Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Am Chem Soc ; 146(15): 10753-10766, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38578841

RESUMEN

Proteolysis targeting chimera (PROTAC) technology is an innovative strategy for cancer therapy, which, however, suffers from poor targeting delivery and limited capability for protein of interest (POI) degradation. Here, we report a strategy for the in situ formulation of antineoplastic Supra-PROTACs via intracellular sulfatase-responsive assembly of peptides. Coassembling a sulfated peptide with two ligands binding to ubiquitin VHL and Bcl-xL leads to the formation of a pro-Supra-PROTAC, in which the ratio of the two ligands is rationally optimized based on their protein binding affinity. The resulting pro-Supra-PROTAC precisely undergoes enzyme-responsive assembly into nanofibrous Supra-PROTACs in cancer cells overexpressing sulfatase. Mechanistic studies reveal that the pro-Supra-PROTACs selectively cause apparent cytotoxicity to cancer cells through the degradation of Bcl-xL and the activation of caspase-dependent apoptosis, during which the rationally optimized ligand ratio improves the bioactivity for POI degradation and cell death. In vivo studies show that in situ formulation enhanced the tumor accumulation and retention of the pro-Supra-PROTACs, as well as the capability for inhibiting tumor growth with excellent biosafety when coadministrating with chemodrugs. Our findings provide a new approach for enzyme-regulated assembly of peptides in living cells and the development of PROTACs with high targeting delivering and POI degradation efficiency.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Quimera Dirigida a la Proteólisis , Antineoplásicos/farmacología , Sulfatasas , Proteolisis , Péptidos , Ubiquitina-Proteína Ligasas
2.
Angew Chem Int Ed Engl ; : e202404703, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38655625

RESUMEN

Self-assembly in living cells represents one versatile strategy for drug delivery; however, it suffers from the limited precision and efficiency. Inspired by viral traits, we here report a cascade targeting-hydrolysis-transformation (THT) assembly of glycosylated peptides in living cells holistically resembling viral infection for efficient cargo delivery and combined tumor therapy. We design a glycosylated peptide via incorporating a ß-galactose-serine residue into bola-amphiphilic sequences. Co-assembling of the glycosylated peptide with two counterparts containing irinotecan (IRI) or ligand TSFAEYWNLLSP (PMI) results in formation of the glycosylated co-assemblies SgVEIP, which target cancer cells via ß-galactose-galectin-1 association and undergo galactosidase-induced morphological transformation. While GSH-reduction causes release of IRI from the co-assemblies, the PMI moieties release p53 and facilitate cell death via binding with protein MDM2. Cellular experiments show membrane targeting, endo-/lysosome-mediated internalization and in situ formation of nanofibers in cytoplasm by SgVEIP. This cascade THT process enables efficient delivery of IRI and PMI into cancer cells secreting Gal-1 and overexpressing ß-galactosidase. In vivo studies illustrate enhanced tumor accumulation and retention of the glycosylated co-assemblies, thereby suppressing tumor growth. Our findings demonstrate an in situ assembly strategy mimicking viral infection, thus providing a new route for drug delivery and cancer therapy in the future.

3.
Biomacromolecules ; 25(4): 2497-2508, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38478850

RESUMEN

Morphology-transformational self-assembly of peptides allows for manipulation of the performance of nanostructures and thereby advancing the development of biomaterials. Acceleration of the morphological transformation process under a biological microenvironment is important to efficiently implement the tailored functions in living systems. Herein, we report redox-regulated in situ seed-induced assembly of peptides via design of two co-assembled bola-amphiphiles serving as a redox-resistant seed and a redox-responsive assembly monomer, respectively. Both of the peptides are able to independently assemble into nanoribbons, while the seed monomer exhibits stronger assembling propensity. The redox-responsive monomer undergoes morphological transformation from well-defined nanoribbons to nanoparticles. Kinetics studies validate the role of the assembled inert monomer as the seeds in accelerating the assembly of the redox-responsive monomer. Alternative addition of oxidants and reductants into the co-assembled monomers promotes the redox-regulated assembly of the peptides facilitated by the in situ-formed seeds. The reduction-induced assembly of the peptide could also be accelerated by in situ-formed seeds in cancer cells with a high level of reductants. Our findings demonstrate that through precisely manipulating the assembling propensity of co-assembled monomers, the in situ seed-induced assembly of peptides could be achieved. Combining the rapid assembly kinetics of the seed-induced assembly with the common presence of redox agents in a biological microenvironment, this strategy potentially offers a new method for developing biomedical materials in living systems.


Asunto(s)
Nanoestructuras , Nanotubos de Carbono , Sustancias Reductoras , Péptidos/química , Nanoestructuras/química , Materiales Biocompatibles , Oxidación-Reducción
4.
Nat Commun ; 15(1): 831, 2024 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-38280857

RESUMEN

Transposon-associated ribonucleoprotein TnpB is known to be the ancestry endonuclease of diverse Cas12 effector proteins from type-V CRISPR system. Given its small size (408 aa), it is of interest to examine whether engineered TnpB could be used for efficient mammalian genome editing. Here, we showed that the gene editing activity of native TnpB from Deinococcus radiodurans (ISDra2 TnpB) in mouse embryos was already higher than previously identified small-sized Cas12f1. Further stepwise engineering of noncoding RNA (ωRNA or reRNA) component of TnpB significantly elevated the nuclease activity of TnpB. Notably, an optimized TnpB-ωRNA system could be efficiently delivered in vivo with single adeno-associated virus (AAV) and corrected the disease phenotype in a tyrosinaemia mouse model. Thus, the engineered miniature TnpB system represents a new addition to the current genome editing toolbox, with the unique feature of the smallest effector size that facilitate efficient AAV delivery for editing of cells and tissues.


Asunto(s)
Edición Génica , Tirosinemias , Ratones , Animales , Sistemas CRISPR-Cas/genética , Tirosinemias/genética , Tirosinemias/terapia , Mamíferos
5.
Angew Chem Int Ed Engl ; 62(49): e202314578, 2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-37870078

RESUMEN

The presence of disordered region or large interacting surface within proteins significantly challenges the development of targeted drugs, commonly known as the "undruggable" issue. Here, we report a heterogeneous peptide-protein assembling strategy to selectively phosphorylate proteins, thereby activating the necroptotic signaling pathway and promoting cell necroptosis. Inspired by the structures of natural necrosomes formed by receptor interacting protein kinases (RIPK) 1 and 3, the kinase-biomimetic peptides are rationally designed by incorporating natural or D -amino acids, or connecting D -amino acids in a retro-inverso (DRI) manner, leading to one RIPK3-biomimetic peptide PR3 and three RIPK1-biomimetic peptides. Individual peptides undergo self-assembly into nanofibrils, whereas mixing RIPK1-biomimetic peptides with PR3 accelerates and enhances assembly of PR3. In particular, RIPK1-biomimetic peptide DRI-PR1 exhibits reliable binding affinity with protein RIPK3, resulting in specific cytotoxicity to colon cancer cells that overexpress RIPK3. Mechanistic studies reveal the increased phosphorylation of RIPK3 induced by RIPK1-biomimetic peptides, elucidating the activation of the necroptotic signaling pathway responsible for cell death without an obvious increase in secretion of inflammatory cytokines. Our findings highlight the potential of peptide-protein hybrid aggregation as a promising approach to address the "undruggable" issue and provide alternative strategies for overcoming cancer resistance in the future.


Asunto(s)
Apoptosis , Péptidos , Apoptosis/fisiología , Muerte Celular , Fosforilación , Péptidos/farmacología , Aminoácidos
6.
Biomater Sci ; 11(16): 5680-5693, 2023 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-37439322

RESUMEN

As a key pathogen of periodontitis, P. gingivalis requires support of the initial colonizing bacterium (S. gordonii preferably) to form symbiotic biofilms on gingival tissues with enhanced antibiotic resistance. Here, we report a new strategy to treat periodontitis biofilms with S. gordonii membrane-coated H2O2 self-supplied nanocomposites (ZnO2/Fe3O4@MV NPs) in a "Jenga" style. Integration of our special MV coatings enables selectively enhanced internalization of the cargos in S. gordonii, thus inducing severe damage to the foundational bacterial layer and collapse/clearance of symbiotic biofilms consequently. This strategy allows us to clear the symbiotic biofilms of S. gordonii and P. gingivalis with active hydroxyl radicals (˙OH) derived from ZnO2-Fe3O4@MV NPs in a H2O2 self-supplied, nanocatalyst-assisted manner. This "Jenga-style" treatment provides a cutting-edge proof of concept for the removal of otherwise robust symbiotic biofilms of periodontitis where the critical pathogens are difficult to target and have antibiotic resistance.


Asunto(s)
Periodontitis , Óxido de Zinc , Humanos , Adhesión Bacteriana , Peróxido de Hidrógeno , Proteínas Bacterianas , Streptococcus gordonii , Periodontitis/microbiología , Biopelículas
8.
Lab Chip ; 22(16): 2944-2953, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35766807

RESUMEN

Pathogenic infections may lead to disruption of homeostasis, thus becoming a serious threat to the human health. Understanding the interactions between bacteria and macrophages is critical for therapeutic development against sepsis or inflammatory bowel disease. Here, we report a technique using droplet biosensors for the detection of nitric oxide (NO) secreted by a single macrophage under inflammatory stimuli. We demonstrated that the limit of detection can be promoted more than two orders of magnitude by our approach, in comparison to the conventional microplate format. The experiments of co-encapsulating single macrophages and different numbers of Escherichia coli (E. coli) enabled fluorescence monitoring of NO secretion by single macrophages over the incubation, and investigation of their interactions inside the isolated droplet for their separate fates. Our approach provides a unique platform to study the bacteria-macrophage interactions at the single cell level.


Asunto(s)
Escherichia coli , Sepsis , Bacterias , Humanos , Macrófagos , Óxido Nítrico
9.
Adv Sci (Weinh) ; 9(18): e2105775, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35460187

RESUMEN

Splicing factor SRSF2 acts as a critical regulator for cell survival, however, it remains unknown whether SRSF2 is involved in myoblast proliferation and myogenesis. Here, knockdown of SRSF2 in myoblasts causes high rates of apoptosis and defective differentiation. Combined conditional knockout and lineage tracing approaches show that Myf5-cre mice lacking SRSF2 die immediately at birth and exhibit a complete absence of mature myofibers. Mutant Myf5-derived cells (tdtomato-positive cells) are randomly scattered in the myogenic and non-myogenic regions, indicating loss of the community effect required for skeletal muscle differentiation. Single-cell RNA-sequencing reveals high heterogeneity of myf5-derived cells and non-myogenic cells are significantly increased at the expense of skeletal muscle cells in the absence of SRSF2, reflecting altered cell fate. SRSF2 is demonstrated to regulate the entry of Myf5 cells into the myogenic program and ensures their survival by preventing precocious differentiation and apoptosis. In summary, SRSF2 functions as an essential regulator for Myf5-derived cells to respond correctly to positional cues and to adopt their myogenic fate.


Asunto(s)
Desarrollo de Músculos , Músculo Esquelético , Animales , Ratones , Ratones Noqueados , Desarrollo de Músculos/genética , Músculo Esquelético/fisiología , Factor 5 Regulador Miogénico/genética , Análisis de Secuencia de ARN
10.
Sci Adv ; 8(7): eabl4923, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35171681

RESUMEN

Malignant tumors will become vulnerable if their uncontrolled biosynthesis and energy consumption engaged in metabolic reprogramming can be cut off. Here, we report finding a glycolytic inhibitor targeting glioblastoma with graphite dots-assisted laser desorption/ionization mass spectrometry as an integrated drug screening and pharmacokinetic platform (GLMSD). We have performed high-throughput virtual screening to narrow an initial library of 240,000 compounds down to the docking of 40 compounds and identified five previously unknown chemical scaffolds as promising hexokinase-2 inhibitors. The best inhibitor (Compd 27) can regulate the reprogrammed metabolic pathway in U87 glioma cells (median inhibitory concentration ~ 11.3 µM) for tumor suppression. Highly effective therapy against glioblastoma has been demonstrated in both subcutaneous and orthotopic brain tumors by synergizing Compd 27 and temozolomide. Our glycolytic inhibitor discovery can inspire personalized medicine targeting reprogrammed metabolisms of malignant tumors. GLMSD enables large, high-quality data for next-generation artificial intelligence-aided drug development.


Asunto(s)
Glioblastoma , Grafito , Inteligencia Artificial , Línea Celular Tumoral , Glioblastoma/metabolismo , Humanos , Espectrometría de Masas , Temozolomida/farmacología , Temozolomida/uso terapéutico
11.
Adv Mater ; 34(17): e2108167, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35132688

RESUMEN

Chemoimmunotherapy using nanotechnology has shown great potential for cancer therapy in the clinic. However, uncontrolled transportation and synergistic responses remain challenges. Here, a self-assembled selenopeptide nanoparticle that strengthens tumor chemoimmunotherapy through the activation of natural killer (NK) cells by the oxidative metabolite of the selenopeptide is developed. With the advantages of the enzyme-induced size-reduction and the reactive-oxygen-species-driven deselenization, this selenopeptide is able to deliver therapeutics, e.g., doxorubicin (DOX), to solid tumors and further activate the NK cells in a programmed manner. Importantly, in vitro and in vivo results prove the mutual promotion between the DOX-induced chemotherapy and the selenopeptide-induced immunotherapy, which synergistically contribute to the improved antitumor efficacy. It is anticipated that the selenopeptide may provide a type of promising stimuli-responsive immune modulator for versatile biomedical applications.


