Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
ACS Biomater Sci Eng ; 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39269431

RESUMEN

Poly(lactide-co-glycolide) (PLGA) is a biocompatible and biodegradable copolymer that has gained high acceptance in biomedical applications. In the present study, PLGA (Mw = 13,900) was synthesized by ring-opening polymerization in the presence of a biocompatible zinc-proline initiator through a green route. Irinotecan (Ir) loaded with efficient PLGA core-lipid shell hybrid nanocarriers (lipomers, LPs) were formulated with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino (polyethylene glycol)-2000] (DSPE-PEG-2000), using soya lecithin, by a nanoprecipitation method, and the fabricated LPs were designated as P-DSPE-Ir and P-DSPE-PEG-Ir, respectively. The formulated LPs were further validated for their physicochemical properties and biological potential for colon cancer application. The potential delivery of a poorly water-soluble chemotherapeutic drug (Ir) was studied for the treatment of colon cancer. LPs were successfully prepared, providing controlled size (80-120 nm) and surface charge (∼ -35 mV), and the sustained release properties and cytotoxicity against CT-26 colon cancer cells were studied. The in vivo biodistribution and tumor site retention in CT-26 xenograft tumor-bearing Balb/C mice showed promising results for tumor uptake and retention for a prolonged time period. Unlike P-DSPE-Ir, the P-DSPE-PEG-Ir LP exhibited significant tumor growth delay as compared to untreated and blank formulation-treated groups in CT-26 (subcutaneous tumor model) after 4 treatments of 10 mg irinotecan/kg dose. The biocompatibility and safety of the LPs were confirmed by an acute toxicity study of the optimized formulation. Overall, this proof-of-concept study demonstrates that the PLGA-based LPs improve the efficacy and bioavailability and decrease neutropenia of Ir to combat colon cancer.

2.
Cell Death Discov ; 10(1): 274, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38851743

RESUMEN

Diverse mechanisms have been established to understand the chemoresistance of hepatocellular carcinoma (HCC), but the contribution of non-coding RNAs is not surveyed well. Here, we aimed to explore the lncRNA-miRNA axis in Hepatitis C and B virus (HCV and HBV) infected HCC to investigate the molecular mechanism of chemoresistance and to identify a potential therapeutic target for HCC. The small RNA transcriptome analysis followed by qRT-PCR validation with the liver tissues of both HCV and HBV infected HCC patients revealed that miR-424-5p, miR-136-3p, miR-139-5p, miR-223-3p, and miR-375-3p were the most downregulated miRNAs in HCC compared to normal (log2 fold change ≤-1.5, Padj ≤ 0.05). In silico pathway analysis with the validated targets of each miRNA revealed that the signalling pathway regulating pluripotency of stem cells is commonly targeted by these five miRNAs. Subsequent validation by 3'UTR-luciferase assay and western blot analysis unveiled that these five miRNAs impeded either same or diverse genes, but all linked to BMP signalling pathway such as BMPR1A/BMPR1B by miR-139-5p, miR-136-3p, and miR-375-3p, and ACVR2A/ACVR2B by miR-424-5p and miR-223-3p. Furthermore, restoration of each miRNA in Huh7/SNU449 cells inhibited phosphorylation of downstream SMAD1/5 and ERK1/2, and attenuated Epithelial-mesenchymal transition, stemness, spheroid formation, chemoresistance, invasion and migration of cells. To investigate the mechanism of suppression of these miRNAs, "DIANA" tool was employed and lncRNA-KCNQ1OT1 was retrieved as interacting partner of all the five miRNAs. In vitro RNA pull-down assay revealed that lncRNA-KCNQ1OT1 physically interacted and sequestered these five miRNAs in the cytoplasm. Hence, KCNQ1OT1 was suppressed in Huh7/SNU449 cells using CRISPR technology and observed regression of oncogenic properties with enhanced chemosensitivity and reduced metastasis in cancer cells. Shrinkage of tumour size and volume in NOD-SCID mice injected with KCNQ1OT1-sgRNA cells further strengthened our observations. Thus, lncRNA-KCNQ1OT1 is the main regulator, which reduces the level of beneficiary miRNAs in the tumour milieu and modulates BMP signalling pathway to promote chemoresistance in HCC, suggesting lncRNA-KCNQ1OT1 might have robust potential to be a therapeutic target in HCC.

3.
ACS Omega ; 9(3): 3807-3826, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38284072

RESUMEN

A poly(d,l-lactide-co-glycolide) (PLGA) copolymer was synthesized using the ring-opening polymerization of d,l-lactide and glycolide monomers in the presence of zinc proline complex in bulk through the green route and was well characterized using attenuated total reflectance-Fourier transform infrared, 1H and 13C nuclear magnetic resonance, gel permeation chromatography, differential scanning calorimetry, X-ray diffraction, matrix-assisted laser desorption/ionization time-of-flight, etc. Furthermore, PLGA-conjugated biotin (PLGA-B) was synthesized using the synthesized PLGA and was employed to fabricate nanoparticles for irinotecan (Ir) delivery. These nanoparticles (PLGA-NP-Ir and PLGA-B-NP-Ir) were tested for physicochemical and biological characteristics. PLGA-B-NP-Ir exhibited a stronger cellular uptake and anticancer activity as compared to PLGA-NP-Ir in CT-26 cancer cells (log p < 0.05). The accumulation and retention of fluorescence-labeled nanoparticles were observed to be better in CT-26-inoculated solid tumors in Balb/c mice. The PLGA-B-NP-Ir-treated group inhibited tumor growth significantly more (log p < 0.001) than the untreated control, PLGA-NP-Ir, and Ir-treated groups. Furthermore, no body weight loss, hematological, and blood biochemical tests demonstrated the nanocarriers' nontoxic nature. This work presents the use of safe PLGA and the demonstration of a proof-of-concept of biotin surface attached PLGA nanoparticle-mediated active targeted Ir administration to combat colon cancer. To treat colon cancer, PLGA-B-NP-Ir performed better due to specific active tumor targeting and greater cellular uptake due to biotin.

4.
J Control Release ; 367: 300-315, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38281670

RESUMEN

Nanoparticle formulations blending optical imaging contrast agents and therapeutics have been a cornerstone of preclinical theranostic applications. However, nanoparticle-based theranostics clinical translation faces challenges on reproducibility, brightness, photostability, biocompatibility, and selective tumor targeting and penetration. In this study, we integrate multimodal imaging and therapeutics within cancer cell-derived nanovesicles, leading to biomimetic bright optotheranostics for monitoring cancer metastasis. Upon NIR light irradiation, the engineered optotheranostics enables deep visualization and precise localization of metastatic lung, liver, and solid breast tumors along with solid tumor ablation. Metastatic cell-derived nanovesicles (∼80 ± 5 nm) are engineered to encapsulate imaging (emissive organic dye and gold nanoparticles) and therapeutic agents (anticancer drug doxorubicin and photothermally active organic indocyanine green dye). Systemic administration of biomimetic bright optotheranostic nanoparticles shows escape from mononuclear phagocytic clearance with (i) rapid tumor accumulation (3 h) and retention (up to 168 h), (ii) real-time monitoring of metastatic lung, liver, and solid breast tumors and (iii) 3-fold image-guided solid tumor reduction. These findings are supported by an improvement of X-ray, fluorescence, and photoacoustic signals while demonstrating a tumor reduction (201 mm3) in comparison with single therapies that includes chemotherapy (134 mm3), photodynamic therapy (72 mm3), and photothermal therapy (88mm3). The proposed innovative platform opens new avenues to improve cancer diagnosis and treatment outcomes by allowing the monitorization of cancer metastasis, allowing the precise cancer imaging, and delivering synergistic therapeutic agents at the solid tumor site.


Asunto(s)
Neoplasias de la Mama , Nanopartículas del Metal , Nanopartículas , Neoplasias , Humanos , Femenino , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/tratamiento farmacológico , Fototerapia/métodos , Biomimética , Oro , Reproducibilidad de los Resultados , Línea Celular Tumoral , Neoplasias/terapia , Nanomedicina Teranóstica/métodos
5.
Cell Rep Phys Sci ; 4(11): 101648, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38021344

RESUMEN

Bioinspired cell-membrane-camouflaged nanohybrids have been proposed to enhance tumor targeting by harnessing their immune escape and self-recognition abilities. In this study, we introduce cancer-cell-derived membrane nanovesicles (CCMVs) integrated with gold nanorods (AuVNRs) in addition to therapeutic and imaging cargos such as doxorubicin and indocyanine green. This approach enhances targeted tumor imaging and enables synergistic chemo-phototherapeutics for solid tumors. CCMVs demonstrate significant tumor penetration and retention, serving as nanotheranostics with accessible surface biomarkers, biomimicking properties, and homologous targeting abilities. By evading uptake by the mononuclear phagocytic system, CCMVs can diffuse into the deep tumor core, leading to precise tumor reduction while preserving the surrounding healthy tissues. Notably, intravenous administration of these theranostic agents ensures biocompatibility, as evidenced by a survival period of approximately two months (up to 63 days) without any observed side effects. Our findings underscore the diagnostic and therapeutic potential of this biomimetic nanotheranostics platform.

6.
Nanotheranostics ; 7(3): 270-280, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37064610

RESUMEN

A series of novel mixed transition metal-Magnesium tartarate complexes of general formulation [MMg(C4H4O6)2 .xH2O] (where M = Mn, Fe, Co, Ni, Cu and Zn) is prepared with bidentate tartarate ligand. The synthesized complexes (C1 to C6) are characterized by various analytical techniques such as Elemental analysis, Thermo gravimetric analysis, FT-IR Spectroscopy, X-ray Diffraction, Magnetic susceptibility study etc. All complexes exhibit the composition MMgL2 where M = Mn(II), Fe(II), Co(II), Ni(II), Cu(II) and Zn(II) and L = bidentate tartarate ligand. Analytical data reveals all complexes possesses 1:1 (metal: ligand) ratio. FT-IR spectral study shows that bidentate tartarate ligand coordinate with metal ion in a bidentate manner through two oxygen atoms. Thermo gravimetric analysis of all complexes shows that degradation curves of complexes agrees with recommended formulae of the complexes. X-ray diffraction technique suggests that all complexes (C1 to C6) are polycrystalline in nature. All newly synthesized metal tartarate complexes and ligand were screened in vitro for their anticancer activity against human breast cancer (MDA-MB-231) cell line. The bioassays of all these complexes showed C3 (Co) and C5 (Cu) Mg-tartarate complexes contains maximum antiproliferative activity at 200 µg/ml concentration on MDA-MB-231 cells as compared to other complexes. MDA-MB-231 cells treated with C3 (Co) and C5 (Cu) Mg-tartarate complexes also showed inhibition in cell migration.


Asunto(s)
Neoplasias de la Mama , Elementos de Transición , Humanos , Femenino , Espectroscopía Infrarroja por Transformada de Fourier , Ligandos , Metales/química , Elementos de Transición/química , Elementos de Transición/farmacología , Neoplasias de la Mama/tratamiento farmacológico
7.
Nat Commun ; 14(1): 1129, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36854749

RESUMEN

Tissue injury to skin diminishes miR-200b in dermal fibroblasts. Fibroblasts are widely reported to directly reprogram into endothelial-like cells and we hypothesized that miR-200b inhibition may cause such changes. We transfected human dermal fibroblasts with anti-miR-200b oligonucleotide, then using single cell RNA sequencing, identified emergence of a vasculogenic subset with a distinct fibroblast transcriptome and demonstrated blood vessel forming function in vivo. Anti-miR-200b delivery to murine injury sites likewise enhanced tissue perfusion, wound closure, and vasculogenic fibroblast contribution to perfused vessels in a FLI1 dependent manner. Vasculogenic fibroblast subset emergence was blunted in delayed healing wounds of diabetic animals but, topical tissue nanotransfection of a single anti-miR-200b oligonucleotide was sufficient to restore FLI1 expression, vasculogenic fibroblast emergence, tissue perfusion, and wound healing. Augmenting a physiologic tissue injury adaptive response mechanism that produces a vasculogenic fibroblast state change opens new avenues for therapeutic tissue vascularization of ischemic wounds.


Asunto(s)
Fibroblastos , Piel , Cicatrización de Heridas , Animales , Humanos , Ratones , Antagomirs/farmacología , Antagomirs/uso terapéutico , Fibroblastos/metabolismo , Fibroblastos/fisiología , Oligonucleótidos/farmacología , Piel/metabolismo , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología
8.
Nanomedicine (Lond) ; 17(11): 753-764, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35575008

RESUMEN

Aim: To evaluate the efficacy of novel methotrexate-loaded nanoparticles (MTX-NPs) in vitro and in vivo in the treatment of breast cancer. Materials & methods: MTX-NPs were tested for cellular uptake, cell viability, cell cycle, cellular wound migration and changes in tumor volume using characterized NPs. Results: The solid lipid NPs (SLNPs) showed strong cellular uptake, increased apoptosis, controlled cytotoxicity at lower IC50 of methotrexate and a sizable reduction in tumor burden. Conclusion: MTX-NP oral formulation can be a promising candidate in breast cancer treatment with improved cellular uptake and in vivo efficacy.


Asunto(s)
Neoplasias de la Mama , Nanopartículas , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Supervivencia Celular , Femenino , Humanos , Liposomas , Metotrexato/farmacología , Ratones
9.
J Invest Dermatol ; 142(3 Pt A): 679-691.e3, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34534575

RESUMEN

Impaired re-epithelialization characterized by hyperkeratotic nonmigratory wound epithelium is a hallmark of nonhealing diabetic wounds. In chronic wounds, the copious release of oncostatin M (OSM) from wound macrophages is evident. OSM is a potent keratinocyte (KC) activator. This work sought to understand the signal transduction pathway responsible for wound re-epithelialization, the primary mechanism underlying wound closure. Daily topical treatment of full-thickness excisional wounds of C57BL/6 mice with recombinant murine OSM improved wound re-epithelialization and accelerated wound closure by bolstering KC proliferation and migration. OSM activated the Jak-signal transducer and activator of transcription pathway as manifested by signal transducer and activator of transcription 3 phosphorylation. Such signal transduction in the human KC induced TP63, the master regulator of KC function. Elevated TP63 induced ITGB1, a known effector of KC migration. In diabetic wounds, OSM was more abundant than the level in nondiabetic wounds. However, in diabetic wounds, OSM activity was compromised by glycation. Aminoguanidine, a deglycation agent, rescued the compromised KC migration caused by glycated OSM. Finally, topical application of recombinant OSM improved KC migration and accelerated wound closure in db/db mice. This work recognizes that despite its abundance at the wound site, OSM is inactivated by glycation, and topical delivery of exogenous OSM is likely to be productive in accelerating diabetic wound closure.


Asunto(s)
Diabetes Mellitus , Repitelización , Animales , Ratones , Ratones Endogámicos C57BL , Oncostatina M , Cicatrización de Heridas/fisiología
10.
Front Oncol ; 11: 651692, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712602

RESUMEN

CD44highCD24low population has been previously reported as cancer stem cells (CSCs) in Oral Squamous Cell Carcinoma (OSCC). Increasing evidence suggests potential involvement of microRNA (miRNA) network in modulation of CSC properties. MiRNAs have thus emerged as crucial players in tumor development and maintenance. However, their role in maintenance of OSCC stem cells remains unclear. Here we report an elevated expression of miR-146a in the CD44highCD24low population within OSCC cells and primary HNSCC tumors. Moreover, over-expression of miR-146a results in enhanced stemness phenotype by augmenting the CD44highCD24low population. We demonstrate that miR-146a stabilizes ß-catenin with concomitant loss of E-cadherin and CD24. Interestingly, CD24 is identified as a novel functional target of miR-146a and ectopic expression of CD24 abrogates miR-146a driven potential CSC phenotype. Mechanistic analysis reveals that higher CD24 levels inhibit AKT phosphorylation leading to ß-catenin degradation. Using stably expressing miR-146a/CD24 OSCC cell lines, we also validate that the miR-146a/CD24/AKT loop significantly alters tumorigenic ability in vivo. Furthermore, we confirmed that ß-catenin trans-activates miR-146a, thereby forming a positive feedback loop contributing to stem cell maintenance. Collectively, our study demonstrates that miR-146a regulates CSCs in OSCC through CD24-AKT-ß-catenin axis.

11.
ACS Appl Bio Mater ; 4(2): 1693-1703, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35014516

RESUMEN

So far, near-infrared (NIR) light responsive nanostructures have been well-defined in cancer nanomedicine. However, poor penetration and retention in tumors are the limiting factors. Here, we report the ultrahigh penetration and retention of carbanosilica (graphene quantum dots, GQDs embedded mesoporous silica) in solid tumors. After NIR light exposure, quick (0.5 h) emission from the tumor area is observed that is further retained up to a week (tested up to 10 days) with a single dose administration of nanohybrids. Emissive and photothermally active GQDs and porous silica shell (about 31% drug loading) make carbanosilica a promising nanotheranostic agent exhibiting 68.75% tumor shrinking compared to without NIR light exposure (34.48%). Generated heat (∼52 °C) alters the permeability of tumor enhancing the accumulation of nanotheranostics into the tumor environment. Successive tumor imaging ensures the prolonged follow-up of image guided tumor regression due to synergistic therapeutic effect of nanohybrids.


Asunto(s)
Neoplasias/terapia , Dióxido de Silicio/uso terapéutico , Grafito/química , Humanos , Nanopartículas/química , Puntos Cuánticos/química , Dióxido de Silicio/farmacología
12.
Sci Rep ; 10(1): 20184, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33214614

RESUMEN

Urolithin A (UA) is a natural compound that is known to improve muscle function. In this work we sought to evaluate the effect of UA on muscle angiogenesis and identify the underlying molecular mechanisms. C57BL/6 mice were administered with UA (10 mg/body weight) for 12-16 weeks. ATP levels and NAD+ levels were measured using in vivo 31P NMR and HPLC, respectively. UA significantly increased ATP and NAD+ levels in mice skeletal muscle. Unbiased transcriptomics analysis followed by Ingenuity Pathway Analysis (IPA) revealed upregulation of angiogenic pathways upon UA supplementation in murine muscle. The expression of the differentially regulated genes were validated using quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC). Angiogenic markers such as VEGFA and CDH5 which were blunted in skeletal muscles of 28 week old mice were found to be upregulated upon UA supplementation. Such augmentation of skeletal muscle vascularization was found to be bolstered via Silent information regulator 1 (SIRT1) and peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1α) pathway. Inhibition of SIRT1 by selisistat EX527 blunted UA-induced angiogenic markers in C2C12 cells. Thus this work provides maiden evidence demonstrating that UA supplementation bolsters skeletal muscle ATP and NAD+ levels causing upregulated angiogenic pathways via a SIRT1-PGC-1α pathway.


Asunto(s)
Cumarinas/farmacología , Músculo Esquelético/efectos de los fármacos , NAD/metabolismo , Sirtuina 1/metabolismo , Adenosina Trifosfato/metabolismo , Administración Oral , Animales , Cumarinas/administración & dosificación , Perfilación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Reproducibilidad de los Resultados
13.
ACS Nano ; 14(10): 12732-12748, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32931251

RESUMEN

Bidirectional cell-cell communication involving exosome-borne cargo such as miRNA has emerged as a critical mechanism for wound healing. Unlike other shedding vesicles, exosomes selectively package miRNA by SUMOylation of heterogeneous nuclear ribonucleoproteinA2B1 (hnRNPA2B1). In this work, we elucidate the significance of exosome in keratinocyte-macrophage crosstalk following injury. Keratinocyte-derived exosomes were genetically labeled with GFP-reporter (Exoκ-GFP) using tissue nanotransfection (TNT), and they were isolated from dorsal murine skin and wound-edge tissue by affinity selection using magnetic beads. Surface N-glycans of Exoκ-GFP were also characterized. Unlike skin exosome, wound-edge Exoκ-GFP demonstrated characteristic N-glycan ions with abundance of low-base-pair RNA and was selectively engulfed by wound macrophages (ωmϕ) in granulation tissue. In vitro addition of wound-edge Exoκ-GFP to proinflammatory ωmϕ resulted in conversion to a proresolution phenotype. To selectively inhibit miRNA packaging within Exoκ-GFPin vivo, pH-responsive keratinocyte-targeted siRNA-hnRNPA2B1 functionalized lipid nanoparticles (TLNPκ) were designed with 94.3% encapsulation efficiency. Application of TLNPκ/si-hnRNPA2B1 to the murine dorsal wound-edge significantly inhibited expression of hnRNPA2B1 by 80% in epidermis compared to the TLNPκ/si-control group. Although no significant difference in wound closure or re-epithelialization was observed, the TLNPκ/si-hnRNPA2B1 treated group showed a significant increase in ωmϕ displaying proinflammatory markers in the granulation tissue at day 10 post-wounding compared to the TLNPκ/si-control group. Furthermore, TLNPκ/si-hnRNPA2B1 treated mice showed impaired barrier function with diminished expression of epithelial junctional proteins, lending credence to the notion that unresolved inflammation results in leaky skin. This work provides insight wherein Exoκ-GFP is recognized as a major contributor that regulates macrophage trafficking and epithelial barrier properties postinjury.


Asunto(s)
Exosomas , Animales , Queratinocitos , Macrófagos , Ratones , Piel , Cicatrización de Heridas
14.
Commun Biol ; 3(1): 284, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32504032

RESUMEN

Developing a nanotheranostic agent with better image resolution and high accumulation into solid tumor microenvironment is a challenging task. Herein, we established a light mediated phototriggered strategy for enhanced tumor accumulation of nanohybrids. A multifunctional liposome based nanotheranostics loaded with gold nanoparticles (AuNPs) and emissive graphene quantum dots (GQDs) were engineered named as NFGL. Further, doxorubicin hydrochloride was encapsulated in NFGL to exhibit phototriggered chemotherapy and functionalized with folic acid targeting ligands. Encapsulated agents showed imaging bimodality for in vivo tumor diagnosis due to their high contrast and emissive nature. Targeted NFGL nanohybrids demonstrated near infrared light (NIR, 750 nm) mediated tumor reduction because of generated heat and Reactive Oxygen Species (ROS). Moreover, NFGL nanohybrids exhibited remarkable ROS scavenging ability as compared to GQDs loaded liposomes validated by antitumor study. Hence, this approach and engineered system could open new direction for targeted imaging and cancer therapy.


Asunto(s)
Doxorrubicina/administración & dosificación , Oro/administración & dosificación , Grafito/administración & dosificación , Liposomas/administración & dosificación , Fototerapia/métodos , Nanomedicina Teranóstica/métodos , Células 3T3 , Animales , Antibióticos Antineoplásicos/administración & dosificación , Neoplasias de la Mama , Línea Celular Tumoral , Humanos , Rayos Infrarrojos , Nanopartículas del Metal/administración & dosificación , Ratones , Puntos Cuánticos/administración & dosificación
15.
Nanoscale ; 12(19): 10664-10684, 2020 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-32374338

RESUMEN

Acidic pH is a crucial intrinsic property of the microenvironment of most solid tumors. Hence, the use of pH sensitive tumor targeting nanoparticles is an attractive approach to enhance the therapeutic efficacy of anti-cancer agents in solid tumors. Chitosan nanoparticles (CHNPs) have been widely explored in the area of cancer drug delivery; nevertheless their true potential as a pH responsive targeted drug delivery vehicle in cancer therapy has not been deciphered yet as most of the research is limited to pH dependent stability and drug release. In the present study, we investigate the direct effect of pH in synergy with RGD peptide based targeting on the therapeutic efficacy of chitosan nanoparticles (RGD-CHNPs) in breast cancer. Furthermore, for the first time we performed a comprehensive study showing the anti-tumor, anti-migratory and anti-angiogenic effect of raloxifene (Rlx) loaded CHNPs in breast cancer. We prepared stable formulations of raloxifene encapsulated CHNPs and RGD-CHNPs by the nontoxic ionic gelation method. pH dependent studies revealed that NPs possess higher stability and zeta potential along with enhanced cellular uptake at acidic pH (as present in solid tumors) compared to physiological pH. Furthermore, RGD conjugation enhanced the in vitro cellular uptake of CHNPs in αvß3 integrin expressing breast cancer cells and induced higher cellular apoptosis in breast cancer cells which was further augmented by lower pH. Moreover, Rlx-RGD-CHNPs significantly inhibited breast cancer cell migration and angiogenesis. In vivo studies showed that Cy5.5 conjugated RGD-CHNPs can distinctly visualize tumors and Rlx-RGD-CHNPs significantly inhibit breast tumor growth without causing any toxic effect to normal tissue as confirmed by hematology and blood biochemical studies. Therefore, RGD-CHNPs could potentially enhance the therapeutic efficacy of chemotherapeutic drugs due to the synergistic effect of pH responsiveness and tumor specific targeting in breast cancer.


Asunto(s)
Neoplasias de la Mama , Quitosano , Nanopartículas , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Oligopéptidos , Clorhidrato de Raloxifeno/uso terapéutico , Microambiente Tumoral
16.
Int J Nanomedicine ; 14: 8285-8302, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31802866

RESUMEN

BACKGROUND: Curcumin has been widely used owing to its various medicinal properties including antitumor effects. However, its clinical application is limited by its instability, poor solubility and low bioavailability. Folic acid (FA)-functionalized nanoformulations may enhance the sustained release of an anticancer drug (curcumin) by tumor-specific targeting to improve therapeutic benefit. This study aims to design a nanoconjugate (NC) comprised of folate-curcumin-loaded gold-polyvinylpyrrolidone nanoparticles (FA-CurAu-PVP NPs) for targeted delivery in breast cancer model systems. METHODS: We developed curcumin-loaded FA-functionalized Au-PVP NCs by layer-by-layer assembly. The folic acid-curcumin Au-PVP NCs (FA-CurAu-PVP NCs) were characterized by ultraviolet-visible spectra, Fourier transform infrared spectroscopy, X-ray powder diffraction and thermogravimetric analysis. In vitro anticancer and antimigratory effects of NCs were examined by performing MTT and wound migration assays. The in vivo antitumor efficacy of NCs was investigated using a preclinical breast cancer orthotopic mouse model. RESULTS: Curcumin (40 µg/mL) was loaded along with conjugation of folate onto Au-PVP NPs to form FA-CurAu-PVP NCs. The size and charge of the NCs were increased gradually through layer-by-layer assembly and showed 80% release of curcumin at acidic pH. The NC did not show aggregation when incubated with human serum and mimicked an intrinsic peroxidase-like property in the presence of 3,3',5,5'-tetramethylbenzidine substrate. The MTT data using these NCs showed efficient anticancer activity at lower doses in estrogen/progesterone receptor (ER/PR)-negative cells compared with ER/PR-positive cells. Furthermore, the NCs did not show cytotoxicity at the investigated concentration in human breast epithelial and mouse fibroblast cell lines. They showed inhibitory effects on cell migration and high antitumor efficacy in in vivo analysis. CONCLUSION: These results suggest that folate-based tumor targeting using CurAu-PVP NCs is a promising approach for tumor-specific therapy of breast cancer without harming normal cells.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Ácido Fólico/química , Oro/química , Nanopartículas del Metal/química , Polímeros/química , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Curcumina/farmacología , Curcumina/uso terapéutico , Portadores de Fármacos/química , Liberación de Fármacos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Nanopartículas del Metal/ultraestructura , Ratones , Nanoconjugados/química , Povidona/química , Suero/metabolismo
17.
ACS Appl Bio Mater ; 2(10): 4324-4334, 2019 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-35021447

RESUMEN

Wound healing is a dynamic and complex process that requires a suitable environment to enhance the rapid healing process. In this context, fabrications of nanofibrous materials with antibiotic and antibacterial properties are becoming extremely important. In this present work, we report on the fabrication and characterization of electro-spun cellulose ether-PVA nanofiber mats loaded with halloysite clay (HNT) and gentamicin sulfate (GS) for faster wound healing applications. The morphology of nanofiber mats was examined by SEM and TEM. The average diameter of the nanofiber mats were in the range of 325 ± 30 nm. The physicochemical characterizations were done by FT-IR and XRD, which reveal the presence of HNT and GS into the nanofibers. The incorporation of halloysite gave good mechanical strength to the nanofiber mats. Swelling studies indicated the hydrophilicity of the mats. In vitro studies revealed that HNTs are nontoxic to L929 fibroblast cells and also promote cell growth and proliferation. The antibacterial property of HNT was also studied. The slow release of GS from the nanofiber mats was observed for a period of 18 days. The in vivo wound healing studies on the wistar rats for 21 days revealed the wound healing faster within 2 weeks by the incorporation of HNT and GS into the nanofiber mats and hence these nanofiber mats show great potential in acute and chronic wound healing applications.

18.
ACS Appl Bio Mater ; 2(8): 3312-3320, 2019 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-35030773

RESUMEN

Selective tissue visualization and localized tumor regression without affecting the surrounding healthy tissues are critical concerns in cancer nanomedicine. Importantly, the complete wrapping of a flimsy matrix like liposome by multifunctional graphene oxide is an interesting engineering idea for nanomedicine design. Moreover, designing a safe and biodegradable nanohybrid with significant theranostic ability is a current need for targeted combined therapies. Here, we report a comprehensive result of in vivo tumor diagnosis and phototriggered tumor regression using a biodegradable red emissive nanotheranostic system, viz., graphene oxide flakes fortified liposome (GOF-Lipo), functionalized with folic acid (FA): GOF-Lipo-FA. Graphene oxide support enhances the stability of drug-loaded liposomes in an extracellular environment that prevents the premature release of loaded anticancer drug from the liposomal cavity. Promising outcomes of tumor regression (∼300 to 25 mm3) from organized cellular and animal studies are demonstrated in this work. These studies reveal superior biocompatibility, deep intracellular localization, 4T1 breast tumor diagnosis, and long time tumor binding ability of an injected emissive nanohybrid. Overall, a single dose of designed multifunctional systems demonstrates the best tumor regression.

19.
Nanoscale ; 10(40): 19082-19091, 2018 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-30288516

RESUMEN

Specific targeting and phototriggered therapy in mouse model have recently emerged as the starting point of cancer theragnosis. Herein, we report a bioresponsive and degradable nanohybrid, a liposomal nanohybrid decorated with red emissive carbon dots, for localized tumor imaging and light-mediated tumor growth inhibition. Unsaturated carbon dots (C-dots) anchored to liposomes convert near-infrared (NIR) light into heat and also produce reactive oxygen species (ROS), demonstrating the capability of phototriggered cancer cell death and tumor regression. The photothermal and oxidative damage of breast tumor by the nonmetallic nanohybrid has also been demonstrated. Designed nanoparticles show excellent aqueous dispersibility, biocompatibility, light irradiated enhanced cellular uptake, release of reactive oxygen species, prolonged and specific tumor binding ability and good photothermal response (62 °C in 5 minutes). Safe and localized irradiation of 808 nm light demonstrates significant tumor growth inhibition and bioresponsive degradation of the fluorescent nanohybrid without affecting the surrounding healthy tissues.


Asunto(s)
Rayos Infrarrojos , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/terapia , Fototerapia/métodos , Puntos Cuánticos , Animales , Línea Celular Tumoral , Femenino , Humanos , Liposomas , Ratones , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Puntos Cuánticos/química , Puntos Cuánticos/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo
20.
Front Oncol ; 8: 72, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29616190

RESUMEN

Advancements in the early detection of cancer coupled with improved surgery, radiotherapy, and adjuvant therapy led to substantial increase in patient survival. Nevertheless, cancer metastasis is the leading cause of death in several cancer patients. The majority of these deaths are associated with metastatic relapse kinetics after a variable period of clinical remission. Most of the cancer recurrences are thought to be associated with the reactivation of dormant disseminated tumor cells (DTCs). In this review, we have summarized the cellular and molecular mechanisms related to DTCs and the role of microenvironmental niche. These mechanisms regulate the dormant state and help in the reactivation, which leads to metastatic outgrowth. Identification of novel therapeutic targets to eliminate these dormant tumor cells will be highly useful in controlling the metastatic relapse-related death with several cancers.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA