Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mol Pharm ; 21(3): 1436-1449, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38291705

ABSTRACT

Small interfering RNAs (siRNAs) have emerged as a powerful tool to manipulate gene expression in vitro. However, their potential therapeutic application encounters significant challenges, such as degradation in vivo, limited cellular uptake, and restricted biodistribution, among others. This study evaluates the siRNA delivery efficiency of three different lipid-substituted polyethylenimine (PEI)-based carriers, named Leu-Fect A-C, to different organs in vivo, including xenograft tumors, when injected into the bloodstream of mice. The siRNA analysis was undertaken by stem-loop RT-PCR, followed by qPCR or digital droplet PCR. Formulating siRNAs with a Leu-Fect series of carriers generated nanoparticles that effectively delivered the siRNAs into K652 and MV4-11 cells, both models of leukemia. The Leu-Fect carriers were able to successfully deliver BCR-Abl and FLT3 siRNAs into leukemia xenograft tumors in mice. All three carriers demonstrated significantly enhanced siRNA delivery into organs other than the liver, including the xenograft tumors. Preferential biodistribution of siRNAs was observed in the lungs and spleen. Among the delivery systems, Leu-Fect A exhibited the highest biodistribution into organs. In conclusion, lipid-substituted PEI-based delivery systems offer improvements in addressing pharmacokinetic challenges associated with siRNA-based therapies, thus opening avenues for their potential translation into clinical practice.


Subject(s)
Leukemia , Neoplasms , Humans , Mice , Animals , RNA, Small Interfering/genetics , Polyethyleneimine , Tissue Distribution , Leukemia/genetics , Leukemia/therapy , Lipids
2.
J Gene Med ; 24(6): e3419, 2022 06.
Article in English | MEDLINE | ID: mdl-35373897

ABSTRACT

INTRODUCTION: Breast cancer continues to be one of the leading causes of death in women, and the lack of treatment options for distant metastasis warrants the need to identify and develop more effective approaches. The aim of this study was to identify and validate targets that are associated with the survival and migration of the breast cancer cells in vitro through RNA interference (RNAi) approach. METHODS: Linoleic-acid-modified polyethylenimine (PEI) polymer was used to screen a short interfering RNA (siRNA) library against numerous cell adhesion and cytoskeleton genes in MDA-MB-231 triple-negative breast cell line, and the functional outcome of silencing was determined by growth and migration inhibition with further target validation studies. RESULTS: Heat shock protein 90B1 (HSP90B1) was identified as a crucial gene that is known to be involved in various breast cancer machineries, including uncontrolled proliferation and brain metastasis. The success of this approach was also due to the use of hyaluronic acid (HA) additive in lipopolymer complexes that showed a profound impact in reducing the cell viability (~50%), migration (~40%), and mRNA transcript levels (~80%) with a physiologically relevant siRNA concentration of 60 nM. The use of Dicer-substrate siRNA proved to be beneficial in target silencing, and a combinational treatment of integrin-ß1 (ITGB1) and HSP90B1 was effective in reducing the migration of the MDA-MB-231 and MDA-MB-436 breast cancer cells. CONCLUSION: This study demonstrates the potential to identify and silence targets using a lipid-modified PEI/siRNA system and highlights the importance of HSP90B1 in the growth and migration of breast cancer cells.


Subject(s)
Breast Neoplasms , Polyethyleneimine , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Heat-Shock Proteins , Humans , Linoleic Acid , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , RNA, Small Interfering/genetics
3.
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
4.
Int J Cancer ; 142(3): 597-606, 2018 02 01.
Article in English | MEDLINE | ID: mdl-28960310

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in variety of cancer cells without affecting most normal cells, which makes it a promising agent for cancer therapy. However, TRAIL therapy is clinically not effective due to resistance induction. To identify novel regulators of TRAIL that can aid in therapy, protein targets whose silencing sensitized breast cancer cells against TRAIL were screened with an siRNA library against 446 human apoptosis-related proteins in MDA-231 cells. Using a cationic lipopolymer (PEI-αLA) for delivery of library members, 16 siRNAs were identified that sensitized the TRAIL-induced death in MDA-231 cells. The siRNAs targeting BCL2L12 and SOD1 were further evaluated based on the novelty and their ability to sensitize TRAIL induced cell death. Silencing both targets sensitized TRAIL-mediated cell death in MDA-231 cells as well as TRAIL resistant breast cancer cells, MCF-7. Combination of TRAIL and siRNA silencing BCL2L12 had no effect in normal human umbilical vein cells and human bone marrow stromal cell. The silencing of BCL2L12 and SOD1 enhanced TRAIL-mediated apoptosis in MDA-231 cells via synergistically activating capsase-3 activity. Hence, here we report siRNAs targeting BCL2L12 and SOD1 as a novel regulator of TRAIL-induced cell death in breast cancer cells, providing a new approach for enhancing TRAIL therapy for breast cancer. The combination of siRNA targeting BCL2L12 and TRAIL can be a highly effective synergistic pair in breast cancer cells with minimal effect on the non-transformed cells.


Subject(s)
Breast Neoplasms/therapy , RNA, Small Interfering/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , MCF-7 Cells , Muscle Proteins/genetics , Polyethyleneimine/administration & dosage , Polyethyleneimine/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , Superoxide Dismutase-1/genetics
5.
ACS Appl Bio Mater ; 6(3): 1105-1121, 2023 03 20.
Article in English | MEDLINE | ID: mdl-36853230

ABSTRACT

Cationic polyethylenimine (PEI)-based nonviral gene carriers have been desirable to overcome the limitations of viral vectors in gene therapy. A range of PEI derivatives were designed, synthesized, and evaluated for nonviral delivery applications of plasmid DNA (pDNA). Linolenic acid, lauric acid, and oleic acid were covalently conjugated with low-molecular-weight PEI (Mw ∼ 1200 Da) via two different linkers, gallic acid (GA) and p-hydroxybenzoic acid (PHPA), that allows a differential loading of lipids per modified amine (3 vs 1, respectively). 1H NMR spectrum confirmed the expected structure of the conjugates as well as the level of lipid substitution. SYBR Green binding assay performed to investigate the 50% binding concentration (BC50) of lipophilic polymers to pDNA revealed increased BC50 with an increased level of lipid substitution. The particle analysis determined that GA- and PHPA-modified lipopolymers gave pDNA complexes with ∼300 and ∼100 nm in size, respectively. At the polymer/pDNA ratio of 5.0, the ζ-potentials of the complexes were negative (-6.55 to -10.6 mV) unlike the complexes with the native PEI (+11.2 mV). The transfection experiments indicated that the prepared lipopolymers showed higher transfection in attachment-dependent cells than in suspension cells based on the expression of the reporter green fluorescent protein (GFP) gene. When loaded with Cy3-labeled pDNA, the lipopolymers exhibited effective cellular uptake in attachment-dependent cells while the cellular uptake was limited in suspension cells. These results demonstrate the potential of lipid-conjugated PEI via GA and PHPA linkers, which are promising for the modification of anchorage-dependent cells.


Subject(s)
Nanoparticles , Polyethyleneimine , Polyethyleneimine/chemistry , Transfection , DNA/chemistry , Nanoparticles/chemistry , Genetic Therapy , Lipids
6.
Acta Biomater ; 148: 279-297, 2022 08.
Article in English | MEDLINE | ID: mdl-35738388

ABSTRACT

Lipid-modified low molecular weight branched polyethyleneimines (PEIs) are promising non-viral gene delivery systems that have been successfully explored for treatment of various diseases. The present study aims to determine in vitro safety of these delivery systems based on assessment of cytotoxicity with peripheral blood mononuclear cells (PBMCs), hemolysis with human red blood cells (RBC) and cytokine secretion from several sources of PBMCs. The viability of cells treated with lipopolymer/pDNA complexes was dependent on the polymer:pDNA ratio used but remained low at therapeutically relevant concentrations for most lipopolymers, except for the propionic acid substituted PEIs. The extent of hemolysis was minimal and below the accepted safety levels with most of the lipopolymers; however, some linoleic acid substituted PEIs yielded significant hemolysis activity. Unlike strong cytokine secretion from PMA/IO stimulated cells, most lipopolymer/pDNA complexes remained non-responsive, showing minimal changes in cytokine secretion (TNF-α, IL-6 and IFN-γ) irrespective of the lipopolymer/pDNA formulations. The 0.6 kDa PEI with lauric acid substituent displayed slight cytokine upregulation, however it remained low relative to the positive controls. This study demonstrated that the lipid modified LMW PEIs are expected to be safe in contact with blood components. However, close attention to lipopolymer concentration and ratio of polymer to pDNA in formulations might be required for individual lipopolymers for optimal safety response in nucleic acid therapies. STATEMENT OF SIGNIFICANCE: This manuscript investigated the safety aspects of various lipid modified low molecular weight polyethylenimine (LMW-PEI) polymers employed for pDNA delivery through in vitro studies. Using peripheral blood mononuclear cells (PBMCs) from multiple sources, we show that the hemolysis ability was minimal for most polymers, although a particular lipid substituent (linoleic acid) at specific ratios exhibited hemolysis. The levels of pro-inflammatory cytokines (TNF-α, IL-6 and IFN-γ) were slightly upregulated only with a lauric acid substituted 0.6PEI, but remained low relative to positive control treatments. We further report the beneficial effect of polyacrylic acid additives on hemolysis and cytokine secretion to a reasonable extent. This study confirms the feasibility of using LMW-PEI as safe delivery agents for various therapeutic purposes.


Subject(s)
Hemolysis , Polyethyleneimine , Gene Transfer Techniques , Humans , Interleukin-6 , Leukocytes, Mononuclear , Linoleic Acid , Molecular Weight , Plasmids , Polyethyleneimine/adverse effects , Transfection , Tumor Necrosis Factor-alpha/genetics
7.
Biomater Biosyst ; 3: 100021, 2021 Sep.
Article in English | MEDLINE | ID: mdl-36824309

ABSTRACT

Uncontrolled proliferation of the myeloid cells due to BCR-ABL fusion has been successfully treated with tyrosine kinase inhibitors (TKIs), which improved the survival rate of Chronic Myeloid Leukemia (CML) patients. However, due to interactions of CML cells with bone marrow microenvironment, sub-populations of CML cells could become resistant to TKI treatment. Since integrins are major cell surface molecules involved in such interactions, the potential of silencing integrin-ß1 on CML cell line K562 cells was explored using short interfering RNA (siRNA) delivered through lipid-modified polyethyleneimine (PEI) polymers. Reduction of integrin-ß1 in K562 cells decreased cell adhesion towards human bone marrow stromal cells and to fibronectin, a major extracellular matrix protein for which integrin-ß1 is a primary receptor. Interaction of K562 cells with fibronectin decreased the sensitivity of the cells to BCR-ABL siRNA treatment, but a combinational treatment with integrin-ß1 and BCR-ABL siRNAs significantly reduced colony forming ability of the cells. Moreover, integrin-ß1 silencing enhanced the detachment of K562 cells from hBMSC samples (2 out of 4 samples), which could make them more susceptible to TKIs. Therefore, the polymeric-siRNA delivery targeting integrin-ß1 could be beneficial to reduce interactions with bone marrow microenvironment, aiding in the response of CML cells to therapeutic treatment.

8.
J Control Release ; 326: 335-349, 2020 10 10.
Article in English | MEDLINE | ID: mdl-32682900

ABSTRACT

Tumor Necrosis Factor (TNF) Related Apoptosis-Inducing Ligand (TRAIL), an immune cytokine of TNF-family, has received much attention in late 1990s as a potential cancer therapeutics due to its selective ability to induce apoptosis in cancer cells. TRAIL binds to cell surface death receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5) and facilitates formation of death-inducing signaling complex (DISC), eventually activating the p53-independent apoptotic cascade. This unique mechanism makes the TRAIL a potential anticancer therapeutic especially for p53-mutated tumors. However, recombinant human TRAIL protein (rhTRAIL) and TRAIL-R agonist monoclonal antibodies (mAb) failed to exert robust anticancer activities due to inherent and/or acquired resistance, poor pharmacokinetics and weak potencies for apoptosis induction. To get TRAIL back on track as a cancer therapeutic, multiple strategies including protein modification, combinatorial approach and TRAIL gene therapy are being extensively explored. These strategies aim to enhance the half-life and bioavailability of TRAIL and synergize with TRAIL action ultimately sensitizing the resistant and non-responsive cells. We summarize emerging strategies for enhanced TRAIL therapy in this review and cover a wide range of recent technologies that will provide impetus to rejuvenate the TRAIL therapeutics in the clinical realm.


Subject(s)
Neoplasms , Receptors, TNF-Related Apoptosis-Inducing Ligand , Apoptosis , Apoptosis Regulatory Proteins , Cell Death , Cell Line, Tumor , Humans , Neoplasms/drug therapy , Prospective Studies , TNF-Related Apoptosis-Inducing Ligand
9.
Stem Cells Dev ; 28(11): 734-744, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30585758

ABSTRACT

Nonviral gene therapy with specific short interfering RNAs (siRNAs) against BCR-Abl can be an alternative and/or supportive therapy of chronic myeloid leukemia (CML) with tyrosine kinase inhibitors (TKIs), given the often observed resistance to TKIs in clinical setting. In this study, we explored the feasibility of BCR-Abl siRNA therapy in CML K562 cells in vitro by employing a cationic polymer derived from cholesterol (Chol) grafted low-molecular weight polyethyleneimine (PEI). The first generation TKI imatinib upregulated the expression of BCR-Abl in K562 cells as expected. Delivery of BCR-Abl siRNA in both drug-sensitive and drug-resistant K562 cells significantly downregulated the mRNA levels in both cell types. Similarly, the BCR-Abl siRNA treatment arrested the growth of both drug-sensitive and drug-resistant K562 cells with no obvious differences despite a large difference in drug responsiveness. The BCR-Abl gene silencing in combination with TKI treatments exhibited significant synergism in drug-resistant K562 cells in generating substantial antileukemic activity, where the TKIs on their own were not effective. The effect of BCR-Abl siRNA and TKIs on non-CML cells (Jurkat and primary fibroblast) was negligible, indicating the specificity of the proposed therapy. This strategy can significantly overcome TKI resistance in CML cells, suggesting a feasible and effective treatment model for CML patients suffering from clinical resistances.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Genes, abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Cell Line, Tumor , Fusion Proteins, bcr-abl/genetics , Genetic Therapy/methods , Humans , Imatinib Mesylate/pharmacology , Jurkat Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , RNA Interference
10.
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
11.
J Control Release ; 310: 141-154, 2019 09 28.
Article in English | MEDLINE | ID: mdl-31430499

ABSTRACT

Despite development of effective tyrosine kinase inhibitors for treatment of chronic myeloid leukemia (CML), some patients do not effectively respond to the therapy and can display resistance in response to the drug therapy. To develop an alternative approach to CML therapy, we are exploring siRNA mediated silencing of the primary CML oncogene, BCR-ABL, by using non-viral (polymeric) delivery systems. In this study, a group of lipopolymers derived from low molecular PEIs substituted with linoleic acid (LA), α-linolenic acid (αLA) and cholesterol (Chol) was investigated for the first time for siRNA delivery to CML primary samples. The delivery efficiency in primary cells was equivalent to CML K562 cell line, and the lipopolymers gave effective internalization of siRNA depending on the nature of lipid substituent. The PEI-αLA (2.5 αLA/PEI), PEI-Chol (2.2 Chol/PEI), and PEI-LA (2.6 LA/PEI) lipopolymers used as BCR-ABL siRNA carriers (at 60 nM siRNA) reduced the BCR-ABL mRNA expression by 17% to 45%, and inhibited the formation of colonies by 24% to 41% in comparison with control siRNA in mononuclear cells. BCR-ABL siRNA treatment reduced the BCR-ABL mRNA expression by 50% in one of two CD34+ samples tested, and combination of BCR-ABL siRNA with imatinib (IM) treatment decreased the colony formation by 65% in one of two samples evaluated. The fact that no single polymer was universally effective in all patient samples may suggest patient-to-patient variability in terms of therapeutic responses to siRNA therapy. These results showed that a low dose of BCR-ABL siRNA could be used with lipopolymers to reduce BCR-ABL mRNA expression, CML cell survival and colony formation. This proof of principle study in CML primary cells can be applied to silencing of other therapeutic targets besides BCR-ABL and a study with larger patient samples is warranted for better identification of effective siRNA carriers.


Subject(s)
Drug Carriers/chemistry , Fusion Proteins, bcr-abl/genetics , Gene Silencing , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Lipids/chemistry , Polyethyleneimine/chemistry , RNA, Small Interfering , Cell Culture Techniques , Cell Line, Tumor , Cholesterol/chemistry , Female , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Microscopy, Confocal , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , alpha-Linolenic Acid/chemistry
12.
J Nanobiotechnology ; 6: 1, 2008 Jan 04.
Article in English | MEDLINE | ID: mdl-18173857

ABSTRACT

This project involved the synthesis of N-hexanoyl chitosan or simply modified chitosan (MC) stabilized iron oxide nanoparticles (MC-IOPs) and the biological evaluation of MC-IOPs. IOPs containing MC were prepared using conventional methods, and the extent of cell uptake was evaluated using mouse macrophages cell line (RAW cells). MC-IOPs were found to rapidly associate with the RAW cells, and saturation was typically reached within the 24 h of incubation at 37 degrees C. Nearly 8.53 +/- 0.31 pg iron/cell were bound or internalized at saturation. From these results, we conclude that MC-IOPs effectively deliver into RAW cells in vitro and we also hope MC-IOPs can be used for MRI enhancing agents in biomedical fields.

13.
Macromol Biosci ; 17(6)2017 06.
Article in English | MEDLINE | ID: mdl-28160423

ABSTRACT

Cell surface integrins, which play important roles in the survival, proliferation, migration, and invasion of cancer cells, are a viable target for treatment of metastatic breast cancer. This line of therapy still remains challenging due to the lack of proper identification and validation of effective targets as well as the lack of suitable therapeutic agents for treatment. The focus is on one such molecular target for this purpose, namely integrin-ß1, and effective lowering of integrin-ß1 levels on a breast cancer model (MDA-MB-231 cells) is achieved by delivering a dicer-substrate short interfering RNA (siRNA) targeting integrin-ß1 with lipid-modified low molecular weight polyethylenimine polymers. Reduction of integrin-ß1 levels leads to reduced adhesion of MDA-MB-231 cells to extracellular matrix component fibronectin as well as to human bone marrow cells. A reduced migration of the breast cancer cells is also observed after integrin-ß1 silencing in "scratch" and "transwell" migration assays. These results highlight the importance of integrin-ß1 for the migration of metastatic breast cancer cells by effectively silencing this target with a practical dose of siRNA.


Subject(s)
Breast Neoplasms/therapy , Gene Transfer Techniques , Integrin beta1/genetics , RNA, Small Interfering/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Gene Silencing , Humans , Neoplasm Metastasis , Polyethyleneimine/chemistry , Polyethyleneimine/therapeutic use , RNA, Small Interfering/therapeutic use
14.
Nanomedicine (Lond) ; 11(9): 1123-38, 2016 May.
Article in English | MEDLINE | ID: mdl-27074102

ABSTRACT

AIM: This study aimed to create fibronectin (FN)-grafted polymeric surfaces to investigate the influence of leukemic cell adhesion on siRNA treatment. MATERIALS & METHODS: FN was grafted on plasma-treated PTFE surfaces using chemical crosslinkers. Adhesion and growth of chronic myeloid leukemia K562 cells on modified surfaces were investigated. The silencing effect of siRNA/lipid-polymers nanoparticles on cells grown on FN-grafted surfaces was evaluated. RESULTS: Crosslinker-mediated immobilization showed significant FN grafting on surfaces, which provided K562 cell adhesion and growth advantage. siRNA nanoparticle silencing was similarly effective on FN-adhered and suspension-growing K562 cells. CONCLUSION: This study provided initial data to develop a cell-adhesive system to investigate therapeutic effects on leukemic cells. The response of chronic myeloid leukemia cells to siRNA nanoparticles was independent on cell attachment.


Subject(s)
Fibronectins/genetics , Gene Transfer Techniques , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , RNA, Small Interfering/genetics , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Cell Adhesion/genetics , Fibronectins/administration & dosage , Fibronectins/chemistry , Genetic Therapy , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Nanoparticles/administration & dosage , Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage , Surface Properties
15.
J Mater Chem B ; 3(19): 3972-3982, 2015 May 21.
Article in English | MEDLINE | ID: mdl-32262619

ABSTRACT

It has been challenging to modify primary cells with non-viral gene delivery. Herein, we developed a ternary nano-formulation for gene delivery to umbilical cord blood and bone marrow derived mesenchymal stem cells (MSC) by using lipid-modified small (1.2 kDa) molecular weight polyethylenimine (PEI1.2). Linoleic acid (LA) was end-capped with carboxyl functionality by coupling with mercaptopropionic acid through thio-ester linkage, and then grafted onto PEI1.2 via N-acylation. The thio-ester LA grafted PEI1.2 (PEI-tLA) displayed a significantly lower (up to 6-fold) DNA binding capability and a higher propensity to dissociate upon polyanionic challenge. The dissociation ability of the complexes was further enhanced by incorporating hyaluronic acid (HA) into plasmid DNA (pDNA) complexes of PEI-tLA. The HA incorporation influenced the surface charge of complexes more so than the hydrodynamic size, but it clearly increased the propensity for dissociation upon a polyanionic challenge. The PEI-tLAs were less toxic on MSC and displayed significantly higher transgene expression in MSC than conventional PEI-LA. Ternary complexes of with HA (pDNA/HA = 2, w/w) further enhanced the efficiency of PEI-tLAs of low (∼2 lipid/PEI) lipid substitution, which was comparable to or higher than commercial transfection reagents. We conclude that PEI-tLA of low lipid substitution can be employed as a gene carrier to design supersensitive nano-formulations.

16.
ACS Appl Mater Interfaces ; 7(44): 24822-32, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26493098

ABSTRACT

High molecular weight (HMW) polyethylenimine (PEI) is one of the most versatile nonviral gene vectors that was extensively investigated over the past two decades. The cytotoxic profile of HMW PEI, however, encouraged a search for safer alternatives. Because of lack of cytotoxicity of low molecular weight (LMW) PEI, enhancing its performance via hydrophobic modifications has been pursued to this end. Since the performance of modified PEIs depends on the nature and extent of substituents, we systematically investigated the effect of hydrophobic modification of LMW (1.2 kDa) PEI with a short propionic acid (PrA). Moderate enhancements in PEI hydrophobicity resulted in enhanced cellular uptake of polyplexes and siRNA-induced silencing efficacy, whereas further increase in PrA substitution abolished the uptake as well as the silencing. We performed all-atom molecular dynamics simulations to elucidate the mechanistic details behind these observations. A new assembly mechanism was observed by the presence of hydrophobic PrA moieties, where PrA migrated to core of the polyplex. This phenomenon caused higher surface hydrophobicity and surface charge density at low substitutions, and it caused deleterious effects on surface hydrophobicity and cationic charge at higher substitutions. It is evident that an optimal balance of hydrophobicity/hydrophilicity is needed to achieve the desired polyplex properties for an efficient siRNA delivery, and our mechanistic findings should provide valuable insights for the design of improved substituents on nonviral carriers.


Subject(s)
Genetic Vectors , Nucleic Acids/chemistry , Polyethyleneimine/chemistry , Carbon/chemistry , Computer Simulation , Gene Silencing , Gene Transfer Techniques , Green Fluorescent Proteins/chemistry , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , K562 Cells , Molecular Dynamics Simulation , Molecular Weight , Propionates/chemistry , Protein Engineering/methods , RNA, Small Interfering/metabolism , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL