Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 14(51): 56440-56453, 2022 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-36525379

RESUMEN

Extrahepatic nucleic acid delivery using polymers typically requires the synthesis and purification of custom monomers, post-synthetic modifications, and incorporation of additional excipients to augment their stability, endosomal escape, and in vivo effectiveness. Here, we report the development of a single-component and excipient-free, polyester-based nucleic acid delivery nanoparticle platform comprising ionizable N-methyldiethanolamine (MDET) and various hydrophobic alkyl diols (Cp) that achieves lung-selective nucleic acid transfection in vivo. PolyMDET and polyMDET-Cp polyplexes displayed high serum and enzymatic stability, while delivering pDNA or mRNA to "hard-to-transfect" innate immune cells. PolyMDET-C4 and polyMDET-C6 mediated high protein expression in lung alveolar macrophages and dendritic cells without inducing tissue damage or systemic inflammatory responses. Improved strategies using readily available starting materials to produce a simple, excipient-free, non-viral nucleic acid delivery platform with lung-selective and innate immune cell tropism has the potential to expedite clinical deployment of polymer-based genetic medicines.


Asunto(s)
Nanopartículas , Poliésteres , ARN Mensajero/genética , ARN Mensajero/química , Transfección , Plásmidos/genética , ADN/química , Polímeros/química , Pulmón/metabolismo , Nanopartículas/química , Inmunidad Innata
2.
Mol Cancer Ther ; 21(10): 1535-1546, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-35930755

RESUMEN

AZD4625 is a potent, selective, and orally bioavailable inhibitor of oncogenic KRASG12C as demonstrated in cellular assays and in vivo in preclinical cell line-derived and patient-derived xenograft models. In vitro and cellular assays have shown selective binding and inhibition of the KRASG12C mutant isoform, which carries a glycine to cysteine mutation at residue 12, with no binding and inhibition of wild-type RAS or isoforms carrying non-KRASG12C mutations. The pharmacology of AZD4625 shows that it has the potential to provide therapeutic benefit to patients with KRASG12C mutant cancer as either a monotherapy treatment or in combination with other targeted drug agents.


Asunto(s)
Antineoplásicos , Cisteína , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Glicina/farmacología , Humanos , Mutación , Isoformas de Proteínas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Med Chem ; 65(9): 6940-6952, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35471939

RESUMEN

KRAS is an archetypal high-value intractable oncology drug target. The glycine to cysteine mutation at codon 12 represents an Achilles heel that has now rendered this important GTPase druggable. Herein, we report our structure-based drug design approach that led to the identification of 21, AZD4625, a clinical development candidate for the treatment of KRASG12C positive tumors. Highlights include a quinazoline tethering strategy to lock out a bio-relevant binding conformation and an optimization strategy focused on the reduction of extrahepatic clearance mechanisms seen in preclinical species. Crystallographic analysis was also key in helping to rationalize unusual structure-activity relationship in terms of ring size and enantio-preference. AZD4625 is a highly potent and selective inhibitor of KRASG12C with an anticipated low clearance and high oral bioavailability profile in humans.


Asunto(s)
Antineoplásicos , Neoplasias Pulmonares , Antineoplásicos/farmacología , Diseño de Fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Quinazolinas/farmacología , Relación Estructura-Actividad
4.
Sci Rep ; 12(1): 2699, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177674

RESUMEN

The development of covalent inhibitors against KRAS G12C represents a major milestone in treatment of RAS-driven cancers, especially in non-small cell lung cancer (NSCLC), where KRAS G12C is one of the most common oncogenic driver. Here we investigated if additional KRAS mutations co-occur with KRAS G12C (c.34G>T) in NSCLC tumours and if such mutation co-occurrence affects cellular response to G12C-specific inhibitors. Analysis of a large cohort of NSCLC patients whose tumours harboured KRAS mutations revealed co-occurring KRAS mutations in up to 8% of tumours with the KRAS c.34G>T mutation. KRAS c.35G>T was the most frequently co-occurring mutation, and could occur on the same allele (in cis) translating to a single mutant KRAS G12F protein, or on the other allele (in trans), translating to separate G12C and G12V mutant proteins. Introducing KRAS c.35G>T in trans in the KRAS G12C lung cancer model NCI-H358, as well as the co-occurrence in cis in the KRAS G12F lung cancer model NCI-H2291 led to cellular resistance to the G12C-specific inhibitor AZ'8037 due to continuing active MAPK and PI3K cascades in the presence of the inhibitor. Overall, our study provides a comprehensive assessment of co-occurring KRAS mutations in NSCLC and in vitro evidence of the negative impact of co-occurring KRAS mutations on cellular response to G12C inhibitors, highlighting the need for a comprehensive KRAS tumour genotyping for optimal patient selection for treatment with a KRAS G12C inhibitor.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Tasa de Mutación , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Estudios Retrospectivos
5.
JCI Insight ; 6(13)2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34236045

RESUMEN

The AP-1 transcription factor c-Jun is required for Ras-driven tumorigenesis in many tissues and is considered as a classical proto-oncogene. To determine the requirement for c-Jun in a mouse model of K-RasG12D-induced lung adenocarcinoma, we inducibly deleted c-Jun in the adult lung. Surprisingly, we found that inactivation of c-Jun, or mutation of its JNK phosphorylation sites, actually increased lung tumor burden. Mechanistically, we found that protein levels of the Jun family member JunD were increased in the absence of c-Jun. In c-Jun-deficient cells, JunD phosphorylation was increased, and expression of a dominant-active JNKK2-JNK1 transgene further increased lung tumor formation. Strikingly, deletion of JunD completely abolished Ras-driven lung tumorigenesis. This work identifies JunD, not c-Jun, as the crucial substrate of JNK signaling and oncogene required for Ras-induced lung cancer.


Asunto(s)
Adenocarcinoma del Pulmón , Carcinogénesis , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas ras/metabolismo , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen , Genes jun/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , MAP Quinasa Quinasa 7/genética , MAP Quinasa Quinasa 7/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-jun/genética , Factor de Transcripción AP-1/metabolismo
6.
AAPS PharmSciTech ; 22(3): 101, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712968

RESUMEN

There is an increasing need to develop improved and non-invasive strategies to treat spinal cord injury (SCI). Nanoparticles (NPs) are an enabling technology to improve drug delivery, modulate inflammatory responses, and restore functional responses following SCI. However, the complex pathophysiology associated with SCI presents several distinct challenges that must be overcome for sufficient NP drug delivery to the spinal cord. The objective of this mini-review is to highlight the physiological challenges and cell types available for modulation and discuss several promising advancements using NPs to improve SCI treatment. We will focus our discussion on recent innovative approaches in NP drug delivery and how the implementation of multifactorial approaches to address the proinflammatory and complex immune dysfunction in SCI offers significant potential to improve outcomes in SCI.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/uso terapéutico , Humanos
7.
Biochem Soc Trans ; 48(6): 2691-2701, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33242077

RESUMEN

Oncogenic mutation in KRAS is one of the most common alterations in human cancer. After decades of extensive research and unsuccessful drug discovery programs, therapeutic targeting of KRAS mutant tumour is at an exciting juncture. The discovery of mutation-specific inhibitors of KRASG12C and early positive findings from clinical trials has raised the hope of finally having a drug to treat a significant segment of KRAS mutant cancer patients. Crucially, it has also re-energized the RAS field to look beyond G12C mutation and find new innovative targeting opportunities. However, the early clinical trial data also indicates that there is significant variation in response among patients and that monotherapy treatment with KRASG12C inhibitors (G12Ci) alone is unlikely to be sufficient to elicit a sustained response. Understanding the molecular mechanism of variation in patient response and identifying possible combination opportunities, which could be exploited to achieve durable and significant responses and delay emergence of resistance, is central to the success of G12Ci therapy. Given the specificity of G12Ci, toxicity is expected to be minimal. Therefore, it might be possible to combine G12Ci with other targeted agents which have previously been explored to tackle KRAS mutant cancer but deemed too toxic, e.g. MEK inhibitor. Ongoing clinical trials will shed light on clinical resistance to G12C inhibitors, however extensive work is already ongoing to identify the best combination partners. This review provides an update on combination opportunities which could be explored to maximize the benefit of this new exciting drug.


Asunto(s)
Regulación de la Expresión Génica , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Autofagia , Línea Celular Tumoral , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Ensayos Clínicos como Asunto , Diseño de Fármacos , Epigénesis Genética , Humanos , Sistema Inmunológico , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas ras/metabolismo
8.
J Tissue Eng ; 11: 2041731419897460, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32180936

RESUMEN

Brain diseases including neurodegenerative disorders and tumours are among the most serious health problems, degrading the quality of life and causing massive economic cost. Nanoparticles that load and deliver drugs and genes have been intensively studied for the treatment of brain diseases, and have demonstrated some biological effects in various animal models. Among other efforts taken in the nanoparticle development, targeting of blood brain barrier, specific cell type or local intra-/extra-cellular space is an important strategy to enhance the therapeutic efficacy of the nanoparticle delivery systems. This review underlies the targeting issue in the nanoparticle development for the treatment of brain diseases, taking key exemplar studies carried out in various in vivo models.

9.
J Med Chem ; 63(9): 4468-4483, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32023060

RESUMEN

Attempts to directly drug the important oncogene KRAS have met with limited success despite numerous efforts across industry and academia. The KRASG12C mutant represents an "Achilles heel" and has recently yielded to covalent targeting with small molecules that bind the mutant cysteine and create an allosteric pocket on GDP-bound RAS, locking it in an inactive state. A weak inhibitor at this site was optimized through conformational locking of a piperazine-quinazoline motif and linker modification. Subsequent introduction of a key methyl group to the piperazine resulted in enhancements in potency, permeability, clearance, and reactivity, leading to identification of a potent KRASG12C inhibitor with high selectivity and excellent cross-species pharmacokinetic parameters and in vivo efficacy.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Quinazolinas/uso terapéutico , Quinolonas/uso terapéutico , Regulación Alostérica , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Células CACO-2 , Línea Celular Tumoral , Diseño de Fármacos , Humanos , Masculino , Ratones Desnudos , Conformación Molecular , Mutación , Piperazinas/síntesis química , Piperazinas/farmacocinética , Proteínas Proto-Oncogénicas p21(ras)/genética , Quinazolinas/síntesis química , Quinazolinas/farmacocinética , Quinolonas/síntesis química , Quinolonas/farmacocinética , Ratas Wistar , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
10.
ACS Appl Bio Mater ; 3(9): 6263-6272, 2020 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-34604713

RESUMEN

Genetic engineering of innate and adaptive immune cells represents a potential solution to treat numerous immune-mediated pathologies. Current immune engineering methods to introduce nucleic acids into cells with high efficiency rely on physical mechanisms such as electroporation, viral vectors, or other chemical methods. Gene delivery using non-viral nanoparticles offers significant flexibility in biomaterial design to tune critical parameters such as nano-bio interactions, transfection efficiency, and toxicity profiles. However, their clinical utility has been limited due to complex synthetic procedures, high toxicity at increased polymer (nitrogen, N) to DNA ratios (phosphate, P) (N/P ratios), poor transfection efficiency and nanoparticle stability in the presence of serum, and short-term gene expression. Here, we describe the development of a simple, polymer-based non-viral gene delivery platform based on simple modifications of polyethylenimine (PEI) that displays potent and serum-independent transfection of innate and adaptive immune cells. Cationic acetylated PEI (Ac-PEI) was synthesized and complexed with plasmid DNA (pDNA) followed by enveloping with an anionic polyelectrolyte layer of poly(ethylene-alt-maleic acid) (PEMA) to form immunoplexes (IPs). Cellular interactions and gene expression could be precisely controlled in murine RAW 264.7 macrophages, murine DC2.4 dendritic cells, and human Jurkat T cells by altering the levels of PEMA envelopment, thus providing a strategy to engineer specific cell targeting into the IP platform. Optimally formulated IPs for immune cell transfection in the presence of serum utilized high N/P ratios to enable high stability, displayed reduced toxicity, high gene expression, and a lengthened duration of gene expression (>3 days) compared to non-enveloped controls. These results demonstrate the potential of engineered IPs to serve as simple, modular, targetable, and efficient non-viral gene delivery platform to efficiently alter gene expression within cells of the immune system.

11.
J Exp Med ; 216(2): 450-465, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30642944

RESUMEN

Lung squamous cell carcinoma (LSCC) and adenocarcinoma (LADC) are the most common lung cancer subtypes. Molecular targeted treatments have improved LADC patient survival but are largely ineffective in LSCC. The tumor suppressor FBW7 is commonly mutated or down-regulated in human LSCC, and oncogenic KRasG12D activation combined with Fbxw7 inactivation in mice (KF model) caused both LSCC and LADC. Lineage-tracing experiments showed that CC10+, but not basal, cells are the cells of origin of LSCC in KF mice. KF LSCC tumors recapitulated human LSCC resistance to cisplatin-based chemotherapy, and we identified LUBAC-mediated NF-κB signaling as a determinant of chemotherapy resistance in human and mouse. Inhibition of NF-κB activation using TAK1 or LUBAC inhibitors resensitized LSCC tumors to cisplatin, suggesting a future avenue for LSCC patient treatment.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Resistencia a Antineoplásicos , Neoplasias Pulmonares/enzimología , Complejos Multienzimáticos/metabolismo , Ubiquitinación , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/enzimología , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Cisplatino/farmacología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Complejos Multienzimáticos/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
12.
ChemMedChem ; 13(22): 2427-2436, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30222248

RESUMEN

Folic acid (FA), vitamin B9 , is a good receptor of drugs triggering cellular uptake via endocytosis. FA is sparingly soluble in water. Herein, a new approach for the formation of FA hydrogel by the hydrolysis of glucono-δ-lactone in PBS buffer under physiological conditions has been reported. The gel has a fibrillar network morphology attributable to intermolecular H-bonding and π-stacking interactions. The thixotropic property of the gel is used for the encapsulation of both hydrophilic [doxorubicin (DOX)] and hydrophobic [camptothecin (CPT)] drugs. The loading of DOX and CPT into the gel is attributed to the H-bonding interaction between FA and drugs. The release of DOX is sustainable at pH 4 and 7, and the Peppas model indicates that at pH 7 the diffusion of the drug is Fickian but it is non-Fickian at pH 4. The release of CPT is monitored by fluorescence spectroscopy, which also corroborates the combined release of both drugs. The metylthiazolyldiphenyltetrazolium bromide assay of FA hydrogel demonstrates nontoxic behavior and that the cytotoxicity of the DOX-loaded FA hydrogel is higher than that of pure DOX, with a minimal effect on normal cells.


Asunto(s)
Antineoplásicos/farmacología , Camptotecina/farmacología , Doxorrubicina/farmacología , Portadores de Fármacos/química , Ácido Fólico/química , Hidrogeles/química , Animales , Antineoplásicos/química , Células CHO , Camptotecina/química , Cricetulus , Difusión , Doxorrubicina/química , Portadores de Fármacos/toxicidad , Liberación de Fármacos , Ácido Fólico/toxicidad , Gluconatos/química , Gluconatos/toxicidad , Células HeLa , Humanos , Hidrogeles/toxicidad , Concentración de Iones de Hidrógeno , Hidrólisis , Interacciones Hidrofóbicas e Hidrofílicas , Lactonas/química , Ratones
13.
Biochem Soc Trans ; 46(5): 1303-1311, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30154091

RESUMEN

Oncogenic mutations in RAS genes underlie the pathogenesis of many human tumours, and there has been intense effort for over 30 years to develop effective and tolerated targeted therapeutics for patients with Ras-driven cancers. This review summarises the progress made in Ras drug discovery, highlighting some of the recent developments in directly targeting Ras through advances in small molecule drug design and novel therapeutic strategies.


Asunto(s)
Terapia Molecular Dirigida/métodos , Neoplasias/genética , Proteínas ras/metabolismo , Animales , Antineoplásicos/uso terapéutico , Diseño de Fármacos , Descubrimiento de Drogas , Genes ras , Humanos , Ratones , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Oligonucleótidos Antisentido/genética , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas p21(ras)/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos
14.
Langmuir ; 34(45): 13461-13471, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-29699394

RESUMEN

Chemically and biochemically functionalized colloidal nanoparticles with appropriate surface chemistry are essential for various biomedical applications. Although a variety of approaches are now available in making such functional nanoparticles and nanobioconjugates, the lack of complementary surface chemistry often leads to poor performance with respect to intended biomedical applications. This feature article will focus on our efforts to make colloidal nanobioconjugates with appropriate/complementary surface chemistry for better performance of a designed nanoprobe with respect to cellular and subcellular targeting applications. In particular, we emphasize polyacrylate-based coating chemistry followed by a conjugation strategy for transforming <10 nm inorganic nanoparticle to colloidal nanoprobe of 20-50 nm hydrodynamic size. We show that a colloidal nanoprobe can be chemically designed to control the cell-nanoparticle interaction, cellular endocytosis, and targeting/labeling of subcellular compartments. Further study should be directed to adapt this surface chemistry to different nanoparticles, fine tune the surface chemistry for targeting/imaging on the subcellular/molecular length scale, and develop a delivery nanocarrier for subcellular compartments.


Asunto(s)
Resinas Acrílicas/química , Coloides/química , Nanoconjugados/química , Nanopartículas/química , Orgánulos/metabolismo , Endocitosis/efectos de los fármacos , Células HeLa , Humanos , Tamaño de la Partícula , Multimerización de Proteína/efectos de los fármacos , Propiedades de Superficie
15.
ACS Appl Mater Interfaces ; 10(2): 1976-1986, 2018 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-29257666

RESUMEN

Visible light photocatalysis by TiO2 requires efficient doping of other elements with red-shifted band edge to the visible region. However, preparation of such TiO2 with tunable doping is challenging. Here we report a method of making nitrogen (N) and fluorine (F) codoped TiO2 nanoparticle with tunable doping between 1 and 7 at. %. The preparation of N, F codoped TiO2 nanoparticle involves reaction of colloidal TiO2 nanorods with an ammonium fluoride-urea mixture at 300 °C, and the extent of N/F doping is tuned by varying the amount of ammonium fluoride-urea and the reaction time. Resultant colloidal N, F codoped TiO2 nanoparticles show doping dependent shifting of the band edge from the UV to near-IR region, visible light induced generation of reactive oxygen species (ROS), and visible light photodegradation of bisphenol A. A colloidal form of doped TiO2 nanoparticle offers labeling of cells, visible light induced ROS generation inside a cell, and successive cell death. This work shows the potential advantage of anisotropic nanoparticle precursor for tunable doping and colloidal form of N, F codoped TiO2 nanoparticle as a visible light photocatalyst.


Asunto(s)
Nanopartículas , Catálisis , Muerte Celular , Flúor , Luz , Nitrógeno , Titanio
16.
ACS Appl Mater Interfaces ; 9(48): 41807-41817, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29135217

RESUMEN

Although the antioxidant property of vitamin C is well-known for protecting cells from oxidative stress, a recent study shows that it can also generate oxidative stress under a high intracellular concentration and induce cell death. However, poor chemical stability and low biological concentration (micromolar) of vitamin C restrict its function primarily as an antioxidant. Here, we report two different nanoparticle forms of vitamin C with its intact chemical stability, glucose-responsive release from nanoparticle, and efficient cell delivery in micro to millimolar concentrations. Nanoparticles are composed of silica-coated Au nanoparticles or lipophilic polyaspartic acid-based polymer micelles which are conjugated with vitamin C via phenylboronic acid. Surface chemistry of nanoparticles is optimized for an efficient cellular interaction/uptake and for cell delivery of vitamin C. We found that vitamin C protects cells from oxidative stress at micromolar concentrations, but at millimolar concentrations, it induces cell death by generating oxidative stress. In particular, high-dose vitamin C produces H2O2, disrupts the cellular redox balance, and induces cell death. This study highlights the concentration-dependent biological performance of vitamin C and the requirement of a high-dose cell delivery approach for enhanced therapeutic benefit.


Asunto(s)
Nanopartículas , Ácido Ascórbico , Muerte Celular , Peróxido de Hidrógeno , Estrés Oxidativo , Especies Reactivas de Oxígeno
17.
Nat Commun ; 6: 6782, 2015 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-25851810

RESUMEN

The c-Jun/AP-1 transcription factor controls key cellular behaviours, including proliferation and apoptosis, in response to JNK and Ras/MAPK signalling. While the JNK pathway has been well characterized, the mechanism of activation by Ras was elusive. Here we identify the uncharacterized ubiquitin ligase Trim7 as a critical component of AP-1 activation via Ras. We found that MSK1 directly phosphorylates Trim7 in response to direct activation by the Ras-Raf-MEK-ERK pathway, and this modification stimulates Trim7 E3 ubiquitin ligase activity. Trim7 mediates Lys63-linked ubiquitination of the AP-1 co-activator RACO-1, leading to RACO-1 protein stabilization. Consequently, Trim7 depletion reduces RACO-1 levels and AP-1-dependent gene expression. Moreover, transgenic overexpression of Trim7 increases lung tumour burden in a Ras-driven cancer model, and knockdown of Trim7 in established xenografts reduces tumour growth. Thus, phosphorylation-ubiquitination crosstalk between MSK1, Trim7 and RACO-1 completes the long sought-after mechanism linking growth factor signalling and AP-1 activation.


Asunto(s)
Proteínas Portadoras/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Transactivadores/metabolismo , Factor de Transcripción AP-1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Quinasas raf/metabolismo , Proteínas ras/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Células HEK293 , Células HeLa , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Fosforilación , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Proteínas de Motivos Tripartitos , Ubiquitinación
18.
Cancer Res ; 75(7): 1181-6, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25716680

RESUMEN

The stability of several oncoproteins, including c-Myc, is regulated by ubiquitin-dependent degradation mediated by the SCF(Fbw7) ubiquitin ligase. This activity is antagonized by the deubiquitinase Usp28, which is highly expressed in murine and human intestinal cancers. Usp28 was previously shown to interact with its substrates via a "piggyback" interaction with Fbw7, which suggested that Fbw7 is required for Usp28 activity. Unexpectedly, we found that genetic deletion of Usp28 rescued the lethality of Fbw7-deficient primary fibroblasts. Moreover, Usp28 inactivation in the intestine (Usp28(ΔIEC)) ameliorated the hyperproliferation and the impaired goblet and Paneth cell differentiation observed in Fbw7(ΔIEC) mice. The aggressive intestinal tumor formation of APC(Min/+); Fbw7(ΔIEC) mice was restrained when Usp28 was inactivated concomitantly. In both fibroblasts and intestinal cells, Usp28 deficiency corrected the accumulation of SCF(Fbw7) substrate proteins, including NICD1, c-Jun, and c-Myc. These findings suggested that Usp28 function does not depend on the presence of Fbw7, but instead independently recognizes and deubiquitylates the same substrates as SCF(Fbw7). Fbw7 binds to a phosphorylated motif termed the phosphodegron and we found that Usp28 also interacted with this same motif, but only when it is unphosphorylated, offering a mechanistic explanation for identical substrate selection by Fbw7 and Usp28. Our results indicate an unusually direct antagonism between an E3 ligase and a deubiquitinase, Fbw7 and Usp28, in modulating intestinal homeostasis and cancer.


Asunto(s)
Proteínas F-Box/metabolismo , Homeostasis , Neoplasias Intestinales/metabolismo , Ubiquitina Tiolesterasa/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Carcinogénesis/metabolismo , Diferenciación Celular , Proliferación Celular , Proteína 7 que Contiene Repeticiones F-Box-WD , Células HCT116 , Humanos , Mucosa Intestinal/metabolismo , Intestinos/patología , Ratones Transgénicos , Ubiquitinación
19.
J Phys Chem Lett ; 6(18): 3688-97, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26722743

RESUMEN

Nanoparticle interacts with live cells depending on their surface chemistry, enters into cell via endocytosis, and is commonly trafficked to an endosome/lysozome that restricts subcellular targeting options. Here we show that nanoparticle surface chemistry can be tuned to alter their cell uptake mechanism and subcellular trafficking. Quantum dot based nanoprobes of 20-30 nm hydrodynamic diameters have been synthesized with tunable surface charge (between +15 mV to -25 mV) and lipophilicity to influence their cellular uptake processes and subcellular trafficking. It is observed that cationic nanoprobe electrostatically interacts with cell membrane and enters into cell via clathrin-mediated endocytosis. At lower surface charge (between +10 mV to -10 mV), the electrostatic interaction with cell membrane becomes weaker, and additional lipid raft endocytosis is initiated. If a lipophilic functional group is introduced on a weakly anionic nanoparticle surface, the uptake mechanism shifts to predominant lipid raft-mediated endocytosis. In particular, the zwitterionic-lipophilic nanoprobe has the unique advantage as it weakly interacts with anionic cell membrane, migrates toward lipid rafts for interaction through lipophilic functional group, and induces lipid raft-mediated endocytosis. While predominate or partial clathrin-mediated entry traffics most of the nanoprobes to lysozome, predominate lipid raft-mediated entry traffics them to perinuclear region, particularly to the Golgi apparatus. This finding would guide in designing appropriate nanoprobe for subcellular targeting and delivery.


Asunto(s)
Clatrina/metabolismo , Endocitosis , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Nanopartículas/metabolismo , Clatrina/química , Endosomas/química , Endosomas/metabolismo , Células HeLa , Humanos , Hidrodinámica , Muramidasa/química , Muramidasa/metabolismo , Nanopartículas/química , Tamaño de la Partícula , Puntos Cuánticos , Propiedades de Superficie
20.
ACS Appl Mater Interfaces ; 7(2): 1340-7, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25537800

RESUMEN

Although cholesterol plays significant biochemical function in the human body, excess of it leads to various disorders, and thus, its control/separation is important in medical science and food industries. However, efficient and selective separation of cholesterol is challenging because cholesterol often exists in microheterogeneous or insoluble forms in remote organ and exists with other chemicals/biochemicals. Here, we have described a colloidal magnetic mesoporous silica (MMS)-based approach for efficient separation of cholesterol in different forms. MMS is functionalized with ß-cyclodextrin for selective binding with cholesterol via host-guest interaction. The colloidal form of MMS offers effective interaction with cholesterol of any form, and magnetic property of MMS offers easier separation of bound cholesterol. Functionalized MMS is efficient in separating cholesterol crystals, water-insoluble cholesterol, and the microheterogeneous form of cholesterol from milk or a cellular environment. Developed material can be used to remove cholesterol from a complex bioenvironment and extended for large-scale cholesterol separation from food.


Asunto(s)
Colesterol/aislamiento & purificación , Leche/química , Dióxido de Silicio/química , beta-Ciclodextrinas/química , Adsorción , Animales , Bovinos , Colesterol/química , Fenómenos Magnéticos , Porosidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA