Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Adv Pharm Bull ; 14(2): 453-468, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39206396

RESUMO

Purpose: Receptor-mediated transcytosis (RMT) is a more specific, highly efficient, and reliable approach to crossing the blood-brain-barrier (BBB) and releasing the therapeutic cargos into the brain parenchyma. Methods: Here, we introduced and characterized a human/mouse-specific novel leptin-derived peptide using in silico, in vitro and in vivo experiments. Results: Based on the bioinformatics analysis and molecular dynamics (MD) simulation, a 14 amino acid peptide sequence (LDP 14) was introduced and its interaction with leptin-receptor (ObR) was analyzed in comparison with an well known leptin-derived peptide, Lep 30. MD simulation data revealed a significant stable interaction between ligand binding domains (LBD) of ObR with LDP 14. Analyses demonstrated suitable cellular uptake of LDP 14 alone and its derivatives (LDP 14-modified G4 PAMAM dendrimer and LDP 14-modified G4 PAMAM/pEGFP-N1 plasmid complexes) via ObR, energy and species dependent manner (preferred uptake by human/mouse cell lines compared to rat cell line). Importantly, our findings illustrated that the entry of LDP 14-modified dendrimers in hBCEC-D3 cells not only is not affected by protein corona (PC) formation, as the main reason for diminishing the cellular uptake, but also PC per se can enhance uptake rate. Finally, fluorescein labeled LDP 14-modified G4 PAMAM dendrimers efficiently accumulated in the mice brain with lower biodistribution in other organs, in our in vivo study. Conclusion: LDP 14 introduced as a novel and highly efficient ligand, which can be used for drugs/genes delivery to brain tissue in different central nervous system (CNS) disorders.

2.
Iran J Pharm Res ; 22(1): e136624, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38084295

RESUMO

Background: Breast cancer is a multifaceted disease characterized by genetic and epigenetic changes that lead to uncontrolled cell growth and metastasis. Early detection and treatment are crucial for managing diseases. Objectives: The objective of this study is to investigate the potential of chimeric peptides for drug delivery and to identify biomarkers associated with breast cancer. Recent studies have shown that the low-density lipoprotein receptor-related protein 1 (LRP-1) receptor has a significant impact on the development of breast cancer. In order to facilitate the identification of biomarkers, we have created a chimeric peptide that has been proven to bind successfully to the LRP-1 receptor. Methods: To identify biomarkers, we utilized advanced computational methods to conduct a meta-analysis of microarray data. Specifically, the g:Profiler and eXpression2Kinases (X2K) databases were utilized to identify gene ontologies and transcription factors. We then used the Human Protein Atlas to identify and assess crucial gene expressions. Results: Our results demonstrated that nucleolar and spindle-associated protein 1 (NUSAP1), melatonin receptor 1A (MELT), and cyclin-dependent kinase 1 (CDK1) are three hub genes that play pivotal roles in the pathogenesis of breast cancer. Conclusions: The research findings provide a deeper understanding of the molecular mechanisms involved in developing breast cancer. These findings have significant implications for developing novel therapies and diagnostics for this disease.

3.
Sci Rep ; 13(1): 17265, 2023 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-37828118

RESUMO

Ovarian cancer (OC) incidence and mortality rates continue to escalate globally. Early detection of OC is challenging due to extensive metastases and the ambiguity of biomarkers in advanced High-Grade Primary Tumors (HGPTs). In the present study, we conducted an in-depth in silico analysis in OC cell lines using the Gene Expression Omnibus (GEO) microarray dataset with 53 HGPT and 10 normal samples. Differentially-Expressed Genes (DEGs) were also identified by GEO2r. A variety of analyses, including gene set enrichment analysis (GSEA), ChIP enrichment analysis (ChEA), eXpression2Kinases (X2K) and Human Protein Atlas (HPA), elucidated signaling pathways, transcription factors (TFs), kinases, and proteome, respectively. Protein-Protein Interaction (PPI) networks were generated using STRING and Cytoscape, in which co-expression and hub genes were pinpointed by the cytoHubba plug-in. Validity of DEG analysis was achieved via Gene Expression Profiling Interactive Analysis (GEPIA). Of note, KIAA0101, RAD51AP1, FAM83D, CEP55, PRC1, CKS2, CDCA5, NUSAP1, ECT2, and TRIP13 were found as top 10 hub genes; SIN3A, VDR, TCF7L2, NFYA, and FOXM1 were detected as predominant TFs in HGPTs; CEP55, PRC1, CKS2, CDCA5, and NUSAP1 were identified as potential biomarkers from hub gene clustering. Further analysis indicated hsa-miR-215-5p, hsa-miR-193b-3p, and hsa-miR-192-5p as key miRNAs targeting HGPT genes. Collectively, our findings spotlighted HGPT-associated genes, TFs, miRNAs, and pathways as prospective biomarkers, offering new avenues for OC diagnostic and therapeutic approaches.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , MicroRNAs , Neoplasias Ovarianas , Humanos , Feminino , Multiômica , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Biologia Computacional , MicroRNAs/genética , MicroRNAs/metabolismo , Perfilação da Expressão Gênica , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/genética , Análise de Sequência com Séries de Oligonucleotídeos , Redes Reguladoras de Genes , Proteínas de Ciclo Celular/metabolismo , Quinases relacionadas a CDC2 e CDC28/genética , Proteínas Associadas aos Microtúbulos/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo
4.
Int J Pharm ; 642: 123095, 2023 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-37268031

RESUMO

Salinomycin is a polyether compound that exhibits strong anticancer activity and is known as the cancer stem cell inhibitor that reached clinical testing. The rapid elimination of nanoparticles from the bloodstream by the mononuclear phagocyte system (MPS), the liver, and the spleen, accompanied by protein corona (PC) formation, restricts in vivo delivery of nanoparticles in the tumor microenvironment (TME). The DNA aptamer (TA1) that successfully targets the overexpressed CD44 antigen on the surface of breast cancer cells suffers strongly from PC formation in vivo. Thus, cleverly designed targeted strategies that lead to the accumulation of nanoparticles in the tumor become a top priority in the drug delivery field. In this work, dual redox/pH-sensitive poly (ß-amino ester) copolymeric micelles modified with CSRLSLPGSSSKpalmSSS peptide and TA1 aptamer, as dual targeting ligands, were synthesized and fully characterized by physico-chemical methods. These biologically transformable stealth NPs were altered into the two ligand-capped (SRL-2 and TA1) NPs for synergistic targeting of the 4T1 breast cancer model after exposure to the TME. The PC formation was reduced sharply in Raw 264.7 cells by increasing the CSRLSLPGSSSKpalmSSS peptide concentration in modified micelles. Surprisingly, in vitro and in vivo biodistribution findings showed that dual targeted micelle accumulation in the TME of 4T1 breast cancer model was significantly higher than that of single modified formulation, along with deep penetration 24 h after intraperitoneal injection. Also, an in vivo treatment study showed remarkable tumor growth inhibition in 4T1 tumor-bearing Balb/c mice, compared to different formulations, with a 10% lower therapeutic dose (TD) of SAL that was confirmed by hematoxylin and eosin staining (H&E) and the TUNEL assay. Overall, in this study, we developed smart transformable NPs in which the body's own engineering systems alter their biological identity, which resulted in a reduction in therapeutic dosage along with a lowered off-target effect.


Assuntos
Nanopartículas , Neoplasias , Animais , Camundongos , Micelas , Distribuição Tecidual , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , Resultado do Tratamento , Peptídeos/farmacologia , Camundongos Endogâmicos BALB C
5.
Bioimpacts ; 13(2): 133-144, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37193076

RESUMO

Introduction: Blood-brain barrier with strictly controlled activity participates in a coordinated transfer of bioactive molecules from the blood to the brain. Among different delivery approaches, gene delivery is touted as a promising strategy for the treatment of several nervous system disorders. The transfer of exogenous genetic elements is limited by the paucity of suitable carriers. As a correlate, designing high-efficiency biocarriers for gene delivery is challenging. This study aimed to deliver pEGFP-N1 plasmid into the brain parenchyma using CDX-modified chitosan (CS) nanoparticles (NPs). Methods: Herein, we attached CDX, a 16 amino acids peptide, to the CS polymer using bifunctional polyethylene glycol (PEG) formulated with sodium tripolyphosphate (TPP), by ionic gelation method. Developed NPs and their nanocomplexes with pEGFP-N1 (CS-PEG-CDX/pEGFP) were characterized using DLS, NMR, FTIR, and TEM analyses. For in vitro assays, a rat C6 glioma cell line was used for cell internalization efficiency. The biodistribution and brain localization of nanocomplexes were studied in a mouse model after intraperitoneal injection using in vivo imaging and fluorescent microscopy. Results: Our results showed that CS-PEG-CDX/pEGFP NPs were uptaken by glioma cells in a dose-dependent manner. In vivo imaging revealed successful entry into the brain parenchyma indicated with the expression of green fluorescent protein (GFP) as a reporter protein. However, the biodistribution of developed NPs was also evident in other organs especially the spleen, liver, heart, and kidneys. Conclusion: Based on our results, CS-PEG-CDX NPs can provide a safe and effective nanocarrier for brain gene delivery into the central nervous system (CNS).

6.
Cancer Lett ; 557: 216077, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-36731592

RESUMO

At the moment, anaplastic changes within the brain are challenging due to the complexity of neural tissue, leading to the inefficiency of therapeutic protocols. The existence of a cellular interface, namely the blood-brain barrier (BBB), restricts the entry of several macromolecules and therapeutic agents into the brain. To date, several nano-based platforms have been used in laboratory settings and in vivo conditions to overcome the barrier properties of BBB. Exosomes (Exos) are one-of-a-kind of extracellular vesicles with specific cargo to modulate cell bioactivities in a paracrine manner. Regarding unique physicochemical properties and easy access to various biofluids, Exos provide a favorable platform for drug delivery and therapeutic purposes. Emerging data have indicated that Exos enable brain penetration of selective cargos such as bioactive factors and chemotherapeutic compounds. Along with these statements, the application of smart delivery approaches can increase delivery efficiency and thus therapeutic outcomes. Here, we highlighted the recent advances in the application of Exos in the context of brain tumors.


Assuntos
Neoplasias Encefálicas , Exossomos , Vesículas Extracelulares , Humanos , Exossomos/patologia , Neoplasias Encefálicas/patologia , Sistemas de Liberação de Medicamentos/métodos , Encéfalo/patologia , Vesículas Extracelulares/patologia
7.
J Control Release ; 349: 67-96, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35779656

RESUMO

The key issue in the treatment of solid tumors is the lack of efficient strategies for the targeted delivery and accumulation of therapeutic cargoes in the tumor microenvironment (TME). Targeting approaches are designed for more efficient delivery of therapeutic agents to cancer cells while minimizing drug toxicity to normal cells and off-targeting effects, while maximizing the eradication of cancer cells. The highly complicated interrelationship between the physicochemical properties of nanoparticles, and the physiological and pathological barriers that are required to cross, dictates the need for the success of targeting strategies. Dual targeting is an approach that uses both purely biological strategies and physicochemical responsive smart delivery strategies to increase the accumulation of nanoparticles within the TME and improve targeting efficiency towards cancer cells. In both approaches, either one single ligand is used for targeting a single receptor on different cells, or two different ligands for targeting two different receptors on the same or different cells. Smart delivery strategies are able to respond to triggers that are typical of specific disease sites, such as pH, certain specific enzymes, or redox conditions. These strategies are expected to lead to more precise targeting and better accumulation of nano-therapeutics. This review describes the classification and principles of dual targeting approaches and critically reviews the efficiency of dual targeting strategies, and the rationale behind the choice of ligands. We focus on new approaches for smart drug delivery in which synthetic and/or biological moieties are attached to nanoparticles by TME-specific responsive linkers and advanced camouflaged nanoparticles.


Assuntos
Nanopartículas , Neoplasias , Sistemas de Liberação de Medicamentos , Humanos , Ligantes , Nanopartículas/química , Neoplasias/tratamento farmacológico , Microambiente Tumoral
8.
Sci Rep ; 12(1): 11774, 2022 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-35821262

RESUMO

Here, we investigated the photothermal effect of gold nanorods (GNRs) on human neuroblastoma CD133+ cancer stem cells (CSCs) via autophagic cell death. GNRs were synthesized using Cetyltrimethylammonium bromide (CTAB), covered with bovine serum albumin (BSA). CD133+ CSCs were enriched from human neuroblastoma using the magnetic-activated cell sorting (MACS) technique. Cells were incubated with GNRs coated with BSA and exposed to 808-nm near-infrared laser irradiation for 8 min to yield low (43 °C), medium (46 °C), and high (49 °C) temperatures. After 24 h, the survival rate and the percent of apoptotic and necrotic CSCs were measured using MTT assay and flow cytometry. The expression of different autophagy-related genes was measured using polymerase chain reaction (PCR) array analysis. Protein levels of P62 and LC3 were detected using an enzyme-linked immunosorbent assay (ELISA). The viability of CSC was reduced in GNR-exposed cells compared to the control group (p < 0.05). At higher temperatures (49 °C), the percent of apoptotic CSCs, but not necrotic cells, increased compared to the lower temperatures. Levels of intracellular LC3 and P62 were reduced and increased respectively when the temperature increased to 49 °C (p < 0.05). These effects were non-significant at low and medium temperatures (43 and 46 °C) related to the control CSCs (p > 0.05). The clonogenic capacity of CSC was also inhibited after photothermal therapy (p < 0.05). Despite these changes, no statistically significant differences were found in terms of CSC colony number at different temperatures regardless of the presence or absence of HCQ. Based on the data, the combination of photothermal therapy with HCQ at 49 °C can significantly abort the CSC clonogenic capacity compared to the control-matched group without HCQ (p < 0.0001). PCR array showed photothermal modulation of CSCs led to alteration of autophagy-related genes and promotion of co-regulator of apoptosis and autophagy signaling pathways. Factors related to autophagic vacuole formation and intracellular transport were significantly induced at a temperature of 49 °C (p < 0.05). We also note the expression of common genes belonging to autophagy and apoptosis signaling pathways at higher temperatures. Data showed tumoricidal effects of laser-irradiated GNRs by the alteration of autophagic response and apoptosis.


Assuntos
Nanotubos , Neuroblastoma , Autofagia , Linhagem Celular Tumoral , Ouro/farmacologia , Humanos , Células-Tronco Neoplásicas , Neuroblastoma/terapia , Soroalbumina Bovina/farmacologia
9.
J Control Release ; 345: 371-384, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35301054

RESUMO

The effective treatment of glioma through conventional chemotherapy is proved to be a great challenge in clinics. The main reason is due to the existence of two physiological and pathological barriers respectively including the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) that prevent most of the chemotherapeutics from efficient delivery to the brain tumors. To address this challenge, an ideal drug delivery system would efficiently traverse the BBB and BBTB and deliver the therapeutics into the glioma cells with high selectivity. Herein, a targeted delivery system was developed based on nanostructured lipid carriers (NLCs) modified with two proteolytically stable D-peptides, D8 and RI-VAP (Dual NLCs). D8 possesses high affinity towards nicotine acetylcholine receptors (nAChRs), overexpressed on brain capillary endothelial cells (BCECs), and can penetrate through the BBB with high efficiency. RI-VAP is a specific ligand of cell surface GRP78 (csGRP78), a specific angiogenesis and cancer cell-surface marker, capable of circumventing the BBTB with superior glioma-homing property. Dual NLCs could internalize into BCECs, tumor neovascular endothelial cells, and glioma cells with high specificity and could penetrate through in vitro BBB and BBTB models with excellent efficiency compared to non-targeted or mono-targeted NLCs. In vivo whole-animal imaging and ex vivo imaging further confirmed the superior targeting capability of Dual NLCs towards intracranial glioma. When loaded with Bortezomib (BTZ), Dual NLCs attained the highest therapeutic efficiency by means of superior in vitro cytotoxicity and apoptosis and prolonged survival rate and efficient anti-glioma behavior in intracranial glioma bearing mice. Collectively, the designed targeting platform in this study could overcome multiple barriers and effectively deliver BTZ to glioma cells, which represent its potential for advanced brain cancer treatment with promising therapeutic outcomes.


Assuntos
Neoplasias Encefálicas , Glioma , Animais , Barreira Hematoencefálica/metabolismo , Bortezomib/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Células Endoteliais/metabolismo , Glioma/tratamento farmacológico , Lipídeos/uso terapêutico , Camundongos
10.
Sci Rep ; 11(1): 23984, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34907215

RESUMO

Unraveling unwanted side effects of nanotechnology-based therapies like photothermal therapy (PTT) is vital in translational nanomedicine. Herein, we monitored the relationship between autophagic response at the transcriptional level by using a PCR array and tumor formation ability by colony formation assay in the human neuroblastoma cell line, SH-SY5Y, 48 h after being exposed to two different mild hyperthermia (43 and 48 °C) induced by PTT. In this regard, the promotion of apoptosis and autophagy were evaluated using immunofluorescence imaging and flow cytometry analyses. Protein levels of Ki-67, P62, and LC3 were measured using ELISA. Our results showed that of 86 genes associated with autophagy, the expression of 54 genes was changed in response to PTT. Also, we showed that chaperone-mediated autophagy (CMA) and macroautophagy are stimulated in PTT. Importantly, the results of this study also showed significant changes in genes related to the crosstalk between autophagy, dormancy, and metastatic activity of treated cells. Our findings illustrated that PTT enhances the aggressiveness of cancer cells at 43 °C, in contrast to 48 °C by the regulation of autophagy-dependent manner.


Assuntos
Autofagia/efeitos dos fármacos , Ouro , Hipotermia Induzida , Nanopartículas Metálicas , Nanotubos/química , Neuroblastoma , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ouro/química , Ouro/farmacologia , Humanos , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Proteínas de Neoplasias/biossíntese , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neuroblastoma/terapia
11.
Sci Rep ; 11(1): 21425, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34728703

RESUMO

Adjuvant-aided combination chemotherapy is one of the most effective ways of cancer treatment by overcoming the multidrug resistance (MDR) and reducing the side-effects of anticancer drugs. In this study, Conferone (Conf) was used as an adjuvant in combination with Doxorubicin (Dox) for inducing apoptosis to MDA-MB-231 cells. Herein, the novel biodegradable amphiphilic ß-cyclodextrin grafted poly maleate-co-PLGA was synthesized by thiol-ene addition and ring-opening process. Micelles obtained from the novel copolymer showed exceptional properties such as small size of around 34.5 nm, CMC of 0.1 µg/mL, and cell internalization of around 100% at 30 min. These novel engineered micelles were used for combination delivery of doxorubicin-conferone with high encapsulation efficiency of near 100% for both drugs. Our results show that combination delivery of Dox and Conf to MDA-MB-231 cells had synergistic effects (CI < 1). According to cell cycle and Annexin-V apoptosis analysis, Dox-Conf loaded micelle significantly induce tumor cell apoptosis (more than 98% of cells population showed apoptosis at IC50 = 0.259 µg/mL). RT-PCR and western-blot tests show that Dox-Conf loaded ßCD-g-PMA-co-PLGA micelle induced apoptosis via intrinsic pathway. Therefore, the unique design of multi-functional pH-sensitive micelles open a new perspective for the development of nanomedicine for combination chemo-adjuvant therapy against malignant cancer.


Assuntos
Neoplasias da Mama/patologia , Cumarínicos/farmacologia , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos , Micelas , beta-Ciclodextrinas/química , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacologia , Apoptose , Neoplasias da Mama/tratamento farmacológico , Ciclo Celular , Proliferação de Células , Cumarínicos/administração & dosagem , Cumarínicos/química , Doxorrubicina/administração & dosagem , Doxorrubicina/química , Feminino , Humanos , Concentração de Íons de Hidrogênio , Células Tumorais Cultivadas
12.
Biomedicines ; 9(11)2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34829766

RESUMO

In recent years, smart/stimuli-responsive hydrogels have drawn tremendous attention for their varied applications, mainly in the biomedical field. These hydrogels are derived from different natural and synthetic polymers but are also composite with various organic and nano-organic fillers. The basic functions of smart hydrogels rely on their ability to change behavior; functions include mechanical, swelling, shaping, hydrophilicity, and bioactivity in response to external stimuli such as temperature, pH, magnetic field, electromagnetic radiation, and biological molecules. Depending on the final applications, smart hydrogels can be processed in different geometries and modalities to meet the complicated situations in biological media, namely, injectable hydrogels (following the sol-gel transition), colloidal nano and microgels, and three dimensional (3D) printed gel constructs. In recent decades smart hydrogels have opened a new horizon for scientists to fabricate biomimetic customized biomaterials for tissue engineering, cancer therapy, wound dressing, soft robotic actuators, and controlled release of bioactive substances/drugs. Remarkably, 4D bioprinting, a newly emerged technology/concept, aims to rationally design 3D patterned biological matrices from synthesized hydrogel-based inks with the ability to change structure under stimuli. This technology has enlarged the applicability of engineered smart hydrogels and hydrogel composites in biomedical fields. This paper aims to review stimuli-responsive hydrogels according to the kinds of external changes and t recent applications in biomedical and 4D bioprinting.

13.
Mol Pharm ; 18(12): 4341-4353, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34779630

RESUMO

The formation of protein corona (PC) around nanoparticles (NPs) has been reported inside biological conditions. This effect can alter delivery capacity toward the targeted tissues. Here, we synthesized folic acid-modified chitosan NPs (FA-CS NPs) using different concentrations of folic acid (5, 10, and 20%). FA-CS NPs were exposed to plasmas of breast cancer patients and healthy donors to evaluate the possibility of PC formation. We also monitored uptake efficiency in in vitro conditions after incubation with human breast cancer cell line MDA-MB-231 and monocyte/macrophage-like Raw264.7 cells. Data showed that the formation of PC around FA-CS NPs can change physicochemical properties coincided with the rise in NP size and negative surface charge. SDS-PAGE electrophoresis revealed differences in the type and content rate of plasma proteins attached to NP surface in a personalized manner. Based on MTT data, the formation of PC around NPs did not exert cytotoxic effects on MDA-MB-231 cells while this phenomenon reduced uptake rate. Fluorescence imaging and flow cytometry analyses revealed reduced cellular internalization rate in NPs exposed to patients' plasma compared to the control group. In contrast to breast MDA-MB-231 cells, Raw264.7 cells efficiently adsorbed the bare and PC-coated NPs from both sources, indicating the involvement of ligand-receptor-dependent and independent cellular engulfment. These data showed that the PC formed on the FA-CS NPs is entirely different in breast cancer patients and healthy counterparts. PC derived from patients' plasma almost abolishes the targeting efficiency of FA-CS NPs even in different mechanisms, while this behavior was not shown in the control group. Surprisingly, Raw264.7 cells strongly adsorbed the PC-coated NPs, especially when these particles were in the presence of patients' sera. It is strongly suggested that the formation of PC around can affect delivering capacity of FA-CS NPs to cancer cells. It seems that the PC-coated FA-CS NPs can be used as an efficient delivery strategy for the transfer of specific biomolecules in immune system disorders.


Assuntos
Neoplasias da Mama/sangue , Neoplasias da Mama/tratamento farmacológico , Quitosana/química , Sistemas de Liberação de Medicamentos , Ácido Fólico/química , Nanopartículas/química , Linhagem Celular Tumoral , Feminino , Humanos , Macrófagos/fisiologia
14.
Genes (Basel) ; 12(7)2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-34356120

RESUMO

The transcription factor high mobility group protein A2 (HMGA2) plays an important role in the pathogenesis of some cancers including breast cancer. Polyamidoamine dendrimer generation 4 is a kind of highly branched polymeric nanoparticle with surface charge and highest density peripheral groups that allow ligands or therapeutic agents to attach it, thereby facilitating target delivery. Here, methotrexate (MTX)- modified polyamidoamine dendrimer generation 4 (G4) (G4/MTX) was generated to deliver specific small interface RNA (siRNA) for suppressing HMGA2 expression and the consequent effects on folate receptor (FR) expressing human breast cancer cell lines (MCF-7, MDA-MB-231). We observed that HMGA2 siRNA was electrostatically adsorbed on the surface of the G4/MTX nanocarrier for constructing a G4/MTX-siRNA nano-complex which was verified by changing the final particle size and zeta potential. The release of MTX and siRNA from synthesized nanocomplexes was found in a time- and pH-dependent manner. We know that MTX targets FR. Interestingly, G4/MTX-siRNA demonstrates significant cellular internalization and gene silencing efficacy when compared to the control. Besides, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay demonstrated selective cell cytotoxicity depending on the folate receptor expressing in a dose-dependent manner. The gene silencing and protein downregulation of HMGA2 by G4/MTX-siRNA was observed and could significantly induce cell apoptosis in MCF-7 and MDA-MB-231 cancer cells compared to the control group. Based on the findings, we suggest that the newly developed G4/MTX-siRNA nano-complex may be a promising strategy to increase apoptosis induction through HMGA2 suppression as a therapeutic target in human breast cancer.


Assuntos
Neoplasias da Mama/genética , Proteína HMGA2/genética , Apoptose/efeitos dos fármacos , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Terapia Baseada em Transplante de Células e Tecidos/métodos , Dendrímeros/metabolismo , Dendrímeros/farmacologia , Dendrímeros/uso terapêutico , Feminino , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Proteína HMGA2/metabolismo , Humanos , Células MCF-7 , Metotrexato/farmacologia , Nylons/farmacologia , RNA Interferente Pequeno/genética
15.
Cell Biosci ; 11(1): 142, 2021 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-34294165

RESUMO

Nowadays, a large population around the world, especially the elderly, suffers from neurological inflammatory and degenerative disorders/diseases. Current drug delivery strategies are facing different challenges because of the presence of the BBB, which limits the transport of various substances and cells to brain parenchyma. Additionally, the low rate of successful cell transplantation to the brain injury sites leads to efforts to find alternative therapies. Stem cell byproducts such as exosomes are touted as natural nano-drug carriers with 50-100 nm in diameter. These nano-sized particles could harbor and transfer a plethora of therapeutic agents and biological cargos to the brain. These nanoparticles would offer a solution to maintain paracrine cell-to-cell communications under healthy and inflammatory conditions. The main question is that the existence of the intact BBB could limit exosomal trafficking. Does BBB possess some molecular mechanisms that facilitate the exosomal delivery compared to the circulating cell? Although preliminary studies have shown that exosomes could cross the BBB, the exact molecular mechanism(s) beyond this phenomenon remains unclear. In this review, we tried to compile some facts about exosome delivery through the BBB and propose some mechanisms that regulate exosomal cross in pathological and physiological conditions.

16.
Mol Pharmacol ; 100(2): 119-143, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33990406

RESUMO

Most commonly recognized as a catabolic pathway, autophagy is a perplexing mechanism through which a living cell can free itself of excess cytoplasmic components, i.e., organelles, by means of certain membranous vesicles or lysosomes filled with degrading enzymes. Upon exposure to external insult or internal stimuli, the cell might opt to activate such a pathway, through which it can gain control over the maintenance of intracellular components and thus sustain homeostasis by intercepting the formation of unnecessary structures or eliminating the already present dysfunctional or inutile organelles. Despite such appropriateness, autophagy might also be considered a frailty for the cell, as it has been said to have a rather complicated role in tumorigenesis. A merit in the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. In fact, several investigations on tumorigenesis have reported diminished levels of autophagic activity in tumor cells, which might result in transition to malignancy. On the contrary, autophagy has been suggested to be a seemingly favorable mechanism to progressed malignancies, as it contributes to survival of such cells. Based on the recent literature, this mechanism might also be activated upon the entry of engineered nanomaterials inside a cell, supposedly protecting the host from foreign materials. Accordingly, there is a good chance that therapeutic interventions for modulating autophagy in malignant cells using nanoparticles may sensitize cancerous cells to certain treatment modalities, e.g., radiotherapy. In this review, we will discuss the signaling pathways involved in autophagy and the significance of the mechanism itself in apoptosis and tumorigenesis while shedding light on possible alterations in autophagy through engineered nanomaterials and their potential therapeutic applications in cancer. SIGNIFICANCE STATEMENT: Autophagy has been said to have a complicated role in tumorigenesis. In the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. On the contrary, autophagy has been suggested to be a favorable mechanism to progressed malignancies. This mechanism might be affected upon the entry of nanomaterials inside a cell. Accordingly, therapeutic interventions for modulating autophagy using nanoparticles may sensitize cancerous cells to certain therapies.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/patologia , Transdução de Sinais , Antineoplásicos/uso terapêutico , Apoptose , Autofagia/efeitos dos fármacos , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Nanotecnologia , Estadiamento de Neoplasias , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
J Control Release ; 328: 932-941, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33129921

RESUMO

As one of the deadliest diseases, cancer frequently resists existing therapeutics because they do not target all cells within a progressing tumor, for example both tumor stem and proliferating cells. This frequently results in enrichment of invasive and metastatic drug-resistant tumor cells subpopulations, cancer recurrence and eventually, patient mortality. Thus, there is an urgent need to identify specific markers, by which the targeted imaging and/or therapeutic "guided missile"-like agents can specifically detect and/or eradicate all cancer cells within a heterogeneous tumor, while leaving the normal cells intact. As a member of heat shock protein 70 (HSP70) superfamily, glucose regulated protein 78 (GRP78) has been documented as a molecular chaperone in the endoplasmic reticulum (ER) which mainly responds to ER stresses in normal cells. There is over-expression of GRP78 on the surface of cancer cells and angiogenic endothelial cells, which makes it a promising target for different types of peptides and antibodies that can be employed for targeted cancer therapy or imaging. In this review, we discuss the biological processes, functional importance and translocation mechanisms of cell surface GRP78 (csGRP78) in tumor cells. As a cancer biomarker, we also review the potential applications of csGRP78 targeted therapy and imaging and finally we suggest a brief roadmap ahead of csGRP78 targeting for targeted theranostic implications.


Assuntos
Proteínas de Membrana , Neoplasias , Chaperona BiP do Retículo Endoplasmático , Células Endoteliais , Glucose , Proteínas de Choque Térmico , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico
18.
Colloids Surf B Biointerfaces ; 194: 111225, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32622253

RESUMO

This study aimed to design an effective targeted combination of doxorubicin (Dox)-Curcumin (Cur) delivery system to eradicate the MDA-MB231 cell line. A novel biodegradable poly ε-Caprolactone-co-maleic anhydride-graft-citric acid copolymer micelle (PCL-co-P(MA-g-CA)) was synthesized through thiolen radical copolymerization and ring-opening polymerization. The unique micelle structure allowed simultaneous loading of hydrophilic Dox and hydrophobic Cur with a loading efficiency of above 98 % for each drug. The physicochemical characterization of copolymeric micelle was analyzed by 1HNMR, 13CNMR, FTIR, DSC, CMC, DLS and SEM. The in vitro cytotoxicity was assessed by MTT assay, cell cycle analysis, annexin V-FITC apoptosis, qRT-PCR and western blot. The final obtained micelles with critical micelle concentration (CMC) of 0.5 µg/mL, and particle size and surface charge was 60 nm and -14.1 mV, respectively. Beside the fast uptake of designed micelle, Dox@Cur loaded micelle showed a synergistic effect with the combination index (CI) value of below 1. Our results revealed that this novel engineered combinatorial micelle induced apoptosis (96 %) which was proved by annexin V and cell cycle. qRT-PCR and western blot assays demonstrated involvement of intrinsic apoptosis pathways in the genetic and protein levels. Finally, the penetration of Dox@Cur loaded micelle was evaluated by 3D in vitro tumor formation. Our findings showed the penetration behavior of micelles is in a concentration-dependent manner. In conclusion, combinational therapy by using Dox and Cur nano-formulation has boosted the cytotoxicity in MDA-MB231 cells by promoting the apoptotic response.


Assuntos
Curcumina , Micelas , Caproatos , Linhagem Celular Tumoral , Ácido Cítrico , Curcumina/farmacologia , Doxorrubicina/farmacologia , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Lactonas , Tamanho da Partícula , Poliésteres
19.
Colloids Surf B Biointerfaces ; 188: 110762, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31911391

RESUMO

Co-delivery of therapeutic agents and small interfering RNA (siRNA) can be achieved by a suitable nanovehicle. In this work, the solubility and bioavailability of curcumin (Cur) were enhanced by entrapment in a polyamidoamine (PAMAM) dendrimer, and a polyplex was formed by grafting Bcl-2 siRNA onto the surface amine groups to produce PAMAM-Cur/Bcl-2 siRNA nanoparticles (NPs). The synthesized polyplex NPs had a particle size of ∼180 nm, and high Cur loading content of ∼82 wt%. Moreover, the PAMAM-Cur/Bcl-2 siRNA NPs showed more effective cellular uptake, and higher inhibition of tumor cell proliferation compared to PAMAM-Cur nanoformulation and free Cur, due to the combined effect of co-delivery of Cur and Bcl-2 siRNA. The newly described PAMAM-Cur/Bcl-2 siRNA polyplex NPs could be a promising co-delivery nanovehicle.


Assuntos
Antineoplásicos/farmacologia , Curcumina/farmacologia , Dendrímeros/farmacologia , Sistemas de Liberação de Medicamentos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/farmacologia , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Curcumina/química , Dendrímeros/química , Ensaios de Seleção de Medicamentos Antitumorais , Células HeLa , Humanos , Tamanho da Partícula , RNA Interferente Pequeno/química , Propriedades de Superfície
20.
Cell Mol Life Sci ; 76(14): 2697-2718, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31030227

RESUMO

The field of tissue engineering (TE) experiences its most exciting time in the current decade. Recent progresses in TE have made it able to translate into clinical applications. To regenerate damaged tissues, TE uses biomaterial scaffolds to prepare a suitable backbone for tissue regeneration. It is well proven that the cell-biomaterial crosstalk impacts tremendously on cell biological activities such as differentiation, proliferation, migration, and others. Clarification of exact biological effects and mechanisms of a certain material on various cell types promises to have a profound impact on clinical applications of TE. Chitosan (CS) is one of the most commonly used biomaterials with many promising characteristics such as biocompatibility, antibacterial activity, biodegradability, and others. In this review, we discuss crosstalk between CS and various cell types to provide a roadmap for more effective applications of this polymer for future uses in tissue engineering and regenerative medicine.


Assuntos
Materiais Biocompatíveis/metabolismo , Quitosana/metabolismo , Medicina Regenerativa , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Humanos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA