Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Biomaterials ; 304: 122384, 2024 01.
Article in English | MEDLINE | ID: mdl-38016334

ABSTRACT

siRNA has demonstrated a promising paradigm for therapy of acute lung injury(ALI). However, the pulmonary mucus layer barrier powerfully hinders the therapeutic efficacy. Herein, we proposed to use dual drive power to enhance the mucus permeation of siRNA by constructing the neutral and targeted selenium nanozymes therapeutic system. The multifunctional selenium nanozymes (CWP-Se@Man) were synthesized by modifying with cationic water-soluble pillar arene (CWP) and mannose (Man). After loading CCR2-siRNA, the CWP-Se@Man reached electroneutrality that co-driven by electroneutrality and targeting, the mucus permeation capacity of CWP-Se@Man enhanced by ∼15 fold, thus effectively penetrate pulmonary mucus layer and deliver CCR2-siRNA into macrophages. Moreover, with optimizing the composition of CWP-Se@Man made of CWP (Slutsky, 2013) [5] or CWP (Ichikado et al., 2012) [6], the therapeutic system CWP (Ichikado et al., 2012) [6]-Se@Man showed better biological activities due to smaller size. In inflamed modes, the CWP-Se@Man nanotherapeutic systems loading CCR2-siRNA not only exerted pronounced anti-inflammatory effect through combining inhibit the chemotactic effect and ROS, but also effectively against ALI after blocking the circulatory effect of ROS and inflammatory cytokines. Therefore, this strategy of dual-driving force penetration mucus renders a unique approach for mediating trans-mucus nucleic acid delivery in lungs, and provide a promising treatment for the acute lung injury therapy.


Subject(s)
Acute Lung Injury , Nanoparticles , Selenium , Humans , Reactive Oxygen Species , RNA, Small Interfering , Acute Lung Injury/drug therapy , Mucus , Lung
2.
Nanomedicine (Lond) ; 18(3): 259-277, 2023 02.
Article in English | MEDLINE | ID: mdl-37125618

ABSTRACT

Aims: Achieving an effective biocompatible system for siRNAs delivery to the tumor site remains a significant challenge. Materials & methods: Selenium nanoparticles (SeNPs) modified by chitosan (CS) and hyaluronic acid (HA) were fabricated for PLK1 siRNAs (siPLK1) delivery to the bladder cancer cells. The HA-CS-SeNP@siPLK1 efficacy was evaluated using in vitro and in vivo models. Results: HA-CS-SeNP@siPLK1 was selectively internalized into T24 cells through clathrin-mediated endocytosis. Treatment with HA-CS-SeNP@siPLK1 successfully silenced the PLK1 gene, inhibited cell proliferation and induced cell cycle arrest in vitro. HA-CS-SeNP@siPLK1 could also inhibit tumor growth in vivo without causing systemic toxicity. Conclusion: Our results suggest that HA-CS-SeNPs may provide a good vehicle for delivering siPLK1 to the bladder tumor site.


siRNAs are small biomolecules shown as novel insights in cancer gene therapy because of their capability to silence target genes. However, achieving an effective biocompatible system for siRNA delivery to the tumor site remains a significant challenge. This work aimed to develop a nanoparticle-based delivery system consisting of selenium nanoparticles modified by chitosan and hyaluronic acid to sustain the release of siRNAs to bladder cancer cells. The results of this study demonstrated that this nanosystem successfully silenced the PLK1 gene and reduced the proliferation in vitro and in vivo. These findings suggest that hyaluronic acid-chitosan-selenium nanoparticles may open a new insight for targeted gene therapy for bladder cancer.


Subject(s)
Chitosan , Nanoparticles , Selenium , Urinary Bladder Neoplasms , Humans , RNA, Small Interfering/genetics , Hyaluronic Acid , Cell Line, Tumor , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism
3.
Adv Mater ; 35(13): e2209672, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36749980

ABSTRACT

Multi-component nanoparticles (mNPs) hold great potential for disease prevention and treatment. However, a major barrier is the lack of versatile platforms to accommodate steps of assembly processes of mNPs. Here the microfluidics-enabled serial assembly (MESA) of mNPs is presented. The microfluidic chip, as a mini-conveyor of initial materials, sequentially enables the assembly of sorafenib supramolecule, electrostatic adsorption of siRNA, and surface assembly of protective lipids. The produced lipid-siRNA-sorafenib nanoparticles (LSS NPs) have ultrahigh encapsulation efficiencies for sorafenib (≈100%) and siRNA (≈95%), which benefit from the accommodation of both fast and slow processes on the chip. Although carrying negative charges, LSS NPs enable cytosolic delivery of agents and high gene silencing efficiency within tumor cells. In vivo, the LSS NPs delivering hypoxia-induced factor (HIF1α)-targeted siRNA efficiently regress tumors of Hep3B xenograft and hepatocellular carcinoma patient-derived primary cells xenograft (PDCX) and finally extend the average survival of PDCX mice to 68 days. Thus, this strategy is promising as a sorafenib/siRNA combination therapy, and MESA can be a universal platform for fabricating complex nanosystems.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Humans , Animals , Mice , Sorafenib , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , RNA, Small Interfering/genetics , Microfluidics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Lipids , Cell Line, Tumor
4.
Biomater Adv ; 133: 112605, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35525767

ABSTRACT

This investigation reports the green approach for developing laser activatable nanoscale-graphene colloids (nGC-CO-FA) for chemo-photothermal combined gene therapy of triple-negative breast cancer (TNBC). The nano colloid was found to be nanometric as characterized by SEM, AFM, and zeta sizer (68.2 ± 2.1 nm; 13.8 ± 1.2 mV). The doxorubicin (Dox) loaded employing hydrophobic interaction/π-π stacking showed >80% entrapment efficiency with a sustained pH-dependent drug release profile. It can efficiently incorporate siRNA and Dox and successfully co-localize them inside TNBC cells to obtain significant anticancer activity as evaluated using CCK-8 assay, apoptosis assay, cell cycle analysis, cellular uptake, fluorescence assay, endosomal escape study, DNA content analysis, and gene silencing efficacy studies. nGC-CO-FA/Dox/siRNA released the Dox in temperature- and a pH-responsive manner following NIR-808 laser irradiation. The synergistic photo-chemo-gene therapy using near infrared-808 nm laser (NIR-808) irradiation was found to be more effective as compared to without NIR-808 laser-treated counterparts (∆T: 37 ± 1.1 °C → to 49.2 ± 3.1 °C; 10 min; 0.5 W/cm2), suggesting the pivotal role of photothermal combined gene-therapy in the treatment of TNBC.


Subject(s)
Hyperthermia, Induced , Triple Negative Breast Neoplasms , Doxorubicin/pharmacology , Genetic Therapy , Humans , Lasers , Phototherapy , RNA, Small Interfering/genetics , Triple Negative Breast Neoplasms/genetics
5.
Mol Pharm ; 18(6): 2105-2121, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33945284

ABSTRACT

The ongoing pandemic of global concern has killed about three million humans and affected around 151 million people worldwide, as of April 30, 2021. Although recently approved vaccines for COVID-19 are engendering hope, finding new ways to cure the viral pandemic is still a quest for researchers worldwide. Major pandemics in history have been of viral origin, such as SARS, MERS, H1NI, Spanish flu, and so on. A larger emphasis has been on discovering potential vaccines, novel antiviral drugs, and agents that can mitigate the viral infection symptoms; however, a relatively new area, RNA interference (RNAi), has proven effective as an antiviral agent. The RNAi phenomenon has been largely exploited to cure cancer, neurodegenerative diseases, and some rare diseases. The U.S. Food and Drug Administration has recently approved three siRNA products for human use that garner significant hope in siRNA therapeutics for coronaviruses. There have been some commentaries and communications addressing this area. We have summarized and illustrated the significance and the potential of the siRNA therapeutics available as of April 30, 2021 to combat the ongoing viral pandemic and the emerging new variants such as B.1.1.7 and B.1.351. Numerous successful in vitro studies and several investigations to address the clinical application of siRNA therapeutics provide great hope in this field. This seminal Review describes the significance of siRNA-based therapy to treat diverse viral infections in addition to the current coronavirus challenge. In addition, we have thoroughly reviewed the patents approved for coronaviruses, the major challenges in siRNA therapy, and the potential approaches to address them, followed by innovation and prospects.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Pandemics/prevention & control , RNA, Small Interfering/therapeutic use , SARS-CoV-2/genetics , Antiviral Agents/history , COVID-19/epidemiology , COVID-19/history , COVID-19/virology , Clinical Trials as Topic , Drug Approval , Drug Evaluation, Preclinical , History, 20th Century , History, 21st Century , Humans , Mutation , Patents as Topic , RNA, Small Interfering/history , SARS-CoV-2/pathogenicity
6.
Int J Nanomedicine ; 15: 9759-9770, 2020.
Article in English | MEDLINE | ID: mdl-33304100

ABSTRACT

BACKGROUND: Delivery of therapeutic small interfering RNA (siRNA) via functionalized nanoparticles holds great promise for cancer therapy. However, developing a safe and efficient delivery carrier of siRNA is a challenging issue. METHODS: RGDfC peptide was used to modify the surface of selenium nanoparticles (SeNPs) to synthesize a biocompatible siRNA delivery vehicle (R-SeNPs), and MEF2D-siRNA was loaded onto R-SeNPs to prepare a functionalized selenium nanoparticle R-Se@MEF2D-siRNA. The chemical properties of R-SeNPs were characterized, and the anticancer efficacy as well as related mechanisms of R-Se@MEF2D-siRNA were further explored. RESULTS: R-Se@MEF2D-siRNA was significantly taken up by SKOV3 cells and could enter SKOV3 cells mainly in the clathrin-associated endocytosis way. The result of in vitro siRNA release demonstrated that R-Se@MEF2D-siRNA could release MEF2D-siRNA quicker in a microenvironment simulating a lysosomal environment in tumor cells compared to a normal physiological environment. The results of qRT-PCR assay proved that R-Se@MEF2D-siRNA could effectively silence the expression of the MEF2D gene in SKOV3 cells. R-Se@MEF2D-siRNA remarkably suppressed the proliferation of SKOV3 cells and further triggered its apoptosis. In addition, R-Se@MEF2D-siRNA had the capability to disrupt mitochondrial membrane potential (MMP) in SKOV3 cells and resulted in the overproduction of reactive oxygen species (ROS), indicating that mitochondrial dysfunction and ROS generation played an important role in the apoptosis of SKOV3 cells induced by R-Se@MEF2D-siRNA. In vivo, R-Se@MEF2D-siRNA also exhibited excellent antitumor activity mainly through decreasing tumor cells proliferation and triggering their apoptosis in tumor-bearing nude mice. CONCLUSION: R-Se@MEF2D-siRNA provides an alternative strategy for ovarian cancer treatment in the clinic.


Subject(s)
Gene Silencing , Nanoparticles/chemistry , Ovarian Neoplasms/therapy , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Selenium/chemistry , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Drug Carriers/chemistry , Female , Humans , MEF2 Transcription Factors/deficiency , MEF2 Transcription Factors/genetics , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Reactive Oxygen Species/metabolism , Tumor Microenvironment/genetics
7.
Biomaterials ; 245: 119976, 2020 07.
Article in English | MEDLINE | ID: mdl-32213362

ABSTRACT

Photothermal therapy (PTT) normally requires to maintain the temperature of tumor lesions above 50 °C, which potentially induces local inflammation and tumor metastasis. To avoid these side effects, it is vital to achieve effective antitumor efficacy at relatively low temperature (42-45 °C) during the PTT treatment. Herein, we design a polydopamine (PDA)-coated nucleic acid nanogel as a therapeutic complex for siRNA-mediated low-temperature PTT. First, siRNAs that target the heat-shock-protein 70 (Hsp70) serve as crosslinkers to guide the DNA-grafted polycaprolactone (DNA-g-PCL) assemble into nanosized hydrogel particles through nucleic acid hybridization. Thereafter, the obtained siRNA-embedded nanogels are further coated with a thin layer of polydopamine, which not only protects the nanogels against enzymatic degradation but also endows the nanogels with excellent photothermal conversion capacity under near infrared (NIR) light irradiation. After surface PEGylation, this triple shield siRNA delivery complex shows the capability of effective ablating the tumor under relatively mild condition.


Subject(s)
Hyperthermia, Induced , Nucleic Acids , Indoles , Nanogels , Phototherapy , Photothermal Therapy , Polyethylene Glycols , Polyethyleneimine , Polymers , RNA, Small Interfering , Temperature
8.
Mater Sci Eng C Mater Biol Appl ; 110: 110594, 2020 May.
Article in English | MEDLINE | ID: mdl-32204058

ABSTRACT

Short interfering RNA (siRNA) has been investigated as a promising modality of cancer treatment due to its capability to target specific target genes for downregulation. However, the successful application of this strategy depends on producing a safe and effective carrier system for delivering siRNA to the tumor. Thus, investigation of siRNA delivery carriers is a fundamental step in the field of siRNA-based therapeutics. In the current research, the surface of selenium nanoparticles (SeNPs) were modified with the tumor-targeted molecular RGDfC peptide with positive charge to synthetize the biocompatible siRNA carrier RGDfC-SeNPs. Subsequently, KLK12-siRNA was loaded onto the surface of RGDfC-SeNPs to create functionalized nanoparticles (RGDfC-Se@siRNA) that we tested for in vitro and in vivo antitumor efficacy. We measured significantly greater particle uptake in HT-29 colorectal cancer cells relative to HUVECs, providing evidence for the targeted delivery of RGDfC-Se@siRNA. We found that RGDfC-Se@siRNA could enter HT-29 cells primarily via clathrin-mediated endocytosis. Further, these particles experienced faster siRNA release in an acidic microenvironment compared to pH 7.4. The results from quantitative PCR and Western blot assays suggested that the target gene of KLK12 in HT-29 cells were obviously silenced by RGDfC-Se@siRNA. The further biological studies showed that treatment with RGDfC-Se@siRNA had ability to suppress the proliferation and migration/invasion of HT-29 cells, and triggered HT-29 cells apoptosis. RGDfC-Se@siRNA could induce the mitochondrial membrane potential (MMP) disruption and enhance the reactive oxygen species (ROS) generation in HT-29 cells, indicating that RGDfC-Se@siRNA induced the HT-29 cells apoptosis possibly by a ROS-mediated mitochondrial dysfunction pathway. Importantly, the in vivo antitumor study also verified that RGDfC-Se@siRNA could significantly suppress the growth of tumor in vivo. In addition, we did not observe any signs of systemic or tissue-specific toxicity after administration of RGDfC-Se@siRNA in mice. As a whole, these findings suggest that RGDfC-Se@siRNA has promising potential as a therapy for colorectal cancer.


Subject(s)
Colorectal Neoplasms , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Kallikreins/biosynthesis , Metal Nanoparticles , Neoplasm Proteins/biosynthesis , Oligopeptides , Selenium , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Female , HT29 Cells , Human Umbilical Vein Endothelial Cells , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/chemistry , Oligopeptides/pharmacology , Selenium/chemistry , Selenium/pharmacology , Xenograft Model Antitumor Assays
9.
Pharm Res ; 37(3): 46, 2020 Feb 03.
Article in English | MEDLINE | ID: mdl-32016611

ABSTRACT

PURPOSE: Short interfering RNA (siRNA) therapy promises a new era in treatment of breast cancers but effective delivery systems are needed for clinical use. Since silencing complementary targets may offer improved efficacy, this study was undertaken to identify non-viral carriers for combinatorial siRNA delivery for more effective therapy. METHODS: A library of lipid-substituted polymers from low molecular weight polyethyleneimine (PEI), linoleic acid (LA) and α-linoleic acid (αLA) with amide or thioester linkages was prepared and investigated for delivering Mcl-1, survivin and STAT5A siRNAs in breast cancer cells. RESULTS: The effective polymers formed 80-190 nm particles with similar zeta-potentials, but the serum stability was greater for complexes formed with amide-linked lipid conjugates. The LA and αLA substitutions, with the low molecular weight PEI (1.2 kDa and 2.0 kDa) were able to deliver siRNA effectively to cells and retarded the growth of breast cancer cells. The amide-linked lipid substituents showed higher cellular delivery of siRNA as compared to thioester linkages. Upon combinational delivery of siRNAs, growth of MCF-7 cells was inhibited to a greater extent with 2.0PEI-LA9 mediated delivery of Mcl-1 combined survivin siRNAs as compared to individual siRNAs. The qRT-PCR analysis confirmed the decrease in mRNA levels of target genes with specific siRNAs and 2.0PEI-LA9 was the most effective polymer for delivering siRNAs (either single or in combination). CONCLUSIONS: This study yielded effective siRNA carriers for combinational delivery of siRNAs. Careful choice of siRNA combinations will be critical since targeting individual genes might alter the expression of other critical mediators.


Subject(s)
Breast Neoplasms/metabolism , Drug Carriers/chemistry , Gene Targeting/methods , Polyethyleneimine/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Cell Line, Tumor , Female , Gene Silencing , Humans , Linoleic Acid , Lipids , MCF-7 Cells , Myeloid Cell Leukemia Sequence 1 Protein , Polyethyleneimine/metabolism , Polymers/chemistry , Polymers/metabolism , STAT5 Transcription Factor/metabolism , Survivin/metabolism , Tumor Suppressor Proteins/metabolism
10.
Drug Deliv ; 27(1): 15-25, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31830840

ABSTRACT

Small interfering RNA (siRNA) exhibits great potential as a novel therapeutic option due to its highly sequence-specific ability to silence genes. However, efficient and safe delivery carriers are required for developing novel therapeutic paradigms. Thus, the successful development of efficient delivery platforms for siRNA is a crucial issue for the development of siRNA-based drugs in cancer treatments. In this study, biocompatible selenium nanoparticles (SeNPs) were loaded with RGDfC peptide to fabricate tumor-targeting gene delivery vehicle RGDfC-SeNPs. Subsequently, RGDfC-SeNPs were loaded with Derlin1-siRNA to fabricate RGDfC-Se@siRNA, which are functionalized selenium nanoparticles. RGDfC-Se@siRNA showed greater uptake in HeLa cervical cancer cells in comparison with that in human umbilical vein endothelial cells (HUVECs), verifying the RGDfC-mediated specific uptake of RGDfC-Se@siRNA. RGDfC-Se@siRNA was capable of entering HeLa cells via clathrin-associated endocytosis, and showed faster siRNA release in a cancer cell microenvironment in comparison with a normal physiological environment. qPCR and western blotting assays both indicated that RGDfC-Se@siRNA exhibited an obvious gene silencing efficacy in HeLa cells. RGDfC-Se@siRNA suppressed the invasion, migration and the proliferation of HeLa cells, and triggered HeLa cell apoptosis. Moreover, RGDfC-Se@siRNA induced the disruption of mitochondrial membrane potentials. Meanwhile, RGDfC-Se@siRNA enhanced the generation of reactive oxygen species (ROS) in HeLa cell, suggesting that mitochondrial dysfunction mediated by ROS might play a significant role in RGDfC-Se@siRNA-induced apoptosis. Interestingly, RGDfC-SeNPs@siRNA exhibited significant antitumor activity in a HeLa tumor-bearing mouse model. Additionally, RGDfC-SeNPs@siRNA is nontoxic to main organ of mouse. The above results indicate that RGDfC-Se@siRNA provides a promising potential for cervical cancer therapy.


Subject(s)
Membrane Proteins/drug effects , Nanoparticles/chemistry , Oligopeptides/pharmacology , RNA, Small Interfering/administration & dosage , Selenium/chemistry , Apoptosis/drug effects , Blotting, Western , Cell Migration Inhibition , Cell Proliferation/drug effects , Female , Gene Silencing/drug effects , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Reactive Oxygen Species/metabolism , Tumor Microenvironment , Uterine Cervical Neoplasms/drug therapy
11.
Hum Gene Ther ; 30(12): 1531-1546, 2019 12.
Article in English | MEDLINE | ID: mdl-31547718

ABSTRACT

Preclinical studies showed that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) therapy is safe and effective to combat cancers, but clinical outcomes have been less than optimal due to short half-life of TRAIL protein, insufficient induction of apoptosis, and TRAIL resistance displayed in many tumors. In this study, we explored co-delivery of a TRAIL expressing plasmid (pTRAIL) and complementary small interfering RNAs (siRNAs) (silencing Bcl2-like 12 [BCL2L12] and superoxide dismutase 1 [SOD1]) to improve the response of breast cancer cells against TRAIL therapy. It is desirable to co-deliver the pDNA along with siRNA using a single delivery agent, but this is challenging given different structures of long/flexible pDNA and short/rigid siRNA. Toward this goal, we identified an aliphatic lipid-grafted low-molecular weight polyethylenimine (PEI) that accommodated both pDNA and siRNA in a single complex. The co-delivery of pTRAIL with BCL2L12- or SOD1-specific siRNAs resulted more significant cell death in different breast cancer cells compared with separate delivery without affecting nonmalignant cells viability. Ternary complexes of lipopolymer with pTRAIL and BCL2L12 siRNA significantly retarded the growth of breast cancer xenografts in mice. The enhanced anticancer activity was attributed to increased in situ secretion of TRAIL and sensitization of breast cancer cells against TRAIL by the co-delivered siRNAs. The lipid-grafted PEIs capable of co-delivering multiple types of nucleic acids can serve as powerful carriers for more effective complementary therapeutics. Graphical Abstract [Figure: see text].


Subject(s)
Breast Neoplasms/genetics , Genetic Therapy , Muscle Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Superoxide Dismutase-1/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Animals , Apoptosis/drug effects , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Silencing/drug effects , Gene Transfer Techniques , Heterografts , Humans , Mice , Muscle Proteins/antagonists & inhibitors , Plasmids/genetics , Plasmids/pharmacology , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Superoxide Dismutase-1/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors
12.
Biomaterials ; 223: 119463, 2019 12.
Article in English | MEDLINE | ID: mdl-31521887

ABSTRACT

The success of photothermal therapy (PTT) is often hampered by the thermo-resistance of tumor cells mediated by over-expressed heat shock proteins (HSPs). Herein, we developed a guanidine-rich, spherical helical polypeptide (DPP) with multivalency-assisted strong membrane penetrating capability, which mediated effective RNAi against tumor glycolysis metabolism to sensitize PTT. ICG was loaded into the internal cavity of DPP, and siRNA against pyruvate kinase M2 (siPKM2) was condensed by DPP to form positively charged nanocomplexes (NCs). The NCs were further coated with human serum albumin to enhance serum stability, prolong blood circulation, and improve tumor targeting. Due to its multivalent topology, DPP exhibited stronger membrane activity yet lower cytotoxicity than its linear analogue (LPP), thus enabling efficient PKM2 silencing in MCF-7 cells in vitro (~75%) and in vivo (~70%). The PKM2 silencing inhibited tumor glycolysis metabolism and further depleted the energy supply for HSPs production, thus overcoming the heat endurance of tumor cells to strengthen ICG-mediated photothermal ablation. Additionally, siPKM2-mediated energy depletion led to tumor cell starvation, which imparted synergistic anti-cancer effect with PTT. This study therefore provides a promising strategy for designing membrane-penetrating siRNA delivery materials, and it renders a unique RNAi-mediated anti-metabolic mechanism in sensitizing PTT and enabling starvation therapy.


Subject(s)
Glycolysis , Neoplasms/therapy , Phototherapy/methods , RNA, Small Interfering/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Carrier Proteins/metabolism , Cell Line, Tumor , Drug Delivery Systems , Drug Resistance, Neoplasm , Female , Gene Silencing , Guanidine/chemistry , Humans , Hyperthermia, Induced , Kinetics , MCF-7 Cells , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Peptides/chemistry , Polymers/chemistry , RNA Interference , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins
13.
Acta Biomater ; 86: 416-428, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30611792

ABSTRACT

Combined photothermal and gene therapy provides a promising modality toward cancer treatment, yet facile integration and controlled codelivery of gene payloads and photothermal conversion agents (PTCAs) remains a great challenge. Inspired by the robust wet adhesion of marine mussels, we present a rationally designed nanosystem constructed by using hybrid mesoporous polydopamine nanoparticles (MPDA) with sub-100 nm sizes and a high photothermal conversion efficiency of 37%. The surface of the particles were modified with tertiary amines by the facile Michael addition/Schiff base reactions of PDA to realize high siRNA loading capacity (10 wt%). Moreover, a successful calcium phosphate (CaP) coating via biomineralization was constructed on the cationic nanoparticle to prohibit premature release of siRNA. The CaP coating underwent biodegradation in weakly-acidic subcellular conditions (lysosomes). The synergistic integration of tertiary amines and catechol moieties on the subsequently exposed surfaces was demonstrated to feature the destabilization/disruption ability toward model cellular membranes via the greatly enhanced interfacial adhesion and interactions. Consequently, sufficient permeability of lysosomal membranes, and in turn, a high lysosomal escape efficiency, was realized, which then resulted in high gene silencing efficiencies via sufficient cytosolic delivery of siRNA. When an efficient knocking down (65%) of survivin (an inhibitor of apoptosis proteins) was combined with a subsequent photothermal ablation, remarkably higher therapeutic efficiencies were observed both in vitro and in vivo, as compared with monotherapy. The system may help to pave a new avenue on the utilization of bio-adhesive surfaces for handling the obstacles of combined photothermal and gene therapy. STATEMENT OF SIGNIFICANCE: Polydopamine (PDA) based porous photothermal-conversion agent (PTCA) with sufficiently high conversion efficiency was employed to deliver photothermal/gene therapy modalities towards cancer treatment. CaP coating via PDA-induced biomineralization was constructed to prohibit premature release of siRNA loaded in the pore space of the nanocarriers. Responsive degradation of CaP also led to the exposure of membrane-lytic surfaces built through the synergistic integration of tertiary amines and catechol moieties, and in turn the significantly enhanced lysosomal escape and cytosol siRNA delivery. Therapeutic targeting of survivin was successfully applied for activation of apoptosis and programmed cell death. Combined photothermal and gene therapy improved therapeutic effectiveness.


Subject(s)
Calcium Phosphates/chemistry , Cell Membrane Permeability , Hyperthermia, Induced , Indoles/chemistry , Nanoparticles/chemistry , Phototherapy , Polymers/chemistry , RNA, Small Interfering/therapeutic use , Animals , Calcification, Physiologic , Cell Death , Combined Modality Therapy , Drug Carriers/chemistry , Endocytosis , Erythrocytes/metabolism , Gene Silencing , Genetic Therapy , Hemolysis , Hep G2 Cells , Humans , Lysosomes/metabolism , Male , Mice, Nude , Nanoparticles/ultrastructure , Porosity
14.
Biomed Pharmacother ; 111: 802-812, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30616079

ABSTRACT

Nanoparticles (NPs) serve to reduce the toxicity, enhance bioactivity, improve targeting, and provide versatile means to control the release profile of the encapsulated moiety. Among different NPs, inorganic NPs of metals like Ag, Au, Ce, Fe, Se, Ti and Zn possess a significant place owing to their unique bioactivities in nanoforms. Selenium (Se) is an essential trace element. It is incorporated into selenoproteins as selenocysteine (Sec) representing the most important part of the active center of their enzymatic activities. Many selenoproteins have oxidoreductase activity and, thus, regulate the physiological redox balance. Se has a narrow therapeutic window and the toxicity margins are very delicate whereas the nanoparticles of Se (SeNPs) possess remarkably reduced toxicity. SeNPs have been explored in various oxidative stress and inflammation mediated disorders like arthritis, cancer, diabetes and nephropathy with potential therapeutic benefits. SeNPs constitute an attractive carrier platform to ferry various drugs to the site of action. Herein we have discussed the significance of nanosizing on the pharmacological activity of Se. The role of SeNPs in pharmacological protection against various inflammatory and oxidative stress mediated conditions is presented. However, it is largely unknown how SeNPs may affect the pharmacokinetics and pharmacodynamics of selenoproteins. Most of the available studies were poorly designed without any comparison to the other Se sources. In the future, detailed studies with inclusion of an appropriate source of Se should be carried out with emphasis on understanding the role of selenoproteins in the observed pharmacological activity.


Subject(s)
Antioxidants/administration & dosage , Nanomedicine/methods , Nanoparticles/administration & dosage , Oxidative Stress/drug effects , Selenium/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Antioxidants/metabolism , Humans , Nanomedicine/trends , Nanoparticles/metabolism , Oxidative Stress/physiology , Selenium/metabolism
15.
ACS Appl Mater Interfaces ; 10(25): 21137-21148, 2018 Jun 27.
Article in English | MEDLINE | ID: mdl-29882656

ABSTRACT

Human telomerase reverse transcriptase (hTERT) has been found to be closely related to tumor transformation, growth, and metastasis. Thus, the delivery of hTERT small interfering RNA (siRNA) is an important approach for cancer gene therapy. However, the single anticancer effect of gene silencing is often limited by poor specificity or low efficiency in siRNA delivery and release. In this work, we present small and thin black phosphorus (BP) nanosheets as a biodegradable delivery system for hTERT siRNA. The BP nanosheets prepared with poly(ethylene glycol) (PEG) and polyethylenimine (PEI) modification (PPBP), exhibited high siRNA loading capacity and robust cell uptake. The PPBP nanosheets also exhibited potent photodynamic therapy/photothermal therapy (PDT/PTT) activities when exposed to different wavelengths of laser irradiation. More importantly, PPBP nanosheets underwent a gradual degradation when presented in a mixture of low pH and reactive oxygen species (ROS)-rich environment. The degradation of PPBP was strengthened especially after local and minimal invasive PDT treatment, because of excessive ROS production. Further delivery and release of siRNA to the cytoplasm for gene silencing was achieved by PEI-aided escape from the acidic lysosome. Thus, PPBP-siRNA efficiently inhibited tumor growth and metastasis by specific delivery of hTERT siRNA and a synergistic combination of targeted gene therapy, PTT and PDT.


Subject(s)
Phosphorus/chemistry , Cell Line, Tumor , Genetic Therapy , Humans , Neoplasms , Polyethyleneimine , RNA, Small Interfering
16.
Int J Nanomedicine ; 13: 1539-1552, 2018.
Article in English | MEDLINE | ID: mdl-29588583

ABSTRACT

BACKGROUND: Small interfering RNA (siRNA) as a new therapeutic modality holds promise for cancer treatment. However, the traditional viral carriers are prone to immunogenicity and risk of insertional mutagenesis. METHODS: In order to provide a tumor-targeted delivery carrier of siRNA in cancer therapy, the hyaluronic acid (HA)-selenium (Se)-polyethylenimine (PEI) nanoparticle (NP) was fabricated by decorating SeNP with HA as a tumor-targeting moiety and by linking the polycationic polymers polyethylenimine PEI onto the surface of SeNP. The siRNA was loaded to the surface of SeNP HA-Se-PEI via the electrostatic interaction between siRNA and PEI to prepare the functionalized SeNP HA-Se-PEI@siRNA. RESULTS: The HA-Se-PEI@siRNA was internalized into the HepG2 cell mainly in a clathrin-mediated endocytosis manner. Owing to the active tumor-targeted effect mediated by HA, HA-Se-PEI@siRNA achieved the obvious higher transfection efficiency, greater gene silencing ability, and stronger cytotoxicity in the HepG2 cell compared with the passive tumor-targeted NP Se-PEI@siRNA. The knockdown of hairy and enhancer of split 5 by HA-Se-PEI@siRNA induced the HepG2 cell cycle arrest at the G0/G1 phase and apoptosis. Furthermore, the treatment with HA-Se-PEI@siRNA resulted in greater antitumor efficacy compared with the Se-PEI@siRNA in vitro and in vivo. In addition, the HA-Se-PEI@siRNA was almost no toxic to the key organs of mice. CONCLUSION: These findings provided an alternative therapeutic route for targeted cancer treatments.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Hyaluronic Acid/chemistry , Liver Neoplasms/drug therapy , Nanoparticles/administration & dosage , Selenium/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Hepatocellular/genetics , Endocytosis/drug effects , Endocytosis/genetics , Gene Silencing/drug effects , Hep G2 Cells , Humans , Hyaluronic Acid/pharmacology , Liver Neoplasms/genetics , Mice, Inbred BALB C , Nanoparticles/chemistry , Polyethyleneimine/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Selenium/administration & dosage , Selenium/pharmacology , Xenograft Model Antitumor Assays
17.
Biotechnol Bioeng ; 115(7): 1866-1877, 2018 07.
Article in English | MEDLINE | ID: mdl-29578571

ABSTRACT

Further quantitative understanding of the biological effects and mechanisms involved in cellular and intracellular delivery of nucleic acid materials is required to produce clinical applications of gene therapy. Several modeling approaches have been used in this field; however, a comprehensive approach that integrates all the key pharmacological issues into a holistic framework that is applicable for in vivo conditions is still lacking. This contribution presents a pharmacokinetic/pharmacodynamic model-based control study of non-viral siRNA delivery describing the dynamics of the delivery process and takes into account the main multi-objective optimization issues such as efficacy and toxicity, as well as the effect of uncertainty in cell doubling time. The methodology developed in this work is used to predict the optimal dosage injection rate and optimal intracellular exposure of siRNAs in order to improve the pharmacological effects before cell division occurs. The present analysis successfully provides quantitative predictions of non-viral siRNA activity paving the path for further experimental work to probe more efficient delivery systems.


Subject(s)
Biological Products/pharmacology , Biological Products/pharmacokinetics , Genetic Therapy/methods , RNA, Small Interfering/pharmacology , RNA, Small Interfering/pharmacokinetics , Biological Products/administration & dosage , Biological Products/toxicity , Cell Line , Cell Survival/drug effects , Gene Silencing , Genetic Therapy/adverse effects , Hepatocytes/drug effects , Hepatocytes/physiology , Humans , Models, Statistical , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/toxicity
18.
Acta Biomater ; 66: 294-309, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29183848

ABSTRACT

Triple-negative breast cancer is an aggressive form of breast cancer with few therapeutic options if it recurs after adjuvant chemotherapy. RNA interference could be an alternative therapy for metastatic breast cancer, where small interfering RNA (siRNA) can silence the expression of aberrant genes critical for growth and migration of malignant cells. Here, we formulated a siRNA delivery system using lipid-substituted polyethylenimine (PEI) and hyaluronic acid (HA), and characterized the size, ζ-potential and cellular uptake of the nanoparticulate delivery system. Higher cellular uptake of siRNA by the tailored PEI/HA formulation suggested better interaction of complexes with breast cancer cells due to improved physicochemical characteristics of carrier and HA-binding CD44 receptors. The siRNAs against specific phosphatases that inhibited migration of MDA-MB-231 cells were then identified using library screen against 267 protein-tyrosine phosphatases, and siRNAs to inhibit cell migration were further validated. We then assessed the combinational delivery of a siRNA against CDC20 to decrease cell growth and a siRNA against several phosphatases shown to decrease migration of breast cancer cells. Combinational siRNA therapy against CDC20 and identified phosphatases PPP1R7, PTPN1, PTPN22, LHPP, PPP1R12A and DUPD1 successfully inhibited cell growth and migration, respectively, without interfering the functional effect of the co-delivered siRNA. The identified phosphatases could serve as potential targets to inhibit migration of highly aggressive metastatic breast cancer cells. Combinational siRNA delivery against cell cycle and phosphatases could be a promising strategy to inhibit both growth and migration of metastatic breast cancer cells, and potentially other types of metastatic cancer. STATEMENT OF SIGNIFICANCE: The manuscript investigated the efficacy of a tailored polymeric siRNA delivery system formulation as well as combinational siRNA therapy in metastatic breast cancer cells to inhibit malignant cell growth and migration. The siRNA delivery was undertaken by non-viral means with PEI/HA. We identified six phosphatases that could be critical targets to inhibit migration of highly aggressive metastatic breast cancer cells. We further report on specifically targeting cell cycle and phosphatase proteins to decrease both malignant cell growth and migration simultaneously. Clinical gene therapy against metastatic breast cancer with effective and safe delivery systems is urgently needed to realize the potential of molecular medicine in this deadly disease and our studies in this manuscript is intended to facilitate this endeavor.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Movement , Combinatorial Chemistry Techniques , Hyaluronic Acid/chemistry , Phosphoprotein Phosphatases/metabolism , RNA, Small Interfering/administration & dosage , Surface-Active Agents/chemistry , Triple Negative Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Gene Silencing , Humans , Hyaluronan Receptors/metabolism , Linoleic Acid/chemistry , Particle Size , Polyethyleneimine/chemistry , Reproducibility of Results , Static Electricity , Triple Negative Breast Neoplasms/metabolism
19.
ACS Appl Mater Interfaces ; 9(8): 6712-6724, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28191840

ABSTRACT

We report here a novel and personalized strategy of selenium/ruthenium nanoparticles modified metal organic frameworks MIL-101(Fe) for delivering pooled small interfering RNAs (siRNAs) to enhance therapy efficacy by silencing multidrug resistance (MDR) genes and interfere with microtubule (MT) dynamics in MCF-7/T (Taxol-resistance) cell. The existence of coordinatively unsaturated metal sites in MIL-101(Fe) can strongly interact with the electron-rich functional groups of cysteine, which can be regarded as the linkage between selenium/ruthenium nanoparticles and MIL-101(Fe). Se@MIL-101 and Ru@MIL-101 loaded with MDR gene-silencing siRNAs via surface coordination can significantly enhance protection of siRNAs against nuclease degradation, increase siRNA cellular uptake, and promote siRNA escape from endosomes/lysosome to silence MDR genes in MCF-7/T cell, resulting in enhanced cytotoxicity through the induction of apoptosis with the signaling pathways of phosphorylation of p53, MAPK, and PI3K/Akt and the dynamic instability of MTs and disrupting normal mitotic spindle formation. Furthermore, in vivo investigation of the nanoparticles on nude mice bearing MCF-7/T cancer xenografts confirmed that Se@MIL-101-(P+V)siRNA nanoparticles can significantly enhance cancer therapeutic efficacy and decrease systemic toxicity in vivo.


Subject(s)
Nanoparticles , Animals , Breast Neoplasms , Cell Line, Tumor , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Metal-Organic Frameworks , Mice , Mice, Nude , Paclitaxel , Phosphatidylinositol 3-Kinases , RNA, Small Interfering , Ruthenium , Selenium
20.
Int J Pharm ; 515(1-2): 315-323, 2016 Dec 30.
Article in English | MEDLINE | ID: mdl-27765724

ABSTRACT

This study aimed to build an innovative system to deliver a systemic small interfering RNA (siRNA) treatment for rheumatoid arthritis. We combined arginine-histidine-cysteine based oligopeptide-modified polymer micelles with siRNA targeting the nuclear factor κB subunit, RelA (siRelA). This is a key molecule in the control of inflammation. We tested the cellular uptake of siRNA and its effects on inflammatory cytokine levels in vitro using synoviocytes, and siRNA distribution and therapeutic effects in vivo in mice with collagen-induced arthritis (CIA). These studies showed that arginine-histidine based oligopeptide modified micelles produced effective cellular siRNA uptake and suppressed inflammatory cytokine levels in synoviocytes. In vivo, these micelles produced marked accumulation of siRNAs in arthritic paws in CIA mice, with much less accumulation in healthy mice. The siRelA-polymer micelle complexes also produced more effective suppression of RelA mRNA expression and inflammatory cytokine levels in the arthritic paws of CIA mice and reduced their clinical symptom scores and paw thickness.


Subject(s)
Arthritis, Experimental/metabolism , NF-kappa B/metabolism , Nanoparticles/chemistry , Oligopeptides/pharmacology , Polymers/chemistry , RNA, Small Interfering/metabolism , Animals , Arginine/pharmacology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/metabolism , Collagen/pharmacology , Cysteine/pharmacology , Cytokines/metabolism , Disease Models, Animal , Histidine/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Male , Mice , Mice, Inbred DBA , Micelles , RNA, Messenger/metabolism , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Synoviocytes/drug effects , Synoviocytes/metabolism , Transcription Factor RelA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL