Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 32(7): 2190-2206, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38796705

RESUMO

X-linked adrenoleukodystrophy (ALD), an inherited neurometabolic disorder caused by mutations in ABCD1, which encodes the peroxisomal ABC transporter, mainly affects the brain, spinal cord, adrenal glands, and testes. In ALD patients, very-long-chain fatty acids (VLCFAs) fail to enter the peroxisome and undergo subsequent ß-oxidation, resulting in their accumulation in the body. It has not been tested whether in vivo base editing or prime editing can be harnessed to ameliorate ALD. We developed a humanized mouse model of ALD by inserting a human cDNA containing the pathogenic variant into the mouse Abcd1 locus. The humanized ALD model showed increased levels of VLCFAs. To correct the mutation, we tested both base editing and prime editing and found that base editing using ABE8e(V106W) could correct the mutation in patient-derived fibroblasts at an efficiency of 7.4%. Adeno-associated virus (AAV)-mediated systemic delivery of NG-ABE8e(V106W) enabled robust correction of the pathogenic variant in the mouse brain (correction efficiency: ∼5.5%), spinal cord (∼5.1%), and adrenal gland (∼2%), leading to a significant reduction in the plasma levels of C26:0/C22:0. This established humanized mouse model and the successful correction of the pathogenic variant using a base editor serve as a significant step toward treating human ALD disease.


Assuntos
Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP , Adrenoleucodistrofia , Dependovirus , Modelos Animais de Doenças , Edição de Genes , Terapia Genética , Animais , Adrenoleucodistrofia/terapia , Adrenoleucodistrofia/genética , Camundongos , Humanos , Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP/genética , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/administração & dosagem , Adenina , Mutação , Fibroblastos/metabolismo , Ácidos Graxos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia
2.
Mol Ther Nucleic Acids ; 35(2): 102199, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38766525

RESUMO

Pathogenic structure variations (SVs) are associated with various types of cancer and rare genetic diseases. Recent studies have used Cas9 nuclease with paired guide RNAs (gRNAs) to generate targeted chromosomal rearrangements, focusing on producing fusion proteins that cause cancer, whereas research on precision genome editing for rectifying SVs is limited. In this study, we identified a novel complex genomic rearrangement (CGR), specifically an EYA1 inversion with a deletion, implicated in branchio-oto-renal/branchio-oto syndrome. To address this, two CRISPR-based approaches were tested. First, we used Cas9 nuclease and paired gRNAs tailored to the patient's genome. The dual CRISPR-Cas9 system induced efficient correction of paracentric inversion in patient-derived fibroblast, and effectively restored the expression of EYA1 mRNA and protein, along with its transcriptional activity required to regulate the target gene expression. Additionally, we used CRISPR activation (CRISPRa), which leads to the upregulation of EYA1 mRNA expression in patient-derived fibroblasts. Moreover, CRISPRa significantly improved EYA1 protein expression and transcriptional activity essential for target gene expression. This suggests that CRISPRa-based gene therapies could offer substantial translational potential for approximately 70% of disease-causing EYA1 variants responsible for haploinsufficiency. Our findings demonstrate the potential of CRISPR-guided genome editing for correcting SVs, including those with EYA1 CGR linked to haploinsufficiency.

3.
Nucleic Acids Res ; 52(10): 5792-5803, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38661210

RESUMO

Nucleotide repeat expansion disorders, a group of genetic diseases characterized by the expansion of specific DNA sequences, pose significant challenges to treatment and therapy development. Here, we present a precise and programmable method called prime editor-mediated correction of nucleotide repeat expansion (PE-CORE) for correcting pathogenic nucleotide repeat expansion. PE-CORE leverages a prime editor and paired pegRNAs to achieve targeted correction of repeat sequences. We demonstrate the effectiveness of PE-CORE in HEK293T cells and patient-derived induced pluripotent stem cells (iPSCs). Specifically, we focus on spinal and bulbar muscular atrophy and spinocerebellar ataxia type, two diseases associated with nucleotide repeat expansion. Our results demonstrate the successful correction of pathogenic expansions in iPSCs and subsequent differentiation into motor neurons. Specifically, we detect distinct downshifts in the size of both the mRNA and protein, confirming the functional correction of the iPSC-derived motor neurons. These findings highlight PE-CORE as a precision tool for addressing the intricate challenges of nucleotide repeat expansion disorders, paving the way for targeted therapies and potential clinical applications.


Assuntos
Edição de Genes , Células-Tronco Pluripotentes Induzidas , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Edição de Genes/métodos , Células HEK293 , Neurônios Motores/metabolismo , Diferenciação Celular/genética , Expansão das Repetições de DNA/genética , Expansão das Repetições de Trinucleotídeos/genética
4.
Commun Biol ; 6(1): 466, 2023 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-37117485

RESUMO

Huntington's disease (HD) is caused by a CAG repeat expansion in the huntingtin (HTT) gene. CRISPR-Cas9 nuclease causes double-strand breaks (DSBs) in the targeted DNA that induces toxicity, whereas CRISPR interference (CRISPRi) using dead Cas9 (dCas9) suppresses the target gene expression without DSBs. Delivery of dCas9-sgRNA targeting CAG repeat region does not damage the targeted DNA in HEK293T cells containing CAG repeats. When this study investigates whether CRISPRi can suppress mutant HTT (mHTT), CRISPRi results in reduced expression of mHTT with relative preservation of the wild-type HTT in human HD fibroblasts. Although both dCas9 and Cas9 treatments reduce mHTT by sgRNA targeting the CAG repeat region, CRISPRi delays behavioral deterioration and protects striatal neurons against cell death in HD mice. Collectively, CRISPRi can delay disease progression by suppressing mHtt, suggesting DNA DSB-free CRISPRi is a potential therapy for HD that can compensate for the shortcoming of CRISPR-Cas9 nuclease.


Assuntos
Doença de Huntington , Camundongos , Humanos , Animais , Doença de Huntington/genética , Doença de Huntington/terapia , Doença de Huntington/metabolismo , Quebras de DNA de Cadeia Dupla , Células HEK293 , Corpo Estriado/metabolismo
5.
Mol Ther ; 31(1): 249-259, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36114670

RESUMO

A variety of cancers have been found to have chromosomal rearrangements, and the genomic abnormalities often induced expression of fusion oncogenes. To date, a pair of engineered nucleases including ZFNs, TALENs, and CRISPR-Cas9 nucleases have been used to generate chromosomal rearrangement in living cells and organisms for disease modeling. However, these methods induce unwanted indel mutations at the DNA break junctions, resulting in incomplete disease modeling. Here, we developed prime editor nuclease-mediated translocation and inversion (PETI), a method for programmable chromosomal translocation and inversion using prime editor 2 nuclease (PE2 nuclease) and paired pegRNA. Using PETI method, we successfully introduced DNA recombination in episomal fluorescence reporters as well as precise chromosomal translocations in human cells. We applied PETI to create cancer-associated translocations and inversions such as NPM1-ALK and EML4-ALK in human cells. Our findings show that PETI generated chromosomal translocation and inversion in a programmable manner with efficiencies comparable of Cas9. PETI methods, we believe, could be used to create disease models or for gene therapy.


Assuntos
Neoplasias , Translocação Genética , Humanos , Rearranjo Gênico , Genoma , Endonucleases , Genômica , Receptores Proteína Tirosina Quinases , Edição de Genes/métodos , Sistemas CRISPR-Cas
6.
Mol Ther Nucleic Acids ; 30: 131-142, 2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36250202

RESUMO

RNA-guided CRISPR-Cas12a endonucleases are promising tools for genome engineering. Here we demonstrate that LbCas12a variants derived from Lachnospiraceae bacterium show a broad PAM preference, recognizing certain non-canonical PAMs with high efficiency. Furthermore, we engineered LbABE8e to carry G532R and/or K595R mutations, altering its original PAM specificities; these variants exhibited superior base editing activity in human cells compared with wild-type LbABE8e at sites with non-canonical PAMs. Based on this finding, we utilized the most effective LbCas12a and LbABE8e variants to demonstrate multiplexed and mutant-allele-specific gene editing in oncogenes, made possible by the variant's recognition of non-canonical PAMs. Importantly, LbCas12a-G532R/K595R and LbABE8e-G532R/K595R with optimized crRNA arrays targeted to triple oncogenic mutations inhibited colon cancer cell proliferation. Taken together, these results demonstrate the potential of engineered LbCas12a and LbABE8e as tools for targeting sites with alternative PAMs for genome engineering and therapeutic editing in cancer cells.

7.
Biochem Biophys Res Commun ; 552: 164-169, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33751933

RESUMO

Human mesenchymal stem cells (MSCs) are known to have anti-inflammatory and immunomodulatory functions; thus, several MSC products have been applied as cell therapy in clinical trials worldwide. Recent studies have demonstrated that MSC spheroids have superior anti-inflammatory and immunomodulatory functions to a single cell suspension. Current methods to prepare MSC spheroids include hanging drop, concave microwell aggregation, spinner flask, and gravity circulation. However, all these methods have limitations such as low scalability, easy cell clumping, low viability, and irregular size distribution. Here, we present a nano-patterned culture plasticware named PAMcell™ 3D plate to overcome these limitations. Nano-sized silica particles (700 nm) coated with RGD peptide were arrayed into fusiform onto the PLGA film. This uniform array enabled the seeded MSCs to grow only on the silica particles, forming uniform-sized semi-spheroids within 48 h. These MSC spheroids have been shown to have enhanced stemness, anti-inflammatory, and immunomodulatory functions, as revealed by the increased expression of stem cell markers (Oct4, Sox2, and Nanog), anti-inflammatory (IL-10, TSG6, and IDO), and immunomodulatory molecules (HGF, VEGF, CXCR4) both at mRNA and protein expression levels. Furthermore, these MSC spheroids demonstrated an increased palliative effect on glycemic control in a multiple low-dose streptozotocin-induced diabetes model compared with the same number of MSC single cell suspensions. Taken together, this study presents a new method to produce uniform-sized MSC spheroids with enhanced anti-inflammatory and immunomodulatory functions in vitro and in vivo.


Assuntos
Tecido Adiposo/citologia , Anti-Inflamatórios/imunologia , Técnicas de Cultura de Células/métodos , Fatores Imunológicos/imunologia , Células-Tronco Mesenquimais/imunologia , Esferoides Celulares/imunologia , Animais , Técnicas de Cultura de Células/instrumentação , Células Cultivadas , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/terapia , Expressão Gênica/imunologia , Humanos , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Esferoides Celulares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo
8.
Mol Cells ; 39(4): 316-21, 2016 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-26923188

RESUMO

The receptor activator of nuclear factor κB (RANK) and its ligand RANKL are key regulators of osteoclastogenesis and well-recognized targets in developing treatments for bone disorders associated with excessive bone resorption, such as osteoporosis. Our previous work on the structure of the RANK-RANKL complex revealed that Loop3 of RANK, specifically the non-canonical disulfide bond at the tip, performs a crucial role in specific recognition of RANKL. It also demonstrated that peptide mimics of Loop3 were capable of interfering with the function of RANKL in osteoclastogenesis. Here, we reported the structure-based design of a smaller peptide with enhanced inhibitory efficiency. The kinetic analysis and osteoclast differentiation assay showed that in addition to the sharp turn induced by the disulfide bond, two consecutive arginine residues were also important for binding to RANKL and inhibiting osteoclastogenesis. Docking and molecular dynamics simulations proposed the binding mode of the peptide to the RANKL trimer, showing that the arginine residues provide electrostatic interactions with RANKL and contribute to stabilizing the complex. These findings provided useful information for the rational design of therapeutics for bone diseases associated with RANK/RANKL function.


Assuntos
Osteogênese/efeitos dos fármacos , Peptidomiméticos/síntese química , Peptidomiméticos/farmacologia , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/química , Animais , Arginina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Desenho de Fármacos , Humanos , Camundongos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Peptidomiméticos/química , Ligação Proteica , Relação Estrutura-Atividade
9.
Biochem Biophys Res Commun ; 461(1): 122-7, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25858319

RESUMO

Proteins in the haloalkaloic acid dehalogenase (HAD) superfamily, which is one of the largest enzyme families, is generally composed of a catalytic core domain and a cap domain. Although proteins in this family show broad substrate specificities, the mechanisms of their substrate recognition are not well understood. In this study, we identified a new substrate binding motif of HAD proteins from structural and functional analyses, and propose that this motif might be crucial for interacting with hydrophobic rings of substrates. The crystal structure of TON_0338, one of the 17 putative HAD proteins identified in a hyperthermophilic archaeon, Thermococcus onnurineus NA1, was determined as an apo-form at 2.0 Å resolution. In addition, we determined the crystal structure TON_0338 in complex with Mg(2+) or N-cyclohexyl-2-aminoethanesulfonic acid (CHES) at 1.7 Å resolution. Examination of the apo-form and CHES-bound structures revealed that CHES is sandwiched between Trp58 and Trp61, suggesting that this Trp sandwich might function as a substrate recognition motif. In the phosphatase assay, TON_0338 was shown to have high activity for flavin mononucleotide (FMN), and the docking analysis suggested that the flavin of FMN may interact with Trp58 and Trp61 in a way similar to that observed in the crystal structure. Moreover, the replacement of these tryptophan residues significantly reduced the phosphatase activity for FMN. Our results suggest that WxxW may function as a substrate binding motif in HAD proteins, and expand the diversity of their substrate recognition mode.


Assuntos
Hidrolases/química , Hidrolases/ultraestrutura , Modelos Químicos , Simulação de Acoplamento Molecular , Monoéster Fosfórico Hidrolases/química , Monoéster Fosfórico Hidrolases/ultraestrutura , Thermococcus/enzimologia , Sequência de Aminoácidos , Sítios de Ligação , Simulação por Computador , Ativação Enzimática , Estabilidade Enzimática , Hidrolases/isolamento & purificação , Conformação Molecular , Dados de Sequência Molecular , Monoéster Fosfórico Hidrolases/isolamento & purificação , Ligação Proteica , Especificidade da Espécie , Relação Estrutura-Atividade , Especificidade por Substrato , Thermococcus/classificação
10.
Nucleic Acids Res ; 42(9): 5937-48, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24682817

RESUMO

Double-stranded ribonucleic acid-activated protein kinase (PKR) downregulates translation as a defense mechanism against viral infection. In fish species, PKZ, a PKR-like protein kinase containing left-handed deoxyribonucleic acid (Z-DNA) binding domains, performs a similar role in the antiviral response. To understand the role of PKZ in Z-DNA recognition and innate immune response, we performed structural and functional studies of the Z-DNA binding domain (Zα) of PKZ from Carassius auratus (caZαPKZ). The 1.7-Å resolution crystal structure of caZαPKZ:Z-DNA revealed that caZαPKZ shares the overall fold with other Zα, but has discrete structural features that differentiate its DNA binding mode from others. Functional analyses of caZαPKZ and its mutants revealed that caZαPKZ mediates the fastest B-to-Z transition of DNA among Zα, and the minimal interaction for Z-DNA recognition is mediated by three backbone phosphates and six residues of caZαPKZ. Structure-based mutagenesis and B-to-Z transition assays confirmed that Lys56 located in the ß-wing contributes to its fast B-to-Z transition kinetics. Investigation of the DNA binding kinetics of caZαPKZ further revealed that the B-to-Z transition rate is positively correlated with the association rate constant. Taking these results together, we conclude that the positive charge in the ß-wing largely affects fast B-to-Z transition activity by enhancing the DNA binding rate.


Assuntos
DNA Forma Z/química , Proteínas de Peixes/química , Carpa Dourada , eIF-2 Quinase/química , Sequência de Aminoácidos , Animais , Domínio Catalítico , Cristalografia por Raios X , Ligação de Hidrogênio , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Cloreto de Sódio/química
11.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 3): 615-26, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24598731

RESUMO

Bacterial pathogens have evolved diverse types of efficient machinery to acquire haem, the most abundant source of iron in the human body, and degrade it for the utilization of iron. Gram-positive bacteria commonly encode IsdG-family proteins as haem-degrading monooxygenases. Listeria monocytogenes is predicted to possess an IsdG-type protein (Lmo2213), but the residues involved in haem monooxygenase activity are not well conserved and there is an extra N-terminal domain in Lmo2213. Therefore, its function and mechanism of action cannot be predicted. In this study, the crystal structure of Lmo2213 was determined at 1.75 Šresolution and its haem-binding and haem-degradation activities were confirmed. Structure-based mutational and functional assays of this protein, designated as an Isd-type L. monocytogenes haem-degrading enzyme (Isd-LmHde), identified that Glu71, Tyr87 and Trp129 play important roles in haem degradation and that the N-terminal domain is also critical for its haem-degrading activity. The haem-degradation product of Isd-LmHde is verified to be biliverdin, which is also known to be the degradation product of other bacterial haem oxygenases. This study, the first structural and functional report of the haem-degradation system in L. monocytogenes, sheds light on the concealed haem-utilization system in this life-threatening human pathogen.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/fisiologia , Heme/metabolismo , Listeria monocytogenes/enzimologia , Oxigenases/química , Oxigenases/fisiologia , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/genética , Biliverdina/química , Biliverdina/metabolismo , Catálise , Cristalografia por Raios X , Heme/química , Humanos , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oxigenases/genética
12.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 3): 694-707, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24598739

RESUMO

Escherichia coli SdiA is a quorum-sensing (QS) receptor that responds to autoinducers produced by other bacterial species to control cell division and virulence. Crystal structures reveal that E. coli SdiA, which is composed of an N-terminal ligand-binding domain and a C-terminal DNA-binding domain (DBD), forms a symmetrical dimer. Although each domain shows structural similarity to other QS receptors, SdiA differs from them in the relative orientation of the two domains, suggesting that its ligand-binding and DNA-binding functions are independent. Consistently, in DNA gel-shift assays the binding affinity of SdiA for the ftsQP2 promoter appeared to be insensitive to the presence of autoinducers. These results suggest that autoinducers increase the functionality of SdiA by enhancing the protein stability rather than by directly affecting the DNA-binding affinity. Structural analyses of the ligand-binding pocket showed that SdiA cannot accommodate ligands with long acyl chains, which was corroborated by isothermal titration calorimetry and thermal stability analyses. The formation of an intersubunit disulfide bond that might be relevant to modulation of the DNA-binding activity was predicted from the proximal position of two Cys residues in the DBDs of dimeric SdiA. It was confirmed that the binding affinity of SdiA for the uvrY promoter was reduced under oxidizing conditions, which suggested the possibility of regulation of SdiA by multiple independent signals such as quorum-sensing inducers and the oxidation state of the cell.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Percepção de Quorum , Transativadores/química , Transativadores/metabolismo , Cristalografia por Raios X , Ligantes , Ligases/química , Ligases/metabolismo , Ligação Proteica , Transdução de Sinais , Especificidade da Espécie
13.
J Mater Chem B ; 1(10): 1453-1460, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-32260785

RESUMO

Nanostructured magnetic materials such as iron oxide and bimetallic nanoparticles can be potentially applied to a variety of fields, including electronics and nanomedicine. To develop these applications, it is important to control their particle size which affects their magnetic properties. In particular, it is a major challenge to synthesize small-sized nanoparticles with high reproducibility. In this study, we synthesized cobalt-platinum nanoparticles (CoPt NPs) in an ambient solution phase using PepA, a bacterial aminopeptidase, as a protein shell, and investigated the physicochemical and magnetic properties of NPs with and without encapsulating proteins. The size of CoPt NPs encapsulated by PepA was stringently controlled from 1.1 to 2.8 nm, and their magnetic property was related to the size. The CoPt NPs with the diameter of 1.1 nm showed a superparamagnetic behavior only at low temperatures, while 2.1 and 2.8 nm CoPt NPs were ferromagnetic below the blocking temperature. PepA had no deleterious effects on the coercivity of CoPt NPs, as evidenced by the marginal effect of PepA on the coercivity of CoPt NPs. This study demonstrated that the particle size and magnetic property of CoPt NPs can be controlled by using PepA as a protein shell. Encapsulation by PepA will aid the development of multifunctional magnetic materials, since the biocompatibility and modification capability of PepA can be synergistically combined with the advanced functionalities of CoPt NPs.

14.
J Mol Biol ; 414(2): 231-42, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21996443

RESUMO

Endorepellin, the C-terminal region of perlecan, inhibits angiogenesis by disrupting actin cytoskeleton and focal adhesions. The C-terminal laminin-like globular domain (LG3) of endorepellin directs most of this antiangiogenic activity. To investigate the angiostatic mechanism and to identify structural determinants, we have solved crystal structures of the LG3 domain in both apo- and calcium-bound forms at resolutions of 1.5 Å and 2.8 Å, respectively. The conserved core has the jellyroll fold characteristic of LG domains. The calcium-induced structural changes seem very restricted, and the calcium binding site appears to be preformed, suggesting that the bound calcium ion, rather than structural rearrangements, contributes to antiangiogenesis. We have identified H4268 on the EF loop as a key residue for the biochemical function of LG3, since its mutation abolishes antiangiogenic activity, and mutant LG3 can no longer form a direct interaction with integrin. Taken together, we propose that these two distinct structural elements contribute to the angiostatic effect of endorepellin.


Assuntos
Inibidores da Angiogênese/química , Proteoglicanas de Heparan Sulfato/química , Fragmentos de Peptídeos/química , Sequência de Aminoácidos , Inibidores da Angiogênese/metabolismo , Inibidores da Angiogênese/farmacologia , Sítios de Ligação , Cálcio/metabolismo , Cristalografia por Raios X , Proteoglicanas de Heparan Sulfato/metabolismo , Proteoglicanas de Heparan Sulfato/farmacologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Homologia de Sequência de Aminoácidos
15.
Proc Natl Acad Sci U S A ; 107(47): 20281-6, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-21059944

RESUMO

The receptor activator of nuclear factor-κB (RANK) and its ligand RANKL, which belong to the tumor necrosis factor (TNF) receptor-ligand family, mediate osteoclastogenesis. The crystal structure of the RANKL ectodomain (eRANKL) in complex with the RANK ectodomain (eRANK) combined with biochemical assays of RANK mutants indicated that three RANK loops (Loop1, Loop2, and Loop3) bind to the interface of a trimeric eRANKL. Loop3 is particularly notable in that it is structurally distinctive from other TNF-family receptors and forms extensive contacts with RANKL. The disulfide bond (C125-C127) at the tip of Loop3 is important for determining the unique topology of Loop3, and docking E126 close to RANKL, which was supported by the inability of C127A or E126A mutants of RANK to bind to RANKL. Inhibitory activity of RANK mutants, which contain loops of osteoprotegerin (OPG), a soluble decoy receptor to RANKL, confirmed that OPG shares the similar binding mode with RANK and OPG. Loop3 plays a key role in RANKL binding. Peptide inhibitors designed to mimic Loop3 blocked the RANKL-induced differentiation of osteoclast precursors, suggesting that they could be developed as therapeutic agents for the treatment of osteoporosis and bone-related diseases. Furthermore, some of the RANK mutations associated with autosomal recessive osteopetrosis (ARO) resulted in reduced RANKL-binding activity and failure to induce osteoclastogenesis. These results, together with structural interpretation of eRANK-eRANKL interaction, provided molecular understanding for pathogenesis of ARO.


Assuntos
Osso e Ossos/metabolismo , Modelos Moleculares , Oligopeptídeos/farmacologia , Osteopetrose/metabolismo , Osteoprotegerina/metabolismo , Peptídeos Cíclicos/farmacologia , Ligante RANK/química , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Cristalografia , Camundongos , Mutagênese Sítio-Dirigida , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteopetrose/genética , Ligante RANK/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/genética
16.
Protein Sci ; 19(6): 1258-63, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20512978

RESUMO

The sigmaE-dependent stress response in bacterial cells is initiated by the DegS- and RseP-regulated intramembrane proteolysis of a membrane-spanning antisigma factor, RseA. RseB binds to RseA and inhibits its sequential cleavage, thereby functioning as a negative modulator of this response. In the crystal structure of the periplasmic domain of RseA bound to RseB, the DegS cleavage site of RseA is unstructured, however, its P1 residue is buried in the hydrophobic pocket of RseB, which suggests that RseB binding blocks the access of DegS to the cleavage site.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Membrana/química , Fatores de Transcrição/química , Sequência de Aminoácidos , Domínio Catalítico , Endopeptidases/química , Endopeptidases/genética , Endopeptidases/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Bactérias Gram-Negativas/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Ligação Proteica , Alinhamento de Sequência , Estresse Fisiológico , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
Mol Cells ; 29(4): 397-405, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20238176

RESUMO

UDP-glucose pyrophosphorylases (UGPase; EC 2.7.7.9) catalyze the conversion of UTP and glucose-1-phosphate to UDP-glucose and pyrophosphate and vice versa. Prokaryotic UGPases are distinct from their eukaryotic counterparts and are considered appropriate targets for the development of novel antibacterial agents since their product, UDP-glucose, is indispensable for the biosynthesis of virulence factors such as lipopolysaccharides and capsular polysaccharides. In this study, the crystal structures of UGPase from Helicobacter pylori (HpUGPase) were determined in apo- and UDP-glucose/Mg(2+)-bound forms at 2.9 A and 2.3 A resolutions, respectively. HpUGPase is a homotetramer and its active site is located in a deep pocket of each subunit. Magnesium ion is coordinated by Asp130, two oxygen atoms of phosphoryl groups, and three water molecules with octahedral geometry. Isothermal titration calorimetry analyses demonstrated that Mg(2+) ion plays a key role in the enzymatic activity of UGPase by enhancing the binding of UGPase to UTP or UDP-glucose, suggesting that this reaction is catalyzed by an ordered sequential Bi Bi mechanism. Furthermore, the crystal structure explains the specificity for uracil bases. The current structural study combined with functional analyses provides essential information for understanding the reaction mechanism of bacterial UGPases, as well as a platform for the development of novel antibacterial agents.


Assuntos
Proteínas de Bactérias/química , Helicobacter pylori/enzimologia , Relação Quantitativa Estrutura-Atividade , UTP-Glucose-1-Fosfato Uridililtransferase/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Calorimetria/métodos , Catálise , Domínio Catalítico/genética , Cristalografia por Raios X , Glucofosfatos/química , Glucofosfatos/metabolismo , Cinética , Magnésio/química , Magnésio/metabolismo , Modelos Químicos , Modelos Moleculares , Estrutura Molecular , Mutação , Conformação Proteica , Estrutura Terciária de Proteína , Especificidade por Substrato , UTP-Glucose-1-Fosfato Uridililtransferase/genética , UTP-Glucose-1-Fosfato Uridililtransferase/metabolismo , Uridina Difosfato Glucose/química , Uridina Difosfato Glucose/metabolismo
18.
Curr Drug Targets ; 11(3): 335-44, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20210758

RESUMO

Z-DNA, the alternative form of double-stranded DNA involved in a variety of nucleotide metabolism, is recognized and stabilized by specific Z-DNA binding proteins (ZBPs). Three ZBPs known in vertebrates -ADAR1, DAI and PKZ- modulate innate immunity, particularly, the IFN-induced immune response. The E3L protein of the vaccinia virus appears to compete with the host ZBP for Z-DNA binding, thereby suppressing the host immune system. ZBPs are, therefore, considered to be attractive therapeutic targets for infectious and immune diseases. Recent advances in computer-aided drug development combined with the high-resolution crystal and NMR structures of ZBPs have enabled us to discover novel candidates as ZBP inhibitors. In this study, we present an overview of Z-DNA and known ZBPs as drug targets, and summarize recent progress in the structure-based identification of ZBP inhibitors.


Assuntos
DNA Forma Z/metabolismo , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Animais , Desenho Assistido por Computador , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Humanos , Doenças do Sistema Imunitário/tratamento farmacológico , Doenças do Sistema Imunitário/fisiopatologia , Infecções/tratamento farmacológico , Infecções/microbiologia , Ligação Proteica
19.
Biochem Biophys Res Commun ; 383(4): 386-91, 2009 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-19268655

RESUMO

Endoglin, an accessory membrane receptor of transforming growth factor-beta (TGF-beta)1, modulates the cellular response to TGF-beta via its interaction with type I and II TGF-beta receptors. It has been considered a promising target for the development of therapeutics and cancer markers. We have established stable CHO cell lines that efficiently secrete soluble endoglin (s-endoglin) fused with human growth hormone. Two oligomeric forms were observed in a homogeneous preparation of s-endoglin, as a dimer and a tetramer. The dimeric s-endoglin enhanced TGF-beta responsiveness in U937 cells, thus proving its potential for therapeutic applications. Small angle X-ray scattering (SAXS) experiments revealed elongated conformations of both dimeric and tetrameric s-endoglins in solution, suggesting that s-endoglin might undergo conformational adaptations upon TGF-beta binding. The current results provide important references and material for high-resolution structural studies and for medical applications of s-endoglin.


Assuntos
Antígenos CD/química , Antígenos CD/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Animais , Antígenos CD/genética , Células CHO , Cricetinae , Cricetulus , Endoglina , Genes Reporter , Humanos , Luciferases/genética , Conformação Proteica , Multimerização Proteica , Receptores de Superfície Celular/genética , Espalhamento de Radiação , Raios X
20.
Artigo em Inglês | MEDLINE | ID: mdl-19255480

RESUMO

PKZ, a PKR-like eIF2alpha kinase, consists of a Z-DNA-specific binding domain (Zalpha) and an eIF2alpha kinase domain. The kinase activity of PKZ is modulated by the binding of Zalpha to Z-DNA. The mechanisms underlying Z-DNA binding and the subsequent stimulation of PKZ raise intriguing questions. Interestingly, the Z-DNA-binding domain of PKZ from goldfish (Carassius auratus; caZalpha(PKZ)) shows limited sequence homology to other canonical Zalpha domains, suggesting that it may have a distinct Z-DNA-recognition mode. In this study, the Z-DNA-binding activity and stoichiometry of caZalpha(PKZ) were confirmed using circular dichroism (CD). In addition, preliminary X-ray studies of the caZalpha(PKZ)-Z-DNA complex are reported as the first step in the determination of its three-dimensional structure. Bacterially expressed recombinant caZalpha(PKZ) was purified and crystallized with Z-DNA at 295 K using the microbatch method. X-ray diffraction data were collected to 1.7 A resolution with an R(merge) of 7.4%. The crystals belonged to the monoclinic space group C2, with unit-cell parameters a = 55.54, b = 49.93, c = 29.44 A, beta = 96.5 degrees . Structural analysis of caZalpha(PKZ)-Z-DNA will reveal the binding mode of caZalpha(PKZ) to Z-DNA and its relevance to other Z-DNA-binding proteins.


Assuntos
DNA Forma Z/química , DNA Forma Z/metabolismo , Carpa Dourada/metabolismo , eIF-2 Quinase/química , eIF-2 Quinase/metabolismo , Animais , Dicroísmo Circular , Cristalização , Cristalografia por Raios X , Ligação Proteica , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...