide induces the ubiquitination of casein kinase 1A1 (CK1α) by the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4(CRBN)), resulting in CK1α degradation. CK1α is encoded by a gene within the common deleted region for del(5q) MDS and haploinsufficient expression sensitizes cells to lenalidomide therapy, providing a mechanistic basis for the therapeutic window of lenalidomide in del(5q) MDS. We found that mouse cells are resistant to lenalidomide but that changing a single amino acid in mouse Crbn to the corresponding human residue enables lenalidomide-dependent degradation of CK1α. We further demonstrate that minor side chain modifications in thalidomide and a novel analogue, CC-122, can modulate the spectrum of substrates targeted by CRL4(CRBN). These findings have implications for the clinical activity of lenalidomide and related compounds, and demonstrate the therapeutic potential of novel modulators of E3 ubiquitin ligases.


Assuntos
Caseína Quinase I/metabolismo , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/fisiopatologia , Talidomida/análogos & derivados , Ubiquitinação/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Caseína Quinase I/genética , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Fatores Imunológicos/farmacologia , Células Jurkat , Células K562 , Lenalidomida , Camundongos , Dados de Sequência Molecular , Peptídeo Hidrolases/química , Proteólise/efeitos dos fármacos , Alinhamento de Sequência , Deleção de Sequência , Especificidade da Espécie , Talidomida/farmacologia , Ubiquitina-Proteína Ligases/metabolismo
17.
Nat Struct Mol Biol ; 21(9): 803-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25108355

RESUMO

The Cul4-Rbx1-DDB1-Cereblon E3 ubiquitin ligase complex is the target of thalidomide, lenalidomide and pomalidomide, therapeutically important drugs for multiple myeloma and other B-cell malignancies. These drugs directly bind Cereblon (CRBN) and promote the recruitment of substrates Ikaros (IKZF1) and Aiolos (IKZF3) to the E3 complex, thus leading to substrate ubiquitination and degradation. Here we present the crystal structure of human CRBN bound to DDB1 and the drug lenalidomide. A hydrophobic pocket in the thalidomide-binding domain (TBD) of CRBN accommodates the glutarimide moiety of lenalidomide, whereas the isoindolinone ring is exposed to solvent. We also solved the structures of the mouse TBD in the apo state and with thalidomide or pomalidomide. Site-directed mutagenesis in lentiviral-expression myeloma models showed that key drug-binding residues are critical for antiproliferative effects.


Assuntos
Inibidores da Angiogênese/farmacologia , Proteínas de Ligação a DNA/metabolismo , Peptídeo Hidrolases/metabolismo , Talidomida/análogos & derivados , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Inibidores da Angiogênese/química , Animais , Cristalografia por Raios X , Proteínas de Ligação a DNA/química , Humanos , Lenalidomida , Camundongos , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Peptídeo Hidrolases/química , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Alinhamento de Sequência , Talidomida/química , Talidomida/farmacologia , Ubiquitina-Proteína Ligases
18.
J Med Chem ; 51(16): 5000-8, 2008 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-18665583

RESUMO

Owing to the emergence of resistant virus, next generation non-nucleoside HIV reverse transcriptase inhibitors (NNRTIs) with improved drug resistance profiles have been developed to treat HIV infection. Crystal structures of HIV-1 RT complexed with benzophenones optimized for inhibition of HIV mutants that were resistant to the prototype benzophenone GF128590 indicate factors contributing to the resilience of later compounds in the series (GW4511, GW678248). Meta-substituents on the benzophenone A-ring had the designed effect of inducing better contacts with the conserved W229 while reducing aromatic stacking interactions with the highly mutable Y181 side chain, which unexpectedly adopted a "down" position. Up to four main-chain hydrogen bonds to the inhibitor also appear significant in contributing to resilience. Structures of mutant RTs (K103N, V106A/Y181C) with benzophenones showed only small rearrangements of the NNRTIs relative to wild-type. Hence, adaptation to a mutated NNRTI pocket by inhibitor rearrangement appears less significant for benzophenones than other next-generation NNRTIs.


Assuntos
Benzofenonas/farmacologia , Farmacorresistência Viral/efeitos dos fármacos , Transcriptase Reversa do HIV/antagonistas & inibidores , Inibidores da Transcriptase Reversa/farmacologia , Alcinos , Substituição de Aminoácidos , Benzofenonas/química , Benzoxazinas/química , Cristalografia por Raios X , Ciclopropanos , Desenho de Fármacos , Transcriptase Reversa do HIV/química , Transcriptase Reversa do HIV/genética , Modelos Moleculares , Nevirapina/química , Nitrilas/farmacologia , Relação Estrutura-Atividade , Sulfonamidas/farmacologia
19.
Cell Cycle ; 7(4): 463-7, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18235237

RESUMO

Nearly 25 years ago the first function of an inositol phosphate, namely Ins(1,4,5)P3, was reported to act as a "second messenger" to mobilize calcium from the endoplasmic reticulum (ER). Since this discovery, many other inositol phosphates and the kinases and phosphatases that generate these inositol phosphates have subsequently been discovered. However, the function of these "higher order" inositol phosphates in biological processes, if any, has remained a mystery. Interest in higher order inositol phosphates, such as Ins(1,3,4,5)P4, was renewed this year following reports of novel roles for these molecules in distinct processes within the immune system ranging from T cell development, B cell development and tolerance induction, as well as neutrophil and mast cell function. In this review, we will touch upon recent advances in inositol phosphate function in mammalian cells. More specifically, we will highlight new studies that have identified novel functions for specific higher order inositol phosphates, such as Ins(1,3,4,5)P4, in the immune system.


Assuntos
Fosfatos de Inositol/imunologia , Linfócitos/imunologia , Mamíferos/imunologia , Modelos Imunológicos , Transdução de Sinais/imunologia , Animais , Linfócitos/metabolismo
20.
J Biol Chem ; 282(38): 28117-25, 2007 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-17616525

RESUMO

Inositol 1,3,4-trisphosphate 5/6-kinase (ITPK1) is a reversible, poly-specific inositol phosphate kinase that has been implicated as a modifier gene in cystic fibrosis. Upon activation of phospholipase C at the plasma membrane, inositol 1,4,5-trisphosphate enters the cytosol and is inter-converted by an array of kinases and phosphatases into other inositol phosphates with diverse and critical cellular activities. In mammals it has been established that inositol 1,3,4-trisphosphate, produced from inositol 1,4,5-trisphosphate, lies in a branch of the metabolic pathway that is separate from inositol 3,4,5,6-tetrakisphosphate, which inhibits plasma membrane chloride channels. We have determined the molecular mechanism for communication between these two pathways, showing that phosphate is transferred between inositol phosphates via ITPK1-bound nucleotide. Intersubstrate phosphate transfer explains how competing substrates are able to stimulate each others' catalysis by ITPK1. We further show that these features occur in the human protein, but not in plant or protozoan homologues. The high resolution structure of human ITPK1 identifies novel secondary structural features able to impart substrate selectivity and enhance nucleotide binding, thereby promoting intersubstrate phosphate transfer. Our work describes a novel mode of substrate regulation and provides insight into the enzyme evolution of a signaling mechanism from a metabolic role.


Assuntos
Fosfatos de Inositol/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Transdução de Sinais , Sequência de Aminoácidos , Membrana Celular/metabolismo , Fibrose Cística/metabolismo , Ativação Enzimática , Humanos , Conformação Molecular , Dados de Sequência Molecular , Fosfotransferases (Aceptor do Grupo Álcool)/química , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Fosfolipases Tipo C/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA