Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Small ; 20(10): e2305076, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37909382

RESUMEN

Chronic diabetic wounds remain a worldwide challenge for both the clinic and research. Given the vicious circle of oxidative stress and inflammatory response as well as the impaired angiogenesis of the diabetic wound tissues, the wound healing process is disturbed and poorly responds to the current treatments. In this work, a nickel-based metal-organic framework (MOF, Ni-HHTP) with excellent antioxidant activity and proangiogenic function is developed to accelerate the healing process of chronic diabetic wounds. The Ni-HHTP can mimic the enzymatic catalytic activities of antioxidant enzymes to eliminate multi-types of reactive species through electron transfer reactions, which protects cells from oxidative stress-related damage. Moreover, this Ni-based MOF can promote cell migration and angiogenesis by activating transforming growth factor-ß1 (TGF-ß1) in vitro and reprogram macrophages to the anti-inflammatory phenotype. Importantly, Ni-HHTP effectively promotes the healing process of diabetic wounds by suppressing the inflammatory response and enhancing angiogenesis in vivo. This study reports a versatile and promising MOF-based nanozyme for diabetic wound healing, which may be extended in combination with other wound dressings to enhance the management of diabetic or non-healing wounds.


Asunto(s)
Diabetes Mellitus Experimental , Estructuras Metalorgánicas , Animales , Especies Reactivas de Oxígeno , Estructuras Metalorgánicas/farmacología , Níquel , Angiogénesis , Cicatrización de Heridas/fisiología , Antioxidantes , Hidrogeles
2.
Nano Lett ; 18(11): 6804-6811, 2018 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-30350653

RESUMEN

Free radicals have emerged as new-type and promising candidates for hypoxic tumor treatment, and further study of their therapeutic mechanism by real-time imaging is of great importance to explore their biomedical applications. Herein, we present a smart free-radical generator AuNC-V057-TPP for hypoxic tumor therapy; the AuNC-V057-TPP not only exhibits good therapeutic effect under both hypoxic and normoxic conditions but also can monitor the release of free radicals in real-time both in vitro and in vivo. What is more, with the mitochondria-targeting ability, the AuNC-V057-TPP is demonstrated with improved antitumor efficacy through enhanced free radical level in mitochondria, which leads to mitochondrial membrane damage and ATP production reduction and finally induces cancer cell apoptosis.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Radicales Libres/metabolismo , Oro , Neoplasias Mamarias Animales , Nanopartículas del Metal , Mitocondrias , Imagen Molecular/métodos , Hipoxia Tumoral , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Femenino , Oro/química , Oro/farmacología , Neoplasias Mamarias Animales/diagnóstico por imagen , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/terapia , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología
3.
Small ; 14(20): e1800292, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29665292

RESUMEN

This study reports a double-targeting "nanofirework" for tumor-ignited imaging to guide effective tumor-depth photothermal therapy (PTT). Typically, ≈30 nm upconversion nanoparticles (UCNP) are enveloped with a hybrid corona composed of ≈4 nm CuS tethered hyaluronic acid (CuS-HA). The HA corona provides active tumor-targeted functionality together with excellent stability and improved biocompatibility. The dimension of UCNP@CuS-HA is specifically set within the optimal size window for passive tumor-targeting effect, demonstrating significant contributions to both the in vivo prolonged circulation duration and the enhanced size-dependent tumor accumulation compared with ultrasmall CuS nanoparticles. The tumors featuring hyaluronidase (HAase) overexpression could induce the escape of CuS away from UCNP@CuS-HA due to HAase-catalyzed HA degradation, in turn activating the recovery of initially CuS-quenched luminescence of UCNP and also driving the tumor-depth infiltration of ultrasmall CuS for effective PTT. This in vivo transition has proven to be highly dependent on tumor occurrence like a tumor-ignited explosible firework. Together with the double-targeting functionality, the pathology-selective tumor ignition permits precise tumor detection and imaging-guided spatiotemporal control over PTT operation, leading to complete tumor ablation under near infrared (NIR) irradiation. This study offers a new paradigm of utilizing pathological characteristics to design nanotheranostics for precise detection and personalized therapy of tumors.


Asunto(s)
Hipertermia Inducida , Nanofibras/química , Neoplasias/patología , Fototerapia , Animales , Muerte Celular , Cobre/química , Células Hep G2 , Humanos , Ácido Hialurónico/química , Hialuronoglucosaminidasa/metabolismo , Luminiscencia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células 3T3 NIH , Nanofibras/ultraestructura , Nanopartículas/química , Nanopartículas/ultraestructura , Células RAW 264.7 , Esferoides Celulares/patología , Esferoides Celulares/ultraestructura , Sulfuros/química , Temperatura
4.
Sci Adv ; 10(28): eadn1745, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38996026

RESUMEN

Rapid drug clearance and off-target effects of therapeutic drugs can induce low bioavailability and systemic side effects and gravely restrict the therapeutic effects of inflammatory bowel diseases (IBDs). Here, we propose an amplifying targeting strategy based on orally administered gallium (Ga)-based liquid metal (LM) nano-agents to efficiently eliminate reactive oxygen and nitrogen species (RONS) and modulate the dysregulated microbiome for remission of IBDs. Taking advantage of the favorable adhesive activity and coordination ability of polyphenol structure, epigallocatechin gallate (EGCG) is applied to encapsulate LM to construct the formulations (LM-EGCG). After adhering to the inflamed tissue, EGCG not only eliminates RONS but also captures the dissociated Ga to form EGCG-Ga complexes for enhancive accumulation. The detained composites protect the intestinal barrier and modulate gut microbiota for restoring the disordered enteral microenvironment, thereby relieving IBDs. Unexpectedly, LM-EGCG markedly decreases the Escherichia_Shigella populations while augmenting the abundance of Akkermansia and Bifidobacterium, resulting in favorable therapeutic effects against the dextran sulfate sodium-induced colitis.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Animales , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Administración Oral , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Catequina/análogos & derivados , Catequina/química , Catequina/administración & dosificación , Catequina/farmacología , Galio/química , Galio/farmacología , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Colitis/tratamiento farmacológico , Humanos , Especies de Nitrógeno Reactivo/metabolismo
5.
Bioact Mater ; 25: 95-106, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36789001

RESUMEN

The excessive reactive oxygen species (ROS) is a hallmark associated with the initiation and progression of inflammatory bowel disease (IBD), which execrably form a vicious cycle of ROS and inflammation to continually promote disease progression. Here, the gold nanoparticles-embedded ceria nanoparticles (Au/CeO2) with enhanced antioxidant activities are designed to block this cycle reaction for treating IBD by scavenging overproduced ROS. The Au/CeO2 with core-shell and porous structure exhibits significantly higher enzymatic catalytic activities compared with commercial ceria nanoparticles, likely due to the effective exposure of catalytic sites, higher content of Ce (III) and oxygen vacancy, and accelerated reduction from Ce (IV) to Ce (III). Being coated with negatively-charged hyaluronic acid, the Au/CeO2@HA facilitates accumulation in inflamed colon tissues via oral administration, reduces pro-inflammatory cytokines, and effectively alleviates colon injury in colitis mice. Overall, the Au/CeO2@HA with good biocompatibility is a promising nano-therapeutic for treating IBD.

6.
Acta Biomater ; 158: 686-697, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36623782

RESUMEN

Selectively generating active free radical (AFR) in tumor microenvironment (TME) can promote irreversible oxidation of biomolecules and damage tumor cells, resulting in effective tumor inhibition. However, therapeutic efficacy of AFR-based tumor suppression approaches is often limited by insufficient amount of H2O2 or O2 within TME. To overcome this obstacle, we design a pH/photothermal dual responsive nanosystem (PFeSA@AS) for combined photothermal and nanocatalytic therapy in the near-infrared biowindow. Here the Fe single-atom dispersed N, S-doped carbon nanosheets (FeSA) nanozyme is dispersed by phospholipid-polyethylene glycol-amine (DSPE-PEG-NH2), and further loads artesunate (AS) via an amide reaction. Upon 808-nm laser irradiation in TME, the AS is released and further be catalyzed by the FeSA nanozyme to produce cytotoxic C-centered AFRs, and further be accelerated due to the photothermal conversion performance of FeSA (23.35%). The nanocatalytic process of FeSA nanozyme is realized by density functional theory (DFT). The tumor inhibition rates of a CT26 xenograft model is 92% through a photothermal-enhanced nanocatalytic synergistic therapy, and negligible systematic toxicity is observed. This work offers a potential paradigm of multifunctional single atomic catalysts (SACs) for enhancing tumor nanocatalytic therapy. STATEMENT OF SIGNIFICANCE: We designed a pH/photothermal dual responsive nanosystem (PFeSA@AS) for nanocatalytic therapy: (1) the nanosystem responsively releases AS under 808-nm laser irradiation in TME; (2) FeSA in the nanosystem can act as heme mimetic to convert AS into high cytotoxic C-centered free radicals for nanocatalytic therapy; (3) the photothermal conversion performance of FeSA further enhances the catalytic process to yield abundant AFR. Both in vitro and in vivo results demonstrate that this nanosystem can efficiently inhibit tumor growth through a photothermal-enhanced nanocatalytic synergistic therapy.


Asunto(s)
Hipertermia Inducida , Neoplasias , Humanos , Fototerapia , Línea Celular Tumoral , Artesunato/farmacología , Peróxido de Hidrógeno/farmacología , Catálisis , Microambiente Tumoral
7.
Biomaterials ; 281: 121358, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34979416

RESUMEN

The overexpression of glutathione (GSH) in cancer cells has long been regarded as the primary obstacle for reactive oxygen species (ROS)-involved anti-tumor therapies. To solve this issue, a ferric ion and selenite-codoped calcium phosphate (Fe/Se-CaP) nanohybrid here is fabricated to catabolize endogenous GSH, instead of directly deleting it, to trigger a ROS storm for tumor suppression. The selenite component in Fe/Se-CaP can catabolize GSH to superoxide anion (O2•-) and hydroxyl radicals (•OH) via cascade catalytic reactions, elevating oxidative stress while destroying antioxidant system. The doped Fe can further catalyze the soaring hydrogen peroxide (H2O2) originated from O2•- to •OH via Fenton reactions. Collectively, Fe/Se-CaP mediated self-augmented catabolism dynamic therapy finally induces apoptosis of cancer cells owing to the significant rise of ROS and, combined with CaP adjuvant, evokes adaptive immune responses to suppress tumor progression, providing an innovative train of thought for ROS-involved anti-tumor therapies.


Asunto(s)
Glutatión , Peróxido de Hidrógeno , Glutatión/metabolismo , Peróxido de Hidrógeno/metabolismo , Hierro , Especies Reactivas de Oxígeno/metabolismo , Ácido Selenioso , Superóxidos/metabolismo
8.
Biomaterials ; 281: 121369, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35026671

RESUMEN

Tumor cells obtain energy supply from different metabolic pathways to maintain survival. In this study, a tumor acidity-responsive spherical nanoparticle (called as LMGC) was designed by attaching glucose oxidase (GOx) and mineralizing calcium carbonate on the surface of liquid metal nanoparticles to integrate the synergistic effect of adenosine triphosphate (ATP) generation inhibition and photothermal therapy (PTT) for enhanced tumor therapy. After GOx catalysis, the process of glycolysis was inhibited, and the increased H2O2 level enhanced the intratumoral oxidative stress. Besides, the gluconic acid production accelerated the degradation of LMGC and promoted Ca2+-mediated mitochondrial dysfunction. The inhibition of glycolysis and mitochondrial metabolism could significantly reduce ATP production and down-regulate heat shock protein (HSP) expression, which would reduce tumor cells heat resistance and improve PTT therapeutic effect. This liquid metal-based ATP inhibition system with enhanced therapeutic effect will find great potential for tumor treatment.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Neoplasias , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , Glucosa Oxidasa/metabolismo , Glucólisis , Humanos , Peróxido de Hidrógeno/metabolismo , Mitocondrias/metabolismo , Neoplasias/tratamiento farmacológico , Terapia Fototérmica
9.
ACS Nano ; 16(4): 5851-5866, 2022 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-35412799

RESUMEN

Conventional cancer targeting methodology needs to be reformed to overcome the intrinsic barriers responsible for poor targeting efficiency. This study describes a concept of self-reinforced cancer targeting (SRCT) by correlating targeting with therapy in a reciprocally enhancing manner. SRCT is achieved on the basis of two prerequisites: (1) target molecules have to be expressed on cancer cell membranes but not on normal cells, and (2) notably, their expression on cancer cells must be actively upregulated in response to cellular attack by cancer treatments. As a proof-of-concept, a GRP78-targeting nanovehicle for chemotherapy was designed. Resultant data showed that chemotherapeutic drugs could effectively elevate GRP78 expression on the plasma membranes of cancer cells while having minimal influence on normal cells. DOX pretreatment of cancer cells and tumor tissues can greatly increase the targeting efficacy and therapeutic performance of the prepared GRP78-targeting nanomedicine while somewhat disfavoring the nontargeting counterpart. In vivo and in vitro results demonstrated that this GRP78-targeting nanomedicine could accurately target cancer cells to not only implement chemotherapy but also induce GRP78 upregulation on cancer cells, eventually benefiting continuous cancer-cell-targeted attack by the nanomedicines remaining in the blood circulation or administered in the next dose. The GRP78-targeting nanomedicine displays much better antitumor performance compared with the nontargeting counterpart. SRCT is expected to advance cancer-targeted therapy based on the positive dependency between targeting and therapeutic modalities.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Retroalimentación , Neoplasias/tratamiento farmacológico , Nanomedicina/métodos , Línea Celular Tumoral
10.
Biomaterials ; 287: 121630, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35816980

RESUMEN

Severe skeletal muscle injuries usually lead to a series of poor recovery issues, such as massive myofibers loss, scar tissue formation, significant muscle function impairment, etc. Here, a silk sericin patch delivering miRNA-29-enriched extracellular vesicles-decorated myoblasts (SPEED) is designed for the rapid regeneration and functional repair after severe skeletal muscle injury. Specifically, miR29-enriched extracellular vesicles (miR29-EVs) are prepared and used to deliver miR29 into primary myoblasts, which promote the myotube formation of myoblasts and increase the expression of myogenic genes while inhibiting the expression of fibrotic genes. Our results indicate that miR29-EVs promote the integration of primary myoblasts and host muscle in a severe mouse tibialis anterior (TA) muscle injury model. Moreover, implantation of SPEED drastically stimulates skeletal muscle regeneration, inhibits fibrosis of injured muscles, and leads to significant improvement of muscle contraction forces and motor ability of mice about 3 weeks after treatment. Subsequently, we further evaluate the transcriptomes of TA muscles and find that SPEED can significantly ameliorate energy metabolism and muscular microenvironment of TA muscles on day 9 after implantation. Additionally, bioinformatic analysis and comprehensive molecular biology studies also reveal that the down-regulation of CDC20-MEF2C signaling axis may participate in the muscle repair process. Together, SPEED may serve as an effective alternative for the rapid repair of severe skeletal muscle injuries in the future.

11.
J Mater Chem B ; 9(37): 7686-7697, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34323245

RESUMEN

The validity and biocompatibility of irrigating agents are imperative for the success of root canal therapy. The imperfections in the currently available irrigants highlight the fact that more advanced technologies and strategies are required for complete disinfection in endodontic treatments. In the present study, a Fenton reaction-enhanced antimicrobial sonodynamic therapy (SDT) platform was fabricated for root canal disinfection. Firstly, mesoporous silica nanoparticles (MSNs) were synthesized, grafted with an amino group and then conjugated with sonosensitizer protoporphyrin IX (PpIX). Iron ions were then anchored (M@P-Fe) to initiate a Fenton reaction. Nanoparticle characterization by size and zeta potential measurements, scanning electron microscopy, transmission electron microscopy and thermogravimetric analysis confirmed that the platform was successfully developed. Reactive oxygen species (ROS) generation assessment, methylene blue degradation and electron spin resonance assays illustrated upon ultrasound (US) irradiation, that augmented ROS, can be produced by US activated PpIX and iron mediated Fenton reactions from low concentration H2O2 (0.01%). In vitro anti-Enterococcus faecalis efficacy was demonstrated by growth curve and colony forming unit measurements. Confocal laser scanning microscopy and scanning electron microscopy observations illustrated the effectiveness of the platform on in situ biofilm eradication in root canal. Owing to the stronger oxidizing capability and short lifetime of ROS, the Fenton reaction-enhanced SDT can induce detrimental oxidative damage to bacteria upon activation of US while avoiding nonspecific toxicity to cells, which was verified by cytotoxicity evaluations using CCK-8 assay and morphology observation of MC3T3-E1 cells. Compared to commonly used NaClO, this nanoplatform displayed desirable anti-bacterial, anti-biofilm abilities and better biocompatibility. These results highlight that the integrated M@P-Fe + US + H2O2 platform is a promising candidate for US enhanced root canal irrigation and disinfection.


Asunto(s)
Antibacterianos/química , Materiales Biocompatibles/química , Peróxido de Hidrógeno/química , Hierro/química , Nanopartículas/química , Dióxido de Silicio/química , Animales , Antibacterianos/farmacología , Materiales Biocompatibles/farmacología , Biopelículas/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cavidad Pulpar/microbiología , Enterococcus faecalis/fisiología , Peróxido de Hidrógeno/farmacología , Azul de Metileno/química , Ratones , Porosidad , Protoporfirinas/química , Especies Reactivas de Oxígeno/metabolismo , Tratamiento del Conducto Radicular/métodos , Staphylococcus aureus/efectos de los fármacos , Ultrasonografía
12.
Small Methods ; 5(7): e2100361, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34927984

RESUMEN

Advances in enzymes involve an efficient biocatalytic process, which has demonstrated great potential in biomedical applications. However, designing a functional carrier for enzymes equipped with satisfactory degradability and loading efficiency, remains a challenge. Here, based on transformable liquid metal (LM), a spinose nanodrum is designed as protein carrier to deliver enzyme for tumor treatment. With the assistance of spines and a special drum-like shape, it is found that the spiny LM can carry much more enzymes than spherical LM under the same condition. Benefiting from the satisfactory enzyme loading efficiency of spiny LM, a plasma amine oxidase immobilized spinose LM nanosystem enveloped with epigallocatechin gallate (EGCG)-Fe3+ (LMPE) is fabricated for photothermal and cascade catalytic tumor therapy. Activated by the acidic condition in the tumor microenvironment, the LMPE can oxidize spermine (Spm) and spermidine (Spd) to generate hydrogen peroxide (H2 O2 ) for Fenton catalytic reaction to produce the lethal hydroxyl radical (•OH) for tumor cell killing. Combined with remarkable photothermal performance of LM, LMPE exhibits significant inhibition of tumor in vivo.


Asunto(s)
Peróxido de Hidrógeno , Microambiente Tumoral , Catálisis , Línea Celular Tumoral , Peróxido de Hidrógeno/metabolismo , Espermina
13.
J Control Release ; 320: 159-167, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-31978443

RESUMEN

Glucose-responsive insulin delivery system mimicking the function of pancreatic ß-cells to maintain blood glucose homeostasis would effectively alleviate diabetes. Here, a new glucose-responsive delivery (ZIF@Ins&GOx) for self-regulated insulin release was constructed by encapsulating insulin and glucose oxidase (GOx) into pH-sensitive zeolitic imidazole framework-8 (ZIF-8) nanocrystals. After entering the cavities of ZIF-8, glucose can be oxidized into gluconic acid by GOx, causing a decrease in local pH. Then, ZIF-8 nanocrystals would be degraded under the acidic microenvironment that in turn triggers the release of insulin in a glucose responsive fashion. In vitro studies indicated that the biological activity of insulin could be protected by the rigid structure of ZIF-8 and the release of insulin could be modulated in response to glucose concentrations. In vivo experiments demonstrated that a single subcutaneous injection of the ZIF@Ins&GOx would facilitate the stabilization of blood glucose level of normoglycemic state for up to 72 h in type 1 diabetes (T1D). The multifunctional insulin delivery system shows a new proof-of-concept for T1D treatment by using ZIF-8 nanocrystals loaded with insulin and enzyme.


Asunto(s)
Glucosa Oxidasa , Insulina , Glucemia , Glucosa , Concentración de Iones de Hidrógeno
14.
Nanoscale ; 12(16): 8890-8897, 2020 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-32266902

RESUMEN

Selectively attenuating the protection offered by heat shock protein 90 (HSP90), which is indispensable for the stabilization of the essential regulators of cell survival and works as a cell guardian under oxidative stress conditions, is a potential approach to improve the efficiency of cancer therapy. Here, we designed a biodegradable nanoplatform (APCN/BP-FA) based on a Zr(iv)-based porphyrinic porous coordination network (PCN) and black phosphorus (BP) sheets for efficient photodynamic therapy (PDT) by enhancing the accumulation of the nanoplatforms in the tumor area and attenuating the protection of cancer cells. Owing to the favorable degradability of BP, the nanosystem exhibited accelerated the release of the HSP90 inhibitor tanespimycin (17-AAG) and an apparent promotion in the reactive oxygen species (ROS) yield of PCN as well as expedited the degradation of the PCN-laden BP nanoplatforms. Both in vitro and in vivo results revealed that the elevated amounts of ROS and reduced cytoprotection in tumor cells were caused by the nanoplatforms. This strategy may provide a promising method for attenuating cytoprotection to aid efficient photodynamic therapy.


Asunto(s)
Estructuras Metalorgánicas/química , Neoplasias/tratamiento farmacológico , Fósforo/química , Fotoquimioterapia/métodos , Animales , Benzoquinonas/química , Benzoquinonas/uso terapéutico , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Ácido Fólico/química , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Lactamas Macrocíclicas/química , Lactamas Macrocíclicas/uso terapéutico , Estructuras Metalorgánicas/farmacocinética , Estructuras Metalorgánicas/uso terapéutico , Ratones , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Neoplasias/metabolismo , Fósforo/farmacocinética , Fósforo/uso terapéutico , Porosidad , Porfirinas/química , Porfirinas/farmacocinética , Porfirinas/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Circonio/química , Circonio/farmacocinética , Circonio/uso terapéutico
15.
Nanoscale ; 12(5): 2966-2972, 2020 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-31971210

RESUMEN

Lactate, the main contributor to the acidic tumor microenvironment, not only promotes the proliferation of tumor cells, but also closely relates to tumor invasion and metastasis. Here, a tumor targeting nanoplatform, designated as Me&Flu@MSN@MnO2-FA, was fabricated for effective tumor suppression and anti-metastasis by interfering with lactate metabolism of tumor cells. Metformin (Me) and fluvastatin sodium (Flu) were incorporated into MnO2-coated mesoporous silicon nanoparticles (MSNs), the synergism between Me and Flu can modulate the pyruvate metabolic pathway to produce more lactate, and concurrently inhibit lactate efflux to induce intracellular acidosis to kill tumor cells. As a result of the restricted lactate efflux, the extracellular lactate concentration is reduced, and the ability of the tumor cells to migrate is also weakened. This ingenious strategy based on Me&Flu@MSN@MnO2-FA showed an obvious inhibitory effect on tumor growth and resistance to metastasis.


Asunto(s)
Fluvastatina , Lactatos/metabolismo , Compuestos de Manganeso , Metformina , Nanopartículas , Neoplasias , Microambiente Tumoral/efectos de los fármacos , Antineoplásicos/química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Fluvastatina/química , Fluvastatina/farmacocinética , Fluvastatina/farmacología , Ácido Fólico/metabolismo , Humanos , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacocinética , Compuestos de Manganeso/farmacología , Metformina/química , Metformina/farmacocinética , Metformina/farmacología , Nanopartículas/química , Nanopartículas/uso terapéutico , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Porosidad , Silicio/química , Silicio/farmacocinética , Silicio/farmacología
16.
Biomaterials ; 224: 119500, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31557591

RESUMEN

Redox homeostasis inside malignant cells is a defense mechanism against the reactive oxygen species (ROS)-induced therapy means, but little importance has been paid to this innate barrier. The present study intends to make cancer cells more sensitive to the ROS-induced therapy by disturbing cellular redox homeostasis. To verify this concept, a porous metal-organic framework (MOF) serves not only as the photodynamic therapy (PDT) agent but also as the carrier to transport alkaloid piperlongumine (PL), a thioredoxin reductase (TrxR) inhibitor used to disturb cellular redox homeostasis. The PL-loaded MOF was further coated with cancer cell membranes to gain homologous tumor-targeting capability. Inside tumor cells, the released PL can effectively block the TrxR-mediated ROS elimination pathway. The resultant data show that compared to traditional PDT alone, the combination of PDT and TrxR inhibition causes profound promotions in cellular ROS level by about 1.6 times, in cytotoxicity by about 2 times, and in cellular apoptosis/necrosis rate by about 3 times. Consequently, this strategy based on the interference with cellular redox homeostasis has demonstrated high potency to improve the anticancer PDT performance, adumbrating a new way to boost the power of ROS-induced therapy.


Asunto(s)
Homeostasis , Nanopartículas/uso terapéutico , Fotoquimioterapia , Células 3T3 , Factor de Transcripción Activador 4/metabolismo , Animales , Línea Celular Tumoral , Dioxolanos/uso terapéutico , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Estructuras Metalorgánicas/química , Ratones , Ratones Endogámicos BALB C , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo
17.
Biomaterials ; 223: 119472, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31499254

RESUMEN

Inflammation during photothermal therapy (PTT) of tumor usually results in adverse consequences. Here, a biomembrane camouflaged nanomedicine (mPDAB) containing polydopamine and ammonia borane was designed to enhance PTT efficacy and mitigate inflammation. Polydopamine, a biocompatible photothermal agent, can effectively convert light into heat for PTT. Ammonia borane was linked to the surface of polydopamine through the interaction of hydrogen bonding, which could destroy redox homoeostasis in tumor cells and reduce inflammation by H2 release in tumor microenvironment. Owing to the same origin of outer biomembranes, mPDAB showed excellent tumor accumulation and low systemic toxicity in a breast tumor model. Excellent PTT efficacy and inflammation reduction made the mPDAB completely eliminate the primary tumors, while also restraining the outgrowth of distant dormant tumors. The biomimetic nanomedicine shows potentials as a universal inflammation-self-alleviated platform to ameliorate inflammation-related disease treatment, including but not limited to PTT for tumor.


Asunto(s)
Amoníaco/química , Boranos/química , Neoplasias de la Mama/tratamiento farmacológico , Hidrógeno , Fototerapia/métodos , Animales , Materiales Biocompatibles , Células COS , Chlorocebus aethiops , Femenino , Gases , Células HeLa , Homeostasis , Humanos , Inflamación , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Membranas Artificiales , Ratones , Nanomedicina/métodos , Trasplante de Neoplasias , Oxidación-Reducción , Recurrencia , Temperatura , Microambiente Tumoral
18.
Biomaterials ; 207: 76-88, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30959408

RESUMEN

Various negative effects accompanying with the instability of bare liquid metal (LM) nanoparticles, including undesirable spontaneous coalescence, continuous photothermal performance deterioration and difficult multi-step functionalization, severely hinder its applications in biomedical area. In this study, we proposed a new concept of immobilized liquid metal nanoparticles based on a surface mesoporous silica coating strategy (LM@MSN). Strikingly, it was found that unsteady and vulnerable LM nanoparticles after immobilization exhibited enhanced stabilization and sustainable photothermal performance even with a long and repeated light irradiation in acidic environments. Moreover, integrating the properties of easy surface functionalization and high drug loading efficiency from silica shell, immobilized LM nanoparticle was further used for photothermal involved combinational therapy. The classical anticancer drug doxorubicin (DOX) was encapsulated in pores of silica shell and the hyaluronic acid (HA) was decorated on LM@MSN to construct LM@MSN/DOX@HA for tumor targeted combination therapy. Both in vitro and in vivo studies proved that LM@MSN/DOX@HA could significantly inhibit solid tumor growth under near infrared (NIR) irradiation by synergistic photothermal/chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Nanopartículas del Metal/química , Antineoplásicos/química , Doxorrubicina/química , Sistemas de Liberación de Medicamentos/métodos , Porosidad , Dióxido de Silicio/química
19.
Adv Mater ; 31(15): e1807211, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30803083

RESUMEN

To engineer patient-derived cells into therapy-purposed biologics is a promising solution to realize personalized treatments. Without using gene-editing technology, a live cell-typed therapeutic is engineered for tumor treatment by artificially reprogramming macrophages with hyaluronic acid-decorated superparamagnetic iron oxide nanoparticles (HIONs). This nanoparticle-assisted cell-reprogramming strategy demonstrates profound advantages, due to the combined contributions from the biological regulation of HIONs and the intrinsic nature of macrophages. Firstly, the reprogrammed macrophages present a substantial improvement in their innate capabilities, such as more effective tumor targeting and more efficient generation of bioactive components (e.g., reactive oxygen species, bioactive cytokines) to suppress tumor growth. Furthermore, this cell therapeutic exhibits cytostatic/proapoptotic effects specific to cancer cells. Secondly, HIONs enable macrophages more resistant to the intratumoral immunosuppressive environment. Thirdly, the macrophages are endowed with a strong ability to prime in situ protumoral M2 macrophages into antitumor M1 phenotype in a paracrine-like manner. Consequently, a synergistic tumor-inhibition effect is achieved. This study shows that engineering nanomaterial-reprogrammed live cells as therapeutic biologics may be a more preferable option to the commonly used approaches where nanomaterials are administrated to induce bioresponse of certain cells in vivo.


Asunto(s)
Reprogramación Celular , Macrófagos/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Resistencia a Antineoplásicos , Femenino , Humanos , Macrófagos/inmunología , Nanopartículas de Magnetita , Ratones , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/terapia , Microambiente Tumoral/inmunología
20.
Biomaterials ; 217: 119303, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31271859

RESUMEN

Here, a highly cooperative liquid metal nanoparticle-enzyme (LM@GOX) was constructed for combinational starvation/photothermal therapy of tumor. It was found that the enzyme activity of glucose oxidase (GOX) could be strengthened along with the increased temperature within a given range and its optimal activity is around about 43-60 °C. Utilizing the photothermal conversion ability of liquid metal (LM), the GOX catalytic efficiency could be photo-controlled with improved starvation therapeutic efficiency. Furthermore, due to the accelerating blood flow during the photothermal therapy (PTT), the hypoxic situation in tumor tissues could also be relieved, which would contribute to conquering the hypoxia-suppressed GOX catalysis. In the meanwhile, the severe thermo-resistance of tumor cells during PTT process could be overcome by GOX induced decrease of adenosine triphosphate (ATP) and heat shock proteins (HSPs) level, eventually leading to an improved therapeutic effect of PTT. Both in vitro and in vivo studies proved that LM@GOX could significantly inhibit the growth of solid tumor under NIR illumination by a win-win cooperative starvation/photothermal therapy.


Asunto(s)
Glucosa Oxidasa/metabolismo , Hipertermia Inducida , Luz , Nanopartículas del Metal/química , Neoplasias/terapia , Fototerapia , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/farmacología , Peso Corporal/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Glucosa/farmacología , Proteínas de Choque Térmico/metabolismo , Humanos , Rayos Infrarrojos , Nanopartículas del Metal/ultraestructura , Ratones Endogámicos BALB C , Imagen Óptica , Carga Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA