Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 295(34): 12290-12304, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32651231

RESUMEN

Agrobacterium tumefaciens infects various plants and causes crown gall diseases involving temporal expression of virulence factors. SghA is a newly identified virulence factor enzymatically releasing salicylic acid from its glucoside conjugate and controlling plant tumor development. Here, we report the structural basis of SghR, a LacI-type transcription factor highly conserved in Rhizobiaceae family, regulating the expression of SghA and involved in tumorigenesis. We identified and characterized the binding site of SghR on the promoter region of sghA and then determined the crystal structures of apo-SghR, SghR complexed with its operator DNA, and ligand sucrose, respectively. These results provide detailed insights into how SghR recognizes its cognate DNA and shed a mechanistic light on how sucrose attenuates the affinity of SghR with DNA to modulate the expression of SghA. Given the important role of SghR in mediating the signaling cross-talk during Agrobacterium infection, our results pave the way for structure-based inducer analog design, which has potential applications for agricultural industry.


Asunto(s)
Agrobacterium tumefaciens/metabolismo , Proteínas Bacterianas/metabolismo , Tumores de Planta/microbiología , Elementos de Respuesta , Transducción de Señal , Agrobacterium tumefaciens/genética , Proteínas Bacterianas/genética
2.
Arch Toxicol ; 95(1): 355-374, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32909075

RESUMEN

Nuclear receptors (NRs) are key regulators of energy homeostasis, body development, and sexual reproduction. Xenobiotics binding to NRs may disrupt natural hormonal systems and induce undesired adverse effects in the body. However, many chemicals of concerns have limited or no experimental data on their potential or lack-of-potential endocrine-disrupting effects. Here, we propose a virtual screening method based on molecular docking for predicting potential endocrine-disrupting chemicals (EDCs) that bind to NRs. For 12 NRs, we systematically analyzed how multiple crystal structures can be used to distinguish actives and inactives found in previous high-throughput experiments. Our method is based on (i) consensus docking scores from multiple structures at a single functional state (agonist-bound or antagonist-bound), (ii) multiple functional states (agonist-bound and antagonist-bound), and (iii) multiple pockets (orthosteric site and alternative sites) of these NRs. We found that the consensus enrichment from multiple structures is better than or comparable to the best enrichment from a single structure. The discriminating power of this consensus strategy was further enhanced by a chemical similarity-weighted scoring scheme, yielding better or comparable enrichment for all studied NRs. Applying this optimized method, we screened 252 fatty acids against peroxisome proliferator-activated receptor gamma (PPARγ) and successfully identified 3 previously unknown fatty acids with Kd = 100-250 µM including two furan fatty acids: furannonanoic acid (FNA) and furanundecanoic acid (FUA), and one cyclopropane fatty acid: phytomonic acid (PTA). These results suggested that the proposed method can be used to rapidly screen and prioritize potential EDCs for further experimental evaluations.


Asunto(s)
Disruptores Endocrinos/metabolismo , Ácidos Grasos/metabolismo , Simulación del Acoplamiento Molecular , PPAR gamma/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Pruebas de Toxicidad , Sitios de Unión , Bases de Datos de Proteínas , Disruptores Endocrinos/química , Disruptores Endocrinos/toxicidad , Ácidos Grasos/química , Ácidos Grasos/toxicidad , Estudios de Factibilidad , Ligandos , PPAR gamma/química , PPAR gamma/efectos de los fármacos , Unión Proteica , Conformación Proteica , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Medición de Riesgo , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie
3.
Mol Cell ; 39(5): 773-83, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20832728

RESUMEN

Eukaryotic mRNA degradation often occurs in a process whereby translation initiation is inhibited and the mRNA is targeted for decapping. In yeast cells, Pat1, Scd6, Edc3, and Dhh1 all function to promote decapping by an unknown mechanism(s). We demonstrate that purified Scd6 and a region of Pat1 directly repress translation in vitro by limiting the formation of a stable 48S preinitiation complex. Moreover, while Pat1, Edc3, Dhh1, and Scd6 all bind the decapping enzyme, only Pat1 and Edc3 enhance its activity. We also identify numerous direct interactions between Pat1, Dcp1, Dcp2, Dhh1, Scd6, Edc3, Xrn1, and the Lsm1-7 complex. These observations identify three classes of decapping activators that function to directly repress translation initiation and/or stimulate Dcp1/2. Moreover, Pat1 is identified as critical in mRNA decay by first inhibiting translation initiation, then serving as a scaffold to recruit components of the decapping complex, and finally activating Dcp2.


Asunto(s)
Estabilidad del ARN/fisiología , ARN de Hongos/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Iniciación de la Cadena Peptídica Traduccional/fisiología , ARN de Hongos/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
4.
Genes Dev ; 24(3): 290-300, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20123908

RESUMEN

The Hippo signaling pathway controls cell growth, proliferation, and apoptosis by regulating the expression of target genes that execute these processes. Acting downstream from this pathway is the YAP transcriptional coactivator, whose biological function is mediated by the conserved TEAD family transcription factors. The interaction of YAP with TEADs is critical to regulate Hippo pathway-responsive genes. Here, we describe the crystal structure of the YAP-interacting C-terminal domain of TEAD4 in complex with the TEAD-interacting N-terminal domain of YAP. The structure reveals that the N-terminal region of YAP is folded into two short helices with an extended loop containing the PXXPhiP motif in between, while the C-terminal domain of TEAD4 has an immunoglobulin-like fold. YAP interacts with TEAD4 mainly through the two short helices. Point mutations of TEAD4 indicate that the residues important for YAP interaction are required for its transforming activity. Mutagenesis reveals that the PXXPhiP motif of YAP, although making few contacts with TEAD4, is important for TEAD4 interaction as well as for the transforming activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Proteínas de Unión al ADN/química , Proteínas Musculares/química , Fosfoproteínas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Factores de Transcripción/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular , Cristalografía por Rayos X , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Ratones , Modelos Moleculares , Proteínas Musculares/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Factores de Transcripción de Dominio TEA , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteínas Señalizadoras YAP
5.
Am J Hum Genet ; 95(5): 590-601, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25439726

RESUMEN

Using a combination of exome sequencing and linkage analysis, we investigated an English family with two affected siblings in their 40s with recessive Charcot-Marie Tooth disease type 2 (CMT2). Compound heterozygous mutations in the immunoglobulin-helicase-µ-binding protein 2 (IGHMBP2) gene were identified. Further sequencing revealed a total of 11 CMT2 families with recessively inherited IGHMBP2 gene mutations. IGHMBP2 mutations usually lead to spinal muscular atrophy with respiratory distress type 1 (SMARD1), where most infants die before 1 year of age. The individuals with CMT2 described here, have slowly progressive weakness, wasting and sensory loss, with an axonal neuropathy typical of CMT2, but no significant respiratory compromise. Segregating IGHMBP2 mutations in CMT2 were mainly loss-of-function nonsense in the 5' region of the gene in combination with a truncating frameshift, missense, or homozygous frameshift mutations in the last exon. Mutations in CMT2 were predicted to be less aggressive as compared to those in SMARD1, and fibroblast and lymphoblast studies indicate that the IGHMBP2 protein levels are significantly higher in CMT2 than SMARD1, but lower than controls, suggesting that the clinical phenotype differences are related to the IGHMBP2 protein levels.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Exoma/genética , Modelos Moleculares , Mutación Missense/genética , Fenotipo , Adulto , Secuencia de Bases , Enfermedad de Charcot-Marie-Tooth/patología , Mapeo Cromosómico , Femenino , Haplotipos/genética , Humanos , Datos de Secuencia Molecular , Linaje , Mapeo de Interacción de Proteínas , Análisis de Secuencia de ADN , Nervio Sural/patología
6.
Genes Dev ; 23(9): 1106-18, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19417105

RESUMEN

Eukaryotic translation termination is mediated by two interacting release factors, eRF1 and eRF3, which act cooperatively to ensure efficient stop codon recognition and fast polypeptide release. The crystal structures of human and Schizosaccharomyces pombe full-length eRF1 in complex with eRF3 lacking the GTPase domain revealed details of the interaction between these two factors and marked conformational changes in eRF1 that occur upon binding to eRF3, leading eRF1 to resemble a tRNA molecule. Small-angle X-ray scattering analysis of the eRF1/eRF3/GTP complex suggested that eRF1's M domain contacts eRF3's GTPase domain. Consistently, mutation of Arg192, which is predicted to come in close contact with the switch regions of eRF3, revealed its important role for eRF1's stimulatory effect on eRF3's GTPase activity. An ATP molecule used as a crystallization additive was bound in eRF1's putative decoding area. Mutational analysis of the ATP-binding site shed light on the mechanism of stop codon recognition by eRF1.


Asunto(s)
Codón de Terminación/metabolismo , Modelos Moleculares , Factores de Terminación de Péptidos/química , Proteínas de Schizosaccharomyces pombe/química , Schizosaccharomyces , Adenosina Trifosfato/metabolismo , GTP Fosfohidrolasas/metabolismo , Orden Génico , Humanos , Mutación , Factores de Terminación de Péptidos/genética , Unión Proteica , Biosíntesis de Proteínas/genética , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Reproducibilidad de los Resultados , Ribosomas/metabolismo , Dispersión del Ángulo Pequeño , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo
7.
Gastroenterology ; 149(4): 886-9.e5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26122142

RESUMEN

Hamartomatous polyposis syndromes (HPS) account for a small but appreciable proportion of inherited gastrointestinal cancer predisposition syndromes; patients with HPS have an increased risk for colon and extracolonic malignancies. We present a unique case of familial juvenile polyposis syndrome associated with gastrointestinal ganglioneuromas of unknown etiology. The patient was tested for HPS-associated genes, but no mutation was detected. Exome sequencing identified a germline heterozygous mutation in SMAD9 (SMAD9(V90M)). This mutation was predicted to be an activating mutation. HEK cells transfected to express SMAD9(V90M) had reduced expression of phosphatase and tensin homolog; this reduction was also observed in a polyp from the patient. We have therefore identified a new susceptibility locus for HPS. Patients with hamartomatous polyposis in the colon associated with ganglioneuromatosis should be referred for genetic assessments.


Asunto(s)
Pólipos del Colon/genética , Neoplasias del Sistema Digestivo/genética , Exoma , Ganglioneuroma/genética , Mutación de Línea Germinal , Neoplasia Endocrina Múltiple Tipo 2b/genética , Fosfohidrolasa PTEN/metabolismo , Síndrome de Peutz-Jeghers/genética , Proteína Smad8/genética , Adulto , Pólipos del Colon/diagnóstico , Pólipos del Colon/enzimología , Análisis Mutacional de ADN , Neoplasias del Sistema Digestivo/diagnóstico , Neoplasias del Sistema Digestivo/enzimología , Regulación hacia Abajo , Femenino , Ganglioneuroma/diagnóstico , Ganglioneuroma/enzimología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Masculino , Neoplasia Endocrina Múltiple Tipo 2b/diagnóstico , Neoplasia Endocrina Múltiple Tipo 2b/enzimología , Fosfohidrolasa PTEN/genética , Síndrome de Peutz-Jeghers/diagnóstico , Síndrome de Peutz-Jeghers/enzimología , Fenotipo , Proteína Smad8/metabolismo , Transfección
8.
Proc Natl Acad Sci U S A ; 110(51): 20765-70, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24319092

RESUMEN

The human pathogen Pseudomonas aeruginosa coordinates the expression of virulence factors by using quorum sensing (QS), a signaling cascade triggered by the QS signal molecule and its receptor, a member of the LuxR family of QS transcriptional factors (LasR). The QS threshold and response in P. aeruginosa is defined by a QS LasR-specific antiactivator (QslA), which binds to LasR and prevents it from binding to its target promoter. However, how QslA binds to LasR and regulates its DNA binding activity in QS remains elusive. Here we report the crystal structure of QslA in complex with the N-terminal ligand binding domain of LasR. QsIA exists as a functional dimer to interact with the LasR ligand binding domain. Further analysis shows that QsIA binding occupies the LasR dimerization interface and consequently disrupts LasR dimerization, thereby preventing LasR from binding to its target DNA and disturbing normal QS. Our findings provide a structural model for understanding the QslA-mediated antiactivation mechanism in QS through protein-protein interaction.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Modelos Moleculares , Multimerización de Proteína , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/metabolismo , Percepción de Quorum/fisiología , Transactivadores/química , Transactivadores/metabolismo , Proteínas Bacterianas/genética , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Humanos , Unión Proteica , Estructura Terciaria de Proteína , Pseudomonas aeruginosa/genética , Transactivadores/genética
9.
Proc Natl Acad Sci U S A ; 109(38): 15479-84, 2012 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-22949660

RESUMEN

Many bacterial pathogens produce diffusible signal factor (DSF)-type quorum sensing (QS) signals in modulation of virulence and biofilm formation. Previous work on Xanthomonas campestris showed that the RpfC/RpfG two-component system is involved in sensing and responding to DSF signals, but little is known in other microorganisms. Here we show that in Burkholderia cenocepacia the DSF-family signal cis-2-dodecenoic acid (BDSF) negatively controls the intracellular cyclic dimeric guanosine monophosphate (c-di-GMP) level through a receptor protein RpfR, which contains Per/Arnt/Sim (PAS)-GGDEF-EAL domains. RpfR regulates the same phenotypes as BDSF including swarming motility, biofilm formation, and virulence. In addition, the BDSF(-) mutant phenotypes could be rescued by in trans expression of RpfR, or its EAL domain that functions as a c-di-GMP phosphodiesterase. BDSF is shown to bind to the PAS domain of RpfR with high affinity and stimulates its phosphodiesterase activity through induction of allosteric conformational changes. Our work presents a unique and widely conserved DSF-family signal receptor that directly links the signal perception to c-di-GMP turnover in regulation of bacterial physiology.


Asunto(s)
Burkholderia cenocepacia/genética , Ácidos Grasos Monoinsaturados/química , Guanosina Monofosfato/química , Percepción de Quorum/genética , Receptores de Superficie Celular/química , Proteínas Bacterianas/metabolismo , Burkholderia cenocepacia/metabolismo , Comunicación Celular , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Dimerización , Modelos Genéticos , Mutagénesis , Mutación , Fenotipo , Unión Proteica , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Virulencia
10.
Biochim Biophys Acta ; 1829(6-7): 580-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23287066

RESUMEN

The regulation of mRNA degradation is critical for proper gene expression. Many major pathways for mRNA decay involve the removal of the 5' 7-methyl guanosine (m(7)G) cap in the cytoplasm to allow for 5'-to-3' exonucleolytic decay. The most well studied and conserved eukaryotic decapping enzyme is Dcp2, and its function is aided by co-factors and decapping enhancers. A subset of these factors can act to enhance the catalytic activity of Dcp2, while others might stimulate the remodeling of proteins bound to the mRNA substrate that may otherwise inhibit decapping. Structural studies have provided major insights into the mechanisms by which Dcp2 and decapping co-factors activate decapping. Additional mRNA decay factors can function by recruiting components of the decapping machinery to target mRNAs. mRNA decay factors, decapping factors, and mRNA substrates can be found in cytoplasmic foci named P bodies that are conserved in eukaryotes, though their function remains unknown. In addition to Dcp2, other decapping enzymes have been identified, which may serve to supplement the function of Dcp2 or act in independent decay or quality control pathways. This article is part of a Special Issue entitled: RNA Decay mechanisms.


Asunto(s)
Endorribonucleasas/genética , Caperuzas de ARN/genética , Estabilidad del ARN/genética , Catálisis , Citoplasma , Endorribonucleasas/química , Eucariontes/enzimología , Eucariontes/genética , Humanos , Conformación Proteica , Estructura Terciaria de Proteína , Análogos de Caperuza de ARN/química , Análogos de Caperuza de ARN/genética , Caperuzas de ARN/química
11.
Protein Expr Purif ; 102: 13-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25050461

RESUMEN

Interferon-inducible protein 16 (IFI16) is a multifunctional p200-family protein that plays pivotal roles in p53-mediated apoptosis, tumor suppression and DNA damage response. Recently, another function of IFI16 in innate immune sensing and response has been uncovered, in which IFI16 recognizes the exogenous DNAs through cooperative binding of DNAs via its two DNA binding domains, HINa and HINb. Although the mechanism by which the HINb domain recognizes DNAs has been elucidated, the molecular basis of the cooperativity between HINa and HINb during DNA recognition process is still not clear. Here we report expression and purification of a truncated human IFI16 protein (HINab-GS) containing HINa in tandem with HINb with the joining region between HINa and HINb replaced by a short GS linker in Escherichia coli. DNA binding activities of HINab-GS to various double-stranded DNAs (dsDNAs) of different lengths were then examined using electrophoretic mobility shift assays. HINab-GS exhibited efficient binding activity to dsDNAs, and its DNA binding affinity correlated positively with the length of dsDNAs. A co-crystallization condition of HINab-GS bound to a 30 bp dsDNA derived from vaccinia virus was also found. Together, our work provides a basis for structurally elucidating the mechanism governing cooperative DNA recognition by IFI16.


Asunto(s)
Clonación Molecular , ADN Viral/metabolismo , Escherichia coli/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Secuencia de Aminoácidos , Cristalización , Ensayo de Cambio de Movilidad Electroforética , Humanos , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/aislamiento & purificación , Fosfoproteínas/química , Fosfoproteínas/aislamiento & purificación , Unión Proteica
12.
Nucleic Acids Res ; 40(21): 11009-22, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22965130

RESUMEN

Mutations in immunoglobulin µ-binding protein 2 (Ighmbp2) cause distal spinal muscular atrophy type 1 (DSMA1), an autosomal recessive disease that is clinically characterized by distal limb weakness and respiratory distress. However, despite extensive studies, the mechanism of disease-causing mutations remains elusive. Here we report the crystal structures of the Ighmbp2 helicase core with and without bound RNA. The structures show that the overall fold of Ighmbp2 is very similar to that of Upf1, a key helicase involved in nonsense-mediated mRNA decay. Similar to Upf1, domains 1B and 1C of Ighmbp2 undergo large conformational changes in response to RNA binding, rotating 30° and 10°, respectively. The RNA binding and ATPase activities of Ighmbp2 are further enhanced by the R3H domain, located just downstream of the helicase core. Mapping of the pathogenic mutations of DSMA1 onto the helicase core structure provides a molecular basis for understanding the disease-causing consequences of Ighmbp2 mutations.


Asunto(s)
Proteínas de Unión al ADN/química , Atrofia Muscular Espinal/genética , Mutación Missense , ARN Helicasas/química , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Factores de Transcripción/química , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , ARN/química , ARN Helicasas/genética , Transactivadores/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Adv Healthc Mater ; 13(16): e2303568, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38319010

RESUMEN

High reactive oxygen species (ROS) levels in tumor microenvironment (TME) impair both immunogenic cell death (ICD) efficacy and T cell activity. Furthermore, tumor escapes immunosurveillance via programmed death-1/programmed death ligand-1 (PD-L1) signal, and the insufficient intracellular hydrogen peroxide weakens ferroptosis efficacy. To tackle the above issues, a glutathione (GSH)/ROS/pH triple-responsive prodrug nanomedicine that encapsulates Fe2O3 nanoparticle via electrostatic interaction is constructed for magnetic resonance imaging (MRI)-guided multi-mode theranostics with chemotherapy/ferroptosis/immunotherapy. The diselenide bond consumes ROS in TME to increase T cells and ICD efficacy, the cleavage of which facilitates PD-L1 antagonist D peptide release to block immune checkpoint. After intracellular internalization, Fe2O3 nanoparticle is released in the acidic endosome for MRI simultaneously with lipid peroxides generation for tumor ferroptosis. Doxorubicin is cleaved from polymers in the condition of high intracellular GSH level accompanied by tumor ICD, which simultaneously potentiates ferroptosis by NADPH oxidase mediated H2O2 self-generation. In vivo results indicate that the nanoplatform strengthens tumor ICD, induces cytotoxic T lymphocytes proliferation, inhibits 4T1 tumor regression and metastasis, and prolongs survival median. In all, a new strategy is proposed in strengthening ICD and T cells activity cascade with ferroptosis as well as immune checkpoint blockade for effective tumor immunotherapy.


Asunto(s)
Ferroptosis , Peróxido de Hidrógeno , Inmunoterapia , Profármacos , Especies Reactivas de Oxígeno , Peróxido de Hidrógeno/química , Profármacos/química , Profármacos/farmacología , Profármacos/uso terapéutico , Ferroptosis/efectos de los fármacos , Animales , Ratones , Especies Reactivas de Oxígeno/metabolismo , Inmunoterapia/métodos , Microambiente Tumoral/efectos de los fármacos , Humanos , Imagen por Resonancia Magnética/métodos , Polímeros/química , Línea Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacología , Nanopartículas/química , Ratones Endogámicos BALB C , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Femenino , Glutatión/metabolismo , Glutatión/química , Nanomedicina Teranóstica/métodos
14.
Adv Mater ; 36(14): e2307923, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38174840

RESUMEN

In vivo optical imaging of trace biomarkers in residual microtumors holds significant promise for cancer prognosis but poses a formidable challenge. Here, a novel hydrogel sensor is designed for ultrasensitive and specific imaging of the elusive biomarker. This hydrogel sensor seamlessly integrates a molecular beacon nanoprobe with fibroblasts, offering both high tissue retention capability and an impressive signal-to-noise ratio for imaging. Signal amplification is accomplished through exonuclease I-mediated biomarker recycling. The resulting hydrogel sensor sensitively detects the biomarker carcinoembryonic antigen with a detection limit of 1.8 pg mL-1 in test tubes. Moreover, it successfully identifies residual cancer nodules with a median diameter of less than 2 mm in mice bearing partially removed primary triple-negative breast carcinomas (4T1). Notably, this hydrogel sensor is proven effective for the sensitive diagnosis of invasive tumors in post-surgical mice with infiltrating 4T1 cells, leveraging the role of fibroblasts in locally enriching tumor cells. Furthermore, the residual microtumor is rapidly photothermal ablation by polydopamine-based nanoprobe under the guidance of visualization, achieving ≈100% suppression of tumor recurrence and lung metastasis. This work offers a promising alternative strategy for visually detecting residual microtumors, potentially enhancing the prognosis of cancer patients following surgical interventions.


Asunto(s)
Hidrogeles , Neoplasias , Humanos , Ratones , Animales
15.
ACS Nano ; 18(12): 8971-8987, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38497600

RESUMEN

Immune checkpoint blockade (ICB) therapy has been approved for breast cancer (BC), but clinical response rates are limited. Recent studies have shown that commensal microbes colonize a variety of tumors and are closely related to the host immune system response. Here, we demonstrated that Fusobacterium nucleatum (F.n), which is prevalent in BC, creates an immunosuppressive tumor microenvironment (ITME) characterized by a high-influx of myeloid cells that hinders ICB therapy. Administering the antibiotic metronidazole in BC can deplete F.n and remodel the ITME. To prevent an imbalance in the systemic microbiota caused by antibiotic administration, we designed a biomimetic nanovehicle for on-site antibiotic delivery inspired by F.n homing to BC. Additionally, ferritin-nanocaged doxorubicin was coloaded into this nanovehicle, as immunogenic chemotherapy has shown potential for synergy with ICB. It has been demonstrated that this biomimetic nanovehicle can be precisely homed to BC and efficiently eliminate intratumoral F.n without disrupting the diversity and abundance of systemic microbiota. This ultimately remodels the ITME, improving the therapeutic efficacy of the PD-L1 blocker with a tumor inhibition rate of over 90% and significantly extending the median survival of 4T1 tumor-bearing mice.


Asunto(s)
Fusobacterium nucleatum , Neoplasias , Animales , Ratones , Antígeno B7-H1 , Biomimética , Antibacterianos , Inmunosupresores , Microambiente Tumoral
16.
EMBO J ; 28(3): 274-85, 2009 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-19153607

RESUMEN

Pdcd4 is a tumour suppressor protein. It inhibits translation through interaction with translation initiator eIF4A, resulting in the suppression of neoplastic transformation and tumour invasion. Here, we present the crystal structures of an N-terminal-truncated Pdcd4 in free form and in complex with eIF4A. Upon binding to eIF4A, Pdcd4 undergoes a marked conformational change to form a heterotrimeric complex with eIF4A, with one Pdcd4 binding to two eIF4A molecules in two different modes. The binding of Pdcd4 to eIF4A is required to inhibit the enzymatic activity of eIF4A, translation initiation, and AP-1-dependent transcription. Both MA3 domains are required to efficiently compete with the C-terminal domain of eIF4G (eIF4Gc) for binding to eIF4A whereas a single MA3 is sufficient to inhibit translation. Our structural and mutational analyses reveal that Pdcd4 inhibits translation initiation by trapping eIF4A in an inactive conformation, and blocking its incorporation into the eIF4F complex.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/metabolismo , Biosíntesis de Proteínas , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cristalografía por Rayos X , ADN Helicasas/antagonistas & inhibidores , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Factor 4A Eucariótico de Iniciación/química , Factor 4G Eucariótico de Iniciación/metabolismo , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Soluciones , Factor de Transcripción AP-1/metabolismo , Transcripción Genética
17.
Adv Healthc Mater ; 12(16): e2203031, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36708144

RESUMEN

DNA hydrogels play an increasingly important role in biomedicine and bioanalysis applications. Due to their high programmability, multifunctionality and biocompatibility, they are often used as effective carriers for packing drugs, cells, or other bioactive cargoes in vitro and in vivo. However, the stability of the DNA hydrogels prevents their in-demand rapid release of cargoes to achieve a full therapeutic effect in time. For bioanalysis, the generation of signals sometimes needs the DNA hydrogel to be rapidly degraded when sensing target molecules. To meet these requirements, stimulus-responsive DNA hydrogels are designed. By responding to different stimuli, self-degradable DNA hydrogels can switch from gel to solution for quantitative bioanalysis and precision cargo delivery. This review summarizes the recently developed innovative methods for designing stimuli-responsive self-degradable DNA hydrogels and showed their applications in the bioanalysis and biomedicines fields. Challenges, as well as prospects, are also discussed.


Asunto(s)
ADN , Hidrogeles , ADN/metabolismo
18.
J Control Release ; 356: 272-287, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36870541

RESUMEN

Abundant cancer-associated fibroblasts (CAFs) in highly fibrotic breast cancer constitute an immunosuppressive barrier for T cell activity and are closely related to the failure of immune checkpoint blockade therapy (ICB). Inspired by the similar antigen-processing capacity of CAFs to professional antigen-presenting cells (APCs), a "turning foes to friends" strategy is proposed by in situ engineering immune-suppressed CAFs into immune-activated APCs for improving response rates of ICB. To achieve safe and specific CAFs engineering in vivo, a thermochromic spatiotemporal photo-controlled gene expression nanosystem was developed by self-assembly of molten eutectic mixture, chitosan andfusion plasmid. After photoactivatable gene expression, CAFs could be engineered as APCs via co-stimulatory molecule (CD86) expression, which effectively induced activation and proliferation of antigen-specific CD8 + T cells. Meanwhile, engineered CAFs could also secrete PD-L1 trap protein in situ for ICB, avoiding potential autoimmune-like disorders caused by "off-target" effects of clinically applied PD-L1 antibody. The study demonstrated that the designed nanosystem could efficiently engineer CAFs, significantly enhance the percentages of CD8+ T cells (4-folds), result in about 85% tumor inhibition rate and 83.3% survival rate at 60 days in highly fibrotic breast cancer, further inducing long-term immune memory effects and effectively inhibiting lung metastasis.


Asunto(s)
Neoplasias de la Mama , Fibroblastos Asociados al Cáncer , Neoplasias Pulmonares , Humanos , Femenino , Inhibidores de Puntos de Control Inmunológico/metabolismo , Antígeno B7-H1 , Fibroblastos Asociados al Cáncer/metabolismo , Inmunoterapia , Neoplasias Pulmonares/metabolismo , Neoplasias de la Mama/metabolismo , Microambiente Tumoral
19.
ACS Nano ; 17(17): 16923-16934, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37606317

RESUMEN

Multidrug resistance (MDR) is a major cause of chemotherapy failure in oncology, and gene therapy is an excellent measure to reverse MDR. However, conventional gene therapy only modulates the expression of MDR-associated proteins but hardly affects their existing function, thus limiting the efficiency of tumor treatment. Herein, we designed a photoactivated DNA nanodrug (MCD@TMPyP4@DOX) to improve tumor chemosensitivity through the downregulation of MDR-related genes and mitochondria-targeted photodynamic therapy (PDT). The self-assembled DNA nanodrug encodes the mucin 1 (MUC1) aptamer and the cytochrome C (CytC) aptamer to facilitate its selective targeting to the mitochondria in tumor cells; the encoded P-gp DNAzyme can specifically cleave the substrate and silence MDR1 mRNA with the help of Mg2+ cofactors. Under near-infrared (NIR) light irradiation, PDT generates reactive oxygen species (ROS) that precisely damage the mitochondria of tumor cells and break single-stranded DNA (ssDNA) to activate MCD@TMPyP4@DOX self-disassembly for release of DOX and DNAzyme. We have demonstrated that this multifunctional DNA nanodrug has high drug delivery capacity and biosafety. It enables downregulation of P-gp expression while reducing the ATP on which P-gp pumps out drugs, improving the latency of gene therapy and synergistically reducing DOX efflux to sensitize tumor chemotherapy. We envision that this gene-modulating DNA nanodrug based on damaging mitochondria is expected to provide an important perspective for sensitizing tumor chemotherapy.


Asunto(s)
ADN Catalítico , Nanopartículas , Resistencia a Antineoplásicos , ADN , ADN de Cadena Simple , Terapia Genética , Mitocondrias , Nanopartículas/uso terapéutico
20.
Nat Commun ; 14(1): 4511, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37500633

RESUMEN

Postoperative tumor recurrence and metastases often lead to cancer treatment failure. Here, we develop a local embedded photodynamic immunomodulatory DNA hydrogel for early warning and inhibition of postoperative tumor recurrence. The DNA hydrogel contains PDL1 aptamers that capture and enrich in situ relapsed tumor cells, increasing local ATP concentration to provide a timely warning signal. When a positive signal is detected, local laser irradiation is performed to trigger photodynamic therapy to kill captured tumor cells and release tumor-associated antigens (TAA). In addition, reactive oxygen species break DNA strands in the hydrogel to release encoded PDL1 aptamer and CpG, which together with TAA promote sufficient systemic antitumor immunotherapy. In a murine model where tumor cells are injected at the surgical site to mimic tumor recurrence, we find that the hydrogel system enables timely detection of tumor recurrence by enriching relapsed tumor cells to increase local ATP concentrations. As a result, a significant inhibitory effect of approximately 88.1% on recurrent tumors and effectively suppressing metastasis, offering a promising avenue for timely and effective treatment of postoperative tumor recurrence.


Asunto(s)
Hidrogeles , Recurrencia Local de Neoplasia , Humanos , Animales , Ratones , Recurrencia Local de Neoplasia/prevención & control , Antígenos de Neoplasias , ADN , Adenosina Trifosfato , Línea Celular Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA