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










Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(19): eadm9561, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38718119

RESUMEN

Lactic acid (LA) accumulation in the tumor microenvironment poses notable challenges to effective tumor immunotherapy. Here, an intelligent tumor treatment microrobot based on the unique physiological structure and metabolic characteristics of Veillonella atypica (VA) is proposed by loading Staphylococcus aureus cell membrane-coating BaTiO3 nanocubes (SAM@BTO) on the surface of VA cells (VA-SAM@BTO) via click chemical reaction. Following oral administration, VA-SAM@BTO accurately targeted orthotopic colorectal cancer through inflammatory targeting of SAM and hypoxic targeting of VA. Under in vitro ultrasonic stimulation, BTO catalyzed two reduction reactions (O2 → •O2- and CO2 → CO) and three oxidation reactions (H2O → •OH, GSH → GSSG, and LA → PA) simultaneously, effectively inducing immunogenic death of tumor cells. BTO catalyzed the oxidative coupling of VA cells metabolized LA, effectively disrupting the immunosuppressive microenvironment, improving dendritic cell maturation and macrophage M1 polarization, and increasing effector T cell proportions while decreasing regulatory T cell numbers, which facilitates synergetic catalysis and immunotherapy.


Asunto(s)
Neoplasias Colorrectales , Inmunoterapia , Microambiente Tumoral , Neoplasias Colorrectales/terapia , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Inmunoterapia/métodos , Animales , Ratones , Humanos , Catálisis , Línea Celular Tumoral , Nanoestructuras/química , Materiales Biomiméticos/química , Administración Oral , Titanio/química , Biomimética/métodos , Ácido Láctico/química , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Compuestos de Bario
2.
Adv Mater ; : e2313097, 2024 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-38643386

RESUMEN

Therapy-induced immunogenic cell death (ICD) can initiate both innate and adaptive immune responses for amplified anti-tumor efficacy. However, dying cell-released ICD signals are prone to being sequestered by the TIM-3 receptors on dendritic cell (DC) surfaces, preventing immune surveillance. Herein, dismantlable coronated nanoparticles (NPs) are fabricated as a type of spatiotemporally controlled nanocarriers for coupling tumor cell-mediated ICD induction to DC-mediated immune sensing. These NPs are loaded with an ICD inducer, mitoxantrone (MTO), and wrapped by a redox-labile anti-TIM-3 (αTIM-3) antibody corona, forming a separable core-shell structure. The antibody corona disintegrates under high levels of extracellular reactive oxygen species in the tumor microenvironment, exposing the MTO-loaded NP core for ICD induction and releasing functional αTIM-3 molecules for DC sensitization. Systemic administration of the coronated NPs augments DC maturation, promotes cytotoxic T cell recruitment, enhances tumor susceptibility to immune checkpoint blockade, and prevents the side effects of MTO. This study develops a promising nanoplatform to unleash the potential of host immunity in cancer therapy.

3.
Adv Sci (Weinh) ; : e2310211, 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38460166

RESUMEN

The precise targeted delivery of therapeutic agents to deep regions of the brain is crucial for the effective treatment of various neurological diseases. However, achieving this goal is challenging due to the presence of the blood-brain barrier (BBB) and the complex anatomy of the brain. Here, a biomimetic self-propelled nanomotor with cascade targeting capacity is developed for the treatment of neurological inflammatory diseases. The self-propelled nanomotors are designed with biomimetic asymmetric structures with a mesoporous SiO2 head and multiple MnO2 tentacles. Macrophage membrane biomimetic modification endows nanomotors with inflammatory targeting and BBB penetration abilities The MnO2 agents catalyze the degradation of H2 O2 into O2 , not only by reducing brain inflammation but also by providing the driving force for deep brain penetration. Additionally, the mesoporous SiO2 head is loaded with curcumin, which actively regulates macrophage polarization from the M1 to the M2 phenotype. All in vitro cell, organoid model, and in vivo animal experiments confirmed the effectiveness of the biomimetic self-propelled nanomotors in precise targeting, deep brain penetration, anti-inflammatory, and nervous system function maintenance. Therefore, this study introduces a platform of biomimetic self-propelled nanomotors with inflammation targeting ability and active deep penetration for the treatment of neurological inflammation diseases.

4.
Nat Commun ; 15(1): 1042, 2024 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-38310127

RESUMEN

Chronic diabetic wounds are at lifelong risk of developing diabetic foot ulcers owing to severe hypoxia, excessive reactive oxygen species (ROS), a complex inflammatory microenvironment, and the potential for bacterial infection. Here we develop a programmed treatment strategy employing live Haematococcus (HEA). By modulating light intensity, HEA can be programmed to perform a variety of functions, such as antibacterial activity, oxygen supply, ROS scavenging, and immune regulation, suggesting its potential for use in programmed therapy. Under high light intensity (658 nm, 0.5 W/cm2), green HEA (GHEA) with efficient photothermal conversion mediate wound surface disinfection. By decreasing the light intensity (658 nm, 0.1 W/cm2), the photosynthetic system of GHEA can continuously produce oxygen, effectively resolving the problems of hypoxia and promoting vascular regeneration. Continuous light irradiation induces astaxanthin (AST) accumulation in HEA cells, resulting in a gradual transformation from a green to red hue (RHEA). RHEA effectively scavenges excess ROS, enhances the expression of intracellular antioxidant enzymes, and directs polarization to M2 macrophages by secreting AST vesicles via exosomes. The living HEA hydrogel can sterilize and enhance cell proliferation and migration and promote neoangiogenesis, which could improve infected diabetic wound healing in female mice.


Asunto(s)
Diabetes Mellitus , Pie Diabético , Microalgas , Femenino , Animales , Ratones , Especies Reactivas de Oxígeno , Antibacterianos/farmacología , Hipoxia , Oxígeno , Cicatrización de Heridas , Hidrogeles
5.
Biomaterials ; 306: 122478, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38266348

RESUMEN

Platelets play a critical role as circulating cells in the human body and contribute to essential physiological processes such as blood clotting, hemostasis, vascular repair, and thrombus formation. Currently, platelets are extensively employed in the development of innovative biomimetic drug delivery systems, offering significant enhancements in circulation time, biocompatibility, and targeted delivery efficiency compared to conventional drug delivery approaches. Leveraging the unique physiological functions of platelets, these platelet-derived drug delivery systems (DDSs) hold great promise for the treatment of diverse diseases, including cancer, cardiovascular diseases, infectious diseases, wound healing and other diseases. This review primarily focuses on the design and characteristics of existing platelet-derived DDSs, including their preparation and characterization methods. Furthermore, this review comprehensively outlines the applications of these materials across various diseases, offering a holistic understanding of their therapeutic potential. This study aimed to provide a comprehensive overview of the potential value of these materials in clinical treatment, serving as a valuable reference for the advancement of novel platelet-derived DDSs and their broader utilization in the field of disease treatment.


Asunto(s)
Enfermedades Cardiovasculares , Enfermedades Transmisibles , Neoplasias , Humanos , Enfermedades Cardiovasculares/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Plaquetas/fisiología , Neoplasias/tratamiento farmacológico , Enfermedades Transmisibles/tratamiento farmacológico
6.
Adv Mater ; 36(3): e2308726, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37842855

RESUMEN

Piezoelectric, pyroelectric, and ferroelectric materials are considered unique biomedical materials due to their dielectric crystals and asymmetric centers that allow them to directly convert various primary forms of energy in the environment, such as sunlight, mechanical energy, and thermal energy, into secondary energy, such as electricity and chemical energy. These materials possess exceptional energy conversion ability and excellent catalytic properties, which have led to their widespread usage within biomedical fields. Numerous biomedical applications have demonstrated great potential with these materials, including disease treatment, biosensors, and tissue engineering. For example, piezoelectric materials are used to stimulate cell growth in bone regeneration, while pyroelectric materials are applied in skin cancer detection and imaging. Ferroelectric materials have even found use in neural implants that record and stimulate electrical activity in the brain. This paper reviews the relationship between ferroelectric, piezoelectric, and pyroelectric effects and the fundamental principles of different catalytic reactions. It also highlights the preparation methods of these three materials and the significant progress made in their biomedical applications. The review concludes by presenting key challenges and future prospects for efficient catalysts based on piezoelectric, pyroelectric, and ferroelectric nanomaterials for biomedical applications.


Asunto(s)
Materiales Biocompatibles , Regeneración Ósea , Materiales Biocompatibles/farmacología , Encéfalo , Catálisis , Proliferación Celular
7.
Nat Commun ; 14(1): 6973, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37914681

RESUMEN

The dense stroma of desmoplastic tumor limits nanotherapeutic penetration and hampers the antitumor immune response. Here, we report a denaturation-and-penetration strategy and the use of tin monosulfide nanoparticles (SnSNPs) as nano-sonosensitizers that can overcome the stromal barrier for the management of desmoplastic triple-negative breast cancer (TNBC). SnSNPs possess a narrow bandgap (1.18 eV), allowing for efficient electron (e-)-hole (h+) pair separation to generate reactive oxygen species under US activation. More importantly, SnSNPs display mild photothermal properties that can in situ denature tumor collagen and facilitate deep penetration into the tumor mass upon near-infrared irradiation. This approach significantly enhances sonodynamic therapy (SDT) by SnSNPs and boosts antitumor immunity. In mouse models of malignant TNBC and hepatocellular carcinoma (HCC), the combination of robust SDT and enhanced cytotoxic T lymphocyte infiltration achieves remarkable anti-tumor efficacy. This study presents an innovative approach to enhance SDT and antitumor immunity using the denaturation-and-penetration strategy, offering a potential combined sono-immunotherapy approach for the cancer nanomedicine field.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas , Neoplasias , Neoplasias de la Mama Triple Negativas , Terapia por Ultrasonido , Humanos , Animales , Ratones , Carcinoma Hepatocelular/terapia , Neoplasias de la Mama Triple Negativas/terapia , Neoplasias Hepáticas/terapia , Neoplasias/terapia , Especies Reactivas de Oxígeno , Nanopartículas/uso terapéutico , Línea Celular Tumoral
8.
Nanoscale ; 15(48): 19407-19422, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-37965689

RESUMEN

Treating cancer remains one of the most formidable challenges in modern medicine, with traditional treatment options often being limited by poor therapeutic outcomes and unacceptable side effects. Nanocatalytic therapy activates tumor-localized catalytic reactions in situ via nontoxic or minimally toxic nanocatalysts responding to unique cues from the tumor microenvironment or external stimuli. In particular, sonocatalytic cancer therapy is a promising approach that has emerged as a potential solution to this problem through the combination of ultrasound waves and catalytic materials to selectively target and destroy cancer cells. Compared to light, ultrasound exhibits higher spatial precision, lower energy attenuation, and superior tissue penetrability, furnishing more energy to catalysts. Multidimensional modulation of nanocatalyst structures and properties is pivotal to maximizing catalytic efficiency given constraints in external stimulative energy as well as substrate types and levels. In this review, we discuss the various theories and mechanisms underlying sonocatalytic cancer therapy, as well as advanced catalysts that have been developed for this application. Additionally, we explore the design of sonocatalytic cancer therapy systems, including the use of heterojunction catalysts and the optimal conditions for achieving maximum therapeutic effects. Finally, we highlight the potential benefits of sonocatalytic cancer therapy over traditional cancer treatments, including its noninvasive nature and lower toxicity.


Asunto(s)
Neoplasias , Humanos , Neoplasias/terapia , Ondas Ultrasónicas , Microambiente Tumoral
9.
Adv Sci (Weinh) ; 10(34): e2303298, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37867225

RESUMEN

Mononuclear phagocytes (MPs) are vital components of host immune defenses against cancer. However, tumor-infiltrating MPs often present tolerogenic and pro-tumorigenic phenotypes via metabolic switching triggered by excessive lipid accumulation in solid tumors. Inspired by viral infection-mediated MP modulation, here enveloped immunometabolic nanoparticles (immeNPs) are designed to co-deliver a viral RNA analog and a fatty acid oxidation regulator for synergistic reshaping of intratumoral MPs. These immeNPs are camouflaged with cancer cell membranes for tumor homing and opsonized with anti-CD163 antibodies for specific MP recognition and uptake. It is found that internalized immeNPs coordinate lipid metabolic reprogramming with innate immune stimulation, inducing M2-to-M1 macrophage repolarization and tolerogenic-to-immunogenic dendritic cell differentiation for cytotoxic T cell infiltration. The authors further demonstrate that the use of immeNPs confers susceptibility to anti-PD-1 therapy in immune checkpoint blockade-resistant breast and ovarian tumors, and thereby provide a promising strategy to expand the potential of conventional immunotherapy.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Linfocitos T Citotóxicos , Neoplasias/terapia , Anticuerpos , Macrófagos , Lípidos
10.
Mater Horiz ; 10(12): 5474-5483, 2023 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-37703055

RESUMEN

Chronic diabetic wounds persistently face the threat of evolving into diabetic foot ulcers owing to severe hypoxia, high levels of reactive oxygen species (ROS), and a complex inflammatory microenvironment. To concurrently surmount these obstacles, we developed an all-round therapeutic strategy based on nanozymes that simultaneously scavenge ROS, generate O2 and regulate the immune system. First, we designed a dynamic covalent bond hybrid of a metal-organic coordination polymer as a synthesis template, obtaining high-density platinum nanoparticle assemblies (PNAs). This compact assembly of platinum nanoparticles not only effectively simulates antioxidant enzymes (CAT, POD) but also, under ultrasound (US), enhances electron polarization through the surface plasmon resonance effect, endowing it with the ability to induce GSH generation by effectively replicating the enzyme function of glutathione reductase (GR). PNAs, by mimicking the activity of CAT and POD, effectively catalyze hydrogen peroxide, alleviate hypoxia, and effectively generate GSH under ultrasound, further enhancing ROS scavenging. Notably, PNAs can regulate macrophage responses in the inflammatory microenvironment, circumventing the use of any additives. It was confirmed that PNAs can enhance cell proliferation and migration, promote neoangiogenesis IN VITRO, and accelerate the healing of infected diabetic wounds IN VIVO. We believe that an all-round therapeutic method based on PNA nanozymes could be a promising strategy for sustained diabetic wound healing.


Asunto(s)
Pie Diabético , Nanopartículas del Metal , Ácidos Nucleicos de Péptidos , Humanos , Hidrogeles , Nanopartículas del Metal/uso terapéutico , Platino (Metal) , Especies Reactivas de Oxígeno , Hipoxia
11.
Nat Commun ; 14(1): 5140, 2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37612298

RESUMEN

The exogenous excitation requirement and electron-hole recombination are the key elements limiting the application of catalytic therapies. Here a tumor microenvironment (TME)-specific self-triggered thermoelectric nanoheterojunction (Bi0.5Sb1.5Te3/CaO2 nanosheets, BST/CaO2 NSs) with self-built-in electric field facilitated charge separation is fabricated. Upon exposure to TME, the CaO2 coating undergoes rapid hydrolysis, releasing Ca2+, H2O2, and heat. The resulting temperature difference on the BST NSs initiates a thermoelectric effect, driving reactive oxygen species production. H2O2 not only serves as a substrate supplement for ROS generation but also dysregulates Ca2+ channels, preventing Ca2+ efflux. This further exacerbates calcium overload-mediated therapy. Additionally, Ca2+ promotes DC maturation and tumor antigen presentation, facilitating immunotherapy. It is worth noting that the CaO2 NP coating hydrolyzes very slowly in normal cells, releasing Ca2+ and O2 without causing any adverse effects. Tumor-specific self-triggered thermoelectric nanoheterojunction combined catalytic therapy, ion interference therapy, and immunotherapy exhibit excellent antitumor performance in female mice.


Asunto(s)
Peróxido de Hidrógeno , Neoplasias , Femenino , Animales , Ratones , Inmunoterapia , Neoplasias/terapia , Presentación de Antígeno , Transporte Biológico , Microambiente Tumoral
12.
Materials (Basel) ; 16(16)2023 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-37629946

RESUMEN

Selective laser melting (SLM) of high-temperature alloys involves intricate interdependencies among key process parameters, such as laser power and scanning speed, affecting properties such as density and tensile strength. However, relying solely on experiential knowledge for process parameter design often hampers the precise attainment of target requirements. To address this challenge, we propose an innovative approach that integrates the analytic hierarchy process (AHP) and weighted particle swarm optimization (WPSO) to recommend SLM process parameters for high-temperature alloy fabrication. Our proposed AHP-WPSO model consists of three main steps. First, a comprehensive historical database is established, capturing the process parameters and performance metrics of high-temperature alloy SLM parts. Utilizing an AHP framework, we compute the performance similarity between target and historical cases, applying rational thresholds to identify analogous cases. When suitable analogs are elusive, the model seamlessly transitions to the second step. Here, the WPSO model optimizes and recommends process parameters according to target specifications. Lastly, our experimental validation of the GH4169 high-temperature alloy through SLM experiments corroborates the effectiveness of our AHP-WPSO model in making process parameter recommendations. The outcomes underscore the model's high accuracy, attaining a recommendation precision of 99.81% and 96.32% when historical analogs are present and absent, respectively. This innovative approach offers a robust and reliable solution to the challenges posed in SLM process parameter optimization for high-temperature alloy applications.

13.
Adv Sci (Weinh) ; 10(26): e2302131, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37409429

RESUMEN

The advent of immunotherapy has marked a new era in cancer treatment, offering significant clinical benefits. Cell membrane as drug delivery materials has played a crucial role in enhancing cancer therapy because of their inherent biocompatibility and negligible immunogenicity. Different cell membranes are prepared into cell membrane nanovesicles (CMNs), but CMNs have limitations such as inefficient targeting ability, low efficacy, and unpredictable side effects. Genetic engineering has deepened the critical role of CMNs in cancer immunotherapy, enabling genetically engineered-CMN (GCMN)-based therapeutics. To date, CMNs that are surface modified by various functional proteins have been developed through genetic engineering. Herein, a brief overview of surface engineering strategies for CMNs and the features of various membrane sources is discussed, followed by a description of GCMN preparation methods. The application of GCMNs in cancer immunotherapy directed at different immune targets is addressed as are the challenges and prospects of GCMNs in clinical translation.


Asunto(s)
Sistemas de Liberación de Medicamentos , Neoplasias , Inmunoterapia , Membrana Celular/genética , Ingeniería Genética , Neoplasias/terapia
14.
Adv Mater ; 35(36): e2301257, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37262365

RESUMEN

Cancer-associated fibroblasts (CAFs) promote cancer stem cell (CSC)-mediated chemoresistance and immunosuppressive tumor microenvironment. However, direct depletion of CAFs may increase cancer invasiveness and metastasis. As a generalized strategy against chemoresistant cancers, Gemini-like homotypic targeting nanoparticles (NPs) are designed for two-pronged CAF transformation and cancer cell elimination. The CAF-targeted NPs couple vitamin B3 metabolic reprogramming to epigenetic modulation of secreted pro-stemness and immunosuppressive factors, thereby diminishing CSC and suppressive immune cell populations to enhance cancer cell drug susceptibility and cytotoxic T cell infiltration. In mouse models of breast, liver, pancreatic and colorectal cancers that are resistant to their respective first-line chemotherapeutics, a single dose of hydrogel co-delivering the Gemini-like NPs can rehabilitate chemosensitivity, induce immune activation, and achieve tumor regression. Moreover, it stimulates robust T cell memory for long-term protection against tumor rechallenge. This study thus represents an innovative approach with broad applicability for overcoming cancer chemoresistance.


Asunto(s)
Antineoplásicos , Fibroblastos Asociados al Cáncer , Neoplasias , Animales , Ratones , Neoplasias/tratamiento farmacológico , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/metabolismo , Linfocitos T Citotóxicos , Vitaminas/metabolismo , Vitaminas/farmacología , Microambiente Tumoral , Línea Celular Tumoral
15.
Angew Chem Int Ed Engl ; 62(41): e202308413, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37380606

RESUMEN

Tumor-associated macrophages (TAMs) play a critical role in the immunosuppressive solid tumor microenvironment (TME), yet in situ engineering of TAMs for enhanced tumor immunotherapy remains a significant challenge in translational immuno-oncology. Here, we report an innovative nanodrug-delivering-drug (STNSP@ELE) strategy that leverages two-dimensional (2D) stanene-based nanosheets (STNSP) and ß-Elemene (ELE), a small-molecule anticancer drug, to overcome TAM-mediated immunosuppression and improve chemo-immunotherapy. Our results demonstrate that both STNSP and ELE are capable of polarizing the tumor-supportive M2-like TAMs into a tumor-suppressive M1-like phenotype, which acts with the ELE chemotherapeutic to boost antitumor responses. In vivo mouse studies demonstrate that STNSP@ELE treatment can reprogram the immunosuppressive TME by significantly increasing the intratumoral ratio of M1/M2-like TAMs, enhancing the population of CD4+ and CD8+ T lymphocytes and mature dendritic cells, and elevating the expression of immunostimulatory cytokines in B16F10 melanomas, thereby promoting a robust antitumor response. Our study not only demonstrates that the STNSP@ELE chemo-immunotherapeutic nanoplatform has immune-modulatory capabilities that can overcome TAM-mediated immunosuppression in solid tumors, but also highlights the promise of this nanodrug-delivering-drug strategy in developing other nano-immunotherapeutics and treating various types of immunosuppressive tumors.


Asunto(s)
Melanoma , Nanopartículas , Neoplasias , Ratones , Animales , Macrófagos Asociados a Tumores , Macrófagos/metabolismo , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Melanoma/patología , Nanopartículas/uso terapéutico , Microambiente Tumoral
16.
Adv Mater ; 35(30): e2211415, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37096955

RESUMEN

Dendritic cells (DCs) are crucial mediators of innate and adaptive antitumor immunity, whereas exogenously and endogenously driven lipid accumulation causes immune tolerance of tumor-associated DCs (TADCs) and thereby diminishes tumor responsiveness to various therapies. Herein, a type of multilevel lipid rewiring nanoparticles (NPs) for TADC revitalization is designed. These self-assembled NPs specifically bind to the lipid transport receptor Msr1 on the TADC surface and orchestrate the restriction of extracellular lipid uptake, cytoplasmic de novo lipid biosynthesis and nuclear lipogenic gene transcription. It is found that the slimming of TADCs via the three-in-one lipid metabolic reprogramming substantially promotes their maturation and rehabilitate their functions in inflammatory cytokine production, cytotoxic T cell recruitment, and tumor inhibition. Significantly, tumor resistance to immune checkpoint blockade therapy is further overcome. The study presents a non-canonical strategy to remodel tumor-infiltrating immune cells and paves a new path for improving the efficacy of cancer immunotherapy.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Neoplasias/patología , Linfocitos T Citotóxicos , Inmunoterapia , Células Dendríticas , Lípidos
17.
Biomaterials ; 296: 122068, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36868032

RESUMEN

Photodynamic therapy (PDT)-mediated antitumor immune response depends on oxidative stress intensity and subsequent immunogenic cell death (ICD) in tumor cells, yet the inherent antioxidant system restricts reactive oxygen species (ROS)-associated oxidative damage, which is highly correlated with the upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) and the downstream products, such as glutathione (GSH). Herein, to overcome this dilemma, we designed a versatile nanoadjuvant (RI@Z-P) to enhance the sensitivity of tumor cells to oxidative stress via Nrf2-specific small interfering RNA (siNrf2). The constructed RI@Z-P could significantly amplify photooxidative stress and achieve robust DNA oxidative damage, activating the stimulator of interferon genes (STING)-dependent immune-sensing to produce interferon-ß (IFN-ß). Additionally, RI@Z-P together with laser irradiation reinforced tumor immunogenicity by exposing or releasing damage-associated molecular patterns (DAMPs), showing the prominent adjuvant effect for promoting dendritic cell (DC) maturation and T-lymphocyte activation and even alleviating the immunosuppressive microenvironment to some extent.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Glutatión/metabolismo , Inmunoterapia , ADN/metabolismo
18.
J Hazard Mater ; 451: 131083, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-36878031

RESUMEN

Photocatalytic oxidation technology holds promise for ideal advanced treatment of antibiotic wastewater. Single-atom catalysts (SACs) are a new hotspot in catalytic science, but the photochemical studies on the removal of antibiotics from water and biocompatibility after entering the environment are scarce. In this work, we prepared a single Mn atom immobilized on N-doped biochar (Mn@N-Biochar) by impregnation calcination method for enhancing photocatalytic degradation of sulfanilamide (SNM) in different types of various water systems. Compared with the original biochar, Mn@N-Biochar showed enhanced SNM degradation and TOC removal capacity. DFT calculation concluded that the electrons of d-orbital (Mn) and p-orbital (N) altered the electronic structure of biochar and enhanced the photoelectric performance. It was shown that Mn@N-Biochar caused negligible systemic inflammation and tissue damage when given orally in mice, and also did not alter cell death and ROS production in human lung, kidney, and liver cells, as compared with biochar. We are convinced that Mn@N-Biochar could enhance the photocatalytic degradation of antibiotics while maintaining biocompatibility, which could be a promising strategy for wastewater treatment.


Asunto(s)
Antibacterianos , Electrones , Humanos , Animales , Ratones , Antibacterianos/farmacología , Sulfanilamida , Carbón Orgánico/farmacología , Carbón Orgánico/química , Agua
19.
Front Pharmacol ; 14: 1052301, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36794282

RESUMEN

Breast cancer is the leading cause of cancer-related fatalities in female worldwide. Effective therapies with low side effects for breast cancer treatment and prevention are, accordingly, urgently required. Targeting anticancer materials, breast cancer vaccines and anticancer drugs have been studied for many years to decrease side effects, prevent breast cancer and suppress tumors, respectively. There are abundant evidences to demonstrate that peptide-based therapeutic strategies, coupling of good safety and adaptive functionalities are promising for breast cancer therapy. In recent years, peptide-based vectors have been paid attention in targeting breast cancer due to their specific binding to corresponding receptors overexpressed in cell. To overcome the low internalization, cell penetrating peptides (CPPs) could be selected to increase the penetration due to the electrostatic and hydrophobic interactions between CPPs and cell membranes. Peptide-based vaccines are at the forefront of medical development and presently, 13 types of main peptide vaccines for breast cancer are being studied on phase III, phase II, phase I/II and phase I clinical trials. In addition, peptide-based vaccines including delivery vectors and adjuvants have been implemented. Many peptides have recently been used in clinical treatments for breast cancer. These peptides show different anticancer mechanisms and some novel peptides could reverse the resistance of breast cancer to susceptibility. In this review, we will focus on current studies of peptide-based targeting vectors, CPPs, peptide-based vaccines and anticancer peptides for breast cancer therapy and prevention.

20.
Biomaterials ; 295: 122031, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36731367

RESUMEN

This study reports an ultrasound-mediated and two-dimensional (2D) porous vermiculite nanosheets (VMT NSs)-based nanocatalyst platform (Arg@VMT@PDA-PEG) that synergistically harnessed the Fenton reaction-based chemodynamic therapy (CDT), 2D semiconductor-based sonodynamic therapy (SDT) and nitric oxide (NO)-based gas therapy for combination cancer therapy. The tumor microenvironment responsive degradation of polydopamine (PDA) shell could not only prevent L-Arg, a NO donor, leakage during blood circulation, but also selectively release the active sites of VMT NSs for catalytic reactions in tumor cells. Additionally, the Fenton reactions mediated by the abundant Fe2+/Fe3+ in VMT NSs could efficiently produce ·OH and consume glutathione (GSH) for CDT. Moreover, the reactive oxygen species (ROS, ·OH and ·O2-) produced by ultrasound-triggered Arg@VMT@PDA-PEG could not only execute SDT but also oxidize L-Arg to NO for synergetic gas therapy. The results show that the transformation of ROS to NO can enhance curative efficacy owing to the ability of NO with much longer life-time in freely diffusing into cells from intercellular space. This biodegradable Arg@VMT@PDA-PEG nanocatalytic platform integrating three different catalytic reactions provides a new therapeutic paradigm for combination cancer therapy.


Asunto(s)
Arginina , Neoplasias , Humanos , Porosidad , Especies Reactivas de Oxígeno , Terapia Combinada , Glutatión , Óxido Nítrico , Línea Celular Tumoral , Microambiente Tumoral , Peróxido de Hidrógeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...