Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 242
Filter
Add more filters

Publication year range
1.
J Nanobiotechnology ; 22(1): 463, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095888

ABSTRACT

Neurodegenerative disorders are complex, progressive, and life-threatening. They cause mortality and disability for millions of people worldwide. Appropriate treatment for neurodegenerative diseases (NDs) is still clinically lacking due to the presence of the blood-brain barrier (BBB). Developing an effective transport system that can cross the BBB and enhance the therapeutic effect of neuroprotective agents has been a major challenge for NDs. Exosomes are endogenous nano-sized vesicles that naturally carry biomolecular cargoes. Many studies have indicated that exosome content, particularly microRNAs (miRNAs), possess biological activities by targeting several signaling pathways involved in apoptosis, inflammation, autophagy, and oxidative stress. Exosome content can influence cellular function in healthy or pathological ways. Furthermore, since exosomes reflect the features of the parental cells, their cargoes offer opportunities for early diagnosis and therapeutic intervention of diseases. Exosomes have unique characteristics that make them ideal for delivering drugs directly to the brain. These characteristics include the ability to pass through the BBB, biocompatibility, stability, and innate targeting properties. This review emphasizes the role of exosomes in alleviating NDs and discusses the associated signaling pathways and molecular mechanisms. Furthermore, the unique biological features of exosomes, making them a promising natural transporter for delivering various medications to the brain to combat several NDs, are also discussed.


Subject(s)
Blood-Brain Barrier , Drug Delivery Systems , Exosomes , Neurodegenerative Diseases , Exosomes/metabolism , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Animals , Drug Delivery Systems/methods , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , MicroRNAs/metabolism , Signal Transduction/drug effects , Drug Carriers/chemistry
2.
Pharm Res ; 39(2): 353-367, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35166995

ABSTRACT

PURPOSE: The invention and application of new immunotherapeutic methods can compensate for the inefficiency of conventional cancer treatment approaches, partly due to the inhibitory microenvironment of the tumor. In this study, we tried to inhibit the growth of cancer cells and induce anti-tumor immune responses by silencing the expression of the ß-catenin in the tumor microenvironment and transmitting interleukin (IL)-15 cytokine to provide optimal conditions for the dendritic cell (DC) vaccine. METHODS: For this purpose, we used folic acid (FA)-conjugated SPION-carboxymethyl dextran (CMD) chitosan (C) nanoparticles (NPs) to deliver anti-ß-catenin siRNA and IL-15 to cancer cells. RESULTS: The results showed that the codelivery of ß-catenin siRNA and IL-15 significantly reduced the growth of cancer cells and increased the immune response. The treatment also considerably stimulated the performance of the DC vaccine in triggering anti-tumor immunity, which inhibited tumor development and increased survival in mice in two different cancer models. CONCLUSIONS: These findings suggest that the use of new nanocarriers such as SPION-C-CMD-FA could be an effective way to use as a novel combination therapy consisting of ß-catenin siRNA, IL-15, and DC vaccine to treat cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Cancer Vaccines/administration & dosage , Dendritic Cells/transplantation , Drug Carriers , Interleukin-15/administration & dosage , Magnetic Iron Oxide Nanoparticles , Melanoma, Experimental/therapy , RNA, Small Interfering/administration & dosage , RNAi Therapeutics , Skin Neoplasms/therapy , beta Catenin/genetics , Animals , Antineoplastic Agents/chemistry , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Dendritic Cells/immunology , Drug Compounding , Female , Gene Expression Regulation, Neoplastic , Interleukin-15/chemistry , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred BALB C , RNA, Small Interfering/genetics , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Burden/drug effects , Tumor Microenvironment
3.
J Drug Deliv Sci Technol ; 67: 102899, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34630635

ABSTRACT

The inexorable coronavirus disease 2019 (COVID-19) pandemic with around 226 million people diagnosed and approximately 4.6 million deaths, is still moving toward more frightening statistics, calling for the urgent need to explore solutions for the current challenges in therapeutic and diagnostic approaches. The challenges associated with existing therapeutics in COVID-19 include lack of in vivo stability, efficacy, and safety. Nanoparticles (NPs) can offer a handful of tools to tackle these problems by enabling efficacious and safe delivery of both virus- and host-directed therapeutics. Furthermore, they can enable maximized clinical outcome while eliminating the chance of resistance to therapy by tissue-targeting and concomitant delivery of multiple therapeutics. The promising application of NPs as vaccine platforms is reflected by the major advances in developing novel COVID-19 vaccines. Two of the most critical COVID-19 vaccines are mRNA-based vaccines delivered via NPs, making them the first FDA-approved mRNA vaccines. Besides, NPs have been deployed as simple, rapid, and precise tools for point of care disease diagnosis. Not enough said NPs can also be exploited in novel ways to expedite the drug discovery process. In light of the above, this review discusses how NPs can overcome the hurdles associated with therapeutic and diagnostic approaches against COVID-19.

4.
Cancer Cell Int ; 21(1): 204, 2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33849536

ABSTRACT

BACKGROUND: Relapse and metastasis in colorectal cancer (CRC) are often attributed to cancer stem-like cells (CSCs), as small sub-population of tumor cells with ability of drug resistance. Accordingly, development of appropriate models to investigate CSCs biology and establishment of effective therapeutic strategies is warranted. Hence, we aimed to assess the capability of two widely used and important colorectal cancer cell lines, HT-29 and Caco-2, in generating spheroids and their detailed morphological and molecular characteristics. METHODS: CRC spheroids were developed using hanging drop and forced floating in serum-free and non-attachment conditions and their morphological features were evaluated by scanning electron microscopy (SEM). Then, the potential of CSCs enrichment in spheroids was compared to their adherent counterparts by analysis of serial sphere formation capacity, real-time PCR of key stemness genes (KLF4, OCT4, SOX2, NANOG, C-MYC) and the expression of potential CRC-CSCs surface markers (CD166, CD44, and CD133) by flow cytometry. Finally, the expression level of some EMT-related (Vimentin, SNAIL1, TWIST1, N-cadherin, E-cadherin, ZEB1) and multi-drug resistant (ABCB1, ABCC1, ABCG2) genes was evaluated. RESULTS: Although with different morphological features, both cell lines were formed CSCs-enriched spheroids, indicated by ability to serial sphere formation, significant up-regulation of stemness genes, SOX2, C-MYC, NANOG and OCT4 in HT-29 and SOX2, C-MYC and KLF4 in Caco-2 spheroids (p-value < 0.05) and increased expression of CRC-CSC markers compared to parental cells (p-value < 0.05). Additionally, HT-29 spheroids exhibited a significant higher expression of both ABCB1 and ABCG2 (p-value = 0.02). The significant up-regulation of promoting EMT genes, ZEB1, TWIST1, E-cadherin and SNAIL1 in HT-29 spheroids (p-value = 0.03), SNAIL1 and Vimentin in Caco-2 spheroids (p-value < 0.05) and N-cadherin down-regulation in both spheroids were observed. CONCLUSION: Enrichment of CSC-related features in HT-29 and Caco-2 (for the first time without applying special scaffold/biochemical) spheroids, suggests spheroid culture as robust, reproducible, simple and cost-effective model to imitate the complexity of in vivo tumors including self-renewal, drug resistance and invasion for in vitro research of CRC-CSCs.

5.
Exp Eye Res ; 204: 108423, 2021 03.
Article in English | MEDLINE | ID: mdl-33453276

ABSTRACT

Retinoblastoma (Rb) is the most common intraocular malignancy in children that accounts for approximately 4% of all pediatric malignancies. Since chemotherapy is a widely practiced treatment for Rb, there is a growing interest in developing new and effective drugs to overcome systemic and local side effects of chemotherapy to improve the quality of life and increase the chances of survival. This study sought to fabricate thiolated chitosan nanoparticles containing topotecan (TPH-TCs-NPs) with a view of enhancing drug loading and release control. This research was also designed to assess the ability of TPH-TCs-NPs to improve cell association, increase treatment efficacy in retinoblastoma cells and xenograft-rat-model of retinoblastoma, and overcome current topotecan hydrochloride (TPH) intravitreal administration challenges, including stability loss and poor cellular uptake. Modified ionic gelation method was optimized to fabricate TPH-TCs-NPs and TPH-TMC-NPs (N-trimethyl chitosan nanoparticles containing TPH). We characterized the NPs and quantified topotecan loading and release against a free TPH standard. The efficacy of TPH-NPs was quantified in human retinoblastoma cells (Y79) by XTT and flow cytometry measurement. In addition, Y79 cells were injected intravitreally in both eyes of immunodeficient wistar albino rats to create a xenograft-rat-model to compare the antitumor effectiveness of TPH-NPs and TPH by intravitreal administration. TPH-NPs complexation was confirmed by EDX, FTIR, and DSC techniques. TPH-TCs-NPs and TPH-TMC-NPs had high encapsulation efficiency (85.23 ± 2 and 73.34 ± 2% respectively). TPH-TCs-NPs showed a mean diameter, polidispersity index, and zeta potential of 25±2 nm, 0.21 ± 0.03 and +12 ± 2 mV, respectively. As a function of dose, TCs and TMC NPs were more efficacious than free topotecan (IC50s 53.17 and 85.88 nM, relative to 138.30 nM respectively, P = 0.012). Kruskal-Wallis test showed a statistically significant difference between the groups. Additionally, a significant difference between the tumor control and TPH-TCs-NPs treated group in xenograft-rat-model ( Range of P-value: 0.026 to 0.035) was shown by Bonferroni post hoc test. The current investigation demonstrated enhanced efficacy and association of TPH-TCs-NPs relative to free TPH in retinoblastoma cells and tumor in vitro and in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Chitosan/administration & dosage , Drug Carriers , Retinal Neoplasms/drug therapy , Retinoblastoma/drug therapy , Topoisomerase I Inhibitors/administration & dosage , Topotecan/administration & dosage , Animals , Calorimetry, Differential Scanning , Chitosan/chemistry , Flow Cytometry , Humans , Intravitreal Injections , Male , Nanoparticles , Neoplasm Transplantation , Particle Size , Rats , Rats, Wistar , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Spectrometry, X-Ray Emission , Spectroscopy, Fourier Transform Infrared , Transplantation, Heterologous , Treatment Outcome , Tumor Cells, Cultured , X-Ray Diffraction
6.
Pharm Dev Technol ; 26(4): 381-389, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33538232

ABSTRACT

Targeted drug delivery is a tool to make treatment more specific, selective, and effective and to prevent unwanted complications. Prostate specific membrane antigen (PSMA) is a useful biomarker in order to monitor and control prostate cancer. Glutamate-Urea-R (Glu-Urea-R) is a PSMA enzyme inhibitor capable of binding to this surface marker of prostate cancer cell in an efficient and special manner. The aim of this project was to develop a docetaxel-loaded nanoparticle of poly (lactic-co-glycolic acid) polyethylene glycol which is cojugated to a urea-based anti-PSMA ligand named glutamate-urea-lysine (glu-urea-lys) for targeted delivery of docetaxel in prostate cancer. The obtained nanoparticles, prepared by nanoprecipitation method, were spheres with a particle size of around 150 nm and zeta potential of -7.08 mV. Uptake studies on the PC3 (as PSMA negative) and LNCaP (as PSMA positive) cells demonstrated that drug uptake was efficient by the PSMA positive cells. IC50 of targeted NPs on LNCaP cell line compared to non-targeted ones was reduced by more than 70% in three different incubation times of 24, 48, and 72 h. In conclusion, the nanoparticles are expected to specifically transport docetaxel to PSMA-positive prostate cancer cells and consequently, enhance the antitumor efficacy of docetaxel on these cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Docetaxel/administration & dosage , Nanoparticles , Prostatic Neoplasms/drug therapy , Antigens, Surface/metabolism , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Docetaxel/pharmacology , Drug Carriers/chemistry , Drug Delivery Systems , Glutamate Carboxypeptidase II/metabolism , Glutamic Acid/chemistry , Humans , Inhibitory Concentration 50 , Male , Particle Size , Polyethylene Glycols/chemistry , Polyglactin 910/chemistry , Prostatic Neoplasms/pathology , Time Factors , Urea/chemistry
7.
J Cell Physiol ; 235(3): 2049-2059, 2020 03.
Article in English | MEDLINE | ID: mdl-31364167

ABSTRACT

RNA interference is one of the prosperous approaches for cancer treatment. However, small interfering RNA (siRNA) delivery to cancer cells has been faced with various challenges restricting their clinical application over the decades. Since ROR1 is an onco-embryonic gene overexpressed in many malignancies, suppression of ROR1 by siRNA can potentially fight cancer. Herein, a delivery system for ROR1 siRNA based on HIV-1 TAT peptide-capped gold nanoparticles (GNPs) was developed to treat breast cancer. Besides, we introduced a new feasible method for conjugating the peptide to the nanoparticles. Since the GNPs have high affinity to the sulfur, the findings demonstrated the peptide successfully conjugated to the nanoparticles via Au-S bonds. As positively charged nanoparticles showed high cellular uptake, we could use a low concentration of nanoparticles led to high efficient gene transfection with negligible cytotoxicity that was confirmed by flow cytometry, confocal microscopy, gel retardation, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Following transfection, downregulation of ROR1 and its targeted gene, CCND1, induced apoptosis in cancer cells. In conclusion, the reported capped GNPs could be potentially utilized for delivering negatively charged therapeutic agents in particular genes.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , tat Gene Products, Human Immunodeficiency Virus/chemistry , Apoptosis/genetics , Cell Line, Tumor , Cyclin D1/genetics , Gene Transfer Techniques , HIV-1/metabolism , Humans , Immobilization/physiology , Transfection/methods
8.
J Cell Physiol ; 235(12): 10068-10080, 2020 12.
Article in English | MEDLINE | ID: mdl-32488862

ABSTRACT

Inhibitory immune checkpoint (ICP) molecules are important immunosuppressive factors in a tumor microenvironment (TME). They can robustly suppress T-cell-mediated antitumor immune responses leading to cancer progression. Among the checkpoint molecules, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) is one of the critical inhibitors of anticancer T-cell responses. Besides, the expression of adenosine receptor (A2AR) on tumor-infiltrating T cells potently reduces their function. We hypothesized that concomitant silencing of these molecules in T cells might lead to enhanced antitumor responses. To examine this assumption, we purified T cells from the tumor, spleen, and local lymph nodes of CT26 colon cancer-bearing mice and suppressed the expression of A2AR and CTLA-4 using the small interfering RNA (siRNA)-loaded polyethylene glycol-chitosan-alginate (PCA) nanoparticles. The appropriate physicochemical properties of the produced nanoparticles (NPs; size of 72 nm, polydispersive index [PDI] < 0.2, and zeta potential of 11 mV) resulted in their high efficiency in transfection and suppression of target gene expression. Following the silencing of checkpoint molecules, various T-cell functions, including proliferation, apoptosis, cytokine secretion, differentiation, and cytotoxicity were analyzed, ex vivo. The results showed that the generated nanoparticles had optimal physicochemical characteristics and significantly suppressed the expression of target molecules in T cells. Moreover, a concomitant blockade of A2AR and CTLA-4 in T cells could synergistically enhance antitumor responses through the downregulation of PKA, SHP2, and PP2Aα signaling pathways. Therefore, this combination therapy can be considered as a novel promising anticancer therapeutic strategy, which should be further investigated in subsequent studies.


Subject(s)
CTLA-4 Antigen/genetics , Colonic Neoplasms/therapy , Nanoparticles/chemistry , Receptor, Adenosine A2A/genetics , Alginates/chemistry , Animals , CD8-Positive T-Lymphocytes/drug effects , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Chitosan/chemistry , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Polyethylene Glycols/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Microenvironment/drug effects
9.
J Cell Physiol ; 235(12): 9702-9717, 2020 12.
Article in English | MEDLINE | ID: mdl-32424937

ABSTRACT

There is an interconnected network between S1P/sphingosine-1-phosphate receptor 1 (S1PR1), IL-6/glycoprotein 130 (GP130), and signal transducer and activator of transcription 3 (STAT3) signaling pathways in the tumor microenvironment, which leads to cancer progression. S1P/S1PR1 and IL-6/GP130 signaling pathways phosphorylate and activate STAT3, and it then induces the expression of S1PR1 and interleukin-6 (IL-6) in a positive feedback loop leading to cancer progression. We hypothesized that blockade of this amplification loop can suppress the growth and development of cancer cells. Therefore, we silenced STAT3 upstream molecules including the S1PR1 and GP130 molecules in cancer cells using small interfering RNA (siRNA)-loaded alginate-conjugated trimethyl chitosan (ATMC) nanoparticles (NPs). The generated NPs had competent properties including the appropriate size, zeta potential, polydispersity index, morphology, high uptake of siRNA, high rate of capacity, high stability, and low toxicity. We evaluated the effects of siRNA loaded ATMC NPs on tumor hallmarks of three murine-derived cancer cell lines, including 4T1 (breast cancer), B16-F10 (melanoma), and CT26 (colon cancer). The results confirmed the tumor-suppressive effects of combinational targeting of S1PR1 and GP130. Moreover, combination therapy could potently suppress tumor growth as assessed by the chick chorioallantoic membrane assay. In this study, we targeted this positive feedback loop for the first time and applied this novel combination therapy, which provides a promising approach for cancer treatment. The development of a potent nanocarrier system with ATMC for this combination was also another aspect of this study, which should be further investigated in cancer animal models in further studies.


Subject(s)
Breast Neoplasms/genetics , Cytokine Receptor gp130/genetics , Melanoma, Experimental/genetics , RNA, Small Interfering/pharmacology , Sphingosine-1-Phosphate Receptors/genetics , Animals , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Chitosan/chemistry , Chitosan/pharmacology , Cytokine Receptor gp130/antagonists & inhibitors , Drug Delivery Systems , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/genetics , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Nanoparticles/chemistry , Proprotein Convertases/genetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , STAT3 Transcription Factor/genetics , Serine Endopeptidases/genetics , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Tumor Microenvironment/drug effects
10.
Immunology ; 159(1): 75-87, 2020 01.
Article in English | MEDLINE | ID: mdl-31587253

ABSTRACT

Dendritic cell (DC) -based cancer immunotherapy is one of the most important anti-cancer immunotherapies, and has been associated with variable efficiencies in different cancer types. It is well-known that tumor microenvironment plays a key role in the efficacy of various immunotherapies such as DC vaccine. Accordingly, the expression of programmed death ligand 1 (PD-L1) on DCs, which interacts with PD-1 on T cells, leads to inhibition of anti-tumor responses following presentation of tumor antigens by DCs to T cells. Therefore, we hypothesized that down-regulation of PD-L1 in DCs in association with silencing of PD-1 on T cells may lead to the enhancement of T-cell priming by DCs to have efficient anti-tumor T-cell responses. In this study, we silenced the expression of PD-L1 in DCs and programmed cell death protein 1 (PD-1) in T cells by small interfering RNA (siRNA) -loaded chitosan-dextran sulfate nanoparticles (NPs) and evaluated the DC phenotypic and functional characteristics and T-cell functions following tumor antigen recognition on DCs, ex vivo. Our results showed that synthesized NPs had good physicochemical characteristics (size 77·5 nm and zeta potential of 14·3) that were associated with efficient cellular uptake and target gene silencing. Moreover, PD-L1 silencing was associated with stimulatory characteristics of DCs. On the other hand, presentation of tumor antigens by PD-L1-negative DCs to PD-1-silenced T cells led to induction of potent T-cell responses. Our findings imply that PD-L1-silenced DCs can be considered as a potent immunotherapeutic approach in combination with PD-1-siRNA loaded NPs, however; further in vivo investigation is required in animal models.


Subject(s)
B7-H1 Antigen/immunology , Breast Neoplasms/therapy , Cancer Vaccines/immunology , Colonic Neoplasms/therapy , Dendritic Cells/transplantation , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/immunology , RNAi Therapeutics , T-Lymphocytes/immunology , Animals , Apoptosis , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Coculture Techniques , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred BALB C , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction , T-Lymphocytes/metabolism
11.
Pharm Res ; 37(10): 196, 2020 Sep 17.
Article in English | MEDLINE | ID: mdl-32944844

ABSTRACT

PURPOSE: Hypoxia-inducible factor (HIF) is one of the critical components of the tumor microenvironment that is involved in tumor development. HIF-1α functionally and physically interacts with CDK1, 2, and 5 and stimulates the cell cycle progression and Cyclin-Dependent Kinase (CDK) expression. Therefore, hypoxic tumor microenvironment and CDK overexpression lead to increased cell cycle progression and tumor expansion. Therefore, we decided to suppress cancer cell expansion by blocking HIF-1α and CDK molecules. METHODS: In the present study, we used the carboxylated graphene oxide (CGO) conjugated with trimethyl chitosan (TMC) and hyaluronate (HA) nanoparticles (NPs) loaded with HIF-1α-siRNA and Dinaciclib, the CDK inhibitor, for silencing HIF-1α and blockade of CDKs in CD44-expressing cancer cells and evaluated the impact of combination therapy on proliferation, metastasis, apoptosis, and tumor growth. RESULTS: The results indicated that the manufactured NPs had conceivable physicochemical properties, high cellular uptake, and low toxicity. Moreover, combination therapy of cancer cells using CGO-TMC-HA NPs loaded with HIF-1α siRNA and Dinaciclib (SCH 727965) significantly suppressed the CDKs/HIF-1α and consequently, decreased the proliferation, migration, angiogenesis, and colony formation in tumor cells. CONCLUSIONS: These results indicate the ability of CGO-TMC-HA NPs for dual drug/gene delivery in cancer treatment. Furthermore, the simultaneous inhibition of CDKs/HIF-1α can be considered as a novel anti-cancer treatment strategy; however, further research is needed to confirm this treatment in vivo. Graphical Abstract The suppression of HIF-1α and CDKs inhibits cancer growth. HIF-1α is overexpressed by the cells present in the tumor microenvironment. The hypoxic environment elevates mitochondrial ROS production and increases p38 MAP kinase, JAK/STAT, ERK, JNK, and Akt/PI3K signaling, resulting in cyclin accumulation and aberrant cell cycle progression. Furthermore, the overexpression of HIF-1α/CDK results in increased expression of genes such as BCL2, Bcl-xl, Ki-67, TGFß, VEGF, FGF, MMP2, MMP9, and, HIF-1α and consequently raise the survival, proliferation, angiogenesis, metastasis, and invasion of tumor cells. In conclusion, HIF-1α-siRNA/Dinaciclib-loaded CGO-TMC-HA NPs can inhibit the tumor expansion by blockage of CDKs and HIF-1α (JAK: Janus kinase, STAT: Signal transducer and activator of transcription, MAPK: mitogen-activated protein kinase, ERK: extracellular signal-regulated kinase, JNK: c-Jun N-terminal kinase, PI3K: phosphatidylinositol 3-kinase).


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasms, Experimental/therapy , Pyridinium Compounds/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chitosan/chemistry , Cyclic N-Oxides , Graphite/chemistry , Hyaluronic Acid/chemistry , Indolizines , Mice , Nanoparticles/chemistry , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pyridinium Compounds/chemistry , Pyridinium Compounds/pharmacokinetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics
12.
Nanomedicine ; 26: 102181, 2020 06.
Article in English | MEDLINE | ID: mdl-32169399

ABSTRACT

As vascular endothelial growth factor in choroidal neovascularization is a major cause of visual loss of the elderlies and diabetics, gene therapy may offer an alternative treatment. However, siRNA instability and inefficient delivery are the main hindrances. To address this issue, we developed a nano-sized siRNA loaded therapeutic delivery system. The chitosan-hyaluronic acid nano-polyplexes were prepared by the modified ionic gelation method. The obtained nano-polyplex with a narrow size distribution, indicated no significant cytotoxicity in the MTT test and proper cellular uptake in confocal images. The RT-PCR analysis indicated remarkable gene silencing on HUVEC cells. The intravitreally administered nano-polyplexes in rabbits overcame both the vitreous and retina barriers and reached the posterior tissues efficiently. Intravitreal injections of the VEGFR-2 siRNA nano-polyplexes significantly reduced the size of the laser-induced choroidal neovascularization, compared to the control group. Consequently, the developed formulation can be a promising candidate for intravitreal delivery of siRNA.


Subject(s)
Chitosan/pharmacology , Choroidal Neovascularization/drug therapy , RNA, Small Interfering/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Chitosan/chemistry , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Drug Compounding , Drug Delivery Systems , Human Umbilical Vein Endothelial Cells , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Intravitreal Injections , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
13.
Nanomedicine ; 29: 102240, 2020 10.
Article in English | MEDLINE | ID: mdl-32553948

ABSTRACT

Overexpression of adenosine in the tumor region leads to suppression of various immune cells, particularly T cells through ligation with adenosine 2a receptor (A2aR). In this study, we intended to increase the efficacy of tumor lysate-loaded DC vaccine by silencing the expression of A2aR on T cells through the application of A2aR-specific siRNA-loaded PEG-chitosan-lactate (PCL) nanoparticles (NPs) in the 4T1 breast tumor-bearing mice. Combination therapy by DC vaccine and siRNA-loaded NPs markedly induced tumor regression and increased survival time of mice. These ameliorative effects were partly via downregulation of immunosuppressive cells, increased function of cytotoxic T lymphocytes, and induction of immune-stimulatory cytokines. Moreover, combination therapy could markedly suppress angiogenesis and metastasis processes. These results imply the efficacy of novel combination therapy for the treatment of breast cancer by using A2aR siRNA-loaded NPs and DC vaccine which can be translated into the initial phase of clinical trials in the near future.


Subject(s)
Breast Neoplasms/therapy , Mammary Neoplasms, Animal/therapy , Nanoparticles/chemistry , Receptor, Adenosine A2A/genetics , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/pharmacology , Cell Line, Tumor , Chitosan/chemistry , Chitosan/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy , Lactic Acid/chemistry , Lactic Acid/pharmacology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
14.
J Cell Physiol ; 234(11): 20554-20565, 2019 11.
Article in English | MEDLINE | ID: mdl-31144311

ABSTRACT

Gene therapy, including small interfering RNA (siRNA) technology, is one of the leading strategies that help to improve the outcomes of the current therapeutic systems against HIV-1 infection. The successful therapeutic application of siRNAs requires their safe and efficient delivery to specific cells. Here, we introduce a superparamagnetic iron oxide nanoparticle (SPION) for delivering siRNA against HIV-1 nef (anti-nef siRNA) into two cell lines, HEK293 and macrophage RAW 264.7. SPIONs were coated with trimethyl chitosan (TMC), and thereafter, different concentrations of SPION-TMC were coated with different ratios of a carboxymethyl dextran (CMD) to modify the physicochemical properties and improve the biological properties of the nanocarriers. The nanoparticles exhibited a spherical shape with an average size of 112 nm. The obtained results showed that the designed delivery route enhanced the uptake of siRNA into both HEK293 and RAW 264.7 cells compared with control groups. Moreover, CMD-TMC-SPIONs containing anti-nef siRNA significantly reduced the expression of HIV-1 nef in HEK293 stable cells. The modified siRNA-loaded SPIONs also displayed no toxicity or apoptosis-inducing effects on the cells. The CMD-TMC-SPIONs are suggested as potential nanocarriers for siRNA delivery in gene therapy of HIV-1 infection.


Subject(s)
Chitosan/chemistry , Dextrans/chemistry , Ferric Compounds/chemistry , Gene Transfer Techniques , Metal Nanoparticles/chemistry , nef Gene Products, Human Immunodeficiency Virus/metabolism , Animals , HEK293 Cells , Humans , Mice , RAW 264.7 Cells , RNA, Small Interfering , nef Gene Products, Human Immunodeficiency Virus/genetics
15.
J Cell Physiol ; 234(4): 4702-4712, 2019 04.
Article in English | MEDLINE | ID: mdl-30191977

ABSTRACT

Chemical burns are a major cause of corneal haze and blindness. Corticosteroids are commonly used after corneal burns to attenuate the severity of the inflammation-related fibrosis. While research efforts have been aimed toward application of novel therapeutics. In the current study, a novel drug delivery system based nanostructured lipid carriers (NLCs) were designed to treat corneal alkaline burn injury. Rapamycin, a potent inhibitor of mammalian target of rapamycin pathway, was loaded in NLCs (rapa-NLCs), and the NLCs were characterized. Cell viability assay, cellular uptake of NLCs, and in vitro evaluation of the fibrotic/angiogenic genes suppression by rapa-NLCs were carried out on human isolated corneal fibroblasts. Immunohistochemistry (IHC) assays were also performed after treatment of murine model of corneal alkaline burn with rapa-NLCs. According to the results, rapamycin was efficiently loaded in NLCs. NLCs could enhance coumarin-6 fibroblast uptake by 1.5 times. Rapa-NLCs efficiently downregulated platelet-derived growth factor and transforming growth factor beta genes in vitro. Furthermore, proliferation of fibroblasts, a major cause of corneal haze after injury, reduced. IHC staining of treated cornea with alpha-smooth muscle actin and CD34 + antibodies showed efficient prevention of myofibroblasts differentiation and angiogenesis, respectively. In conclusion, ocular delivery of rapamycin using NLCs after corneal injury may be considered as a promising antifibrotic/angiogenic treatment approach to preserve patient eyesight.


Subject(s)
Burns, Chemical/drug therapy , Cell Proliferation/drug effects , Cornea/drug effects , Corneal Injuries/drug therapy , Corneal Opacity/drug therapy , Drug Carriers , Eye Burns/drug therapy , Fibroblasts/drug effects , Lipids/chemistry , Nanoparticles , Sirolimus/administration & dosage , Administration, Ophthalmic , Animals , Burns, Chemical/etiology , Burns, Chemical/metabolism , Burns, Chemical/pathology , Cells, Cultured , Cornea/metabolism , Cornea/pathology , Corneal Injuries/chemically induced , Corneal Injuries/metabolism , Corneal Injuries/pathology , Corneal Neovascularization/chemically induced , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Corneal Neovascularization/prevention & control , Corneal Opacity/chemically induced , Corneal Opacity/metabolism , Corneal Opacity/pathology , Disease Models, Animal , Drug Compounding , Eye Burns/chemically induced , Eye Burns/metabolism , Eye Burns/pathology , Fibroblasts/metabolism , Fibrosis , Humans , Male , Mice, Inbred BALB C , Nanomedicine , Sirolimus/chemistry , Sodium Hydroxide , Wound Healing/drug effects
16.
J Cell Physiol ; 233(10): 7165-7177, 2018 10.
Article in English | MEDLINE | ID: mdl-29741783

ABSTRACT

CD73 facilitates tumor growth by upregulation of the adenosine (immunosuppressive factor) in the tumor microenvironment, however, its precise molecular mechanisms is not precisely understood. Regarding the importance of angiogenesis in tumor development and spreading, we decided to assign the anti-angiogenic effects of CD73 suppression. We used chitosan lactate (ChLa) nanoparticles (NPs) to deliver CD73-specific small interfering RNA (siRNA) into cancer cells. Our results showed that treatment of the 4T1 cells with CD73-specific siRNA-loaded NPs led to potent inhibition of cancer cell proliferation and cell cycle arrest, in vitro. This growth arrest was correlated with downregulation of angiogenesis-related molecules including vascular endothelial growth factor (VEGF)-A, VEGF-R2, interleukin (IL)-6, and transforming growth factor (TGF)-ß. Moreover, administration of NPs loaded with CD73-siRNA into 4T1 breast cancer-bearing mice led to tumor regression and increased mice survival time accompanied with downregulation of angiogenesis (VEGF-A, VEGF-R2, VE-Cadherin, and CD31) and lymphangiogenesis (VEGF-C and LYVE-1)-related genes in the tumor site. Furthermore, the expression of angiogenesis promoting factors including IL-6, TGF-ß, signal transducer, and activator of transcription (STAT)3, hypoxia inducible factor (HIF)-1α, and cyclooxygenase (COX)2 was decreased after the CD73 suppression in mice. Moreover, analysis of leukocytes derived from the tumor samples, spleen, and regional lymph nodes showed that they had lower capability for secretion of angiogenesis promoting factors after CD73-silencing. These results indicate that suppression of tumor development by downregulation of CD73 is in part related to angiogenesis arrest. These findings imply a promising strategy for inhibiting tumor growth accompanied with suppressing the angiogenesis process.


Subject(s)
5'-Nucleotidase/genetics , Angiogenesis Inhibitors/pharmacology , Breast Neoplasms/genetics , Neovascularization, Pathologic/genetics , Animals , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , RNA, Small Interfering/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
18.
Nanomedicine ; 14(6): 1891-1903, 2018 08.
Article in English | MEDLINE | ID: mdl-29885900

ABSTRACT

Recent insights into the nanomedicine have revealed that nanoplatforms enhance the efficacy of carrier in therapeutic applications. Here, multifunctional nanoplatforms were utilized in miRNA-101 delivery and NIR thermal therapy to induce apoptosis in breast cancer cells. Au nanorods (NRs) or nanospheres (NSs) covered with graphene oxide (GO) were prepared and functionalized with polyethylene glycol as a stabilizer and poly-L-arginine (P-L-Arg) as a targeting agent. In nanoplatforms, coupling Au@GO prepared stable structures with higher NIR reactivity. P-L-Arg substantially enhanced the cellular uptake and gene retardation of stuffs coated by them. However, rod-shape nanoplatforms indicated better performance in cellular uptake and gene transfection than spherical ones. NIR thermal therapy was implemented to improve gene release and in synergy with miRNA-101 activated the apoptotic pathway and decreased the viability of breast cancer cell (<20%). Briefly, presented delivery systems are potentially efficient in distinguishing cancer cells, miRNA internalization and controlling apoptosis of cancer cells.


Subject(s)
Breast Neoplasms/therapy , Gold/chemistry , Graphite/chemistry , Hyperthermia, Induced , MicroRNAs/administration & dosage , Nanotubes , Phototherapy , Cell Proliferation , Combined Modality Therapy , Drug Delivery Systems , Female , Humans , MicroRNAs/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL