Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Colloids Surf B Biointerfaces ; 234: 113724, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38183870

ABSTRACT

Both ursolic acid (UA) and sorafenib (Sora) have been generally utilized in cancer treatment, and the combination of the two has also shown a good anti-tumor effect. However, single-agent therapy for Hepatocellular carcinoma (HCC) has the disadvantages of multi-drug resistance, poor water solubility and low bioavailability, and the application of traditional nanocarrier materials is limited due to their low drug loading and low carrier-related toxicity. Therefore, we prepared US NPs with different proportions of UA and Sora by solvent exchange method for achieving synergistic HCC therapy. US NPs had suitable particle size, good dispersibility and storage stability, which synergistically inhibited the proliferation of HepG2 cells, SMMC7721 cells and H22 cells. In addition, we also proved that US NPs were able to suppress the migration of HepG2 cells and SMMC7721 cells and reduce the adhesion ability and colony formation ability of these cells. According to the results, US NPs could degrade the membrane potential of mitochondrial, participate in cell apoptosis, and synergistically induce autophagy. Collectively, the carrier-free US NPs provide new strategies for HCC treatment and new ideas for the development of novel nano-drug delivery systems containing UA and Sora.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Humans , Sorafenib/pharmacology , Sorafenib/therapeutic use , Carcinoma, Hepatocellular/pathology , Ursolic Acid , Pharmaceutical Preparations , Liver Neoplasms/pathology , Cell Line, Tumor
2.
J Colloid Interface Sci ; 656: 177-188, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-37989051

ABSTRACT

The effectiveness of chemotherapeutic agents for hepatocellular carcinoma (HCC) is unsatisfactory because of tumor heterogeneity, multidrug resistance, and poor target accumulation. Therefore, multimodality-treatment with accurate drug delivery has become increasingly popular. Herein, a cell penetrating peptide-aptamer dual modified-nanocomposite (USILA NPs) was successfully constructed by coating a cell penetrating peptide and aptamer onto the surface of sorafenib (Sora), ursolic acid (UA) and indocyanine green (ICG) condensed nanodrug (USI NPs) via one-pot assembly for targeted and synergistic HCC treatment. USILA NPs showed higher cellular uptake and cytotoxicity in HepG2 and H22 cells, with a high expression of epithelial cell adhesion molecule (EpCAM). Furthermore, these NPs caused more significant mitochondrial membrane potential reduction and cell apoptosis. These NPs could selectively accumulate at the tumor site of H22 tumor-bearing mice and were detected with the help of ICG fluorescence; moreover, they retarded tumor growth better than monotherapy. Thus, USILA NPs can realize the targeted delivery of dual drugs and the integration of diagnosis and treatment. Moreover, the effects were more significant after co-administration of iRGD peptide, a tumor-penetrating peptide with better penetration promoting ability or programmed cell death ligand 1 (PD-L1) antibody for the reversal of the immunosuppressive state in the tumor microenvironment. The tumor inhibition rates of USILA NPs + iRGD peptide or USILA NPs + PD-L1 antibody with good therapeutic safety were 72.38 % and 67.91 % compared with control, respectively. Overall, this composite nanosystem could act as a promising targeted tool and provide an effective intervention strategy for enhanced HCC synergistic treatment.


Subject(s)
Carcinoma, Hepatocellular , Cell-Penetrating Peptides , Liver Neoplasms , Nanoparticles , Mice , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Pharmaceutical Preparations , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Cell-Penetrating Peptides/chemistry , B7-H1 Antigen/therapeutic use , Nanoparticles/chemistry , Cell Line, Tumor , Tumor Microenvironment
3.
J Control Release ; 361: 727-749, 2023 09.
Article in English | MEDLINE | ID: mdl-37591461

ABSTRACT

CRISPR/Cas9 genome editing is a promising therapeutic technique, which makes precise and rapid gene editing technology possible on account of its high sensitivity and efficiency. CRISPR/Cas9 system has been proved to able to effectively disrupt and modify genes, which shows great potential for cancer treatment. Current researches proves that virus vectors are capable of effectively delivering the CRISPR/Cas9 system, but immunogenicity and carcinogenicity caused by virus transmission still trigger serious consequences. Therefore, the greatest challenge of CRISPR/Cas9 for cancer therapy lies on how to deliver it to the target tumor site safely and effectively. Non-viral delivery systems with specific targeting, high loading capacity, and low immune toxicity are more suitable than viral vectors, which limited by uncontrollable side effects. Their medical advances and applications have been widely concerned. Herein, we present the molecule mechanism and different construction strategies of CRISPR/Cas9 system for editing genes at the beginning of this research. Subsequently, several common CRISPR/Cas9 non-viral deliveries for cancer treatment are introduced. Lastly, based on the main factors limiting the delivery efficiency of non-viral vectors proposed in the existing researches and literature, we summarize and discuss the main methods to solve these limitations in the existing tumor treatment system, aiming to introduce further optimization and innovation of the CRISPR/Cas9 non-viral delivery system suitable for cancer treatment.


Subject(s)
CRISPR-Cas Systems , Neoplasms , Gene Editing , Technology , Neoplasms/genetics , Neoplasms/therapy
4.
Colloids Surf B Biointerfaces ; 229: 113468, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37515961

ABSTRACT

Thrombus is one of the culprits for global health problems. However, most current antithrombotic drugs are limited by restricted targeting ability and a high risk of systemic bleeding. A hybrid cell membrane-coated biomimetic nanosystem (PM/RM@PLGA@P/R) was constructed in this paper to fulfil the targeted delivery of ginsenoside (Rg1) and perfluorohexane (PFH). Poly lactic-co-glycolic acid (PLGA) is used as carriers to coat Rg1 and PFH. Thanks to the camouflage of erythrocyte membrane (RM) and platelet membrane (PM), the nanosystem in question possesses remarkable features including immune escape and self-targeting. Therefore, a compact nano-core with PLGA@P/R was formed, with a hybrid membrane covering the surface of the core, forming a "core-shell" structure. With its "core-shell" structure, this nanoparticle fancifully combines the advantages of both PFH (the low-intensity focused ultrasound (LIFU)-responsive phase-change thrombolysis) and Rg1(the antioxidant, anti-inflammatory and anticoagulant abilities). Meanwhile, PM/RM@PLGA@P/R nanoparticles exhibits superior in-vitro performance in terms of ROS scavenging, anticoagulant activity and immune escape compared with those without cell membranes (PLGA@P/R). Furthermore, in the animal experiment in which the tail vein thrombosis model was established by injecting k-carrageenan, the combined treatment of LIFU and PM/RM@PLGA@P/R showed a satisfactory antithrombotic efficiency (88.20 %) and a relatively higher biological safety level. This strategy provides new insights into the development of more effective and safer targeted biomimetic nanomedicines for antithrombotic treatments, possessing potential application in synergistic therapy field.


Subject(s)
Ginsenosides , Nanoparticles , Thrombosis , Animals , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/chemistry , Erythrocyte Membrane , Ginsenosides/pharmacology , Biomimetics , Thrombosis/drug therapy , Anticoagulants , Nanoparticles/chemistry
5.
Talanta ; 250: 123753, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-35932717

ABSTRACT

MicroRNAs (miRNAs) have been identified as important biomarkers with great significance for diagnosis and treatment of various diseases. However, their unique properties, such as small size, high sequence homology, and low abundance, make quantitative analysis of miRNAs extremely challenging. Herein, we reported a cascade catalytic hairpin assembly (CCHA) for sensitive and selective detection of miRNA with three kinds of hairpin probes (HP1, HP2, and HP3). In the presence of target miRNA, a series of toehold-mediated intermolecular DNA strand displacement and hybridization was activated among HP1, HP2, and HP3 to assembly numbers of DNA nanoobjects. During this period, the fluorescence response was greatly intensified to indicate the presence and expression level of interested target miRNA. We have demonstrated that the proposed method exhibits a high assay sensitivity to detect low concentration target and an excellent sequence specificity to distinguish even a single-nucleotide difference in vitro. Moreover, we also demonstrated that our design enables the intracellular imaging of miRNA in live cancer and normal cells. These results showing the promising potential of our CCHA for powerful biosensing, clinic diagnosis, or prognosis.


Subject(s)
Biosensing Techniques , MicroRNAs , Biosensing Techniques/methods , Chromosomal Proteins, Non-Histone , DNA/genetics , Limit of Detection , MicroRNAs/analysis , MicroRNAs/genetics , Nucleotides
6.
Am J Chin Med ; 50(3): 749-772, 2022.
Article in English | MEDLINE | ID: mdl-35450513

ABSTRACT

The endothelium covers the internal lumen of the entire circulatory system and plays an important modulatory role in vascular homeostasis. Endothelium dysfunction, characterized by a vasoconstrictive, pro-inflammatory, and pro-coagulant state, usually manifests as a significant pathological process of vascular diseases, including hypertension, atherosclerosis (AS), stroke, diabetes mellitus, coronary artery disease, and cancer. Therefore, there is an urgent necessity to seek promising therapeutic drugs or remedies to ameliorate endothelial dysfunction-induced vascular ailments and complications. Recently, much attention has been attached to ginsenosides, the most significant active components of ginseng, which have always been referred to as "all-healing" and widely used for its extensively medicinal value. Surprisingly, ginsenosides have diverse biological activity which might be related to inflammation, apoptosis, oxidative stress, and angiogenesis. In this review, a brief introduction about endothelial dysfunction and ginsenosides was demonstrated, and the emphasis was put on summarizing multi-faceted pharmacological effects and underlying molecular mechanisms of ginsenosides on the endothelium, including vasorelaxation, anti-oxidation, anti-inflammation, and angio-modulation. Beyond that, nanotechnology to improve efficacy and the existing clinical trials of ginsenosides were concluded. Hopefully, our work will give suggestions for promoting clinical application of traditional Chinese medicine, e.g., hypertension, AS, diabetes, ischemic stroke, and cancer. This review provides a comprehensive base of knowledge for ginsenosides to prevention and treatment of vascular injury- related diseases with clinical significance.


Subject(s)
Ginsenosides , Hypertension , Neoplasms , Panax , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Humans , Hypertension/drug therapy , Neoplasms/drug therapy , Pharmaceutical Preparations
7.
Colloids Surf B Biointerfaces ; 209(Pt 2): 112177, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34749194

ABSTRACT

Rapamycin (RAPA) functions as effectively clinical immunosuppressive agent, its significant tumor growth suppression effect via various pathways in diverse cancers, especially combined with photothermal therapy, is gaining a burgeoning attention. However, its critical defects, low solubility and poor stability, have severely hampered its further application. Herein, RAPA, indocyanine green (ICG) and epigallocatechin gallate (EGCG) serving as chemotherapeutic drug, photosensitizer and biomimetic coatings, respectively, were co-assembled into carrier-free, high biocompatible ICG-RAPA-EGCG nanoparticles (IRE NPs) for synergistic cancer therapy. Particularly, the bioinspired EGCG coatings not only improved the stability of IRE NPs under physiological conditions to avert NPs disassembly and drug release, but also maintained the photostability of ICG to achieve excellent photothermal response. The results indicated that the as-prepared IRE NPs displayed good monodispersity and enhanced stability at various stored media after introducing of EGCG. Compared with monotherapy of RAPA or ICG, IRE NPs showed higher dose-dependent toxicity in MCF-7 cells, HepG2 cells and HeLa cells, especially plus near-infrared laser irradiation. Furthermore, IRE NPs exhibited quicker uptake in cells, higher accumulation in tumor region (even in 48 h) than free ICG and effectively inhibited tumor growth without side effect in H22 tumor-bearing mice. Collectively, the carrier-free IRE NPs provided a simply alternative approach to fabricate RAPA/photosensitizer co-loaded nanoparticles for combinatorial tumor therapy.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Animals , Biomimetics , Cell Line, Tumor , HeLa Cells , Humans , Indocyanine Green , Mice , Photosensitizing Agents , Phototherapy , Photothermal Therapy , Polyphenols , TOR Serine-Threonine Kinases
8.
Int J Pharm ; 605: 120784, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34111544

ABSTRACT

Diabetes is a metabolic disease caused by insufficient insulin secretion, action or resistance, in which insulin plays an irreplaceable role in the its treatment. However, traditional administration of insulin requires continuous subcutaneous injections, which is accompanied by inevitable pain, local tissue necrosis and hypoglycemia. Herein, a green and safe nanoformulation with unique permeability composed of insulin and ginsenosides is developed for transdermal delivery to reduce above-mentioned side effects. The ginsenosides are self-assembled to form shells to protect insulin from hydrolysis and improve the stability of nanoparticles. The nanoparticles can temporarily permeate into cells in 5 min and promptly excrete from the cell for deeper penetration. The insulin permeation is related to the disorder of stratum corneum lipids caused by ginsenosides. The skin acting as drug depot mantains the nanoparticles released continuously, therefore the body keeps euglycemic for 48 h. Encouraged by its long-lasting and effective transdermal therapy, ginsenosides-based nano-system is expected to deliver other less permeable drugs like proteins and peptides and benefit those who are with chronic diseases that need long-term medication.


Subject(s)
Ginsenosides , Nanoparticles , Administration, Cutaneous , Drug Delivery Systems , Insulin , Permeability , Skin
9.
Acta Pharm Sin B ; 11(1): 246-257, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33532190

ABSTRACT

Nanotechnology has emerged as an ideal approach for achieving the efficient chemo agent delivery. However, the potential toxicity and unclear internal metabolism of most nano-carriers was still a major obstacle for the clinical application. Herein, a novel "core‒shell" co-assembly carrier-free nanosystem was constructed based on natural sources of ursolic acid (UA) and polyphenol (EGCG) with the EpCAM-aptamer modification for hepatocellular carcinoma (HCC) synergistic treatment. As the nature products derived from food-plant, UA and EGCG had good anticancer activities and low toxicity. With the simple and "green" method, the nanodrugs had the advantages of good stability, pH-responsive and strong penetration of tumor tissues, which was expected to increase tumor cellular uptake, long circulation and effectively avoid the potential defects of traditional carriers. The nanocomplex exhibited the low cytotoxicity in the normal cells in vitro, good biosafety of organic tissues and efficient tumor accumulation in vivo. Importantly, UA combined with EGCG showed the immunotherapy by activating the innate immunity and acquired immunity resulting in significant synergistic therapeutic effect. The research could provide new ideas for the research and development of self-assembly delivery system in the future, and offer effective intervention strategies for clinical HCC treatment.

10.
Biochem Pharmacol ; 186: 114471, 2021 04.
Article in English | MEDLINE | ID: mdl-33587918

ABSTRACT

Atherosclerosis (AS), characterized by pathological constriction of blood vessels due to chronic low-grade inflammation and lipid deposition, is a leading cause of human morbidity and mortality worldwide. Cell adhesion molecules (CAMs) have the ability to regulate the inflammatory response and endothelial function, as well as potentially driving plaque rupture, which all contribute to the progression of AS. Moreover, recent advances in the development of clinical agents in the cardiovascular field are based on CAMs, which show promising results in the fight against AS. Here, we review the current literature on mechanisms by which CAMs regulate atherosclerotic progression from the earliest induction of inflammation to plaques formation. In particular, we focused on therapeutic strategies based on CAMs inhibitors that prevent leukocyte from migrating to endothelium, including high-affinity antibodies and antagonists, nonspecific traditional medicinal formulas and lipid lowering drugs. The CAMs-based drug delivery nanosystem and the available data on the more reasonable and effective clinical application of CAMs inhibitors have been emphasized, raising hope for further progress in the field of AS therapy.


Subject(s)
Atherosclerosis/drug therapy , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion/drug effects , Drug Delivery Systems/methods , Nanostructures/administration & dosage , Animals , Atherosclerosis/metabolism , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/metabolism , Humans
11.
ACS Appl Mater Interfaces ; 12(51): 57362-57372, 2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33301289

ABSTRACT

The rapid development of CRISPR/Cas9 systems has opened up tantalizing prospects to sensitize cancers to chemotherapy using efficient targeted genome editing, but safety concerns and possible off-target effects of viral vectors remain a major obstacle for clinical application. Thus, the construction of novel nonviral tumor-targeting nanodelivery systems has great potential for the safe application of CRISPR/Cas9 systems for gene-chemo-combination therapy. Here, we report a polyamidoamine-aptamer-coated hollow mesoporous silica nanoparticle for the co-delivery of sorafenib and CRISPR/Cas9. The core-shell nanoparticles had good stability, enabled ultrahigh drug loading, targeted delivery, and controlled-release of the gene-drug combination. The nanocomplex showed >60% EGFR-editing efficiency without off-target effects in all nine similar sites, regulating the EGFR-PI3K-Akt pathway to inhibit angiogenesis, and exhibited a synergistic effect on cell proliferation. Importantly, the co-delivery nanosystem achieved efficient EGFR gene therapy and caused 85% tumor inhibition in a mouse model. Furthermore, the nanocomplex showed high accumulation at the tumor site in vivo and exhibited good safety with no damage to major organs. Due to these properties, the nanocomplex provides a versatile delivery approach for efficient co-loading of gene-drug combinations, allowing for precise gene editing and synergistic inhibition of tumor growth without apparent side effects on normal tissues.


Subject(s)
CRISPR-Cas Systems , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Sorafenib/therapeutic use , Animals , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/toxicity , CRISPR-Associated Protein 9/genetics , Cell Line, Tumor , Drug Carriers/chemistry , Drug Carriers/toxicity , Drug Liberation , Epithelial Cell Adhesion Molecule/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Editing , Genes, erbB-1 , Humans , Mice , Nanoparticles/toxicity , Polyamines/chemistry , Polyamines/toxicity , Porosity , Signal Transduction/drug effects , Silicon Dioxide/toxicity
12.
Acta Biomater ; 111: 373-385, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32413580

ABSTRACT

Nanosized drug delivery systems have emerged to improve the therapeutic performance of anticancer drugs. Here, an amphiphile-based nanoparticle consisting of amphiphilic prodrug N-[3b-acetoxy-urs-12-en-28-oyl]-amino-2-methylpiperazine was developed (UP12 NPs) with uniform sizes (~100 nm), which possessed the advantages of small molecules and nanomedicine. The positively charged UP12 NPs significantly enhanced the cellular drug uptake on HepG2 cells than negatively charged UA NPs. Meanwhile, UP12 and these therapeutic amphiphile-based nanoparticles could induce cell apoptosis more efficiently than that of UA and UA NPs. Moreover, molecular docking demonstrated that the UP12 and intercellular adhesion molecule 1 (ICAM-1) could dock well. UP12 and UP12 NPs significantly decreased the mRNA expression of ICAM-1 and inhibited the migration and adhesion of liver cancer cells (HepG2 cells), which indicated that UP12 might be one of the potential ICAM-1 inhibitors. In vivo, UP12 NPs enhanced tumor accumulation, inhibited tumor lung metastasis and showed good biocompatibility. Overall, UP12 or UP12 NPs could be developed as prospective drugs for cancer metastasis therapy via ICAM-1 mediated cell adhesion. STATEMENT OF SIGNIFICANCE: In this study, we fabricated the therapeutic amphiphile-based nanoparticles by assembly of ursolic acid piperazine derivative N-[3b-acetoxy-urs-12-en-28-oyl]-amino-2-methylpiperazine (name as UP12 NPs) with low cytotoxicity. UP12 NPs exhibited spherical morphology and uniform sizes. Particularly, these therapeutic amphiphile-based nanoparticles significantly enhanced tumor accumulation and inhibited tumor lung metastases via intercellular adhesion molecule 1 (ICAM-1) mediated cell adhesion.


Subject(s)
Carcinoma, Hepatocellular , Intercellular Adhesion Molecule-1 , Liver Neoplasms , Nanoparticles , Carcinoma, Hepatocellular/drug therapy , Cell Adhesion , Cell Line, Tumor , Drug Delivery Systems , Humans , Liver Neoplasms/drug therapy , Molecular Docking Simulation , Prospective Studies
13.
Cancer Biol Med ; 16(3): 498-513, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31565480

ABSTRACT

OBJECTIVE: Cancer metastasis remains the primary cause of cancer-related death worldwide. In a previous study, we found that levels of BTB/POZ domain-containing protein KCTD12 are lower in metastatic melanoma cells than in parental melanoma cells. The purpose of this study was to identify the roles of KCTD12 in cancer metastasis. METHODS: The Cancer Genome Atlas (TCGA) datasets were used to evaluate the relationship between KCTD12 and skin cutaneous melanoma (SKCM) prognosis. The effects of endogenous KCTD12 on biological behaviors were examined using the MTT assay. The impacts of KCTD12 on melanoma stemness were explored using spheroid formation assay. KCTD12 knockout A375 cells were generated to confirm the inhibitory effect of KCTD12 on CD271, and a mouse metastatic model was used to determine the impact of KCTD12 on melanoma metastasis in vivo. RESULTS: KCTD12 levels were lower in lung metastatic cells than in paired parental melanoma cells, and low KCTD12 expression indicated a poor prognosis in SKCM. Cancer metastasis-related capacities were higher in lung metastatic cells than in parental melanoma cells. Moreover, KCTD12 knockdown enhanced tumor growth and metastasis both in vitro and in vivo. Mechanistically, the interaction between KCTD12 and CD271 might be responsible for the stemness transformation after KCTD12 knockdown. CONCLUSIONS: This study identifies for the first time the role of the interaction between KCTD12 and CD271 in inducing melanoma cell stemness transformation. Moreover, KCTD12 repression enhances melanoma cell growth, adhesion, migration and invasion.

14.
J Mater Chem B ; 7(44): 6914-6923, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31482166

ABSTRACT

The combination of chemotherapy and photothermal therapy displays improved anti-cancer effects and lower systematic toxicity of a free drug compared with monotherapy. In this study, we designed innovative, carrier-free nanodrugs (PTX/ICG NDs) composed of the chemotherapeutic agent paclitaxel (PTX) and the photosensitizer indocyanine green (ICG) via self-assembly. The nanodrugs not only incorporated two different modalities into one delivery system for combined chemo-photothermal therapy but also enhanced the solubility of PTX without the need for any carrier. The as-prepared PTX/ICG NDs exhibited the merits of a relatively uniform size of 140 ± 1.4 nm, surface charge of -36 ± 2.2 mV, and high drug loading content of PTX. The combination strategy exerted a synergistic effect on the cytotoxicity of cancer cells in vitro, which could be attributed to the high cellular uptake and sustained release of PTX. Furthermore, an in vivo study indicated that PTX/ICG NDs showed higher accumulation in the tumor site than free ICG and possessed strong synergistic chemo-photothermal therapy efficacy against tumors in H22 tumor-bearing mice. Taken together, our study demonstrates that PTX/ICG NDs hold promise to become an alternative chemo-photothermal therapy agent to treat cancers.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Delivery Systems , Indocyanine Green/chemistry , Nanoparticles , Paclitaxel/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Cell Line, Tumor , Cell Survival , Coloring Agents/chemistry , Drug Design , Humans , Mice , Neoplasms, Experimental , Paclitaxel/chemistry , Rats
15.
Eur J Med Chem ; 179: 916-935, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31306818

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors. Traditional chemotherapy drugs are hard to reach a satisfactory therapeutic effect since advanced HCC is highly chemo-resistant. Sorafenib is an oral multikinase inhibitor that can suppress tumor cell proliferation, angiogenesis and induce cancer cell apoptosis. However, the poor solubility, rapid metabolism and low bioavailability of sorafenib greatly restricted its further clinical application. During the past decade, numerous sorafenib derivatives have been designed and synthesized to overcome its disadvantages and improve its clinical performance. This article focuses on the therapeutic effects and mechanisms of various sorafenib derivatives with modifications on the N-methylpicolinamide group, urea group, central aromatic ring or others. More importantly, this review summarizes the current status of the structure-activity relationship (SAR) of reported sorafenib derivatives, which can provide some detailed information of future directions for further structural modifications of sorafenib to discovery new anti-tumor drugs with improved clinical performance.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Sorafenib/pharmacology , Animals , Antineoplastic Agents/chemistry , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Chemistry, Pharmaceutical , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Molecular Structure , Protein Kinase Inhibitors/chemistry , Sorafenib/chemistry
16.
Talanta ; 187: 272-278, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-29853047

ABSTRACT

Herein, we describe a novel trigger-induced DNA nanoassembly method using only one loop-stem shaped hairpin probe (HP) that consists of three different functional regions as a single building unit. The Region I is designed complementary to the trigger, while the Region II and Region III are projected to complementary with each other. When hybridized with the trigger, a toehold mediated strand displacement (TMSD) occurred on the strand of Region I, leading to the release of Region III for further hybridization with the Region II on another HP molecule and in turn inducing a stepwise growth of HP with the aid of polymerase. Unlike the conventional assembly approaches that rely on the sophisticated sequence design and complex operation, the single-HP nanoassembly is easy and fast. Moreover, because many HPs are opened during the assembly process, we exemplified the nanoassembly strategy by re-designing a new labeled hairpin probe to analyze the Kras oncogene with a high sensitivity and specificity. The present study demonstrated a novel promising DNA nanoassembly strategy for biological applications.

17.
Analyst ; 142(23): 4438-4445, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-29082392

ABSTRACT

Occurrence and application of oligonucleotide probes have promoted great progress in the biochemical analysis field due to their unique biological and chemical properties. In this work, a long-stem shaped multifunctional molecular beacon (LS-MMB) that is responsive to a cancer-related gene, p53, is well-prepared. By designing the probe with long-paired bases at its two ends and short-paired bases between the middle region and the 3' end, the LS-MMB is intelligently endowed with the ability to recognize the target analyte, serve as the polymerization primer/template, and signal the hybridization event synchronously, which is distinctly advantageous over the traditional molecular beacons (MBs). Moreover, it is excitingly found that the LS-MMB can be employed to exert intramolecular and intermolecular interactions for strand displacement amplification (SDA) without the involvement of any assistant probes; this therapy results in a really easy and rapid sensing system that provides an extremely low background noise and high target output signal. In this case, an excellent sensitivity and specificity to detect target gene down to picomolar level and resolution to even one nucleotide variation are achieved, respectively. In addition, the application potential for real genomic DNA analysis is realized. We envision that the probe of LS-MMB can act as a universal platform for biosensing and biomedical research.


Subject(s)
Biosensing Techniques , DNA/analysis , Nucleic Acid Amplification Techniques , Nucleic Acid Hybridization , Oligonucleotide Probes/chemistry , A549 Cells , Genes, p53 , Humans , Polymerization
18.
Biosens Bioelectron ; 94: 626-631, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28371752

ABSTRACT

As one of the most exciting building blocks, DNA has gained increasing attention in the construction of promising nanostructures for various biological and medical purposes. In this contribution, we have developed an easily constructed DNA nanoassembly-based biosensing system that consists of one signal hairpin probe (SHP) and one label-free hairpin probe (LHP) for target p53 gene analysis. The probes of SHP and LHP were designed to be incapable of interacting with each other in the absence of the p53 gene. When the target gene is introduced, the 3' end of SHP (or LHP) hybridizes with the middle region of LHP (or SHP), leading to polymerase-sustained DNA nanoassembly. Because one target species can exhaust many building scaffolds to execute the programmable nanoassembly in one-pot approach, the fluorescence intensity of SHP is greatly enhanced in the presence of target gene in a simple and robust manner. The practical applicability was successfully demonstrated by screening target gene extracted from cancer cells. We believe this intriguing sensing strategy and desirable assay ability would provide new opportunities to develop versatile biochemical analysis methods.


Subject(s)
Biosensing Techniques/methods , DNA/isolation & purification , Nanostructures/chemistry , Tumor Suppressor Protein p53/isolation & purification , Humans , Nucleic Acid Hybridization/methods , Tumor Suppressor Protein p53/chemistry
19.
Analyst ; 142(4): 613-620, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28119962

ABSTRACT

In this work, we have proposed a chain anadiplosis-structured DNA nanowire by using two well-defined assembly strands (AS1 and AS2). The presence of a target analyte would drive the single-stranded AS1 dissociate from the pre-formatted nanowire, converting into a fully double-stranded form responsible for extensive accumulation of G-rich cleavage fragment1 (GCF1) because of an autonomously performed polymerization/nicking/displacement process. In turn, the produced GCF1 is able to hybridize with the un-peeled AS2, allowing the replication over AS2 to occur and generate large amounts of G-rich cleavage fragment2 (GCF2) with the ability to hybridize with the un-peeled AS1, thereafter initiating new enzymatic reactions for further collection of GCF1. Because the reactions occur repeatedly, the assembled nanowires gradually dissociated and completely collapsed in the end, achieving the goal of substantial signal amplification for the colorimetric readout of the target analytes. The sensing feasibility is firstly verified by one trigger primer (TP), and then exemplified with the detection of the target, the kras oncogene, with high sensitivity and specificity. As a proof-of-concept strategy, the intelligent signal readout pathway and desired assay ability provide unique insights into the materials research and biological studies.


Subject(s)
Biosensing Techniques , Colorimetry , DNA/chemistry , Nanowires , Humans , Nucleic Acid Amplification Techniques , Nucleic Acids , Proto-Oncogene Proteins p21(ras)/analysis
20.
Biomaterials ; 120: 57-65, 2017 03.
Article in English | MEDLINE | ID: mdl-28033500

ABSTRACT

Facile assembly of intelligent DNA nanoobjects with the ability to exert in situ visualization of intracellular microRNAs (miRNAs) has long been concerned in the fields of DNA nanotechnology and basic medical study. Here, we present a driving primer (DP)-triggered polymerization-mediated metastable assembly (PMA) strategy to prepare a well-ordered metastable DNA nanoarchitecture composed of only two hairpin probes (HAPs), which has never been explored by assembly methods. Its structural features and functions are characterized by atomic force microscope (AFM) and gel electrophoresis. Even if with a metastable molecular structure, this nanoarchitecture is relatively stable at physiological temperature. The assembly strategy can be expanded to execute microRNA-21 (miRNA-21) in situ imaging inside cancer cells by labelling one of the HAPs with fluorophore and quencher. Compared with the conventional fluorescence probe-based in situ hybridization (FISH) technique, confocal images revealed that the proposed DNA nanoassembly can not only achieve greatly enhanced imaging effect within cancer cells, but also reflect the miRNA-21 expression level sensitively. We believe that the easily constructed DNA nanoarchitecture and in situ profiling strategy are significant progresses in DNA assembly and molecule imaging in cells.


Subject(s)
DNA/ultrastructure , MicroRNAs/chemistry , MicroRNAs/ultrastructure , Microscopy, Fluorescence/methods , Molecular Imaging/methods , Nanoparticles/ultrastructure , Neoplasms, Experimental/ultrastructure , Crystallization/methods , DNA/chemistry , Fluorescent Dyes , Humans , MCF-7 Cells , Nanoparticles/chemistry , Neoplasms, Experimental/chemistry , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...