Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Death Differ ; 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879724

RESUMEN

Development of the cerebellum requires precise regulation of granule neuron progenitor (GNP) proliferation. Although it is known that primary cilia are necessary to support GNP proliferation, the exact molecular mechanism governing primary cilia dynamics within GNPs remains elusive. Here, we establish the pivotal roles for the centrosomal kinase TTBK2 (Tau tubulin kinase-2) and the E3 ubiquitin ligase HUWE1 in GNP proliferation. We show that TTBK2 is highly expressed in proliferating GNPs under Sonic Hedgehog (SHH) signaling, coinciding with active GNP proliferation and the presence of primary cilia. TTBK2 stabilizes primary cilia by inhibiting their disassembly, thereby promoting GNP proliferation in response to SHH. Mechanistically, we identify HUWE1 as a novel centrosomal E3 ligase that facilitates primary cilia disassembly by targeting TTBK2 degradation. Disassembly of primary cilia serves as a trigger for GNP differentiation, allowing their migration from the external granule layer (EGL) of the cerebellum to the internal granule layer (IGL) for subsequent maturation. Moreover, we have established a link between TTBK2 and SHH-type medulloblastoma (SHH-MB), a tumor characterized by uncontrolled GNP proliferation. TTBK2 depletion inhibits SHH-MB proliferation, indicating that TTBK2 may be a potential therapeutic target for this cancer type. In summary, our findings reveal the mechanism governing cerebellar development and highlight a potential anti-cancer strategy for SHH-MB.

2.
Int J Mol Sci ; 24(15)2023 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-37569637

RESUMEN

Mitochondria (MITO) and peroxisomes (PEXO) are the major organelles involved in the oxidative metabolism of cells, but detailed examination of their dynamics and functional adaptations during skeletal muscle (SKM) development (myogenesis) is still lacking. In this study, we found that during myogenesis, MITO DNA, ROS level, and redox ratio increased in myotubes, but the membrane potential (Δψm) and ATP content reduced, implying that the MITO efficiency might reduce during myogenesis. The PEXO number and density both increased during myogenesis, which probably resulted from the accumulation and increased biogenesis of PEXO. The expression of PEXO biogenesis factors was induced during myogenesis in vitro and in utero, and their promoters were also activated by MyoD. Knockdown of the biogenesis factors Pex3 repressed not only the PEXO density and functions but also the levels of MITO genes and functions, suggesting a close coupling between PEXO biogenesis and MITO functions. Surprisingly, Pex3 knockdown by the CRISPRi system repressed myogenic differentiation, indicating critical involvement of PEXO biogenesis in myogenesis. Taken together, these observations suggest that the dynamics and functions of both MITO and PEXO are coupled with each other and with the metabolic changes that occur during myogenesis, and these metabolic couplings are critical to myogenesis.


Asunto(s)
Fibras Musculares Esqueléticas , Peroxisomas , Peroxisomas/metabolismo , Diferenciación Celular/genética , Fibras Musculares Esqueléticas/metabolismo , Mitocondrias/metabolismo , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo
3.
Nat Commun ; 14(1): 3050, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37237031

RESUMEN

Activation of tumor-intrinsic innate immunity has been a major strategy for improving immunotherapy. Previously, we reported an autophagy-promoting function of the deubiquitinating enzyme TRABID. Here, we identify a critical role of TRABID in suppressing anti-tumor immunity. Mechanistically, TRABID is upregulated in mitosis and governs mitotic cell division by removing K29-linked polyubiquitin chain from Aurora B and Survivin, thereby stabilizing the entire chromosomal passenger complex. TRABID inhibition causes micronuclei through a combinatory defect in mitosis and autophagy and protects cGAS from autophagic degradation, thereby activating the cGAS/STING innate immunity pathway. Genetic or pharmacological inhibition of TRABID promotes anti-tumor immune surveillance and sensitizes tumors to anti-PD-1 therapy in preclinical cancer models in male mice. Clinically, TRABID expression in most solid cancer types correlates inversely with an interferon signature and infiltration of anti-tumor immune cells. Our study identifies a suppressive role of tumor-intrinsic TRABID in anti-tumor immunity and highlights TRABID as a promising target for sensitizing solid tumors to immunotherapy.


Asunto(s)
Neoplasias , Nucleotidiltransferasas , Proteasas Ubiquitina-Específicas , Animales , Masculino , Ratones , Autofagia , Inmunidad Innata , Mitosis , Neoplasias/tratamiento farmacológico , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo
4.
ACS Appl Mater Interfaces ; 15(19): 23593-23601, 2023 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-37157130

RESUMEN

Understanding the physicochemical modulation of functional molecules is the primary step in exploring novel stimuli-responsive materials, and preventing the π-π stacking configuration of π-conjugated molecules has been an effective strategy of vapochromic material development, such as of nanoporous frameworks. Nevertheless, the more complicated synthetic strategy should in fact be applied in many circumstances. In this study, we explore a facile supramolecular strategy where the commodity plastic, syndiotactic-poly(methyl methacrylate) (st-PMMA), is utilized to wrap C60 to form the inclusion complex. The structural characterization revealed that C60s in the st-PMMA supramolecular helix had a lower coordination number (CN = 2) compared to the face-centered-cubic packing of pure C60s (CN = 12). Since the st-PMMA/C60 helical complex has structural flexibility, the π-π stacking structure of C60 was further interrupted by the intercalation of toluene vapors, and the complete isolation of C60 in the complex induced the desired vapochromic behavior. Furthermore, the aromatic interaction between C60 and aromatic solvent vapors enabled the st-PMMA/C60 inclusion complex to selectively encapsulate chlorobenzene, toluene, etc., and induce the color change. The st-PMMA/C60 inclusion complex exhibited a transparent film of sufficient structural integrity such that it can still induce a reversible color change after several cycles. As a result, a new strategy has been discovered for the development of novel vapochromic materials via host-guest chemistry.

5.
Autophagy ; 18(12): 2830-2850, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35316161

RESUMEN

Centrosome amplification is a phenomenon frequently observed in human cancers, so centrosome depletion has been proposed as a therapeutic strategy. However, despite being afflicted with a lack of centrosomes, many cancer cells can still proliferate, implying there are impediments to adopting centrosome depletion as a treatment strategy. Here, we show that TFEB- and TFE3-dependent autophagy activation contributes to acentrosomal cancer proliferation. Our biochemical analyses uncover that both TFEB and TFE3 are novel PLK4 (polo like kinase 4) substrates. Centrosome depletion inactivates PLK4, resulting in TFEB and TFE3 dephosphorylation and subsequent promotion of TFEB and TFE3 nuclear translocation and transcriptional activation of autophagy- and lysosome-related genes. A combination of centrosome depletion and inhibition of the TFEB-TFE3 autophagy-lysosome pathway induced strongly anti-proliferative effects in cancer cells. Thus, our findings point to a new strategy for combating cancer.Abbreviations: AdCre: adenoviral Cre recombinase; AdLuc: adenoviral luciferase; ATG5: autophagy related 5; CQ: chloroquine; DAPI: 4',6-diamidino-2-phenylindole; DKO: double knockout; GFP: green fluorescent protein; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LAMP2: lysosomal associated membrane protein 2; LTR: LysoTracker Red; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MITF: melanocyte inducing transcription factor; PLK4: polo like kinase 4; RFP: red fluorescent protein; SASS6: SAS-6 centriolar assembly protein; STIL: STIL centriolar assembly protein; TFEB: transcription factor EB; TFEBΔNLS: TFEB lacking a nuclear localization signal; TFE3: transcription factor binding to IGHM enhancer 3; TP53/p53: tumor protein p53.


Asunto(s)
Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Centrosoma , Neoplasias , Humanos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Proliferación Celular , Centrosoma/metabolismo , Lisosomas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Serina-Treonina Quinasas
6.
Biochem J ; 478(4): 911-926, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33527978

RESUMEN

M-cadherin is a skeletal muscle-specific transmembrane protein mediating the cell-cell adhesion of myoblasts during myogenesis. It is expressed in the proliferating satellite cells and highly induced by myogenic regulatory factors (MRFs) during terminal myogenic differentiation. Several conserved cis-elements, including 5 E-boxes, 2 GC boxes, and 1 conserved downstream element (CDE) were identified in the M-cadherin proximal promoter. We found that E-box-3 and -4 close to the transcription initiation site (TIS) mediated most of its transactivation by MyoD, the strongest myogenic MRF. Including of any one of the other E-boxes restored the full activation by MyoD, suggesting an essential collaboration between E-boxes. Stronger activation of M-cadherin promoter than that of muscle creatine kinase (MCK) by MyoD was observed regardless of culture conditions and the presence of E47. Furthermore, MyoD/E47 heterodimer and MyoD ∼ E47 fusion protein achieved similar levels of activation in differentiation medium (DM), suggesting high affinity of MyoD/E47 to E-boxes 3/4 under DM. We also found that GC boxes and CDE positively affected MyoD mediated activation. The CDE element was predicted to be the target of the chromatin-modifying factor Meis1/Pbx1 heterodimer. Knockdown of Pbx1 significantly reduced the expression level of M-cadherin, but increased that of N-cadherin. Using ChIP assay, we further found significant reduction in MyoD recruitment to M-cadherin promoter when CDE was deleted. Taken together, these observations suggest that the chromatin-modifying function of Pbx1/Meis1 is critical to M-cadherin promoter activation before MyoD is recruited to E-boxes to trigger transcription.


Asunto(s)
Cadherinas/genética , Elementos E-Box/genética , Regulación de la Expresión Génica/genética , Desarrollo de Músculos/genética , Regiones Promotoras Genéticas/genética , Animales , Secuencia de Bases , Células Cultivadas , Secuencia Conservada , Fibroblastos , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/fisiología , Proteína MioD/metabolismo , Mioblastos , Factor de Transcripción 1 de la Leucemia de Células Pre-B/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico
7.
Biomater Sci ; 7(8): 3404-3417, 2019 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-31251311

RESUMEN

For those patients with HER2-overexpressing breast cancer, treatment with PEGylated liposomal doxorubicin (PLD) is inefficacious due to the intrinsic low sensitivity to doxorubicin. A very large increase in drug accumulation by active targeting may enhance the therapeutic efficacy of PLD. We established a humanized bispecific antibody (BsAb; mPEG × HER2) which has dual specificity for methoxy-polyethylene glycol (mPEG) and human epidermal growth factor receptor 2 (HER2) to enhance the specificity, internalization and anticancer activity of PLD for cancer cells that overexpress HER2. One-step formulation of PLD with mPEG × HER2 converted the PLD into HER2 targeted liposomes that were stable at 4 °C in PBS as well as at 37 °C in the presence of serum. αHER2/PLD induced receptor-mediated endocytosis and enhanced doxorubicin accumulation in MCF7/HER2 (HER2-amplified) breast cancer cells. αHER2/PLD also displayed more than 200-fold increased cytotoxicity to MCF7/HER2 cells and 28-fold increased cytotoxicity to drug-resistant MDA-MB-361 cells with a physical deletion of the TOP2A gene. αHER2/PLD specifically accumulated doxorubicin in the nucleus of cancer cells in tumor-bearing mice and produced significantly greater antitumor activity against MCF7/HER2 (P < 0.0001) and MDA-MB-361 (P < 0.05) tumors as compared to untargeted PLD. Furthermore, the cardiotoxicity of αHER2/PLD was similar to that of PLD in human cardiomyocytes and in mice. Our results indicate that the one-step formulation of PLD by mPEG × HER2 is a simple method to confer tumor specificity, increase drug internalization and enhance the anticancer activity of PLD against HER2-overexpressing and doxorubicin-resistant breast cancer.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Nanopartículas/química , Polietilenglicoles/química , Animales , Antineoplásicos/química , Transporte Biológico , Doxorrubicina/química , Doxorrubicina/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/toxicidad , Composición de Medicamentos , Humanos , Células MCF-7 , Ratones , Miocitos Cardíacos/efectos de los fármacos , Polietilenglicoles/metabolismo , Polietilenglicoles/toxicidad , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo
8.
Redox Biol ; 20: 321-333, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30391825

RESUMEN

PGC-1α is a key regulator of oxidative metabolism facilitating the expression of genes critical for the function and biogenesis of the two key oxidative organelles, mitochondria and peroxisomes, in skeletal muscle (SKM) and other organs. Our recent studies have found that the transcription factor Bhlhe40 negatively regulates PGC-1α gene expression and its coactivational activity, therefore, this factor should have profound influence on the biogenesis and metabolic activity of mitochondria and peroxisomes. Here we found that both the number and activity of peroxisomes were increased upon knockdown of Bhlhe40 expression but were repressed by its over-expression. Mitochondrial efficiency was significantly reduced by Bhlhe40 knockdown, resulting in the burst of ROS. Over-expression of a constitutively active PGC-1α-interactive domain (named as VBH135) of Bhlhe40 mimicked the effects of its knockdown on peroxisomes but simultaneously reduced ROS level. Furthermore, the efficiency, but not the number, of mitochondria was also increased by VBH135, suggesting differential regulation of peroxisomes and mitochondria by Bhlhe40. Unsaturated fatty acid oxidation, insulin response, and oxidative respiration were highly enhanced in Bhlhe40 knockdown or VBH135 over-expressed cells, suggesting the importance of Bhlhe40 in the regulation of unsaturated fatty acid and glucose oxidative metabolism. Expression profiling of genes important for either organelle also supports differential regulation of peroxisomes and mitochondria by Bhlhe40. These observations have established the important role of Bhlhe40 in SKM oxidative metabolism as the critical regulator of peroxisome and mitochondrion biogenesis and functions, and thus should provide a novel route for developing drugs targeting SKM metabolic diseases.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Homeodominio/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Desarrollo de Músculos/genética , Mioblastos/metabolismo , Peroxisomas/genética , Peroxisomas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores , Catalasa/metabolismo , Ácidos Grasos/metabolismo , Expresión Génica , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Ratones , Oxidación-Reducción , Consumo de Oxígeno , ARN Interferente Pequeño/genética , Ratas , Especies Reactivas de Oxígeno/metabolismo
9.
Anal Chem ; 89(11): 6082-6090, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28485140

RESUMEN

Sensitive quantification of the pharmacokinetics of poly(ethylene glycol) (PEG) and PEGylated molecules is critical for PEGylated drug development. Here, we developed a sensitive sandwich enzyme-linked immunosorbent assay (ELISA) for PEG by tethering an anti-PEG antibody (AGP3) via tethers with different dimensions on the surface of 293T cells (293T/S-αPEG, short-type cells; 293T/L-αPEG, long-type cells; 293T/SL-αPEG, hybrid-type cells) to improve the binding capacity and detection limit for free PEG and PEGylated molecules. The binding capacity of hybrid-type cells for PEG-like molecules (CH3-PEG5K-FITC (FITC = fluorescein isothiocyanate) and eight-arm PEG20K-FITC) was at least 10-80-fold greater than that of 293T cells expressing anti-PEG antibodies with uniform tether lengths. The detection limit of free PEG (OH-PEG3K-NH2 and CH3-PEG5K-NH2) and PEG-like molecule (CH3-PEG5K-FITC, CH3-PEG5K-SHPP, and CH3-PEG5K-NIR797) was14-137 ng mL-1 in the hybrid-type cell-based sandwich ELISA. 293T/SL-αPEG cells also had significantly higher sensitivity for quantification of a PEGylated protein (PegIntron) and multiarm PEG macromolecules (eight-arm PEG20K-NH2 and eight-arm PEG40K-NH2) at 3.2, 16, and 16 ng mL-1, respectively. Additionally, the overall binding capacity of 293T/SL-αPEG cells for PEGylated macromolecules was higher than that of 293T/S-αPEG or 293T/L-αPEG cells. Anchoring anti-PEG antibodies on cells via variable-length tethers for cell-based sandwich ELISA, therefore, provides a sensitive, high-capacity method for quantifying free PEG and PEGylated molecules.


Asunto(s)
Anticuerpos/metabolismo , Membranas/metabolismo , Polietilenglicoles/análisis , Reactivos de Enlaces Cruzados/química , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos
10.
PLoS One ; 11(8): e0160418, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27494183

RESUMEN

Molecular weight markers that can tolerate denaturing conditions and be auto-detected by secondary antibodies offer great efficacy and convenience for Western Blotting. Here, we describe M&R LE protein markers which contain linear epitopes derived from the heavy chain constant regions of mouse and rabbit immunoglobulin G (IgG Fc LE). These markers can be directly recognized and stained by a wide range of anti-mouse and anti-rabbit secondary antibodies. We selected three mouse (M1, M2 and M3) linear IgG1 and three rabbit (R1, R2 and R3) linear IgG heavy chain epitope candidates based on their respective crystal structures. Western blot analysis indicated that M2 and R2 linear epitopes are effectively recognized by anti-mouse and anti-rabbit secondary antibodies, respectively. We fused the M2 and R2 epitopes (M&R LE) and incorporated the polypeptide in a range of 15-120 kDa auto-detecting markers (M&R LE protein marker). The M&R LE protein marker can be auto-detected by anti-mouse and anti-rabbit IgG secondary antibodies in standard immunoblots. Linear regression analysis of the M&R LE protein marker plotted as gel mobility versus the log of the marker molecular weights revealed good linearity with a correlation coefficient R2 value of 0.9965, indicating that the M&R LE protein marker displays high accuracy for determining protein molecular weights. This accurate, regular and auto-detected M&R LE protein marker may provide a simple, efficient and economical tool for protein analysis.


Asunto(s)
Anticuerpos/análisis , Western Blotting/métodos , Epítopos/inmunología , Animales , Anticuerpos/inmunología , Biomarcadores/análisis , Epítopos/genética , Inmunoglobulina G/genética , Ratones , Peso Molecular , Conejos
11.
Elife ; 42015 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-26609813

RESUMEN

Vertebrate centrioles normally propagate through duplication, but in the absence of preexisting centrioles, de novo synthesis can occur. Consistently, centriole formation is thought to strictly rely on self-assembly, involving self-oligomerization of the centriolar protein SAS-6. Here, through reconstitution of de novo synthesis in human cells, we surprisingly found that normal looking centrioles capable of duplication and ciliation can arise in the absence of SAS-6 self-oligomerization. Moreover, whereas canonically duplicated centrioles always form correctly, de novo centrioles are prone to structural errors, even in the presence of SAS-6 self-oligomerization. These results indicate that centriole biogenesis does not strictly depend on SAS-6 self-assembly, and may require preexisting centrioles to ensure structural accuracy, fundamentally deviating from the current paradigm.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centriolos/metabolismo , Biogénesis de Organelos , Multimerización de Proteína , Línea Celular , Células Epiteliales/fisiología , Humanos
12.
PLoS One ; 10(7): e0133470, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26186692

RESUMEN

The development of effective adjuvant is the key factor to boost the immunogenicity of tumor cells as a tumor vaccine. In this study, we expressed membrane-bound granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-18 (IL-18) as adjuvants in tumor cells to stimulate immune response. B7 transmembrane domain fused GM-CSF and IL-18 was successfully expressed in the cell membrane and stimulated mouse splenocyte proliferation. Co-expression of GM-CSF and IL-18 reduced tumorigenesis (P<0.05) and enhanced tumor protective efficacy (P<0.05) significantly in comparison with GM-CSF alone. These results indicated that the combination of GM-CSF andIL-18 will enhance the immunogenicity of a cell-based anti-tumor vaccine. This membrane-bound approach can be applied to other cytokines for the development of novel vaccine strategies.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Carcinogénesis/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Interleucina-18/inmunología , Animales , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Membrana Celular/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Interleucina-18/genética , Ratones , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
13.
Mol Cell Biol ; 35(14): 2518-29, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25963661

RESUMEN

PGC-1α is a transcriptional coactivator promoting oxidative metabolism in many tissues. Its expression in skeletal muscle (SKM) is induced by hypoxia and reactive oxidative species (ROS) generated during exercise, suggesting that PGC-1α might mediate the cross talk between oxidative metabolism and cellular responses to hypoxia and ROS. Here we found that PGC-1α directly interacted with Bhlhe40, a basic helix-loop-helix (bHLH) transcriptional repressor induced by hypoxia, and protects SKM from ROS damage, and they cooccupied PGC-1α-targeted gene promoters/enhancers, which in turn repressed PGC-1α transactivational activity. Bhlhe40 repressed PGC-1α activity through recruiting histone deacetylases (HDACs) and preventing the relief of PGC-1α intramolecular repression caused by its own intrinsic suppressor domain. Knockdown of Bhlhe40 mRNA increased levels of ROS, fatty acid oxidation, mitochondrial DNA, and expression of PGC-1α target genes. Similar effects were also observed when the Bhlhe40-mediated repression was rescued by a dominantly active form of the PGC-1α-interacting domain (PID) from Bhlhe40. We further found that Bhlhe40-mediated repression can be largely relieved by exercise, in which its recruitment to PGC-1α-targeted cis elements was significantly reduced. These observations suggest that Bhlhe40 is a novel regulator of PGC-1α activity repressing oxidative metabolism gene expression and mitochondrion biogenesis in sedentary SKM.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Homeodominio/metabolismo , Músculo Esquelético/metabolismo , Regiones Promotoras Genéticas/genética , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Línea Celular , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Histona Desacetilasas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Masculino , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Músculo Esquelético/citología , Mioblastos/metabolismo , Oxidación-Reducción , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Factores de Transcripción/genética
14.
Vet J ; 205(1): 107-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25975854

RESUMEN

The envelope glycoprotein E2 of classical swine fever virus (CSFV) is widely used as a marker for measuring vaccine efficacy and antibody titer. The glycosylation profile of E2 may affect the immunogenicity of the vaccine and the timing of re-vaccination. In this study, a human embryonic kidney cell line was used to secrete fully-glycosylated CSFV E2, which was then coated onto ELISA plates without purification or adjustment. The resulting E2-secreting medium-direct-coating (E2-mDc) ELISA was successfully used to measure anti-E2 antibody titers in vaccinated and field pig sera samples. Compared with a virus neutralization test (as standard), the E2-mDc ELISA was found to be more accurate (90%) than a commercial CSFV antibody diagnostic kit (62%). In conclusion, the mammalian cell-secreted antigen can provide cheap, accurate and effective assays for vaccine efficacy and disease diagnoses.


Asunto(s)
Anticuerpos Antivirales/análisis , Virus de la Fiebre Porcina Clásica/inmunología , Medios de Cultivo , Ensayo de Inmunoadsorción Enzimática/métodos , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Células HEK293 , Humanos , Porcinos , Proteínas del Envoltorio Viral/metabolismo
15.
ScientificWorldJournal ; 2015: 740815, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25839056

RESUMEN

Glucuronidation is a major metabolism process of detoxification for carcinogens, 4-(methylnitrosamino)-1-(3-pyridy)-1-butanone (NNK) and 1,2-dimethylhydrazine (DMH), of reactive oxygen species (ROS). However, intestinal E. coli ß-glucuronidase (eßG) has been considered pivotal to colorectal carcinogenesis. Specific inhibition of eßG may prevent reactivating the glucuronide-carcinogen and protect the intestine from ROS-mediated carcinogenesis. In order to develop specific eßG inhibitors, we found that 59 candidate compounds obtained from the initial virtual screening had high inhibition specificity against eßG but not human ßG. In particular, we found that compounds 7145 and 4041 with naphthalenylidene-benzenesulfonamide (NYBS) are highly effective and selective to inhibit eßG activity. Compound 4041 (IC50 = 2.8 µM) shows a higher inhibiting ability than compound 7145 (IC50 = 31.6 µM) against eßG. Furthermore, the molecular docking analysis indicates that compound 4041 has two hydrophobic contacts to residues L361 and I363 in the bacterial loop, but 7145 has one contact to L361. Only compound 4041 can bind to key residue (E413) at active site of eßG via hydrogen-bonding interactions. These novel NYBS-based eßG specific inhibitors may provide as novel candidate compounds, which specifically inhibit eßG to reduce eßG-based carcinogenesis and intestinal injury.


Asunto(s)
Simulación por Computador , Descubrimiento de Drogas/métodos , Proteínas de Escherichia coli/antagonistas & inhibidores , Glucuronidasa/antagonistas & inhibidores , Simulación del Acoplamiento Molecular/métodos , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/enzimología , Proteínas de Escherichia coli/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Glucuronidasa/química , Glucuronidasa/metabolismo , Humanos , Estructura Secundaria de Proteína
16.
Mol Cancer Ther ; 13(12): 2852-63, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25277385

RESUMEN

Beta-glucuronidase (ßG) is a potential biomarker for cancer diagnosis and prodrug therapy. The ability to image ßG activity in patients would assist in personalized glucuronide prodrug cancer therapy. However, whole-body imaging of ßG activity for medical usage is not yet available. Here, we developed a radioactive ßG activity-based trapping probe for positron emission tomography (PET). We generated a (124)I-tyramine-conjugated difluoromethylphenol beta-glucuronide probe (TrapG) to form (124)I-TrapG that could be selectively activated by ßG for subsequent attachment of (124)I-tyramine to nucleophilic moieties near ßG-expressing sites. We estimated the specificity of a fluorescent FITC-TrapG, the cytotoxicity of tyramine-TrapG, and the serum half-life of (124)I-TrapG. ßG targeting of (124)I-TrapG in vivo was examined by micro-PET. The biodistribution of (131)I-TrapG was investigated in different organs. Finally, we imaged the endogenous ßG activity and assessed its correlation with therapeutic efficacy of 9-aminocamptothecin glucuronide (9ACG) prodrug in native tumors. FITC-TrapG showed specific trapping at ßG-expressing CT26 (CT26/mßG) cells but not in CT26 cells. The native TrapG probe possessed low cytotoxicity. (124)I-TrapG preferentially accumulated in CT26/mßG but not CT26 cells. Meanwhile, micro-PET and whole-body autoradiography results demonstrated that (124)I-TrapG signals in CT26/mßG tumors were 141.4-fold greater than in CT26 tumors. Importantly, Colo205 xenografts in nude mice that express elevated endogenous ßG can be monitored by using infrared glucuronide trapping probes (NIR-TrapG) and suppressed by 9ACG prodrug treatment. (124)I-TrapG exhibited low cytotoxicity allowing long-term monitoring of ßG activity in vivo to aid in the optimization of prodrug targeted therapy.


Asunto(s)
Glucuronidasa/metabolismo , Glucurónidos/uso terapéutico , Radioisótopos de Yodo , Tomografía de Emisión de Positrones , Profármacos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Tomografía de Emisión de Positrones/métodos , Sensibilidad y Especificidad , Distribución Tisular , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Biomaterials ; 35(37): 9930-9940, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25212525

RESUMEN

Methoxy PEGylated nanoparticles (mPEG-NPs) are increasingly used for cancer imaging and therapy. Here we describe a general and simple approach to confer tumor tropism to any mPEG-NP. We demonstrate this approach with humanized bispecific antibodies (BsAbs) that can bind to both mPEG molecules on mPEG-NPs and to EGFR or HER2 molecules overexpressed on the surface of cancer cells. Simple mixing of BsAbs with mPEG-NPs can mediate preferential binding of diverse mPEG-NPs to cancer cells that overexpress EGFR or HER2 under physiological conditions and significantly increase cancer cell killing by liposomal doxorubicin to EGFR(+) and HER2(+) cancer cells. BsAbs modification also enhanced accumulation of fluorescence-labeled NPs and significantly increased the anticancer activity of drug-loaded NPs to antigen-positive human tumors in a mouse model. Anti-mPEG BsAbs offer a simple one-step method to confer tumor specificity to mPEG-NPs for enhanced tumor accumulation and improved therapeutic efficacy.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Anticuerpos Biespecíficos , Anticuerpos Monoclonales Humanizados , Doxorrubicina/análogos & derivados , Nanopartículas , Polietilenglicoles , Animales , Antibióticos Antineoplásicos/uso terapéutico , Anticuerpos Biespecíficos/química , Anticuerpos Monoclonales Humanizados/química , Células 3T3 BALB , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Doxorrubicina/uso terapéutico , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Receptores ErbB/análisis , Femenino , Humanos , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Imagen Óptica , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Polietilenglicoles/uso terapéutico , Receptor ErbB-2/análisis
18.
PLoS One ; 9(7): e102569, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25036759

RESUMEN

Developing a high-throughput method for the effecient selection of the highest producing cell is very important for the production of recombinant protein drugs. Here, we developed a novel transiently protein-anchored system coupled with fluorescence activated cell sorting (FACS) for the efficient selection of the highest producing cell. A furin cleavage peptide (RAKR) was used to join a human anti-epithelial growth factor antibody (αEGFR Ab) and the extracellular-transmembrane-cytosolic domains of the mouse B7-1 antigen (B7). The furin inhibitor can transiently switch secreted αEGFR Ab into a membrane-anchored form. After cell sorting, the level of membrane αEGFR Ab-RAKR-B7 is proportional to the amount of secreted αEGFR Ab in the medium. We further selected 23 αEGFR Ab expressing cells and demonstrated a high correlation (R2 = 0.9165) between the secretion level and surface expression levels of αEGFR Ab. These results suggested that the novel transiently protein-anchored system can easily and efficiently select the highest producing cells, reducing the cost for the production of biopharmaceuticals.


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Animales , Anticuerpos/metabolismo , Antígeno B7-1/metabolismo , Línea Celular , Citosol/metabolismo , Receptores ErbB/metabolismo , Furina/metabolismo , Células HEK293 , Humanos , Ratones , Unión Proteica/fisiología , Transporte de Proteínas/fisiología , Proteínas Recombinantes de Fusión/metabolismo
19.
PLoS One ; 9(5): e97367, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24824752

RESUMEN

Single-chain variable fragments (scFvs) serve as an alternative to full-length monoclonal antibodies used in research and therapeutic and diagnostic applications. However, when recombinant scFvs are overexpressed in bacteria, they often form inclusion bodies and exhibit loss of function. To overcome this problem, we developed an scFv secretion system in which scFv was fused with osmotically inducible protein Y (osmY), a bacterial secretory carrier protein, for efficient protein secretion. Anti-EGFR scFv (αEGFR) was fused with osmY (N- and C-termini) and periplasmic leader sequence (pelB) to generate αEGFR-osmY, osmY-αEGFR, and pelB-αEGFR (control), respectively. In comparison with the control, both the osmY-fused αEGFR scFvs were soluble and secreted into the LB medium. Furthermore, the yield of soluble αEGFR-osmY was 20-fold higher, and the amount of secreted protein was 250-fold higher than that of osmY-αEGFR. In addition, the antigen-binding activity of both the osmY-fused αEGFRs was 2-fold higher than that of the refolded pelB-αEGFR from inclusion bodies. Similar results were observed with αTAG72-osmY and αHer2-osmY. These results suggest that the N-terminus of osmY fused with scFv produces a high yield of soluble, functional, and secreted scFv, and the osmY-based bacterial secretion system may be used for the large-scale industrial production of low-cost αEGFR protein.


Asunto(s)
Sistemas de Secreción Bacterianos/inmunología , Reactores Biológicos , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Microbiología Industrial/métodos , Proteínas de Unión Periplasmáticas/metabolismo , Anticuerpos de Cadena Única/biosíntesis , Western Blotting , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Receptores ErbB/metabolismo , Escherichia coli/genética , Señales de Clasificación de Proteína/genética , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/metabolismo
20.
Mol Cell Endocrinol ; 383(1-2): 147-58, 2014 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-24361750

RESUMEN

Valproic acid (VPA) has been shown to increase the reprogramming efficiency of induced pluripotent stem cells (iPSC) from somatic cells, but the mechanism by which VPA enhances iPSC induction has not been defined. Here we demonstrated that VPA directly activated Oct4 promoter activity through activation of the PI3K/Akt/mTOR signaling pathway that targeted the proximal hormone response element (HRE, -41∼-22) in this promoter. The activating effect of VPA is highly specific as similar compounds or constitutional isomers failed to instigate Oct4 promoter activity. We further demonstrated that the upstream 2 half-sites in this HRE were essential to the activating effect of VPA and they were targeted by a subset of nuclear receptors, such as COUP-TFII and TR2. These findings show the first time that NRs are implicated in the VPA stimulated expression of stem cell-specific factors and should invite more investigation on the cooperation between VPA and NRs on iPSC induction.


Asunto(s)
Factor de Transcripción COUP II/genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Musculares/efectos de los fármacos , Miembro 1 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Ácido Valproico/farmacología , Animales , Secuencia de Bases , Factor de Transcripción COUP II/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Reprogramación Celular , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Datos de Secuencia Molecular , Células Musculares/citología , Células Musculares/metabolismo , Miembro 1 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/agonistas , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Ácido Valproico/análogos & derivados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...