Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.594
Filtrar
1.
Sci Rep ; 14(1): 18311, 2024 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-39112669

RESUMEN

Finding a novel drug delivery system (DDS) represents one of the most challenging endeavors in cancer therapy. Hence, in this study, we developed a new biocompatible and biodegradable zinc-based nanoscale metal-organic framework (Zn-NMOF) coated with folic acid (FA) functionalized chitosan (CS) to facilitate targeted delivery of doxorubicin (D), a standard chemotherapeutic agent, into breast cancer cells. The synthesis of the NMOF-CS-FA-D nanocomposite preceded its comprehensive characterization via FT-IR, DLS, XRD, SEM, and TEM analyses. Subsequent in vitro studies were conducted on MCF-7 breast cancer cells and HFF-1 normal cells, encompassing assessments of cell viability, expression levels of apoptotic and autophagy genes, cell cycle arrest, and apoptotic analyses. The size of the NMOF-CS-FA-D particles was determined to be less than 80 nm, with a drug loading efficiency of 72 ± 5%. The release kinetics of DOX from the nanocomposite were investigated, revealing controlled release behavior at pH 7.4 and accelerated release at pH 5.0, which is conducive to drug delivery into cancer cells. In vitro results indicated a 17.39% ± 6.34 cell viability after 24 h of treatment with a 40 nM concentration of the NMOF-CS-FA-D nanocomposite. Furthermore, the expression levels of Caspase-9 and BAX, key apoptotic genes, along with BECLIN1, an autophagy gene, were found to increase by two-fold, four-fold, and two-fold, respectively, following 5 h of treatment with the nanocomposite. Additionally, analysis of cell cycle distribution revealed 15.4 ± 2% of cells in the sub-G1 phase, indicative of apoptotic cells, and 31.9% of cells undergoing early and late apoptosis in MCF-7 cells. Collectively, these findings underscore the potential of the NMOF-CS-FA-D nanocomposite in inhibiting cancer cell proliferation with low side effects.


Asunto(s)
Apoptosis , Neoplasias de la Mama , Quitosano , Doxorrubicina , Estructuras Metalorgánicas , Nanocompuestos , Zinc , Humanos , Nanocompuestos/química , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Células MCF-7 , Zinc/química , Zinc/farmacología , Quitosano/química , Femenino , Doxorrubicina/farmacología , Doxorrubicina/química , Doxorrubicina/administración & dosificación , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ácido Fólico/química , Ácido Fólico/farmacología , Sistemas de Liberación de Medicamentos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Liberación de Fármacos , Portadores de Fármacos/química , Caspasa 9/metabolismo , Caspasa 9/genética , Autofagia/efectos de los fármacos
2.
Anal Chim Acta ; 1319: 342963, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39122276

RESUMEN

BACKGROUND: NO2- and S2- are two kinds of common toxic anions widely distributed in environmental water, soil and food products. Human beings have suffered a lot of diseases from intake of excessive NO2- or S2-, i.e., infantile methemoglobin, cancer and even to death. Although tremendous efforts have been afforded to monitor NO2- and S2-, most were high instrument-depended with complex processing procedures. To keep food safety and to protect human health, it will be a huge challenge to develop a convenient and efficient way to monitor S2- and NO2- in practice. RESULTS: A kind of folic acid capping Bi3+-doped Ag quantum dots (FA@Bi3+-Ag QDs) was developed for the first time by one-pot homogeneous reduced self-assembly. Not only did FA@Bi3+-Ag QDs possess intrinsic fluorescent property, it expressed synergistic peroxidase-like activity to catalyze the redox of 3,3',5,5'-tetramethylbenzidine (TMB) and H2O2 with Km/vmax of 0.087 mM/6.61 × 10-8 M s-1 and 6.42 mM/6.25 × 10-7 M s-1 respectively. Interestingly, trace S2- could exclusively alter its fluorescent property and peroxidase-like activity, exhibiting significant hypochromic and "turn-on" fluorescent effects. While trace NO2- could make FA@Bi3+-Ag QDs-TMB-H2O2 system hyperchromic. Under the optimized conditions, FA@Bi3+-Ag QDs were applied for dual-mode recognition of S2- and visual sensing of NO2- in real food samples with satisfactory recoveries, i.e., 100.7-107.9 %/95.8-104.7 % and 97.2-104.8 % respectively. The synergistic enzyme-mimic mechanism of FA@Bi3+-Ag QDs and its selective response mechanisms to S2- and NO2- were also proposed. SIGNIFICANCE: This represents the first nanozyme-based FA@Bi3+-Ag QDs system for dual-mode recognition of S2- and visual sensing of NO2-, well meeting the basic requirement in drinking water set by WHO. It will offer a promising way for multi-mode monitoring of different pollution using the same nanozyme-based sensor.


Asunto(s)
Ácido Fólico , Puntos Cuánticos , Plata , Puntos Cuánticos/química , Ácido Fólico/química , Plata/química , Nitritos/análisis , Nitritos/química , Peróxido de Hidrógeno/química , Humanos , Bencidinas/química , Límite de Detección , Oxidación-Reducción
3.
Int J Biol Sci ; 20(10): 3823-3841, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39113706

RESUMEN

Macrophages show high plasticity and play a vital role in the progression of metabolic dysfunction-associated steatohepatitis (MASH). X-box binding protein 1 (XBP1), a key sensor of the unfolded protein response, can modulate macrophage-mediated pro-inflammatory responses in the pathogenesis of MASH. However, how XBP1 influences macrophage plasticity and promotes MASH progression remains unclear. Herein, we formulated an Xbp1 siRNA delivery system based on folic acid modified D-α-tocopheryl polyethylene glycol 1000 succinate nanoparticles (FT@XBP1) to explore the precise role of macrophage-specific Xbp1 deficiency in the progression of MASH. FT@XBP1 was specifically internalized into hepatic macrophages and subsequently inhibited the expression of spliced XBP1 both in vitro and in vivo. It promoted M1-phenotype macrophage repolarization to M2 macrophages, reduced the release of pro-inflammatory factors, and alleviated hepatic steatosis, liver injury, and fibrosis in mice with fat-, fructose- and cholesterol-rich diet-induced MASH. Mechanistically, FT@XBP1 promoted macrophage polarization toward the M2 phenotype and enhanced the release of exosomes that could inhibit the activation of hepatic stellate cells. A promising macrophage-targeted siRNA delivery system was revealed to pave a promising strategy in the treatment of MASH.


Asunto(s)
Ácido Fólico , Macrófagos , ARN Interferente Pequeño , Proteína 1 de Unión a la X-Box , Animales , Masculino , Ratones , Estrés del Retículo Endoplásmico/efectos de los fármacos , Hígado Graso/metabolismo , Ácido Fólico/química , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Nanopartículas/química , Proteína 1 de Unión a la X-Box/metabolismo
4.
Eur J Pharm Biopharm ; 201: 114386, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38950717

RESUMEN

The goal of the study was to fabricate folic acid functionalized docetaxel (DOC)/erlotinib (ERL)-loaded solid lipid nanoparticles (SLNs) to synergistically increase the anticancer activity against triple-negative breast cancer. DOC/ERL-SLNs were prepared by the high shear homogenization - ultrasound dispersion method (0.1 % w/v for DOC, and 0.3 %w/v for ERL) and optimized using Plackett Burman Design (PBD) followed by Box Behnken Design (BBD). The optimized SLNs demonstrated particle size < 200 nm, PDI < 0.35, and negative zeta potential with entrapment and loading efficiency of ∼80 and ∼4 %, respectively. The SLNs and folic acid functionalized SLNs (FA-SLNs) showed sustained release for both drugs, followed by Higuchi and Korsemeyer-Peppas drug release models, respectively. Further, the in vitro pH-stat lipolysis model demonstrated an approximately 3-fold increase in the bioaccessibility of drugs from SLNs compared to suspension. The TEM images revealed the spherical morphology of the SLNs. DOC/ERL loaded SLNs showed dose- and time-dependent cytotoxicity and exhibited a synergism at a molar ratio of 1:3 in TNBC with a combination index of 0.35 and 0.37, respectively. FA-DOC/ERL-SLNs showed enhanced anticancer activity as evidenced by MMP and ROS assay and further inhibited the colony-forming ability and the migration capacity of TNBC cells. Conclusively, the study has shown that SLNs are encouraging systems to improve the pharmaceutical attributes of poorly bioavailable drugs.


Asunto(s)
Docetaxel , Liberación de Fármacos , Sinergismo Farmacológico , Clorhidrato de Erlotinib , Lípidos , Nanopartículas , Tamaño de la Partícula , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Docetaxel/administración & dosificación , Docetaxel/farmacología , Docetaxel/farmacocinética , Humanos , Nanopartículas/química , Clorhidrato de Erlotinib/administración & dosificación , Clorhidrato de Erlotinib/farmacología , Clorhidrato de Erlotinib/farmacocinética , Línea Celular Tumoral , Femenino , Lípidos/química , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Portadores de Fármacos/química , Supervivencia Celular/efectos de los fármacos , Ácido Fólico/química , Liposomas
5.
Int J Nanomedicine ; 19: 6603-6618, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38979533

RESUMEN

Objective: Ovarian cancer cells are prone to acquire tolerance to chemotherapeutic agents, which seriously affects clinical outcomes. The development of novel strategies to enhance the targeting of chemotherapeutic agents to overcome drug resistance and minimize side effects is significant for improving the clinical outcomes of ovarian cancer patients. Methods: We employed folic acid (FA)-modified ZIF-90 nanomaterials (FA-ZIF-90) to deliver the chemotherapeutic drug, cisplatin (DDP), via dual targeting to improve its targeting to circumvent cisplatin resistance in ovarian cancer cells, especially by targeting mitochondria. FA-ZIF-90/DDP could rapidly release DDP in response to dual stimulation of acidity and ATP in tumor cells. Results: FA-ZIF-90/DDP showed good blood compatibility. It was efficiently taken up by human ovarian cancer cisplatin-resistant cells A2780/DDP and aggregated in the mitochondrial region. FA-ZIF-90/DDP significantly inhibited the mitochondrial activity and metastatic ability of A2780/DDP cells. In addition, it effectively induced apoptosis in A2780/DDP cells and overcame cisplatin resistance. In vivo experiments showed that FA-ZIF-90/DDP increased the accumulation of DDP in tumor tissues and significantly inhibited tumor growth. Conclusion: FA-modified ZIF-90 nanocarriers can improve the tumor targeting and anti-tumor effects of chemotherapeutic drugs, reduce toxic side effects, and are expected to be a novel therapeutic strategy to reverse drug resistance in ovarian cancer.


Asunto(s)
Antineoplásicos , Apoptosis , Cisplatino , Resistencia a Antineoplásicos , Ácido Fólico , Imidazoles , Neoplasias Ováricas , Zeolitas , Femenino , Cisplatino/farmacología , Cisplatino/química , Cisplatino/farmacocinética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Humanos , Resistencia a Antineoplásicos/efectos de los fármacos , Animales , Zeolitas/química , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Ácido Fólico/química , Ácido Fólico/farmacología , Imidazoles/química , Imidazoles/farmacología , Imidazoles/administración & dosificación , Apoptosis/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Mitocondrias/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Portadores de Fármacos/química , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Biomaterials ; 311: 122701, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38981152

RESUMEN

Cuproptosis in antitumor therapy faces challenges from copper homeostasis efflux mechanisms and high glutathione (GSH) levels in tumor cells, hindering copper accumulation and treatment efficacy. Herein, we propose a strategy of "adding fuel to the flames" for potent antitumor therapy through a self-accelerating cycle of ferroptosis-cuproptosis. Disulfiram (DSF) loaded hollow mesoporous copper-iron sulfide (HMCIS) nanoparticle with conjugation of polyethylene glycol (PEG) and folic acid (FA) (i.e., DSF@HMCIS-PEG-FA) was developed to swiftly release DSF, H2S, Cu2+, and Fe2+ in the acidic tumor microenvironment (TME). The hydrogen peroxide (H2O2) levels and acidity within tumor cells enhanced by the released H2S induce acceleration of Fenton (Fe2+) and Fenton-like (Cu2+) reactions, enabling the powerful tumor ferroptosis efficacy. The released DSF acts as a role of "fuel", intensifying catalytic effect ("flame") in tumor cells through the sustainable Fenton chemistry (i.e., "add fuel to the flames"). Robust ferroptosis in tumor cells is characterized by serious mitochondrial damage and GSH depletion, leading to excess intracellular copper that triggers cuproptosis. Cuproptosis disrupts mitochondria, compromises iron-sulfur (Fe-S) proteins, and elevates intracellular oxidative stress by releasing free Fe3+. These interconnected processes form a self-accelerating cycle of ferroptosis-cuproptosis with potent antitumor capabilities, as validated in both cancer cells and tumor-bearing mice.


Asunto(s)
Antineoplásicos , Cobre , Disulfiram , Ferroptosis , Ferroptosis/efectos de los fármacos , Animales , Disulfiram/farmacología , Disulfiram/química , Humanos , Ratones , Cobre/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Línea Celular Tumoral , Hierro/metabolismo , Hierro/química , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Ácido Fólico/química , Ácido Fólico/metabolismo , Polietilenglicoles/química , Ratones Endogámicos BALB C , Peróxido de Hidrógeno/metabolismo
7.
Biomacromolecules ; 25(8): 4762-4779, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-38989826

RESUMEN

Injectable hydrogels with good antimicrobial and antioxidant properties, self-healing characteristics, suitable mechanical properties, and therapeutic effects have great practical significance for developing treatments for pressing healthcare challenges. Herein, we have designed a novel, self-healing injectable hydrogel composite incorporating cross-linked biofunctional nanomaterials by mixing alginate aldehyde (Ox-Alg), quaternized chitosan (QCS), adipic acid dihydrazide (ADH), and copper oxide nanosheets surface functionalized with folic acid as the bioligand (F-CuO). Gelation was achieved under physiological conditions via the dynamic Schiff base cross-linking mechanism. The developed nanocomposite injectable hydrogel demonstrated the fast self-healing ability essential to bear deformation and outstanding antibacterial properties along with ROS scavenging ability. Furthermore, the optimized formulation of our F-CuO-embedded injectable hydrogel exhibited excellent cytocompatibility, blood compatibility, and in vitro wound healing performance. Taken together, the F-CuO nanosheet cross-linked injectable hydrogel composite presented herein offers a promising candidate biomaterial with multifunctional properties to develop solutions for addressing clinical challenges.


Asunto(s)
Quitosano , Cobre , Hidrogeles , Nanocompuestos , Quitosano/química , Nanocompuestos/química , Hidrogeles/química , Hidrogeles/farmacología , Cobre/química , Cobre/farmacología , Alginatos/química , Cicatrización de Heridas/efectos de los fármacos , Animales , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/administración & dosificación , Humanos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Ácido Fólico/química , Ácido Fólico/farmacología , Adipatos/química , Ratones , Adhesivos/química , Adhesivos/farmacología
8.
Biomaterials ; 311: 122706, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39032219

RESUMEN

Medicine intervention is the major clinical treatment used to relieve the symptoms and delay the progression of rheumatoid arthritis (RA), but is limited by its poor targeted delivery and short therapeutic duration. Herein, we developed an injectable and bioadhesive gelatin-based (Gel) hydrogel as a local depot of leonurine (Leon)-loaded and folate-functionalized polydopamine (FA-PDA@Leon) nanoparticles for anti-inflammation and chondroprotection in RA. The nanoparticles could protect Leon and facilitate its entry into the M1 phenotype macrophage for intracellular delivery of Leon, while the hydrogel tightly adhered to the tissues in the joint cavity and prolonged the retention of FA-PDA@Leon nanoparticles, thus achieving higher availability and therapeutic efficiency of Leon. In vitro and in vivo experiments demonstrated that the Gel/FA-PDA@Leon hydrogel could strongly suppress the inflammatory response by down-regulating the JAK2/STAT3 signaling pathway in macrophages and protect the chondrocytes from ferritinophagy/ferroptosis. This contributed to maintaining the structural integrity of articular cartilage and accelerating the joint functional recovery. This work provides an effective and convenient strategy to achieve higher bioavailability and long-lasting therapeutic duration of medicine intervention in arthritis diseases.


Asunto(s)
Artritis Reumatoide , Ferroptosis , Hidrogeles , Inflamación , Nanopartículas , Polímeros , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Hidrogeles/química , Ratones , Nanopartículas/química , Inflamación/tratamiento farmacológico , Inflamación/patología , Ferroptosis/efectos de los fármacos , Polímeros/química , Células RAW 264.7 , Nanomedicina/métodos , Indoles/química , Indoles/farmacología , Indoles/administración & dosificación , Masculino , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ácido Fólico/química , Gelatina/química , Humanos , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo
9.
Sci Rep ; 14(1): 16941, 2024 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-39043763

RESUMEN

The nonspecific nature of cancer drug delivery often results in substantial toxic side effects during treatments for breast cancer. To mitigate these negative outcomes, our approach involves loading methotrexate (MTX) within carbon quantum dots (CQDs) synthesized from folic acid, which are then enveloped in exosomal membranes obtained from breast cancer cells (Ex@MTX-CQDs). Analysis utilizing nanoparticle tracking techniques has demonstrated that these Ex@MTX-CQDs maintain the physical and biochemical properties of their exosomal precursors. The release profile of MTX indicated a restricted release percentage (less than 10%) under normal physiological conditions, which is contrasted by a more consistent release rate (approximately 65%) when emulating the conditions found within tumor tissues. The toxicological assessments have confirmed that the presence of exosomes combined with leftover folic acid significantly improves the delivery efficacy of MTX directly to the cancerous cells through the binding to folate and heparan sulfate proteoglycan receptors. This process results in increased disruption of the mitochondrial membrane potential and subsequently triggers apoptosis, ultimately leading to the destruction of cancerous cells. Our research could potentially contribute to the further innovation and application of nanocarriers derived from biological sources for the targeted treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama , Carbono , Exosomas , Ácido Fólico , Metotrexato , Puntos Cuánticos , Humanos , Ácido Fólico/química , Puntos Cuánticos/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Exosomas/metabolismo , Femenino , Metotrexato/farmacología , Metotrexato/administración & dosificación , Metotrexato/química , Carbono/química , Portadores de Fármacos/química , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Apoptosis/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Ondas Ultrasónicas , Células MCF-7 , Potencial de la Membrana Mitocondrial/efectos de los fármacos
10.
Talanta ; 278: 126528, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38996560

RESUMEN

PPy nanoparticles are widely employed as PTT agents, because of their exceptional near-infrared absorption properties. Nonetheless, the efficacy of PTT with PPy nanoparticles is hindered by a challenge, specifically, a lack of precise targeting. In this study, a PTT imaging agent was developed by combining NCQDs having bright green fluorescent properties with PPy nanoparticles along with the masking of folic acid to overcome the challenge of targeting. The synthesized PPy:NCQDs:FA nanocomposite, characterized by extraordinary photothermal property, was utilized for imaging of folate receptor positive (FA+) MCF-7 cancer cells through the emission of green fluorescence by NCQDs incorporated within the nanocomposite. Additionally, these nanoparticles demonstrated a good level of cell viability, exceeding 82 %, even at a concentration of 600 µg mL-1. Even the in vivo toxicity inspection of the nanocomposite exemplified no observed acute toxicity at experimental dosages of 1 and 3 mg per kg body weight. By subjecting MCF-7 cells, inoculated with 100 µg mL-1 of nanocomposite, to NIR laser irradiation for 5 min, a significant decline in cell viability was witnessed, establishing the photothermal therapeutic potency of the nanocomposite. The death of cancer cells induced by nanocomposite was verified through MTT assay, imaging of cells by NCQDs alone, with nanocomposite, and by live/dead cell Calcein AM/PI staining assay. Quantification of induced apoptosis post-laser treatment is conducted through staining with Annexin V-FITC/PI. These findings establish potential use of PPy:NCQDs:FA nanocomposite as versatile theranostic agents, capable of targeted bioimaging and treatment for cancer cells exhibiting folate receptors.


Asunto(s)
Carbono , Supervivencia Celular , Ácido Fólico , Nanopartículas , Nitrógeno , Terapia Fototérmica , Polímeros , Pirroles , Puntos Cuánticos , Humanos , Puntos Cuánticos/química , Puntos Cuánticos/toxicidad , Ácido Fólico/química , Ácido Fólico/farmacología , Células MCF-7 , Carbono/química , Polímeros/química , Nanopartículas/química , Nanopartículas/toxicidad , Supervivencia Celular/efectos de los fármacos , Pirroles/química , Pirroles/farmacología , Nitrógeno/química , Animales , Imagen Óptica , Ratones , Femenino
11.
Biosens Bioelectron ; 262: 116550, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-38976958

RESUMEN

Circulating tumor cell (CTC) has been a valuable biomarker for the diagnosis of breast cancer, while folate receptor is a kind of cell surface receptor glycoprotein which is overexpressed in breast cancer. In this work, we have designed and fabricated an electrochemical biosensor for sensitive detection of folate receptor-positive CTCs based on mild reduction assisted CRISPR/Cas system. Specifically, folate functionalized magnetic beads are firstly prepared to capture CTCs owing to the strong affinity between folate and the folate receptors on the surface of cells. Then, the cell membranes are treated by mild reduction so as to expose a large number of free sulfhydryl groups, which can be coupled with maleimide-DNA to introduce the signal amplified CRISPR/Cas12a system. After the trans-cleavage activity of CRISPR/Cas12a is activated, the long chain DNA modified with electroactive molecules methylene blue can be randomly cleaved into short DNA fragments, which are then captured on the graphite electrode through the host-guest recognition with cucurbit [7]uril, generating highly amplified electrochemical signal corresponding to the number of CTCs. The electrochemical biosensor not only demonstrates the sensitivity with a low detection limit of 2 cells/mL, but also highlights its excellent selectivity and stability in complex environment. Therefore, our biosensor may provide an alternative tool for the analysis of CTCs.


Asunto(s)
Técnicas Biosensibles , Sistemas CRISPR-Cas , Técnicas Electroquímicas , Límite de Detección , Células Neoplásicas Circulantes , Humanos , Técnicas Biosensibles/métodos , Células Neoplásicas Circulantes/patología , Células Neoplásicas Circulantes/química , Técnicas Electroquímicas/métodos , Neoplasias de la Mama/sangre , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/patología , Femenino , Línea Celular Tumoral , Ácido Fólico/química , ADN/química
12.
ACS Appl Mater Interfaces ; 16(32): 41810-41818, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39084852

RESUMEN

Anticancer chemo-immunotherapy has gained considerable attention across various scientific domains as a prospective approach for the comprehensive eradication of malignant tumors. Recent research has particularly been focused on traditional anthracycline chemo drugs, such as doxorubicin and mitoxantrone. These compounds trigger apoptosis in tumor cells and evoke immunogenic cell death (ICD). ICD is a pivotal initiator of the cancer-immunity cycle by facilitating the release of damage-associated molecular patterns (DAMPs). The resultant DAMPs released from cancer cells effectively activate the immune system, resulting in an increase in tumor-infiltrating T cells. In this study, we have innovated a co-delivery strategy involving folate-modified liposomes to deliver doxorubicin and monophosphoryl lipid A (MPLA) simultaneously to tumor tissue. The engineered liposomes exploit the overexpression of folate receptors within the tumor tissues. Delivered doxorubicin initiates ICD at the tumor cells, further enhancing the immunogenic stimulus. Additionally, MPLA helps T cell priming by activating antigen-presenting cells. This intricate interplay culminates in a synergistic effect, ultimately resulting in an augmented and potentiated anticancer chemo-immunotherapeutic liposomal treatment.


Asunto(s)
Doxorrubicina , Muerte Celular Inmunogénica , Inmunoterapia , Lípido A , Liposomas , Receptor Toll-Like 4 , Liposomas/química , Doxorrubicina/farmacología , Doxorrubicina/química , Animales , Muerte Celular Inmunogénica/efectos de los fármacos , Humanos , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/metabolismo , Ratones , Lípido A/análogos & derivados , Lípido A/química , Lípido A/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/terapia , Línea Celular Tumoral , Femenino , Antineoplásicos/química , Antineoplásicos/farmacología , Ácido Fólico/química
13.
Food Chem ; 456: 139964, 2024 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-38876059

RESUMEN

Folate is an essential component of many metabolic processes, and folate deficiency is known to cause various disorders. Folate and folic acid, a synthetic and chemically stable form of folate, enriched diet are typically used to overcome this deficiency. Folic acid and folate however, are susceptible to harsh environment and folates enrichment using nanoparticles is an intensively studied strategy in food industry. This review highlights the current methods and types of matrices utilized to develop folic acid/folate carrying nanoparticles. The folic acid/folate loaded nanoparticles prevent cargo degradation during gut absorption and under harsh food processing conditions including, high temperatures, UV light, and autoclaving. The data demonstrates that nanofortifcation of folates using proteins and biopolymers effectively enhances the bioavailability of the cargo. The encapsulation of folic acid in biopolymers by emulsion, spray drying and ionic gelation represent simplistic methods that can be easily scaled up with applications in food industry.


Asunto(s)
Ácido Fólico , Nanopartículas , Ácido Fólico/química , Nanopartículas/química , Humanos , Animales
14.
ACS Appl Bio Mater ; 7(7): 4454-4470, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38857443

RESUMEN

Liposomes as drug-delivery systems have been researched and applied in multiple scientific reports and introduced as patented products with interesting therapeutic properties. Despite various advantages, this drug carrier faces major difficulties in its innate stability, cancer cell specificity, and control over the release of hydrophobic drugs, particularly quercetin, a naturally derived drug that carries many desirable characteristics for anticancer treatment. To improve the effectiveness of liposomes to deliver quercetin by tackling and mitigating the mentioned hurdles, we developed a strategy to establish the ability to passively target cancerous cells, as well as to increase the bioavailability of loaded drugs by incorporating poly(ethylene glycol), gelatin, and folic acid moieties to modify the liposomal system's surface. This research developed a chemically synthesized gelatin, poly(ethylene glycol), and folic acid as a single polymer to coat drug-loaded liposome systems. Liposomes were coated with gelatin-poly(ethylene glycol)-folic acid by electrostatic interaction, characterized by their size, morphology, ζ potential, drug loading efficiency, infrared structures, differential scanning calorimetry spectra, and drug-releasing profiles, and then evaluated for their cytotoxicity to MCF-7 breast cancer cells, as well as cellular uptake, analyzed by confocal imaging to further elaborate on the in vitro behavior of the coated liposome. The results indicated an unusual change in size with increased coating materials, followed by increased colloidal stability, ζ potential, and improved cytotoxicity to cancer cells, as shown by the cellular viability test with MCF-7. Cellular uptake also confirmed these results, providing data for the effects of biopolymer coating, while confirming that folic acid can increase the uptake of liposome by cancer cells. In consideration of such results, the modified gelatin-poly(ethylene glycol)-folic acid-coated liposome can be a potential system in delivering the assigned anticancer compound. This modified biopolymer showed excellent properties as a coating material and should be considered for further practical applications in the future.


Asunto(s)
Antineoplásicos , Supervivencia Celular , Ensayos de Selección de Medicamentos Antitumorales , Ácido Fólico , Gelatina , Liposomas , Ensayo de Materiales , Tamaño de la Partícula , Polietilenglicoles , Quercetina , Humanos , Liposomas/química , Polietilenglicoles/química , Gelatina/química , Ácido Fólico/química , Ácido Fólico/análogos & derivados , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Quercetina/química , Quercetina/farmacología , Quercetina/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Células MCF-7 , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/síntesis química , Proliferación Celular/efectos de los fármacos , Estructura Molecular
15.
Int J Nanomedicine ; 19: 5227-5243, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38855734

RESUMEN

Purpose: This study aimed to construct targeting drug-loading nanocomposites (FA-FePt/DDP nanoliposomes) to explore their potential in ovarian cancer therapy and molecular magnetic resonance imaging (MMRI). Methods: FA-FePt-NPs were prepared by coupling folate (FA) with polyethylene-glycol (PEG)-coated ferroplatinum nanoparticles and characterized. Then cisplatin (DDP) was encapsulated in FA-FePt-NPs to synthesize FA-PEG-FePt/DDP nanoliposomes by thin film-ultrasonic method and high-speed stirring, of which MMRI potential, magnetothermal effect, and the other involved performance were analyzed. The therapeutic effect of FA-FePt/DDP nanoliposomes combined with magnetic fluid hyperthermia (MFH) on ovarian cancer in vitro and in vivo was evaluated. The expression levels of Bax and epithelial-mesenchymal transition related proteins were detected. The biosafety was also preliminarily observed. Results: The average diameter of FA-FePt-NPs was about 30 nm, FA-FePt/DDP nanoliposomes were about 70 nm in hydrated particle size, with drug slow-release and good cell-specific targeted uptake. In an alternating magnetic field (AMF), FA-FePt/DDP nanoliposomes could rapidly reach the ideal tumor hyperthermia temperature (42~44 °C). MRI scan showed that FA-FePt-NPs and FA-FePt/DDP nanoliposomes both could suppress the T2 signal, indicating a good potential for MMRI. The in vitro and in vivo experiments showed that FA-FePt/DDP-NPs in AMF could effectively inhibit the growth of ovarian cancer by inhibiting cancer cell proliferation, invasion, and migration, and inducing cancer cell apoptosis, much better than that of the other individual therapies; molecularly, E-cadherin and Bax proteins in ovarian cancer cells and tissues were significantly increased, while N-cadherin, Vimentin, and Bcl-2 proteins were inhibited, effectively inhibiting the malignant progression of ovarian cancer. In addition, no significant pathological injury and dysfunction was observed in major visceras. Conclusion: We successfully synthesized FA-FePt/DDP nanoliposomes and confirmed their good thermochemotherapeutic effect in AMF and MMRI potential on ovarian cancer, with no obvious side effects, providing a favorable strategy of integrated targeting therapy and diagnosis for ovarian cancer.


Asunto(s)
Antineoplásicos , Cisplatino , Ácido Fólico , Liposomas , Imagen por Resonancia Magnética , Neoplasias Ováricas , Polietilenglicoles , Femenino , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/terapia , Liposomas/química , Cisplatino/farmacología , Cisplatino/química , Cisplatino/administración & dosificación , Cisplatino/farmacocinética , Animales , Ácido Fólico/química , Ácido Fólico/farmacología , Ácido Fólico/farmacocinética , Humanos , Imagen por Resonancia Magnética/métodos , Polietilenglicoles/química , Línea Celular Tumoral , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Ratones , Platino (Metal)/química , Platino (Metal)/farmacología , Hipertermia Inducida/métodos , Nanocompuestos/química , Ratones Desnudos , Ratones Endogámicos BALB C , Nanopartículas del Metal/química , Campos Magnéticos , Tamaño de la Partícula
16.
Carbohydr Polym ; 340: 122328, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-38857995

RESUMEN

This article presents a novel approach to treating prostate cancer using a nanocarrier composed of folic acid (FA), ß-cyclodextrin (ß-CD), and magnetic graphene oxide (MGO) as a theranostic agent. The carrier is designed to improve the solubility and bioavailability of curcumin, a potential therapeutic substance against prostate cancer. Folic acid receptors overexpressed on the surface of solid tumors, including prostate cancer, may facilitate targeted drug delivery to tumor cells while avoiding nonspecific effects on healthy tissues. The anticancer efficacy of Folic acid-curcumin@ß-CD-MGO in vitro was also examined on LNCaP (an androgen-dependent) and PC3 (an androgen-independent) prostate cancer cells. The relaxivity of nanoparticles in MRI images was also investigated as a diagnostic factor. The results showed a concentration-dependent inhibitory effect on cell proliferation, induction of oxidative damage, and apoptotic effects. Also, nanoparticle relaxometry shows that this agent can be used as a negative contrast agent in MRI images. Overall, this study represents a promising theranostic agent to improve the delivery and trace of curcumin and enhance its therapeutic potential in the treatment of prostate cancer.


Asunto(s)
Proliferación Celular , Curcumina , Ácido Fólico , Grafito , Neoplasias de la Próstata , Nanomedicina Teranóstica , beta-Ciclodextrinas , Curcumina/química , Curcumina/farmacología , Masculino , Grafito/química , Grafito/farmacología , Humanos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , beta-Ciclodextrinas/química , Nanomedicina Teranóstica/métodos , Ácido Fólico/química , Ácido Fólico/farmacología , Proliferación Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Portadores de Fármacos/química , Imagen por Resonancia Magnética/métodos , Nanopartículas/química , Receptores de Folato Anclados a GPI/metabolismo , Liberación de Fármacos , Nanopartículas de Magnetita/química
17.
Biomed Eng Online ; 23(1): 53, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858706

RESUMEN

BACKGROUND: Metastasis is one of the main factors leading to the high mortality rate of gastric cancer. The current monitoring methods are not able to accurately monitor gastric cancer metastasis. METHODS: In this paper, we constructed a new type of hollow Mn 3 O 4 nanocomposites, Mn 3 O 4 @HMSN-Cy7.5-FA, which had a size distribution of approximately 100 nm and showed good stability in different liquid environments. The in vitro magnetic resonance imaging (MRI) results show that the nanocomposite has good response effects to the acidic microenvironment of tumors. The acidic environment can significantly enhance the contrast of T 1 -weighted MRI. The cellular uptake and endocytosis results show that the nanocomposite has good targeting capabilities and exhibits good biosafety, both in vivo and in vitro. In a gastric cancer nude mouse orthotopic metastatic tumor model, with bioluminescence imaging's tumor location information, we realized in vivo MRI/fluorescence imaging (FLI) guided precise monitoring of the gastric cancer orthotopic and metastatic tumors with this nanocomposite. RESULTS: This report demonstrates that Mn 3 O 4 @HMSN-Cy7.5-FA nanocomposites is a promising nano-diagnostic platform for the precision diagnosis and therapy of gastric cancer metastasis in the future. CONCLUSIONS: In vivo MRI/FLI imaging results show that the nanocomposites can achieve accurate monitoring of gastric cancer tumors in situ and metastases. BLI's tumor location information further supports the good accuracy of MRI/FLI dual-modality imaging. The above results show that the MHCF NPs can serve as a good nano-diagnostic platform for precise in vivo monitoring of tumor metastasis. This nanocomposite provides more possibilities for the diagnosis and therapy of gastric cancer metastases.


Asunto(s)
Ácido Fólico , Imagen por Resonancia Magnética , Nanocompuestos , Metástasis de la Neoplasia , Neoplasias Gástricas , Neoplasias Gástricas/diagnóstico por imagen , Neoplasias Gástricas/patología , Animales , Nanocompuestos/química , Ratones , Línea Celular Tumoral , Humanos , Ácido Fólico/química , Compuestos de Manganeso/química , Imagen Óptica , Ratones Desnudos , Óxidos
18.
J Cell Mol Med ; 28(11): e18389, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38864691

RESUMEN

Chemotherapy resistance remains a significant challenge in treating ovarian cancer effectively. This study addresses this issue by utilizing a dual drug-loaded nanomicelle system comprising albendazole (ABZ) and paclitaxel (PTX), encapsulated in a novel carrier matrix of D-tocopheryl polyethylene glycol 1000 succinate vitamin E (TPGS), soluplus and folic acid. Our objective was to develop and optimize this nanoparticulate delivery system using solvent evaporation techniques to enhance the therapeutic efficacy against ovarian cancer. The formulation process involved pre-formulation, formulation, optimization, and comprehensive characterization of the micelles. Optimization was conducted through a 32 factorial design, focusing on the effects of polymer ratios on particle size, zeta potential, polydispersity index (PDI) and entrapment efficiency (%EE). The optimal formulation demonstrated improved dilution stability, as indicated by a critical micelle concentration (CMC) of 0.0015 mg/mL for the TPGS-folic acid conjugate (TPGS-FOL). Extensive characterization included differential scanning calorimetry (DSC), nuclear magnetic resonance (NMR), and Fourier-transform infrared spectroscopy (FTIR). The release profile exhibited an initial burst followed by sustained release over 90 h. The cytotoxic potential of the formulated micelles was superior to that of the drugs alone, as assessed by MTT assays on SKOV3 ovarian cell lines. Additionally, in vivo studies confirmed the presence of both drugs in plasma and tumour tissues, suggesting effective targeting and penetration. In conclusion, the developed TPGS-Fol-based nanomicelles for co-delivering ABZ and PTX show promising results in overcoming drug resistance, enhancing solubility, sustaining drug release, and improving therapeutic outcomes in ovarian cancer treatment.


Asunto(s)
Albendazol , Micelas , Neoplasias Ováricas , Paclitaxel , Femenino , Paclitaxel/farmacología , Paclitaxel/administración & dosificación , Paclitaxel/uso terapéutico , Paclitaxel/química , Albendazol/química , Albendazol/farmacología , Albendazol/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Humanos , Animales , Línea Celular Tumoral , Portadores de Fármacos/química , Polietilenglicoles/química , Vitamina E/química , Ácido Fólico/química , Ratones , Liberación de Fármacos , Tamaño de la Partícula , Polivinilos/química , Polímeros/química , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Int J Biol Macromol ; 272(Pt 1): 132804, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38825272

RESUMEN

Cellulose nanofibres (CNFs), also known as nano-fibrillated cellulose, have emerged as highly promising sustainable biomaterials owing to their numerous advantages, including high accessibility, long-term sustainability, low toxicity, and mechanical properties. Recently, marine organisms have been explored as novel and environmentally friendly sources of cellulose fibers (CFs) due to their easy cultivation, extraction and biocompatibility. Dinoflagellates, a group of marine phytoplankton, have gained particular attention due to their unique cellulosic morphology and lignin-free biomass. Previously, we showed that the unique amorphous nature of dinoflagellate-derived cellulose offers various benefits. This study further explores the potential of dinoflagellate-derived CFs as a sustainable and versatile CNF source. Extracted dinoflagellate cellulose is effectively converted into CNFs via one-step TEMPO oxidation without significant polymer degradation. In addition, the biological compatibility of the CNFs is improved by amine-grafting using putrescine and folic acid. The products are characterised by conductometric titration, zeta potential measurements, TGA, GPC, FTIR, SEM/TEM, XRD, and XPS. Finally, in a proof-of-principle study, the application of the functionalised CNFs in drug delivery is tested using methylene blue as a drug model. Our findings suggest that dinoflagellate-derived CNFs provide an eco-friendly platform that can be easily functionalised for various applications, including drug delivery.


Asunto(s)
Celulosa , Dinoflagelados , Nanofibras , Dinoflagelados/química , Celulosa/química , Nanofibras/química , Óxidos N-Cíclicos/química , Ácido Fólico/química
20.
Sci Rep ; 14(1): 13985, 2024 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-38886450

RESUMEN

Crocin is a carotenoid compound in saffron with anti-cancer properties. However, its therapeutic application is limited by its low absorption, bioavailability, and stability, which can be overcome through nanocarrier delivery systems. This study used surface-modified Nano-crystalline cellulose (NCC) to deliver crocin to cancer cells. NCC modified with CTAB were loaded with crocin and then conjugated with folic acid (NCF-CR-NPs). The synthesized nanoparticles (NPs) were characterized using FTIR, XRD, DLS, and FESEM. The crystallinity index of NCC was 66.64%, higher than microcrystalline cellulose (61.4%). The crocin loading and encapsulation efficiency in NCF-CR-NPs were evaluated. Toxicity testing by MTT assay showed that NCF-CR-NPs had higher toxicity against various cancer cell lines, including colon cancer HT-29 cells (IC50 ~ 11.6 µg/ml), compared to free crocin. Fluorescent staining, flow cytometry, and molecular analysis confirmed that NCF-CR-NPs induced apoptosis in HT-29 cells by increasing p53 and caspase 8 expression. The antioxidant capacity of NCF-CR-NPs was also evaluated using ABTS and DPPH radical scavenging assays. NCF-CR-NPs exhibited high free radical scavenging ability, with an IC50 of ~ 46.5 µg/ml for ABTS. In conclusion, this study demonstrates the potential of NCF-CR-NPs to deliver crocin to cancer cells effectively. The NPs exhibited enhanced anti-cancer and antioxidant activities compared to free crocin, making them a promising nanocarrier system for crocin-based cancer therapy.


Asunto(s)
Apoptosis , Carotenoides , Celulosa , Ácido Fólico , Nanopartículas , Carotenoides/química , Carotenoides/farmacología , Ácido Fólico/química , Ácido Fólico/farmacología , Humanos , Celulosa/química , Nanopartículas/química , Apoptosis/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Células HT29 , Portadores de Fármacos/química , Antioxidantes/farmacología , Antioxidantes/química , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Supervivencia Celular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...