Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Int J Mol Sci ; 25(8)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38674017

RESUMEN

The mainstays of lung cancer pathogenesis are cell cycle progression dysregulation, impaired apoptosis, and unregulated cell proliferation. While individual microRNA (miR) targeting or delivering is a promising approach that has been extensively studied, combination of miR targeting can enhance therapeutic efficacy and overcome limitations present in individual miR regulations. We previously reported on the use of a miR-143 and miR-506 combination via transient transfections against lung cancer. In this study, we evaluated the effect of miR-143 and miR-506 under stable deregulations in A549 lung cancer cells. We used lentiviral transductions to either up- or downregulate the two miRs individually or in combination. The cells were sorted and analyzed for miR deregulation via qPCR. We determined the miR deregulations' effects on the cell cycle, cell proliferation, cancer cell morphology, and cell motility. Compared to the individual miR deregulations, the combined miR upregulation demonstrated a miR-expression-dependent G2 cell cycle arrest and a significant increase in the cell doubling time, whereas the miR-143/506 dual downregulation demonstrated increased cellular motility. Furthermore, the individual miR-143 and miR-506 up- and downregulations exhibited cellular responses lacking an apparent miR-expression-dependent response in the respective analyses. Our work here indicates that, unlike the individual miR upregulations, the combinatorial miR treatment remained advantageous, even under prolonged miR upregulation. Finally, our findings demonstrate potential advantages of miR combinations vs. individual miR treatments.


Asunto(s)
Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , MicroARNs , Regulación hacia Arriba , MicroARNs/genética , Humanos , Proliferación Celular/genética , Células A549 , Movimiento Celular/genética , Regulación hacia Arriba/genética , Ciclo Celular/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Línea Celular Tumoral , Apoptosis/genética
2.
J Pharmacol Exp Ther ; 385(1): 35-49, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36746610

RESUMEN

Existing vascular endothelial growth factor-oriented antiangiogenic approaches are known for their high potency. However, significant side effects associated with their use drive the need for novel antiangiogenic strategies. The small GTPase RhoA is an established regulator of actin cytoskeletal dynamics. Previous studies have highlighted the impact of endothelial RhoA pathway on angiogenesis. Rho-associate kinase (ROCK), a direct RhoA effector, is potently inhibited by Fasudil, a clinically relevant ROCK inhibitor. Here, we aimed to target the RhoA signaling in endothelial cells by generating Fasudil-encapsulated CD31-targeting liposomes as a potential antiangiogenic therapy. The liposomes presented desirable characteristics, preferential binding to CD31-expressing HEK293T cells and to endothelial cells, inhibited stress fiber formation and cytoskeletal-related morphometric parameters, and inhibited in vitro angiogenic functions. Overall, this work shows that the nanodelivery-mediated endothelial targeting of RhoA signaling can offer a promising strategy for angiogenesis inhibition in vascular-related diseases. SIGNIFICANCE STATEMENT: Systemic administration of antiangiogenic therapeutics induces side effects to non-targeted tissues. This study, among others, has shown the impact of the RhoA signaling in the endothelial cells and their angiogenic functions. Here, to minimize potential toxicity, this study generated CD31-targeting liposomes with encapsulated Fasudil, a clinically relevant Rho kinase inhibitor, and successfully targeted endothelial cells. In this proof-of-principle study, the efficient Fasudil delivery, its impact on the endothelial signaling, morphometric alterations, and angiogenic functions verify the benefits of site-targeted antiangiogenic therapy.


Asunto(s)
Células Endoteliales , Factor A de Crecimiento Endotelial Vascular , Humanos , Células Endoteliales/metabolismo , Células HEK293 , Liposomas , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Arch Biochem Biophys ; 648: 12-19, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29679536

RESUMEN

Curcumin reduces Hodgkin's lymphoma (HL) cell growth in vitro, but its unfavorable pharmacokinetics highlight the need for novel in vivo delivery systems. Thus, we explored whether formulation of curcumin in solid lipid nanoparticles (SLN-curc) or d-α-Tocopheryl polyethylene glycol 1000 succinate (TPGS) nanoparticles (TPGS-curc) could enhance its efficacy in mice. Curcumin formulated in SLN and in TPGS resulted in higher curcumin plasma levels in mice. Compared to vehicle-treated controls, SLN-curc and TPGS-curc reduced HL xenograft growth by 50.5% (p < 0.02) and 43.0% (p < 0.04), respectively, while curcumin reduced it by 35.8% (p < 0.05). In addition, SLN-curc reduced the expression of proteins involved in cell proliferation and apoptosis (XIAP and Mcl-1) in HL tumor extracts. In HL cells in culture, curcumin decreased the expression of relevant anti-inflammatory cytokines (IL-6 and TNF-α) in a concentration-dependent manner. Moreover, when given in combination with bleomycin, doxorubicin and vinblastine, curcumin showed an additive growth inhibitory effect. In conclusion, SLNs appear as an appropriate and effective drug delivery system for curcumin. Given the efficacy of SLN-curc and the enhanced growth inhibitory effect when combined with chemotherapeutic drugs, we speculate that curcumin, when appropriately formulated, is a promising adjuvant agent for the treatment of HL and merits further evaluation.


Asunto(s)
Curcumina/química , Curcumina/farmacología , Enfermedad de Hodgkin/tratamiento farmacológico , Lípidos/química , Nanopartículas/química , Animales , Línea Celular Tumoral , Curcumina/farmacocinética , Curcumina/uso terapéutico , Composición de Medicamentos , Femenino , Interleucina-6/metabolismo , Ratones , Distribución Tisular , Factor de Necrosis Tumoral alfa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Carcinogenesis ; 37(4): 420-429, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26905586

RESUMEN

Given that glioblastoma multiforme (GBM) is associated with poor prognosis, new agents are urgently needed. We developed phospho-glycerol-ibuprofen-amide (PGIA), a novel ibuprofen derivative, and evaluated its safety and efficacy in preclinical models of GBM, and its mechanism of action using human GBM cells and animal tumor models. Furthermore, we explored whether formulating PGIA in polymeric nanoparticles could enhance its levels in the brain. PGIA was 3.7- to 5.1-fold more potent than ibuprofen in suppressing the growth of human GBM cell lines. PGIA 0.75× IC50 inhibited cell proliferation by 91 and 87% in human LN-229 and U87-MG GBM cells, respectively, and induced strong G1/S arrest.In vivo, compared with control, PGIA reduced U118-MG and U87-MG xenograft growth by 77 and 56%, respectively (P< 0.05), and was >2-fold more efficacious than ibuprofen. Normal human astrocytes were resistant to PGIA, indicating selectivity. Mechanistically, PGIA reduced cyclin D1 levels in a time- and concentration-dependent manner in GBM cells and in xenografts. PGIA induced cyclin D1 degradation via the proteasome pathway and induced dephosphorylation of GSK3ß, which was required for cyclin D1 turnover. Furthermore, cyclin D1 overexpression rescued GBM cells from the cell growth inhibition by PGIA. Moreover, the formulation of PGIA in poly-(L)-lactic acid poly(ethylene glycol) polymeric nanoparticles improved its pharmacokinetics in mice, delivering PGIA to the brain. PGIA displays strong efficacy against GBM, crosses the blood-brain barrier when properly formulated, reaching the target tissue, and establishes cyclin D1 as an important molecular target. Thus, PGIA merits further evaluation as a potential therapeutic option for GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Ciclina D1/metabolismo , Glioblastoma/patología , Ibuprofeno/análogos & derivados , Animales , Línea Celular Tumoral , Ibuprofeno/química , Ratones
5.
Small ; 12(35): 4837-4848, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27432595

RESUMEN

Dual stimuli-sensitive mixed polymeric micelles (MM) are developed for co-delivery of the endogenous tumor suppressor miRNA-34a and the chemotherapeutic agent doxorubicin (Dox) into cancer cells. The novelty of the system resides in two stimuli-sensitive prodrugs, a matrix metalloproteinase 2 (MMP2)-sensitive Dox conjugate and a reducing agent (glutathione, GSH)-sensitive miRNA-34a conjugate, self-assembled in a single particle decorated with a polyethylene glycol corona for longevity, and a cell-penetrating peptide (TATp) for enhanced intracellular delivery. The MMP2-sensitivity of the system results in threefold higher cytotoxicity in MMP2-overexpressing HT1080 cells compared to low MMP2-expressing MCF7 cells. Cellular internalization of Dox increases by more than 70% after inclusion of TATp to the formulation. MMP2-sensitive MM also inhibits proliferation and migration of HT1080 cells. Moreover, GSH-sensitive MM allows for an efficient downregulation of Bcl2, survivin, and notch1 (65%, 55%, and 46%, respectively) in HT1080 cells. Combination of both conjugates in dual sensitive MM reduces HT1080 cell viability to 40% and expression of Bcl2 and survivin. Finally, 50% cell death is observed in 3D models of tumor mass. The results confirm the potential of the MM to codeliver miRNA-34a and doxorubicin triggered by dual stimuli inherent of tumor tissues.


Asunto(s)
Doxorrubicina/uso terapéutico , Sistemas de Liberación de Medicamentos , Técnicas de Transferencia de Gen , Micelas , MicroARNs/administración & dosificación , Neoplasias/tratamiento farmacológico , Tamaño de la Partícula , Polímeros/química , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , MicroARNs/metabolismo , Neoplasias/genética , Neoplasias/patología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
6.
Pharm Res ; 33(12): 2943-2953, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27528390

RESUMEN

PURPOSE: Cisplatin, is recognized as a first line therapeutic for the treatment of non-small cell lung cancer (NSCLC). Cisplatin resistance is identified as the most detrimental complication during treatment and has been associated with upregulation of several genes, such as the anti-apoptotic gene survivin. In this study, we have evaluated the cytotoxic activity of lipid (C6 and C8)-modified platinum compounds in combination with a survivin-silencing siRNA against cisplatin resistant tumors. METHODS: We synthesized and characterized several lipid-modified platinum compounds and evaluated their cytotoxic activity alone or in combination with survivin-silencing siRNA in vitro and in vivo against A549DDP cells and in vivo in tumor xenograft model. RESULTS: The lipid-modified compounds exhibited significantly stronger cytotoxic activity in vitro compared to cisplatin, with CDDP-C6 and CDDP-C8 producing the most pronounced effect, in both A549 and A549DDP cells. Pre-treatment of the A549DDP cells with survivin-silencing siRNA enhanced the cytotoxic activity of these compounds. In vivo, the co-treatment of the survivin-silencing siRNA and CDDP-C8 produced the strongest tumor growth inhibition effect (64.5%, p < 0.05) on a cancer mouse model of chemoresistant lung cancer. In contrast, cisplatin treatment exhibited no significant tumor growth inhibition (4.5%, no p). CONCLUSIONS: Co-treatment of lipid-modified compounds and survivin-silencing siRNA can constitute a reliable alternative to cisplatin treatment for cisplatin-resistant lung tumors that merit further evaluation.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cisplatino/análogos & derivados , Cisplatino/uso terapéutico , Proteínas Inhibidoras de la Apoptosis/genética , Lípidos/química , Neoplasias Pulmonares/tratamiento farmacológico , ARN Interferente Pequeño/genética , Células A549 , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/genética , Proliferación Celular , Resistencia a Antineoplásicos , Femenino , Silenciador del Gen , Humanos , Ácido Hialurónico/química , Neoplasias Pulmonares/genética , Ratones Desnudos , Nanopartículas/química , Polietilenglicoles/química , Polietileneimina/química , Survivin
7.
Nanomedicine ; 12(3): 589-600, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26656632

RESUMEN

In this study, we have formulated redox-responsive epidermal growth factor receptor (EGFR)-targeted type B gelatin nanoparticles as a targeted vector for systemic delivery of gemcitabine therapy in pancreatic cancer. The gelatin nanoparticles were formed by ethanol-induced desolvation process to encapsulate the bound drug. The surface of the nanoparticles was decorated either with poly(ethylene glycol) (PEG) chains to impart enhanced circulation time or with EGFR targeting peptide to confer target specificity. Our in vitro studies in Panc-1 human pancreatic ductal adenocarcinoma cells confirm that gemcitabine encapsulated in EGFR-targeted gelatin nanoparticles, released through disulfide bond cleavage, had a significantly improved cytotoxic profile. Further, the in vivo anticancer activity was evaluated in an orthotopic pancreatic adenocarcinoma tumor bearing SCID beige mice, which confirmed that EGFR-targeted gelatin nanoparticles could efficiently deliver gemcitabine to the tumor leading to higher therapeutic benefit as compared to the drug in solution. FROM THE CLINICAL EDITOR: The treatment of pancreatic cancer remains unsatisfactory, with an average 5-year survival of less than 5%. New treatment modalities are thus urgently needed. In this study, the authors presented their formulation of redox-responsive epidermal growth factor receptor (EGFR)-targeted type B gelatin nanoparticles as a carrier for gemcitabine. In-vitro and in-vivo experiments showed encouraging results. It is hoped that the findings would provide a novel and alternative drug delivery platform for the future.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antimetabolitos Antineoplásicos/administración & dosificación , Desoxicitidina/análogos & derivados , Receptores ErbB/metabolismo , Gelatina/metabolismo , Nanopartículas/metabolismo , Páncreas/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/uso terapéutico , Sistemas de Liberación de Medicamentos , Gelatina/química , Humanos , Ratones , Ratones SCID , Nanopartículas/química , Páncreas/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Gemcitabina
8.
Clin Immunol ; 160(1): 46-58, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25842185

RESUMEN

Cell secreted exosomes (30-100nm vesicles) play a major role in intercellular communication due to their ability to transfer proteins and nucleic acids from one cell to another. Depending on the originating cell type and the cargo, exosomes can have immunosuppressive or immunostimulatory effects, which have potential application as immunotherapies for cancer and autoimmune diseases. Cellular components shed from tumor cells or antigen presenting cells (APCs), such as dendritic cells, macrophages and B cells, have been shown to be efficiently packaged in exosomes. In this review, we focus on the application of exosomes as nanocarriers and immunological agents for cancer and autoimmune immunotherapy. APC-derived exosomes demonstrate effective therapeutic efficacy for the treatment of cancer and experimental autoimmune diseases such as rheumatoid arthritis, inflammatory bowel disease, and multiple sclerosis. In addition to their intrinsic immunomodulating activity, exosomes have many advantages over conventional nanocarriers for drug and gene delivery.


Asunto(s)
Enfermedades Autoinmunes/terapia , Portadores de Fármacos , Exosomas , Inmunoterapia/métodos , Neoplasias/terapia , Células Presentadoras de Antígenos/inmunología , Humanos , Inflamación/terapia
9.
Pharm Res ; 32(5): 1663-75, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25392229

RESUMEN

PURPOSE: The purpose of the study was to evaluate the metabolism, pharmacokinetics and efficacy of phospho-NSAIDs in Ces1c-knockout mice. METHODS: Hydrolysis of phospho-NSAIDs by Ces1c was investigated using Ces1c-overexpressing cells. The rate of phospho-NSAID hydrolysis was compared between wild-type, Ces1c+/- and Ces1c-/- mouse plasma in vitro, and the effect of plasma Ces1c on the cytotoxicity of phospho-NSAIDs was evaluated. Pharmacokinetics of phospho-sulindac was examined in wild-type and Ces1c-/- mice. The impact of Ces1c on the efficacy of phospho-sulindac was investigated using lung and pancreatic cancer models in vivo. RESULTS: Phospho-NSAIDs were extensively hydrolyzed in Ces1c-overexpressing cells. Phospho-NSAID hydrolysis in wild-type mouse plasma was 6-530-fold higher than that in the plasma of Ces1c-/- mice. Ces1c-expressing wild-type mouse serum attenuated the in vitro cytotoxicity of phospho-NSAIDs towards cancer cells. Pharmacokinetic studies of phospho-sulindac using wild-type and Ces1c-/- mice demonstrated 2-fold less inactivation of phospho-sulindac in the latter. Phospho-sulindac was 2-fold more efficacious in inhibiting the growth of lung and pancreatic carcinoma in Ces1c -/- mice, as compared to wild-type mice. CONCLUSIONS: Our results indicate that intact phospho-NSAIDs are the pharmacologically active entities and phospho-NSAIDs are expected to be more efficacious in humans than in rodents due to their differential expression of carboxylesterases.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antineoplásicos/uso terapéutico , Aspirina/análogos & derivados , Hidrolasas de Éster Carboxílico/genética , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Ibuprofeno/análogos & derivados , Organofosfatos/uso terapéutico , Compuestos Organofosforados/uso terapéutico , Sulindac/análogos & derivados , Animales , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacocinética , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Aspirina/metabolismo , Aspirina/farmacocinética , Aspirina/uso terapéutico , Hidrolasas de Éster Carboxílico/sangre , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Ibuprofeno/metabolismo , Ibuprofeno/farmacocinética , Ibuprofeno/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organofosfatos/metabolismo , Organofosfatos/farmacocinética , Compuestos Organofosforados/metabolismo , Compuestos Organofosforados/farmacocinética , Sulindac/metabolismo , Sulindac/farmacocinética , Sulindac/uso terapéutico
10.
J Pharmacol Exp Ther ; 351(1): 61-6, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25047517

RESUMEN

Esterase hydrolysis of drugs can accelerate their elimination, thereby limiting their efficacy. Polyethylene glycol (PEG) covalently attached to drugs (pegylation) is known to improve the efficiency of many drugs. Using as a test agent the novel phospho-ibuprofen (PI), we examined whether pegylation of PI could abrogate its hydrolytic degradation by esterases; PI, known to inhibit colon cancer growth, has a carboxylic ester hydrolyzable by carboxylesterases (CES). We covalently attached mPEG-2000 to PI (PI-PEG) and studied its stability by exposing it to cells overexpressing CES and by administering it to mice. We also evaluated PI-PEG's anticancer efficacy in human colon cancer xenografts and in Apc(min/+) mice. PI-PEG was stable in the presence of cells overexpressing CES1 or CES2, whereas PI was extensively hydrolyzed (90.2 ± 0.7%, 14.3 ± 1.1%, mean ± S.E.M.). In mice, PI was nearly completely hydrolyzed. Intravenous administration of PI-PEG resulted in significant levels in blood and in colon cancer xenografts (xenograft values in parentheses): area under the curve for 0-24 hours = 2351 (2621) (nmol/g) × h; Cmax = 1965 (886) nmol/g; Tmax = 0.08 (2) hour. The blood levels of ibuprofen, its main hydrolytic product, were minimal. Compared with controls, PI-PEG inhibited the growth of the xenografts by 74.8% (P < 0.01) and reduced intestinal tumor multiplicity in Apc(min/+) mice by 73.1% (P < 0.01), prolonging their survival (100% versus 55.1% of controls; P = 0.013). Pegylation protects PI from esterase hydrolysis and improves its pharmacokinetics. In preclinical models of colon cancer, PI-PEG is a safe and efficacious agent that merits further evaluation.


Asunto(s)
Antineoplásicos/farmacocinética , Ibuprofeno/análogos & derivados , Organofosfatos/farmacocinética , Polietilenglicoles/química , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Células HEK293 , Humanos , Hidrólisis , Ibuprofeno/efectos adversos , Ibuprofeno/síntesis química , Ibuprofeno/farmacocinética , Ibuprofeno/uso terapéutico , Ratones , Organofosfatos/efectos adversos , Organofosfatos/síntesis química , Organofosfatos/uso terapéutico , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Pharm Res ; 30(6): 1471-82, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23483440

RESUMEN

PURPOSE: Formulate phospho-sulindac (P-S, OXT-328) in a Pluronic hydrogel to be used as a topical anti-inflammatory agent and study its efficacy, safety and pharmacokinetics in an arthritis model. METHODS: LEW/crlBR rats with Freund's adjuvant-induced arthritis were treated with P-S formulated in Pluronic hydrogel (PSH). We determined the clinical manifestations of arthritis including the locomotor activity of the rats; evaluated joints for inflammation, bone resorption, cartilage damage, COX-2 expression and NF-κB activation; assayed plasma IL-6 and IL-10 levels; and studied the pharmacokinetics of P-S in rats after topical or oral administration. RESULTS: PSH applied at the onset of arthritis or when arthritis was fully developed, suppressed it by 56-82%, improved the locomotor activity of the rats 2.1-4.4 fold, suppressed synovial inflammation, bone resorption, cartilage damage, NF-κB activation and COX-2 expression but not plasma IL-6 and IL-10 levels. There were no side effects. PSH produced rapidly high local levels of P-S with <14% of P-S reaching the circulation, while orally administered P-S was rapidly metabolized generating much lower joint levels of P-S. CONCLUSIONS: Topical application of PSH is efficacious and safe in the treatment of Freund's adjuvant-induced arthritis; has a favorable pharmacokinetic profile; and likely acts by suppressing key pro-inflammatory signaling pathways.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Hidrogel de Polietilenoglicol-Dimetacrilato/administración & dosificación , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacocinética , Compuestos Organofosforados/administración & dosificación , Compuestos Organofosforados/farmacocinética , Sulindac/análogos & derivados , Sulindac/administración & dosificación , Sulindac/farmacocinética , Administración Oral , Administración Tópica , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacocinética , Artritis Experimental/sangre , Artritis Experimental/inducido químicamente , Artritis Experimental/metabolismo , Resorción Ósea/sangre , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Cartílago/efectos de los fármacos , Cartílago/metabolismo , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Femenino , Inflamación/sangre , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-10/sangre , Interleucina-10/metabolismo , Interleucina-6/sangre , Interleucina-6/metabolismo , Articulaciones/efectos de los fármacos , Articulaciones/metabolismo , Actividad Motora/efectos de los fármacos , FN-kappa B/metabolismo , Ratas , Ratas Endogámicas Lew , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/metabolismo
12.
J Vis Exp ; (197)2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37590510

RESUMEN

The lymphatic system participates in the regulation of immune surveillance, lipid absorption, and tissue fluid balance. The isolation of murine lymphatic endothelial cells is an important process for lymphatic research, as it allows the performance of in vitro and biochemical experiments on the isolated cells. Moreover, the development of Cre-lox technology has enabled the tissue-specific deficiency of genes that cannot be globally targeted, leading to the precise determination of their role in the studied tissues. The dissection of the role of certain genes in lymphatic physiology and pathophysiology requires the use of lymphatic-specific promoters, and thus, the experimental verification of the expression levels of the targeted genes. Methods for efficient isolation of lymphatic endothelial cells from wild-type or transgenic mice enable the use of ex vivo and in vitro assays to study the mechanisms regulating the lymphatic functions and the identification of the expression levels of the studied proteins. We have developed, standardized and present a protocol for the efficient isolation of murine dermal lymphatic endothelial cells (DLECs) via magnetic bead purification based on LYVE-1 expression. The protocol outlined aims to equip researchers with a tool to further understand and elucidate important players of lymphatic endothelial cell functions, especially in facilities where fluorescence-activated cell sorting equipment is not available.


Asunto(s)
Células Endoteliales , Vasos Linfáticos , Ratones , Animales , Ratones Transgénicos , Disección , Líquido Extracelular
13.
J Control Release ; 361: 212-235, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37517543

RESUMEN

The characteristics of the primary tumor blood vessels and the tumor microenvironment drive the enhanced permeability and retention (EPR) effect, which confers an advantage towards enhanced delivery of anti-cancer nanomedicine and has shown beneficial effects in preclinical models. Increased vascular permeability is a landmark feature of the tumor vessels and an important driver of the EPR. The main focus of this review is the endothelial regulation of vascular permeability. We discuss current challenges of targeting vascular permeability towards clinical translation and summarize the structural components and mechanisms of endothelial permeability, the principal mediators and signaling players, the targeted approaches that have been used and their outcomes to date. We also critically discuss the effects of the tumor-infiltrating immune cells, their interplay with the tumor vessels and the impact of immune responses on nanomedicine delivery, the impact of anti-angiogenic and tumor-stroma targeting approaches, and desirable nanoparticle design approaches for greater translational benefit.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Antineoplásicos/química , Sistemas de Liberación de Medicamentos , Neoplasias/patología , Nanopartículas/química , Permeabilidad , Nanomedicina , Microambiente Tumoral
14.
Int J Pharm ; 639: 122950, 2023 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-37059241

RESUMEN

Nanocarrier-mediated administration of chemotherapeutic drugs can increase the therapeutic index of drugs by reducing off-target site toxicity. Ligand-targeted drug delivery can be utilized to deliver chemotherapeutic drugs to cancer cells selectively and specifically. Here we report the evaluation of a lyophilized formulation of a liposome containing a peptidomimetic-doxorubicin conjugate for targeted delivery of doxorubicin to HER2-positive cancer cells. The lyophilized liposomal formulation exhibited improved release of peptidomimetic-doxorubicin conjugate at pH 6.5 compared to 7.4 and improved cellular uptake in cancer cells at pH 6.5. In vivo studies indicated that pH-sensitive formulation exhibited site-specific formulation delivery and improved anticancer efficacy than free doxorubicin. The findings suggested that combining a lyophilized pH-sensitive liposomal formulation containing trehalose as lyoprotectant with a targeting ligand coupled cytotoxic agent is a potential method for cancer chemotherapy while maintaining long-term stability at 4 °C of the liposome formulation.


Asunto(s)
Neoplasias Pulmonares , Peptidomiméticos , Humanos , Liposomas , Antibióticos Antineoplásicos , Ligandos , Línea Celular Tumoral , Doxorrubicina , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Pulmonares/tratamiento farmacológico
15.
Pharm Res ; 29(6): 1435-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22072052

RESUMEN

PURPOSE: To incorporate phospho-ibuprofen (P-I), a lipophilic, water insoluble novel anti-cancer agent, into pegylated liposomes and upon formulation optimization to evaluate its antitumor activity in vitro and in vivo. METHODS: P-I loaded liposomes were prepared using the thin-film hydration method, and characterized for size, zeta potential, drug content and drug release. We examined their physical stability by particle size changes; their lyophilization ability in the presence of cryoprotectants; and their antitumor activity in vitro in human cancer cell lines and in vivo in a xenograft murine model. RESULTS: P-I was successfully loaded into liposomes consisting of soy-PC and PEG(2000)-PE. These liposomes were <150 nm in diameter; exhibited prolonged stability in suspension and can be lyophilized using sucrose as cryoprotectant. P-I liposomes inhibited the growth of human cancer cell lines in vitro and in vivo of xenograft in nude mice to a greater extent than free P-I. CONCLUSIONS: High levels of P-I can be incorporated into liposomes which can be lyophilized in the presence of sucrose and showed good stability upon storage. Moreover, these drug-incorporating liposomes were capable of inhibiting the growth of xenografted tumors in mice more effectively than free P-I. These results justify further development of the P-I liposomes.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Ibuprofeno/análogos & derivados , Lípidos/química , Organofosfatos/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Química Farmacéutica , Neoplasias del Colon/patología , Crioprotectores/química , Relación Dosis-Respuesta a Droga , Composición de Medicamentos , Estabilidad de Medicamentos , Femenino , Liofilización , Humanos , Ibuprofeno/administración & dosificación , Ibuprofeno/química , Ibuprofeno/farmacología , Concentración 50 Inhibidora , Inyecciones Intraperitoneales , Cinética , Liposomas , Ratones , Ratones SCID , Organofosfatos/administración & dosificación , Organofosfatos/química , Tamaño de la Partícula , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Solubilidad , Propiedades de Superficie , Tecnología Farmacéutica/métodos , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Pharmaceutics ; 14(12)2022 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-36559106

RESUMEN

In the generational evolution of nano-based drug delivery carriers, active targeting has been a major milestone for improved and selective drug accumulation in tissues and cell types beyond the existing passive targeting capabilities. Among the various active targeting moieties, chlorotoxin, a peptide extracted from scorpions, demonstrated promising tumor cell accumulation and selection. With lung cancer being among the leading diagnoses of cancer-related deaths in both men and women, novel therapeutic methodologies utilizing nanotechnology for drug delivery emerged. Given chlorotoxin's promising biological activity, we explore its potential against lung cancer and its utilization for active targeting against this cancer's tumor cells. Our analysis indicates that despite the extensive chlorotoxin's research against glioblastoma, lung cancer research with the molecule has been limited, despite some promising early results.

17.
Methods Mol Biol ; 2193: 77-83, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32808260

RESUMEN

Wound regeneration is a complex process, which necessitates proper coordination among the inflammatory response, vascularization, matrix formation, and reformation of epithelial tissue. It is a unique process, where healing and regeneration take place simultaneously. Matrix formation is the first critical stage that starts the communication between the keratinocytes, fibroblasts, and integrins. This, in turn, stimulates the differentiation of monocytes into macrophages, to produce cytokines for fibroblasts. This phenomenon is the crucial part for the keratinocyte migration and epithelialization to fill the wound. To understand the complex procedure of wound regeneration, there is a need for easy, convenient, and low-cost approaches that will simulate the wound-repairing process. Scratch assay or cellular migration assay is one of the most convenient and affordable approaches, commonly used by the scientific community. In this chapter, we present the fundamental principles of the experimental procedures required for the Scratch assay.


Asunto(s)
Ensayos de Migración Celular/métodos , Movimiento Celular/fisiología , Técnicas In Vitro/métodos , Cicatrización de Heridas/fisiología , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Integrinas/genética , Macrófagos/metabolismo , Macrófagos/patología , Regeneración/genética , Cicatrización de Heridas/genética
18.
Pharmaceutics ; 13(7)2021 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-34371766

RESUMEN

Despite the apparent advantages for long-term treatment and local therapies against intestinal diseases, the oral delivery of nucleic acids has been challenging due to unfavorable physiological conditions for their stability. In this study, a novel nanodelivery system of PEG-PCL nanoparticles with encapsulated nucleic acids-mannosylated PEI (Man-PEI) complexes was developed for intestinal delivery. We complexed model nucleic acids with Man-PEI at the optimal N/P ratio of 20:1 for in vitro and in vivo analyses. Cells were transfected in vitro and analyzed for gene expression, receptor-mediated uptake, and PEG-PCL nanoparticles' toxicity. We also evaluated the nucleic acid's stability in the nanocarrier during formulation, and under simulated gastrointestinal environments or the presence of nucleases. Finally, we assessed the biodistribution for the PEG-PCL nanoparticles with encapsulated complexes and their ability to transfect intestinal cells in vivo. Nucleic acids complexed with Man-PEI were protected from degradation against nucleases. In comparison to the parent compound PEI, Man-PEI transfected the cells with an overall higher potency. Competition assay indicated receptor-mediated endocytosis promoted by mannose receptors. The PEG-PCL nanoparticles with Man-PEI/plasmid complexes indicated minimal cytotoxicity. The nanocarrier successfully protected the complexes in a simulated gastric fluid environment and released them in a simulated intestinal fluid environment, promoted by the presence of lipases. The oral administration of the PEG-PCL nanoparticles with encapsulated Man-PEI/plasmid complexes transfected intestinal cells with the plasmid in vivo, while presenting a time-dependent progression through the intestines. Conclusively, our carrier system can deliver genetic material to the GI tract and actively target mannose receptor overexpressing cells.

19.
Front Oncol ; 11: 705634, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956857

RESUMEN

Angiogenesis is a key mechanism for tumor growth and metastasis and has been a therapeutic target for anti-cancer treatments. Intensive vascular growth is concomitant with the rapidly proliferating tumor cell population and tumor outgrowth. Current angiogenesis inhibitors targeting either one or a few pro-angiogenic factors or a range of downstream signaling molecules provide clinical benefit, but not without significant side effects. miRNAs are important post-transcriptional regulators of gene expression, and their dysregulation has been associated with tumor progression, metastasis, resistance, and the promotion of tumor-induced angiogenesis. In this mini-review, we provide a brief overview of the current anti-angiogenic approaches, their molecular targets, and side effects, as well as discuss existing literature on the role of miRNAs in angiogenesis. As we highlight specific miRNAs, based on their activity on endothelial or cancer cells, we discuss their potential for anti-angiogenic targeting in cancer as adjuvant therapy and the importance of angiogenesis being evaluated in such combinatorial approaches.

20.
Oncol Rep ; 45(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33649787

RESUMEN

Lung cancer (LC) and pancreatic cancer (PC) are the first and fourth leading causes of cancer­related deaths in the US. Deregulated cell cycle progression is the cornerstone for rapid cell proliferation, tumor development, and progression. Here, we provide evidence that a novel combinatorial miR treatment inhibits cell cycle progression at two phase transitions, through their activity on the CDK4 and CDK1 genes. Following transfection with miR­143 and miR­506, we analyzed the differential gene expression of CDK4 and CDK1, using qPCR or western blot analysis, and evaluated cell cycle inhibition, apoptosis and cytotoxicity. The combinatorial miR­143/506 treatment downregulated CDK4 and CDK1 levels, and induced apoptosis in LC cells, while sparing normal lung fibroblasts. Moreover, the combinatorial miR treatment demonstrated a comparable activity to clinically tested cell cycle inhibitors in inhibiting cell cycle progression, by presenting substantial inhibition at the G1/S and G2/M cell cycle transitions. More importantly, the miR­143/506 treatment presented a broader application, effectively downregulating CDK1 and CDK4 levels, and reducing cell growth in PC cells. These findings suggest that the miR­143/506 combination acts as a promising approach to inhibit cell cycle progression for cancer treatment with minimal toxicity to normal cells.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Neoplasias Pulmonares/metabolismo , MicroARNs/metabolismo , Neoplasias Pancreáticas/metabolismo , Aminopiridinas/farmacología , Antineoplásicos/farmacología , Proteína Quinasa CDC2/antagonistas & inhibidores , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Procesos de Crecimiento Celular , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Regulación hacia Abajo , Fibroblastos/citología , Fibroblastos/metabolismo , Flavonoides/farmacología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , MicroARNs/administración & dosificación , MicroARNs/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Piperidinas/farmacología , Purinas/farmacología , Transfección , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda