Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Control Release ; 321: 629-640, 2020 05 10.
Article in English | MEDLINE | ID: mdl-32135224

ABSTRACT

Liver fibrosis leads to over one million deaths annually worldwide. Hepatic stellate cells (HSCs) have been identified as the main executors of liver fibrosis. Unfortunately, no drug has yet been approved for clinical use against liver fibrosis, largely because the tested drugs have been unable to access HSCs and efficiently remove the collagen accumulation involved in fibrogenesis. Here, we designed an efficient HSC-targeting lipid delivery system that carried dual siRNAs intended to both inhibit collagen synthesis and promote collagen degradation, with the goal of realizing enhanced anti-liver fibrosis by bidirectional regulation of collagen accumulation. The delivery system was constructed by using amphiphilic cationic hyperbranched lipoids (C15-PA) for siRNA complexation and helper lipoids (cholesterol-polyethylene glycol-vitamin A, Chol-PEG-VA) for HSCs targeting. The generated vitamin A-decorated and hyperbranched lipoid-based lipid nanoparticles (VLNPs) showed excellent gene-binding ability and transfection efficiency, and enhanced the delivery of siRNAs to HSCs. Fibrotic mice treated with dual siRNA-loaded VLNPs showed a great reduction in the collagen accumulation seen in this model; the enhanced effect of bidirectional regulation reduced the collagen accumulation level in treated mice to almost that seen in normal mice. There was no notable sign of toxicity or tissue inflammation in mice exposed to repeated intravenous administration of the dual siRNA-loaded VLNPs. In conclusion, our results indicate that biocompatible VLNPs designed to exploit precise targeting and an effective bidirectional regulation strategy hold promise for treating liver fibrosis.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , Nanoparticles , Animals , Collagen , Liver/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis/therapy , Mice , RNA, Small Interfering
2.
Biomaterials ; 230: 119616, 2020 02.
Article in English | MEDLINE | ID: mdl-31837823

ABSTRACT

As hepatic stellate cells (HSCs) are essential for hepatic fibrogenesis, HSCs targeted nano-drug delivery system is a research hotspot in liver fibrosis therapy. However, the excessive deposition of fibrosis collagen (mainly collagen I) in the space of Disse associated with hepatic fibrogenesis would significantly hinder nano-formulation delivery to HSCs. Here, we have prepared a collagenase I and retinol co-decorated polymeric micelle that possess nanodrill-like and HSCs-target function based on poly-(lactic-co-glycolic)-b-poly (ethylene glycol)-maleimide (PLGA-PEG-Mal) (named CRM) for liver fibrosis therapy. Upon encountering collagen I barrier, CRM exerted a nanodrill-like function, efficiently degrading pericellular collagen I and showing greater uptake by human HSCs than other micelle formulations. Besides, CRM could realize excellent accumulation in the fibrotic liver and accurate targeting to activated HSCs in mouse hepatic fibrosis model. Moreover, CRM loaded with nilotinib (CRM/NIL), a second-generation tyrosine kinase inhibitor used in the treatment of liver fibrosis, showed optimal antifibrotic activity. This work suggests that CRM with dual function is an efficient carrier for liver fibrosis drug delivery and collagenase I decorating could be a new strategy for building more efficient HSCs targeted nano-drug delivery system.


Subject(s)
Liver Cirrhosis , Micelles , Extracellular Matrix , Hepatic Stellate Cells/pathology , Humans , Liver/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology
3.
Chem Commun (Camb) ; 54(59): 8218-8221, 2018 Jul 19.
Article in English | MEDLINE | ID: mdl-29985496

ABSTRACT

A highly hydrophilic polymer equipped with guanidinium groups was used to load aromatic ring-containing hydrophobic agent doxorubicin (DOX) via π-π interaction. The results have shown that the delivery system exhibited enhanced cellular uptake and antitumor efficiency compared with free drugs. This study opens new avenues for the application of hydrophilic polymers in drug delivery.


Subject(s)
Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Drug Carriers/chemistry , Guanidines/chemistry , Polymers/chemistry , Animals , Antineoplastic Agents/chemistry , Doxorubicin/chemistry , Drug Carriers/chemical synthesis , Drug Liberation , Female , Fluorescence , Humans , Hydrophobic and Hydrophilic Interactions , MCF-7 Cells , Mice, Inbred BALB C , Polymers/chemical synthesis
4.
Biomater Sci ; 6(8): 2144-2155, 2018 Jul 24.
Article in English | MEDLINE | ID: mdl-29923556

ABSTRACT

Finding a cure for breast cancer currently remains a medical challenge in due to the failure of common treatment methods to inhibit invasion and metastasis of cancer cells, which eventually leads to recurrence of breast cancer. Many secreted proteins are overexpressed and play crucial roles in tumorigenesis and development. The Golgi apparatus is a key protein processing and secretion factory in which metastasis-associated proteins are modified, transported and secreted; thus, regulating the Golgi apparatus of tumor cells is a viable strategy to inhibit tumor metastasis. Herein, celecoxib (CLX) and Brefeldin A (BFA) were encapsulated into the biocompatible polymer PLGA-PEG to form nanoparticles that act on the Golgi apparatus to treat metastatic breast cancer; CLX is a specific COX-2 inhibitor which accumulates in the Golgi apparatus, and BFA is a protein transport inhibitor fusing the Golgi apparatus into endoplasmic reticulum. The optimized CLX and BFA co-loaded nanoparticles (CBNPs) possessed good physicochemical properties. CBNPs efficiently damaged the Golgi apparatus within 30 min and showed enhanced cytotoxicity of CLX and BFA toward murine metastatic breast cancer 4T1 cells. The migration and invasion abilities of the cells were dramatically suppressed by the CBNPs. Further, the expression and secretion of metastasis-associated proteins such as matrix metalloproteinase-9 (MMP-9) and vascular endothelial growth factor (VEGF) were remarkably decreased. Our findings showed that co-delivering CLX and BFA to regulate the Golgi apparatus may be an efficient strategy to inhibit breast cancer growth and suppress tumor cell metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Brefeldin A/pharmacology , Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Drug Delivery Systems , Golgi Apparatus/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biocompatible Materials/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Brefeldin A/administration & dosage , Brefeldin A/chemistry , Celecoxib/administration & dosage , Celecoxib/chemistry , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/chemistry , Drug Screening Assays, Antitumor , Female , Golgi Apparatus/metabolism , HeLa Cells , Humans , Mice , Nanoparticles/chemistry , Particle Size , Polyethylene Glycols/chemistry , Polyglactin 910/chemistry , Tumor Cells, Cultured
5.
J Control Release ; 283: 113-125, 2018 08 10.
Article in English | MEDLINE | ID: mdl-29857004

ABSTRACT

Liver fibrosis refers to excessive accumulation of hepatic collagen, which is primarily produced by activated hepatic stellate cells (HSCs). No effective drugs are clinically available to treat this condition, reflecting the fact that antifibrotic drugs do not specifically target activated HSCs. Here, we report the synthesis and evaluation of poly (lactide-co-glycolide)-polyspermine-poly (ethylene glycol)-vitamin A (PLGA-PSPE-PEG-VA), and activated HSC-targeted, biocompatible amphiphilic polymers for co-delivery of chemical (silibinin) and genetic (siCol1α1) drugs that synergistically suppress collagen I accumulation in fibrogenesis. PLGA-PSPE-PEG-VA self-assembled into core-shell polymeric micelles (PVMs) at low concentrations. After loading with silibinin and siCol1α1, the resulting chemical/genetic drug-loaded PVMs (CGPVMs) exhibited a small particle size and a slightly positive surface. CGPVMs had very low cytotoxicity and hemolytic activity in vitro and were well tolerated in mice, with no liver toxicity or inflammation. Importantly, CGPVMs effectively accumulated in fibrotic livers and specifically targeted activated HSCs. As expected CGPVMs more efficiently decreased collagen I production and ameliorated liver fibrosis compared with chemical drug (silibinin)-loaded PVMs (CPVMs) or genetic drug (siCol1α1)-loaded PVMs (GPVMs) only. These results indicate that CGPVMs are a promising tool for targeted delivery of chemogenes to activated HSCs in the treatment of liver fibrosis.


Subject(s)
Collagen Type I/genetics , Liver Cirrhosis/therapy , Micelles , RNA, Small Interfering/administration & dosage , Silybin/administration & dosage , Vitamin A/administration & dosage , Vitamins/administration & dosage , Animals , Biocompatible Materials , Cell Line , Cell Survival/drug effects , Collagen Type I/metabolism , Drug Carriers/administration & dosage , Gene Transfer Techniques , Genetic Therapy , Hemolysis/drug effects , Humans , Male , Mice, Inbred C57BL , Polymers/administration & dosage , Rabbits , Rats
6.
Int J Pharm ; 535(1-2): 180-193, 2018 Jan 15.
Article in English | MEDLINE | ID: mdl-29129572

ABSTRACT

Cationic polymers have been widely used as one of the most promising non-viral vehicles for gene delivery due to their potential safety and ease of large-scale production. Here, we report the design and synthesis of a series of novel biodegradable fluorinated poly(ß-amino ester)s (FPBAEs) by simple Michael-addition reaction as safe and efficient gene carrier. The results of transfection efficacy assay demonstrated the optimal FPBAE could mediated much higher GFP expression than the commercial transfection agents, polyethyleneimine (PEI, Mw = 25K) and Lipo 2000, as well as the non-fluorinated poly(ß-amino ester)s (PBAE) on both HeLa and HEK-293T cell lines (higher than 70 and 90%, respectively), which was largely attributed to fluorination. Moreover, MTT and hemolysis assay indicated a preferable biocompatibility of FPBAE compared with PEI 25K owing to the low molecular weight and the presence of cleavable ester bonds. Taken together, the novel polymer FPBAE with both excellent gene transfection efficacy and much lower toxicity could serve as a desirable gene vector.


Subject(s)
Biocompatible Materials/chemistry , Fluorocarbon Polymers/chemistry , Gene Transfer Techniques , Polymers/chemistry , Cell Proliferation/genetics , Cell Survival/genetics , DNA/administration & dosage , DNA/genetics , HEK293 Cells , HeLa Cells , Hemolysis , Humans , Transfection , Tumor Suppressor Protein p53/genetics
7.
Chem Commun (Camb) ; 53(62): 8790-8793, 2017 Aug 11.
Article in English | MEDLINE | ID: mdl-28736782
8.
ACS Appl Mater Interfaces ; 9(13): 11337-11344, 2017 Apr 05.
Article in English | MEDLINE | ID: mdl-28291320

ABSTRACT

Multidrug resistance (MDR) greatly impedes the therapeutic efficacy of chemotherapeutic agents. Overexpression of ATP-binding cassette (ABC) transporters, such as P-gp, on the surface of tumor cells is a major mechanism in MDR. In this study, we fabricated manganese dioxide (MnO2)/doxorubicin (DOX)-loaded albumin nanoparticles (BMDN) for magnetic resonance imaging and reversing MDR in resistant tumor. BMDN facilitated the delivery of DOX into MDR tumor cells through their MDR reversal effects including enhanced cellular uptake, reduced drug efflux, and decreased hypoxic tumor microenvironment. BMDN also acted as an effective MRI contrast agent, thereby causing good in vitro and in vivo T1-weighted imaging.


Subject(s)
Contrast Media/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Antineoplastic Agents , Cell Line, Tumor , Doxorubicin , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Magnetic Resonance Imaging , Manganese Compounds , Oxides , Theranostic Nanomedicine
9.
Int J Nanomedicine ; 12: 855-869, 2017.
Article in English | MEDLINE | ID: mdl-28182160

ABSTRACT

BACKGROUND: The natural compound curcumin (Cur) can regulate growth inhibition and apoptosis in various cancer cell lines, although its clinical applications are restricted by extreme water insolubility and instability. To overcome these hurdles, we fabricated a Cur-coordinated reactive oxygen species (ROS)-responsive nanoparticle using the interaction between boronic acid and Cur. MATERIALS AND METHODS: We synthesized a highly biocompatible 4-(hydroxymethyl) phenylboronic acid (HPBA)-modified poly(ethylene glycol) (PEG)-grafted poly(acrylic acid) polymer (PPH) and fabricated a Cur-coordinated ROS-responsive nanoparticle (denoted by PPHC) based on the interaction between boronic acid and Cur. The mean diameter of the Cur-coordinated PPHC nanoparticle was 163.8 nm and its zeta potential was -0.31 mV. The Cur-coordinated PPHC nanoparticle improved Cur stability in physiological environment and could timely release Cur in response to hydrogen peroxide (H2O2). PPHC nanoparticles demonstrated potent antiproliferative effect in vitro in A549 cancer cells. Furthermore, the viability of cells treated with PPHC nanoparticles was significantly increased in the presence of N-acetyl-cysteine (NAC), which blocks Cur release through ROS inhibition. Simultaneously, the ROS level measured in A549 cells after incubation with PPHC nanoparticles exhibited an obvious downregulation, which further proved that ROS depression indeed influenced the therapeutic effect of Cur in PPHC nanoparticles. Moreover, pretreatment with phosphate-buffered saline (PBS) significantly impaired the cytotoxic effect of Cur in A549 cells in vitro while causing less damage to the activity of Cur in PPHC nanoparticle. CONCLUSION: The Cur-coordinated nanoparticles developed in this study improved Cur stability, which could further release Cur in a ROS-dependent manner in cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Curcumin/pharmacology , Drug Delivery Systems , Lung Neoplasms/drug therapy , Nanoparticles/administration & dosage , Reactive Oxygen Species/metabolism , Antineoplastic Agents/chemistry , Curcumin/chemistry , Humans , Hydrogen Peroxide/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Nanoparticles/chemistry , Oxidants/pharmacology , Polymers/chemistry , Tumor Cells, Cultured
10.
Biomaterials ; 113: 266-278, 2017 01.
Article in English | MEDLINE | ID: mdl-27842254

ABSTRACT

A novel biomimetic drug delivery system (BDDS) inspired by the pH-dependent ferric ion-transport and release manner of transferrin (Tf) was developed for combating multidrug-resistant breast cancer. Tf-inspired carrier was synthesized by modifying bovine serum albumin (BSA) with histamine (HA) through amide reaction to provide superior specific coordination sites for ferric ion-drug complexes, and self-assembled into nanoparticles (NPs) induced by coordination bond. Tf-inspired NPs were prepared via environment-friendly method, and well redispersed in saline after lyophilization. When internalized into tumor cells by SPARC (secreted protein acidic and rich in cysteine) mediated endocytosis, Tf-inspired NPs bypassed and decreased the P-glycoprotein-mediated drug efflux and led to more effective treatment of multidrug-resistant breast cancer compared with free drugs both in vitro and in vivo due to the enhanced cellular uptake and rapid pH-responsive drug release. Moreover, Tf-inspired NPs exhibited good biocompatibility and low systemic toxicity. Thus, our results demonstrate that Tf-inspired NPs based on coordination bond represent as a smart drug delivery strategy to combat multidrug-resistant cancer and have great potential for clinical applications in cancer therapy.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Delayed-Action Preparations/chemistry , Doxorubicin/administration & dosage , Nanoparticles/chemistry , Transferrin/analogs & derivatives , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Breast/drug effects , Breast/pathology , Breast Neoplasms/pathology , Cattle , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Female , Histamine/analogs & derivatives , Humans , Hydrogen-Ion Concentration , Mice, Inbred BALB C , Mice, Nude , Serum Albumin, Bovine/chemistry
11.
Int J Pharm ; 513(1-2): 612-627, 2016 Nov 20.
Article in English | MEDLINE | ID: mdl-27682215

ABSTRACT

This report demonstrated a one-step assembly for co-delivering chemotherapeutics and therapeutic nucleic acids, constructed by integrating drug molecules into a nucleic acid condensing polymeric prodrug through degradable linkages. Demethylcantharate was selected as the model drug and pre-modified by esterifying its two carboxylic groups with 2-hydroxyethyl acrylate. The synthesized demethylcantharate diacrylate was then used to polymerize with linear polyethyleneimine (PEI 423) through a one-step Michael-addition reaction. The obtained cationic polymeric demethylcantharate prodrug was used to pack Akt1 shRNA into complexes through a one-step assembly. The formed complexes could release the parent drug demethylcantharate and Akt1 shRNA through the hydrolysis of ester bonds. Cellular assays involving cell uptake, cytotoxicity, and cell migration indicated that demethylcantharate and Akt1 shRNA co-delivered in the present form significantly and synergistically suppress the growth and metastasis of three human cancer cells. This work suggests that incorporating drug molecules into a nucleic acid-packing cationic polymer as a polymeric prodrug in a degradable form is a highly convenient and efficient way to co-deliver drugs and nucleic acids for cancer therapy.


Subject(s)
Acrylates/chemistry , Antineoplastic Agents/chemistry , Cantharidin/analogs & derivatives , Polymers/chemistry , Prodrugs/chemistry , Proto-Oncogene Proteins c-akt/genetics , Acrylates/administration & dosage , Acrylates/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cantharidin/administration & dosage , Cantharidin/chemistry , Cantharidin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , DNA/administration & dosage , DNA/chemistry , DNA/pharmacology , Drug Liberation , Green Fluorescent Proteins/genetics , Humans , Hydrolysis , Polymerization , Polymers/administration & dosage , Polymers/pharmacology , Prodrugs/administration & dosage , Prodrugs/pharmacology , RNA, Small Interfering/chemistry
12.
Int J Pharm ; 513(1-2): 591-601, 2016 Nov 20.
Article in English | MEDLINE | ID: mdl-27682214

ABSTRACT

Lung cancer has been a leading cause of cancer mortality worldwide and aerosol-mediated gene therapy endows numerous advantages compared to other traditional modalities. Here, we reported a folic acid (FA)-modified hyperbranched polyspermine (HPSPE) with prominent biocompatibility for lung cancer cell targeted gene therapy. FA was decorated to the HPSPE via an amidation reaction and the physicochemical properties of nanoplexes formed with DNA were characterized. Gel electrophoresis study elucidated that the designed polymer was capable to condense DNA and protect it from degradation by DNase I. Cell viability and transfection efficiency assay in vitro and in vivo indicated its increased transfection performance with lower toxicity. Furthermore, reduced tumor numbers and down-regulation of Akt1 protein after aerosol treatment containing FA-HPSPE/shAkt1 complexes proved its therapeutic potential for lung cancer suppression. Results obtained in this study suggested that FA-HPSPE with highly biocompatibility and targeting capability while forming complexes with shAkt1 and administrated through noninvasive aerosol could be prospective for inhibiting lung tumorigenesis.


Subject(s)
Antineoplastic Agents/administration & dosage , DNA/administration & dosage , Folic Acid/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Spermine/analogs & derivatives , Spermine/administration & dosage , Aerosols , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , DNA/chemistry , DNA/pharmacology , Folic Acid/chemistry , Folic Acid/pharmacology , Folic Acid Transporters/genetics , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Male , Mice, Inbred C57BL , Spermine/chemistry , Spermine/pharmacology
13.
Int J Pharm ; 506(1-2): 207-13, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27113867

ABSTRACT

Based on development of nano-delivery system, co-delivery of chemotherapeutic drug and small interfering RNA (siRNA) has exerted a promising advantage in cancer therapy. In this work, the superiority of synergistic therapy and safety of the hierarchical targeted co-delivery system loaded with siRNA and lonidamine (LND) were evaluated. The in vivo tumor accumulation ability and cancer growth inhibition effect of the polymer-blend nanocarriers were evaluated by a H22 subcutaneous sarcoma model. Moreover, hematoxylin and eosin (H&E) staining and transferase-mediated dUTP nick end-labeling (TUNEL) staining of tumor sections from each group were compared to assess the therapeutic efficacy. The dual-loaded nanocarriers had better tumor accumulation ability, remarkably inhibited growth of solid tumor in a synergistic manner, even significantly decreased hepatotoxicity of LND, and had good in vivo biocompatibility whereas LND alone showed serious hepatotoxicity. We believed that the dual-loaded hierarchical targeted delivery system with high effectiveness and biocompatibility would provide a promising approach for cancer combination therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Indazoles/administration & dosage , RNA, Small Interfering/administration & dosage , Sarcoma/drug therapy , Animals , Antineoplastic Agents/pharmacology , Drug Carriers/chemistry , Drug Delivery Systems , In Situ Nick-End Labeling , Indazoles/pharmacology , Male , Mice , Mice, Inbred ICR , Nanoparticles , Polymers/chemistry , Sarcoma/genetics , Xenograft Model Antitumor Assays
14.
Int J Pharm ; 506(1-2): 79-86, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27102990

ABSTRACT

Polyamine content, which is associated with tumor growth, can be regulated by ornithine decarboxylase (ODC) and S-adenosyl methionine decarboxylase (SAMDC), two key enzymes in polyamine biosynthesis. Here we aim to develop a pH-responsive cationic poly(agmatine) based on a polyamine analogue-agmatine that can dually function as a gene delivery vector as well as an anticancer agent by inhibiting ODC after intracellular degradation. The core-shell nanoparticles, formed by poly(agmatine)/SAMDC siRNA complex as a core, were coated with bovine serum albumin for better in vivo circulation stability and tumor targeting. When the nanoparticles were taken up by tumor cells via endocytosis and degraded in endosome, the released agmatine and SAMDC siRNA can synergistically inhibit polyamines biosynthesis, inducing inhibition of tumor proliferation. Our study offered a potential way in tumor therapy based on polyamine metabolism.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell Proliferation/drug effects , Neoplasms/drug therapy , Polyamines/metabolism , Adenosylmethionine Decarboxylase/metabolism , Cell Line, Tumor , Endocytosis/drug effects , Endosomes/metabolism , Gene Transfer Techniques , Hep G2 Cells , Humans , MCF-7 Cells , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasms/metabolism , Ornithine Decarboxylase/metabolism , RNA, Small Interfering/metabolism , Serum Albumin, Bovine/chemistry
15.
Biomaterials ; 73: 149-59, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26409000

ABSTRACT

Combination therapy is usually considered as a promising strategy owing to its advantages such as reduced doses, minimized side effects and improved therapeutic efficiency in a variety of diseases including diabetes. Here we synthesized a new highly intracellular stimuli-sensitive chitosan-graft-metformin (CS-MET) prodrug by imine reaction between oxidative chitosan and metformin for type 2 diabetes (T2D) therapy. Hypothetically, CS-MET functions dually as an anti-diabetes prodrug as well as a gene delivery vector without superfluous materials. CS-MET formed nanocomplexes with therapeutic gene through electrostatic interactions and entered cells by Organic Cation Transporter (OCT)-independent endocytosis. The incorporation of metformin into chitosan has been found to increase endosomal escape via the proton sponge effect. When vector carrying a short-hairpin RNA (shRNA) silencing sterol regulatory element-binding protein (SREBP), a major transcription factor involved in de novo lipogenisis, it reduced the SREBP mRNA and proteins efficiently. Furthermore, by intraperitoneal injection, CS-MET/shSREBP nanocomplexes effectively knocked down SREBP in livers of western-type diet (WD)-induced obese C57BL/6J mice, markedly reversed insulin resistance and alleviated the fatty liver phenotype without obvious toxic effects. Thus we were able to show that the intracellular stimuli-sensitive CS-MET prodrug renders a potential platform to increase the anti-diabetes activity with synergistic enhancement of gene therapy.


Subject(s)
Diabetes Mellitus, Type 2/therapy , Nanostructures/chemistry , Polymers/chemistry , Prodrugs/chemistry , Animals , Biocompatible Materials/chemistry , Cell Line, Tumor , Chitosan/administration & dosage , Endocytosis , Fatty Liver/metabolism , Genetic Therapy/methods , Genetic Vectors , Glucose Tolerance Test , Hep G2 Cells , Homeostasis , Humans , Imines/chemistry , Lipids/chemistry , Metformin/administration & dosage , Metformin/chemistry , Mice , Mice, Inbred C57BL , Mice, Obese , Microscopy, Confocal , Oxygen/chemistry , Phenotype , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Static Electricity , Sterol Regulatory Element Binding Protein 1/chemistry
16.
Biomaterials ; 61: 178-89, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26004233

ABSTRACT

The mitochondria-mediated apoptosis pathway is an effective option for cancer therapy due to the presence of cell-suicide weapons in mitochondria. However, anti-apoptotic proteins that are over-expressed in the mitochondria of many malignant tumors, such as Bcl-2 protein, could allow the cancer cells to evade apoptosis, greatly reducing the efficacy of this type of chemotherapy. Here, we constructed a hierarchical targeted delivery system that can deliver siRNA and chemotherapeutic agents sequentially to tumor cells and mitochondria. In detail, the copolymer TPP-CP-LND (TCPL) was synthesized by the mitochondria-targeting ligand triphenylphosphine (TPP) and therapeutic drug lonidamine (LND) conjugated to the polyethyleneimine in chitosan-graft-PEI (CP), and then complexed with siRNA. Followed, the complexes were coated with poly(acrylic acid)-polyethylene glycol-folic acid (PPF) copolymer to form a hierarchical targeted co-delivery system (TCPL/siRNA/PPF NPs). The TCPL/siRNA/PPF NPs had a neutral surface charge, were stable in plasma and exhibited pH-responsive shell separation. Remarkably, the TCPL/siRNA/PPF NPs simultaneously released siBcl-2 into the cytoplasm and delivered LND to mitochondria in the same cancer cell after FA-directed internalization, and even synergistically activated mitochondria apoptosis pathway. This work demonstrated the potential of RNA-interference and mitochondria-targeted chemotherapeutics to collaboratively stimulate the mitochondria apoptosis pathway for cancer therapy.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Indazoles/administration & dosage , Mitochondria/physiology , Nanocapsules/chemistry , RNA, Small Interfering/administration & dosage , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Diffusion , HeLa Cells , Humans , Indazoles/chemistry , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Nanocapsules/ultrastructure , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...