Asunto(s)
Nanopartículas , Neoplasias , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Humanos , Inmunoterapia , Células Asesinas Naturales , Nanomedicina , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
12.
Analyst ; 146(18): 5533-5541, 2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34515705

RESUMEN

It is a pre-requisite to ionize analyte molecules efficiently for detection by laser desorption/ionization mass spectrometry. Here, we report a conceptual demonstration of cationizing neutral small molecules which are typically difficult to be ionized with the traditional organic matrices due to their low proton/cation affinity values. Our strategy features generating radical cations from site-specifically carboxylated 10-(4-carboxyphenyl)-10H-phenothiazine-3,7-dicarboxylic acid (PTZ(A)2-Ph(A)) with a laser, and anchoring the chlorine ion from NaCl through covalent bond-like bridging interactions with the N/S atoms in the heterocyclic structure. This "Maverick" design allows a dramatic change of the energy landscape of analyte sodiation with an enhanced efficiency. We have synthesized two families of compounds based on the model structures of phenothiazine (PTZ) and phenoxazine (PXZ) and their carboxylated derivatives, and performed comparison between them or against the traditional organic matrices in a systematic format. We have demonstrated that PTZ(A)2-Ph(A) is outstanding as a novel MALDI matrix for the detection of oligosaccharides and amino acids, with an ultra-clean background baseline and high signal-to-noise ratios (up to dozens of times better than the traditional matrices). This work provides a new method for the cationization of neutral small molecules in a distinct mechanism, inspiring the development of next-generation matrices for sensitive detection of hard-to-be-ionized molecules by MALDI MS.


Asunto(s)
Oligosacáridos , Protones , Rayos Láser , Fenotiazinas , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Angew Chem Int Ed Engl ; 60(47): 25128-25134, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34549872

RESUMEN

Therapeutic peptides have been widely concerned, but their efficacy is limited by the inability to penetrate cell membranes, which is a key bottleneck in peptide drugs delivery. Herein, an in vivo self-assembly strategy is developed to induce phase separation of cell membrane that improves the peptide drugs internalization. A phosphopeptide KYp is synthesized, containing an anticancer peptide [KLAKLAK]2 (K) and a responsive moiety phosphorylated Y (Yp). After interacting with alkaline phosphatase (ALP), KYp can be dephosphorylated and self-assembles in situ, which induces the aggregation of ALP and the protein-lipid phase separation on cell membrane. Consequently, KYp internalization is 2-fold enhanced compared to non-responsive peptide, and IC50 value of KYp is approximately 5 times lower than that of free peptide. Therefore, the in vivo self-assembly induced phase separation on cell membrane promises a new strategy to improve the drug delivery efficacy in cancer therapy.


Asunto(s)
Membrana Celular/química , Péptidos/aislamiento & purificación , Fosfatasa Alcalina/metabolismo , Membrana Celular/metabolismo , Humanos , Péptidos/química , Péptidos/metabolismo , Conformación Proteica
14.
Nutr Metab (Lond) ; 18(1): 27, 2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33691715

RESUMEN

BACKGROUND: Cancer-associated cachexia is a multifactorial syndrome defined by progressive weight loss with ongoing loss of adipose tissue and skeletal muscle. Adipose loss occurs in the early stage of cachexia and is associated with reduced quality of life and survival time. Although numerous lncRNAs are regarded as novel regulators in adipose metabolism, the role of lncRNAs that selectively modulate the development of adipose loss in cachexia remains limited. METHODS: In this study, we analyzed microarray data of lncRNAs in adipose loss and further explored the function and mechanism of MALAT1 in adipose loss. First, we explored the expression and function of MALAT1 in adipose cell by quantitative PCR and RNA knockdown. Subsequently, the mechanism of MALAT1 involvement in adipose loss was analyzed via RNA-seq, bioinformatics analysis and reporter gene assay. Finally, we explored the clinical significance of MALAT1 through correlation analysis. RESULTS: Cellular experiments revealed that knocking down MALAT1 significantly inhibited the process of adipogenesis. RNA-seq data showed that numerous adipogenic genes were downregulated upon MALAT1 knockdown. A protein-protein interaction network analysis identified PPAR-γ as the central node transcription factor, the inhibition of which explains the downregulation of numerous adipogenic genes. A reporter gene assay suggested that MALAT1 can regulate the gene expression of PPAR-γ at the transcriptional level. Moreover, MALAT1 was weakly expressed in the subcutaneous white adipose tissue of cancer-associated cachexia patients and was related to low fat mass index and poor prognosis in cancer patients. CONCLUSIONS: This study indicated that MALAT1 is associated with adipose loss in cancer-associated cachexia by regulating adipogenesis through PPAR-γ, which may potentially be a novel target for the diagnosis and treatment of cancer-associated cachexia in the clinic.

15.
Biomater Sci ; 8(22): 6175-6189, 2020 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-33026364

RESUMEN

Peptides have shown great potential in cancer treatment due to their good biocompatibility and low toxicity. However, the bioavailability and adverse immune response of peptides limit their further translation from bench to bedside. Over the past few decades, various peptide-based nanomaterials have been developed for drug delivery and cancer treatment. Compared with therapeutic peptides alone, self-assembled peptide nanomaterials have obvious advantages, such as improved stability and biodistribution for high-performance cancer therapy. In this review, we have described the synthesis, self-assembly and the anti-cancer application of therapeutic peptides and their conjugates, particularly polymer-peptide conjugates (PPCs).


Asunto(s)
Nanoestructuras , Neoplasias , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Péptidos , Polímeros , Distribución Tisular
16.
Talanta ; 220: 121371, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-32928398

RESUMEN

Appropriate prescription of antibiotics requires the pharmacokinetic knowledge of the drugs and their metabolites in blood, and their distribution/retention in organ tissues. Here we report that highly crystalline graphite dots (GDs) allow for quantitative profiling of antibiotic metabolites in a spatial-temporal manner, in combination with matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI MSI). GDs matrix features an ultra-clean background base line and high efficiency in ionization of small molecules, thus enabling quantification of sulfamethoxazole (SMZ) and its metabolites with limit of detection (LOD) in the femtomole range. Distinctly different from the other MS techniques, our approach is tolerant to high levels of salt contaminants in the complexed biological samples, thus minimizing the sample purification requirements and allowing for tests in small volumes. We have demonstrated quantitative measurements of the dynamic concentration changes of SMZ (m/z 276.27) and two metabolites, N4-hydroxy-SMZ (m/z 292.28) and N4-acetyl-SMZ (m/z 318.31) with only 1 µL mouse blood sample for each test. High-resolution distribution patterns of SMZ metabolites have directly been visualized a on the liver subsegments. Therefore, it allows for simultaneously acquisition of pharmacokinetic data in the blood combined with detailed hepatic zonation of SMZ metabolites for the first time. As a rapid, high-throughput platform to monitor small molecules in vivo, our approach of GDs-assisted MADLI MSI will foster the medical research on the antibiotic usage and drug development.


Asunto(s)
Antibacterianos , Grafito , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Animales , Antibacterianos/análisis , Antibacterianos/metabolismo , Rayos Láser , Ratones , Espectrofotometría
17.
Stem Cell Reports ; 15(4): 941-954, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-32888503

RESUMEN

Satellite cells are main muscle stem cells that could provide myonuclei for myofiber growth and synaptic-specific gene expression during the early postnatal development. Here, we observed that splicing factor SRSF1 is highly expressed in myoblasts and its expression is closely related with satellite cell activation and proliferation. By genetic deletion of SRSF1 in myogenic progenitors, we found that SRSF1 is critical for satellite cell proliferation in vitro and in vivo. Most notably we also observed that SRSF1 is required for the functional neuromuscular junction (NMJ) formation, as SRSF1-deficient mice fail to form mature pretzel-like NMJs, which leads to muscle weakness and premature death in mice. Finally, we demonstrated that SRSF1 contributes to muscle innervation and muscle development likely by regulating a restricted set of tissue-specific alternative splicing events. Thus, our data define a unique role for SRSF1 in postnatal skeletal muscle growth and function in mice.


Asunto(s)
Diferenciación Celular , Unión Neuromuscular/citología , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Factores de Empalme Serina-Arginina/metabolismo , Empalme Alternativo/genética , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Proliferación Celular , Eliminación de Gen , Ratones Endogámicos C57BL , Ratones Noqueados , Atrofia Muscular/patología , Factores de Empalme Serina-Arginina/deficiencia
18.
J Mater Chem B ; 8(34): 7792-7800, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32747886

RESUMEN

There is increasing demand for anti-doping drug monitoring in sports and food safety checks by developing sensitive and fast analytical methods. Here we report the development of hybrid Ir/SiNW as a new MALDI matrix for the detection of small molecules. This matrix is characterized by sufficient UV absorption, low-noise background, and high efficiency in ionization of small molecules. Sensitive detection of clenbuterol (LOD: 0.18 pmol) and a variety of other small molecules has been achieved using the Ir/SiNW matrix with a reproducible performance. Compared to the individual components separately, the matrix of hybrid Ir/SiNW synthesized via in situ growth can promote the MS signal intensity by up to 10 fold under identical experimental conditions. We provide a unique mechanism for the high performance of the hybrid Ir/SiNW matrix with the characteristic properties of hydrogen atom transfer and enhanced protonation at the interface of the hybrid nanostructures. Our approach of using a hybrid Ir/SiNW matrix enables detection of clenbuterol quantitatively in complicated biological samples and in vivo experiments, promising a useful tool for food security and anti-doping drug monitoring in sports.


Asunto(s)
Clenbuterol/análisis , Iridio/química , Límite de Detección , Espectrometría de Masas/métodos , Nanocables/química , Silicio/química
19.
Commun Biol ; 3(1): 210, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32372053

RESUMEN

Splicing factor SRSF2 is frequently mutated or up-regulated in human cancers. Here, we observe that hepatocyte-specific deletion of Srsf2 trigger development of hepatocellular carcinoma (HCC) in mice, which also involves inflammation and fibrosis. Importantly, we find that, when compensatory hepatocyte proliferation is impaired, activation of hepatic progenitor cells (HPCs) play an important role in liver regeneration and tumor formation. Moreover, the cells of HCC- bearing livers display both HPC and hepatocyte markers, with gene expression profiling suggesting HPC origin and embryonic origin. Mechanically, we demonstrate that levels of oncofetal genes insulin-like growth factor 2 (Igf2) and H19 are significantly increased in the tumors, likely due to decreased DNA methylation of the Igf2/H19 locus. Consequently, signaling via the Igf2 pathway is highly activated in the tumors. Thus, our data demonstrate that loss of Srsf2 triggers HPC-mediated regeneration and activation of oncofetal genes, which altogether promote HCC development and progression in mice.


Asunto(s)
Carcinoma Hepatocelular/genética , Eliminación de Gen , Hepatocitos , Neoplasias Hepáticas/genética , Factores de Empalme Serina-Arginina/deficiencia , Células Madre/metabolismo , Regulación Neoplásica de la Expresión Génica
20.
ACS Nano ; 14(3): 3640-3650, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32119522

RESUMEN

The shape of a drug delivery system impacts its in vivo behavior such as circulation time, accumulation, and penetration. Considering the advantages of functional dyes in bioapplications, we synthesize a class of nanoaggregates based on BF2-azadipyrromethene (aza-BODIPY) dyes, which can realize long blood circulation and deep tumor penetration simultaneously in vivo through morphological transformation modulated by a near-infrared (NIR) laser. First, when the temperature increases, the wormlike nanofibers of the aza-BODIPY-1 aggregate, possessing a long blood circulation time, can be transformed into spherical nanoparticles, which are conducive to increasing the penetration in the solid tumor. Second, without any postmodification, the nanofibers exhibit an outstandingly narrow absorption band in the NIR spectral range, so that they possess ideal photothermal properties. Through 655 nm laser irradiation, the intrinsic photothermal effect causes a local temperature increase to ∼48 °C, realizing the transformation of 1-NFs to 1-NPs. Third, the morphological transformation is real-time detected by photoacoustic (PA) imaging. By monitoring the change of the PA signal at a specific wavelength, the in vivo deformation process of nanomaterials can be traced. Consequently, the in situ morphology transformation of aza-BODIPY-based nanomaterials can simultaneously realize long blood circulation and deep penetration, resulting in the enhanced antitumor outcome.


Asunto(s)
Compuestos de Boro/química , Neoplasias de la Mama/diagnóstico por imagen , Colorantes Fluorescentes/química , Rayos Láser , Nanopartículas/química , Animales , Compuestos de Boro/administración & dosificación , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/síntesis química , Humanos , Rayos Infrarrojos , Inyecciones Intravenosas , Células MCF-7 , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/administración & dosificación , Imagen Óptica , Tamaño de la Partícula , Procesos Fotoquímicos , Propiedades de Superficie , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...