Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
NPJ Precis Oncol ; 8(1): 58, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38429411

RESUMO

Abnormal lipid metabolism promotes hepatocellular carcinoma (HCC) progression, which engenders therapeutic difficulties owing to unclear mechanisms of the phenomenon. We precisely described a special steatotic HCC subtype with HBV-related cirrhosis and probed its drivers. Hematoxylin-eosin (HE) staining of 245 HCC samples revealed a special HCC subtype (41 cases) characterized by HBV-related cirrhosis and intratumoral steatosis without fatty liver background, defined as steatotic HCC with HBV-related cirrhosis (SBC-HCC). SBC-HCC exhibits a larger tumor volume and worse prognosis than non-SBC-HCC. Screening for driver genes promoting fatty acid (FA) biosynthesis in the Gao's HBV-related cirrhosis HCC cases and GSE121248' HBV-related HCC cases revealed that high expression of SOCS5 predicts increased FA synthesis and that SOCS5 is upregulated in SBC-HCC. Through proteomics, metabolomics, and both in vivo and in vitro experiments, we demonstrated that SOCS5 induces lipid accumulation to promote HCC metastasis. Mechanistically, through co-IP and GST-pulldown experiments, we found that the SOCS5-SH2 domain, especially the amino acids Y413 and D443, act as critical binding sites for the RBMX-RRM domain. SOCS5-RBMX costimulates the promoter of SREBP1, inducing de novo lipogenesis, while mutations in the SH2 domain, Y413, and D443 reverse this effect. These findings precisely identified SBC-HCC as a special steatotic HCC subtype and highlighted a new mechanism by which SOCS5 promotes SBC-HCC metastasis.

2.
Pac Symp Biocomput ; 29: 521-533, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38160304

RESUMO

Advances in molecular characterization have reshaped our understanding of low-grade glioma (LGG) subtypes, emphasizing the need for comprehensive classification beyond histology. Lever-aging this, we present a novel approach, network-based Subnetwork Enumeration, and Analysis (nSEA), to identify distinct LGG patient groups based on dysregulated molecular pathways. Using gene expression profiles from 516 patients and a protein-protein interaction network we generated 25 million sub-networks. Through our unsupervised bottom-up approach, we selected 92 subnetworks that categorized LGG patients into five groups. Notably, a new LGG patient group with a lack of mutations in EGFR, NF1, and PTEN emerged as a previously unidentified patient subgroup with unique clinical features and subnetwork states. Validation of the patient groups on an independent dataset demonstrated the robustness of our approach and revealed consistent survival traits across different patient populations. This study offers a comprehensive molecular classification of LGG, providing insights beyond traditional genetic markers. By integrating network analysis with patient clustering, we unveil a previously overlooked patient subgroup with potential implications for prognosis and treatment strategies. Our approach sheds light on the synergistic nature of driver genes and highlights the biological relevance of the identified subnetworks. With broad implications for glioma research, our findings pave the way for further investigations into the mechanistic underpinnings of LGG subtypes and their clinical relevance.Availability: Source code and supplementary data are available at https://github.com/bebeklab/nSEA.


Assuntos
Neoplasias Encefálicas , Glioma , Humanos , Prognóstico , Biologia Computacional , Glioma/genética , Glioma/patologia , Algoritmos , Mapas de Interação de Proteínas , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia
3.
Angew Chem Int Ed Engl ; 62(45): e202307664, 2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37718311

RESUMO

Protein therapeutics targeting intracellular machineries hold profound potential for disease treatment, and hence robust cytosolic protein delivery technologies are imperatively demanded. Inspired by the super-negatively charged, nucleotide-enriched structure of nucleic acids, adenylated pro-proteins (A-proteins) with dramatically enhanced negative surface charges have been engineered for the first time via facile green synthesis. Then, thymidine-modified polyethyleneimine is developed, which exhibits strong electrostatic attraction, complementary base pairing, and hydrophobic interaction with A-proteins to form salt-resistant nanocomplexes with robust cytosolic delivery efficiencies. The acidic endolysosomal environment enables traceless restoration of the A-proteins and consequently promotes the intracellular release of the native proteins. This strategy shows high efficiency and universality for a variety of proteins with different molecular weights and isoelectric points in mammalian cells. Moreover, it enables highly efficient delivery of CRISPR-Cas9 ribonucleoproteins targeting fusion oncogene EWSR1-FLI1, leading to pronounced anti-tumor efficacy against Ewing sarcoma. This study provides a potent and versatile platform for cytosolic protein delivery and gene editing, and may benefit the development of protein pharmaceuticals.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Animais , Pareamento de Bases , Proteínas/genética , Endossomos , Mamíferos
4.
Biomater Sci ; 11(16): 5653-5662, 2023 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-37431292

RESUMO

Cationic polymers used for nucleic acid delivery often suffer from complicated syntheses, undesired intracellular cargo release and low serum stability. Herein, a series of ternary polymers were synthesized via facile green chemistry to achieve efficient plasmid DNA and mRNA delivery in serum. During the one-pot synthesis of the ternary polymer, acetylphenylboric acid (APBA), polyphenol and low-molecular weight polyethyleneimine (PEI 1.8k) were dynamically cross-linked with each other due to formation of an imine between PEI 1.8k and APBA and formation of a boronate ester between APBA and polyphenol. Series of polyphenols, including ellagic acid (EA), epigallocatechin gallate (EGCG), nordihydroguaiaretic acid (NDGA), rutin (RT) and rosmarinic acid (RA), and APBA molecules, including 2-acetylphenylboric acid (2-APBA), 3-acetylphenylboric acid (3-APBA) and 4-acetylphenylboric acid (4-APBA), were screened and the best-performing ternary polymer, 2-PEI-RT, constructed from RT and 2-APBA, was identified. The ternary polymer featured efficient DNA condensation to favor cellular internalization, and the acidic environment in endolysosomes triggered effective degradation of the polymer to promote cargo release. Thus, 2-PEI-RT showed robust plasmid DNA transfection efficiencies in various tumor cells in serum, outperforming the commercial reagent PEI 25k by 1-3 orders of magnitude. Moreover, 2-PEI-RT mediated efficient cytosolic delivery of Cas9-mRNA/sgRNA to enable pronounced CRISPR-Cas9 genome editing in vitro. Such a facile and robust platform holds great potential for non-viral nucleic acid delivery and gene therapy.


Assuntos
Ácidos Nucleicos , Polímeros , Polímeros/química , Técnicas de Transferência de Genes , RNA Guia de Sistemas CRISPR-Cas , Transfecção , Polietilenoimina/química , DNA/genética , Plasmídeos , Polifenóis
5.
J Clin Transl Hepatol ; 11(3): 572-583, 2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-36969886

RESUMO

Background and Aims: Syntaxin 5 (STX5) is a member of the syntaxin or target-soluble SNAP receptor (t-SNARE) family and plays a critical role in autophagy. However, its function and molecular mechanism in tumor cell migration are still unknown. The role of STX5 in influencing hepatocellular carcinoma (HCC) is an important topic in our research. Methods: By using quantitative reverse transcription polymerase chain reaction (qPCR), western blotting, and immunohistochemical analysis of RNA and protein in tissues, we comprehensively evaluated data sets from public databases and clinical patient cohorts for STX5. The correlation of STX5 expression with the clinicopathological characteristics of HCC patients were assessed. In addition, we predicted signal pathways from differentially expressed genes (DEGs) and the Cancer Genome Atlas (TCGA) databases, and confirmed the prediction using integrated transcriptome and RNA-seq. We further investigated the underlying mechanisms of STX5 in the migration and adhesion of HCC cells both in vitro and in vivo. Results: In the TCGA dataset and our patient cohort, STX5 levels were significantly higher in HCC tissues than in adjacent normal liver tissues. At the same time, high expression of STX5 predicted worse prognosis in patients with liver cancer. High expression of STX5 indicates the decrease of adhesion and the increase of migration of HCC cells, and the conversion of epithelial-mesenchymal transition (EMT) in vitro via PI3K/mTOR pathway activation. Conversely, when Sirolimus, a phosphoinositide 3-kinase (PI3K)/AKT/mechanistic target of rapamycin (mTOR) inhibitor acts on cells simultaneously, STX5 overexpression-mediated enhancement of HCC metastasis is reversed. Double-negative regulation of STX5 and mTOR further enhanced the inhibitory effect of STX5 on HCC metastasis. In vivo, STX5 knockdown inhibited the metastasis of HCC cells. Conclusions: Our study demonstrates a novel research result that STX5 promotes HCC metastasis through PI3K/mTOR pathway. We believe that combined inhibition of STX5 and mTOR is a potential treatment for effectively prolonging patient survival and inhibiting HCC metastasis.

6.
Acta Biomater ; 152: 355-366, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36084925

RESUMO

Protein therapy targeting the intracellular machinery holds great potentials for disease treatment, and therefore, effective cytosolic protein delivery technologies are highly demanded. Herein, we developed reactive oxygen species (ROS)-degradable, branched poly(ß-amino ester) (PBAE) with built-in phenylboronic acid (PBA) in the backbone and terminal-pendent arginine for the efficient cytosolic protein delivery. The PBAE could form stable and cell-ingestible nanocomplexes (NCs) with proteins via electrostatic interaction, nitrogen-boronate (N-B) coordination, and hydrogen bonding, while it can be degraded into small segments by the over-produced H2O2 in tumor cells to enable cytoplasmic protein release. As thus, PBAE exhibited high efficiency in delivering varieties of proteins with distinct molecular weights (12.4-430 kDa) and isoelectric points (4.7-10.5) into tumor cells, including enzymes, toxins, and antibodies. Moreover, PBAE mediated efficient delivery of saporin into tumor cells in vivo, provoking pronounced anti-tumor outcomes. This study provides a robust and versatile platform for cytosolic protein delivery, and the elaborately tailored PBAE may find promising applications for protein-based biological research and disease management. STATEMENT OF SIGNIFICANCE: Cytosolic delivery of native proteins holds great therapeutic potentials, which however, is limited by the lack of robust delivery carriers that can simultaneously feature strong protein encapsulation yet effective intracellular protein release. Herein, ROS-degradable, branched poly(ß-amino ester) (PBAE) with backbone-embedded phenylboronic acid (PBA) and terminal-pendent arginine was developed to synchronize these two processes. PBA and arginine moieties allowed PBAE to encapsulate proteins via N-B coordination, electrostatic interaction, hydrogen bonding, and salt bridging, while PBA could be oxidized by over-produced H2O2 inside cancer cells to trigger PBAE degradation and intracellular protein release. As thus, the top-performing PBAE mediated efficient cytosolic delivery of various proteins including enzymes, toxins, and antibodies. This study provides a powerful platform for cytosolic protein delivery, and may find promising utilities toward intracellular protein therapy against cancer and other diseases such as inflammation.


Assuntos
Nanopartículas , Neoplasias , Arginina , Ácidos Borônicos , Ésteres , Humanos , Peróxido de Hidrogênio , Nitrogênio , Polímeros , Espécies Reativas de Oxigênio , Saporinas
7.
Adv Mater ; 34(35): e2110560, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35789055

RESUMO

Protein drugs targeting intracellular machineries have shown profound therapeutic potentials, but their clinical utilities are greatly hampered by the lack of efficient cytosolic delivery techniques. Existing strategies mainly rely on nanocarriers or conjugated cell-penetrating peptides (CPPs), which often have drawbacks such as materials complexity/toxicity, lack of cell specificity, and endolysosomal entrapment. Herein, a unique carrier-free approach is reported for mediating cancer-selective and endocytosis-free cytosolic protein delivery. Proteins are sequentially modified with 4-nitrophenyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) benzyl carbonate as the H2 O2 -responsive domain and 3,4-dihydroxy-l-phenylalanine as the substrate of l-type amino acid transporter 1 (LAT1). Thus, the pro-protein can be directly transported into tumor cells by overexpressed LAT1 on cell membranes, bypassing endocytosis and endolysosomal entrapment. In the cytosol, overproduced H2 O2 restores the protein structure and activity. Using this technique, versatile proteins are delivered into tumor cells with robust efficiency, including toxins, enzymes, CRISPR-Cas9 ribonucleoprotein, and antibodies. Furthermore, intravenously injected pro-protein of saporin shows potent anticancer efficacy in 4T1-tumor-bearing mice, without provoking systemic toxicity. Such a facile and versatile pro-protein platform may benefit the development of protein pharmaceuticals.


Assuntos
Transportador 1 de Aminoácidos Neutros Grandes , Neoplasias Cutâneas , Animais , Transporte Biológico , Citosol/metabolismo , Endocitose , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Camundongos
8.
BMC Cancer ; 22(1): 379, 2022 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-35397536

RESUMO

BACKGROUND: The combined application of immune cells and specific biomarkers related to the tumor immune microenvironment has a better predictive value for the prognosis of HCC. The purpose of this study is to construct a new prognostic model based on immune-related genes that regulate cross-talk between immune and tumor cells to assess the prognosis and explore possible mechanisms. METHOD: The immune cell abundance ratio of 424 cases in the TCGA-LIHC database is obtained through the CIBERSORT algorithm. The differential gene analysis and cox regression analysis is used to screen IRGs. In addition, the function of IRGs was preliminarily explored through the co-culture of M2 macrophages and HCC cell lines. The clinical validation, nomogram establishment and performing tumor microenvironment score were validated. RESULTS: We identified 4 immune cells and 9 hub genes related to the prognosis. Further, we identified S100A9, CD79B, TNFRSF11B as an IRGs signature, which is verified in the ICGC and GSE76427 database. Importantly, IRGs signature is closely related to the prognosis, tumor microenvironment score, clinical characteristics and immunotherapy, and nomogram combined with clinical characteristics is more conducive to clinical promotion. In addition, after co-culture with M2 macrophages, the migration capacity and cell pseudopod of MHCC97H increased significantly. And CD79B and TNFRSF11B were significantly down-regulated in MHCC97H, Huh7 and LM3, while S100A9 was up-regulated. CONCLUSION: We constructed an IRGs signature and discussed possible mechanisms. The nomogram established based on IRGs can accurately predict the prognosis of HCC patients. These findings may provide a suitable therapeutic target for HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Nomogramas , Prognóstico , Microambiente Tumoral/genética
9.
Adv Mater ; 34(8): e2108116, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34894367

RESUMO

Cytosolic protein delivery is a prerequisite for protein-based biotechnologies and therapeutics on intracellular targets. Polymers that can complex with proteins to form nano-assemblies represent one of the most important categories of materials, because of the ease of nano-fabrication, high protein loading efficiency, no need for purification, and maintenance of protein bioactivity. Stable protein encapsulation and efficient intracellular liberation are two critical yet opposite processes toward cytosolic delivery, and polymers that can resolve these two conflicting challenges are still lacking. Herein, hyperbranched poly(ß-amino ester) (HPAE) with backbone-embedded phenylboronic acid (PBA) is developed to synchronize these two processes, wherein PBA enhanced protein encapsulation via nitrogen-boronate (N-B) coordination while triggered polymer degradation and protein release upon oxidation by H2 O2 in cancer cells. Upon optimization of the branching degree, charge density, and PBA distribution, the best-performing A2-B3-C2-S2 -P2 is identified, which mediates robust delivery of various native proteins/peptides with distinct molecular weights (1.6-430 kDa) and isoelectric points (4.1-10.3) into cancer cells, including enzymes, toxins, antibodies, and CRISPR-Cas9 ribonucleoproteins (RNPs). Moreover, A2-B3-C2-S2 -P2 mediates effective cytosolic delivery of saporin both in vitro and in vivo to provoke remarkable anti-tumor efficacy. Such a potent and universal platform holds transformative potentials for protein pharmaceuticals.


Assuntos
Ésteres , Polímeros , Citosol/metabolismo , Ésteres/análise , Ésteres/metabolismo , Polímeros/química , Proteínas/química
10.
Biomater Sci ; 9(19): 6425-6433, 2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34582529

RESUMO

Multifunctional coatings have gained significant attention for their promising potential to address the issue of medical device-related infections. However, they usually have multiple components in one layer which decreases the density of functional groups on surfaces and hence reduces the biological properties. Herein, we report a mono-component and sulfonate-based anionic polypeptide coating with on-demand antibacterial activity, antifouling property, and biocompatibility. The anionic polypeptide was prepared by ring-opening polymerization of L-cysteine-based N-carboxyanhydride (NCA) with allyl groups and a subsequent thiol-ene reaction to incorporate the sulfonate pendants. It adopted a 17.1-19.5% ß-sheet conformation and self-assembled into a spherical nanoparticle. The polypeptide coating showed excellent in vitro antibacterial activity against both Gram-positive (i.e., S. aureus) and Gram-negative bacteria (i.e., E. coli) with >99% killing efficacy after acidic solution treatment and prominent antifouling property and biocompatibility after weak base treatment. An in vivo study revealed that the sulfonate-based polypeptide-coated polydimethylsiloxane (PDMS) exhibited good anti-infection property and histocompatibility.


Assuntos
Anti-Infecciosos , Staphylococcus aureus , Antibacterianos/farmacologia , Materiais Revestidos Biocompatíveis , Escherichia coli , Peptídeos
11.
Biomater Sci ; 9(13): 4639-4647, 2021 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-34036971

RESUMO

Cytosolic protein delivery holds great potential for the development of protein-based biotechnologies and therapeutics. Currently, cytosolic protein delivery is mainly achieved with the assistance of various carriers. Herein, we present a universal and effective strategy for carrier-free cytosolic protein delivery via metabolic glycoengineering and bioorthogonal click reactions. Ac4ManNAz (AAM), an azido-modified N-acetylmannosamine analogue, was first employed to label tumor cell surfaces with abundant azido groups via glycometabolism. Then, proteins including RNase A, cytochrome C (Cyt C), and bovine serum albumin (BSA) were covalently modified with dibenzocyclooctyne (DBCO). Based on the highly efficient bioorthogonal click reactions between DBCO and azido, DBCO-modified proteins could be efficiently internalized by azido-labeled cancer cells. RNase A-DBCO could largely maintain its enzymatic activity and, thus, led to notable anti-tumor efficacy in HeLa and B16F10 cells in vitro and in B16F10 xenograft tumors in vivo. This study therefore provides a simple and powerful approach for carrier-free protein delivery and would have broad applicability in anti-tumor protein therapy.


Assuntos
Química Click , Neoplasias , Humanos , Neoplasias/tratamento farmacológico
12.
J Biomed Nanotechnol ; 17(2): 169-195, 2021 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-33785090

RESUMO

In recent years, emerging immunotherapy has been included in various malignant tumor treatment standards. Temperature has been considered to affect different pathophysiological reactions such as inflammation and cancer for a long time. However, in tumor immunology research, temperature is still rarely considered a significant variable. In this review, we discuss the effects of room temperature, body temperature, and the local tumor temperature on the tumor immune microenvironment from multiple levels and perspectives, and we discuss changes in the body's local and whole-body temperature under tumor conditions. We analyze the current use of ablation treatment-the reason for the opposite immune effect. We should pay more attention to the therapeutic potential of temperature and create a better antitumor microenvironment that can be combined with immunotherapy.


Assuntos
Neoplasias , Microambiente Tumoral , Humanos , Imunoterapia , Neoplasias/terapia , Temperatura
13.
Biomater Sci ; 9(7): 2670-2678, 2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33605949

RESUMO

Non-viral gene delivery vectors with high transfection efficiency both in vitro and in vivo and low cytotoxicity are highly desirable for clinical applications. Herein, a series of guanidine-rich polypeptides bearing hydrophobic amino acid pendants was efficiently prepared via the 1,3-dipolar cycloaddition between azido decorated polypeptide and propargyl functionalized guanidinium and N-acetylamino acids. CD analysis indicated α-helical conformations of all resulting polypeptides in aqueous solution. The guanidine-rich polypeptide/DNA complexes showed significantly enhanced cellular internalization and high cell viability (>90%) in different mammalian cell lines (i.e., HeLa and RAW 264.7) at concentrations of the best performance. The top-performing guanidine-rich polypeptide containing 10% N-acetyl-l-valine pendants outperformed the commercial transfection reagent PEI by 400 times in vitro and 6 times in vivo. This study provides a new guidance for future molecular design of non-viral gene vectors with high delivery efficiency and low cytotoxicity.


Assuntos
Aminoácidos , Técnicas de Transferência de Genes , Animais , Terapia Genética , Guanidina , Humanos , Peptídeos , Polietilenoimina , Transfecção
14.
Biomater Sci ; 9(7): 2721-2731, 2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33617610

RESUMO

Antimicrobial peptides (AMPs) have been considered as a promising new tool to combat the antimicrobial resistance (AMR) crisis. However, the high toxicity and high cost of AMPs hampered their further development. Herein, a series of star poly(L-lysine) (PLL) homo- and copolymers with excellent antimicrobial activity and improved biocompatibility were prepared by the combination of ultra-fast ring opening polymerization (ROP) and side-chain modification. The amine-terminated polyamidoamine dendrimer (Gx-PAMAM) mediated ROP of Nε-tert-butyloxycarbonyl-L-lysine N-carboxyanhydride (Boc-L-Lys-NCA) and γ-benzyl-L-glutamic acid-based N-carboxyanhydride (PBLG-NCA) was able to prepare star PLL homo- and copolymers with 400 residues within 50 min. While the star PLL homopolymers exhibited low minimum inhibitory concentration (MIC = 50-200 µg mL-1) against both Gram-positive and Gram-negative bacteria (i.e., S. aureus and E. coli), they showed high toxicity against various mammalian cell lines. The star PLL copolymers with low contents of hydrophobic and hydroxyl groups showed enhanced antimicrobial activity (MIC = 25-50 µg mL-1) and improved mammalian cell viability. Both SEM and CLSM results indicated the antimicrobial mechanism of membrane disruption.


Assuntos
Antibacterianos , Anti-Infecciosos , Animais , Antibacterianos/farmacologia , Anti-Infecciosos/farmacologia , Escherichia coli , Bactérias Gram-Negativas , Bactérias Gram-Positivas , Peptídeos , Staphylococcus aureus
15.
Biomater Sci ; 9(4): 1301-1312, 2021 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-33350407

RESUMO

The discrepancy of surface receptors on cancerous and non-cancerous cells has been regarded as the mainstay of cancer-targeted therapy. However, due to the heterogeneity of tumor cells and the insufficient levels of receptors on the tumor cell surface, the success of cancer cell-targeted therapies is largely limited. Histone deacetylase/cathepsin l-responsive acetylated azidomannose (DCL-AAM) was previously developed to effectively and selectively label cancer cell surfaces with reactive azido groups via sugar metabolism. Herein, the labeling kinetics and generality of DCL-AAM were systematically investigated in varieties of tumor cells in vitro and in SKOV3 xenograft tumors in vivo. Based on this, dibenzocyclooctyne-cisplatin (DBCO-Pt) prodrug was developed, and DCL-AAM-mediated metabolic labeling of SKOV3 cells enhanced the tumor accumulation of DBCO-Pt ∼2 fold via bioorthogonal click chemistry, potentiating the anti-tumor efficacy of cisplatin yet alleviating the systemic toxicity. This work, therefore, provides the experimental and theoretical support for the future design of sugar metabolism-based targeted delivery systems and may provide a promising candidate for the treatment of cancers lacking appropriate biomarkers.


Assuntos
Neoplasias , Pró-Fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Química Click , Humanos , Neoplasias/tratamento farmacológico
16.
Biomaterials ; 255: 120166, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32544718

RESUMO

The anticancer performance of nanomedicine is largely impeded by insufficient intratumoral penetration. Herein, tumor microenvironment (TME)-amendatory and self-adaptive nanoclusters (NCs) capable of cancer-associated fibroblasts (CAFs) depletion and size/charge conversion were engineered to mediate light-assisted, hierarchical intratumoral penetration. Particularly, large-sized NCs (~50 nm) were prepared via self-assembly of FAP-α-targeting peptide-modified, 1O2-sensitive polymers, which were further used to envelope small-sized dendrimer (~5 nm) conjugated with Ce6 and loaded with DOX (DC/D). After systemic administration, the NCs efficiently targeted CAFs and generated lethal levels of 1O2 upon light irradiation, which depleted CAFs and concomitantly dissociated the NCs to liberate small-sized, positively charged DC/D. Such stroma attenuation and NCs transformation collectively facilitated the delivery of DC/D into deeper regions of CAF-rich tumors, where DOX and 1O2 provoked synergistic anti-cancer efficacies. This study provides an effective approach to facilitate the tumor penetration of nanomedicine by concurrently and spatiotemporally reconfiguring the nano-properties and remodeling the TME.


Assuntos
Nanomedicina , Microambiente Tumoral , Linhagem Celular Tumoral , Polímeros
17.
J Mater Chem B ; 6(46): 7750-7759, 2018 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-32254897

RESUMO

The resistance of tumor cells is a major cause of chemotherapy failure in cancer patients. Photodynamic therapy (PDT) as a noninvasive treatment strategy with high specificity is a promising method for the treatment of cancer. In this study, a CD44 and N-cadherin dual targeting drug delivery system in combination with mesoporous titanium dioxide nanoparticle (MTN)-based PDT has been successfully constructed for overcoming drug resistance. Hyaluronic acid (HA) and ADH-1 (a cyclic pentapeptide) were grafted onto the surface of MTN to construct ADH-1-HA-MTN, and doxorubicin (DOX) was selected as a model drug. HA can both trap DOX in the wells of MTN and target CD44-overexpressing tumor cells. ADH-1 blocks the EMT process of tumor cells by selectively inhibiting the function of N-cadherin. Besides, a large number of reactive oxygen species (ROS) were generated by MTN under X-ray irradiation, which could provide a cancer cell killing effect. Cytotoxicity tests showed that ADH-1-HA-MTN/DOX was more toxic to tumor cells than its non-ADH-1 modified counterparts. Western blotting analysis showed that ADH-1-HA-MTN/DOX overcame the drug resistance of tumor cells by preventing the process of epithelial-mesenchymal transition. Taken together, ADH-1-HA-MTN may be a promising targeted drug delivery system to overcome the drug resistance of tumors.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA