Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.780.307
Filtrar
1.
Methods Mol Biol ; 2848: 187-196, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39240524

RESUMEN

In several ocular diseases, degeneration of retinal neurons can lead to permanent blindness. Transplantation of stem cell (SC)-derived RGCs has been proposed as a potential therapy for RGC loss. Although there are reports of successful cases of SC-derived RGC transplantation, achieving long-distance regeneration and functional connectivity remains a challenge. To address these hurdles, retinal organoids are being used to study the regulatory mechanism of stem cell transplantation. Here we present a modified protocol for differentiating human embryonic stem cells (ESCs) into retinal organoids and transplanting organoid-derived RGCs into the murine eyes.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias Humanas , Células Ganglionares de la Retina , Humanos , Animales , Ratones , Células Madre Embrionarias Humanas/citología , Células Ganglionares de la Retina/citología , Trasplante de Células Madre/métodos , Organoides/citología , Organoides/trasplante , Técnicas de Cultivo de Célula/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Retina/citología , Células Madre Embrionarias/citología
2.
Methods Mol Biol ; 2848: 37-58, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39240515

RESUMEN

Several protocols have been established for the generation of lens organoids from embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and other cells with regenerative potential in humans or various animal models. It is important to examine how well the regenerated lens organoids reflect lens biology, in terms of its development, homeostasis, and aging. Toward this goal, the iSyTE database (integrated Systems Tool for Eye gene discovery; https://research.bioinformatics.udel.edu/iSyTE/ ), a bioinformatics resource tool that contains meta-analyzed gene expression data in wild-type lens across different embryonic, postnatal, and adult stages, can serve as a resource for comparative analysis. This article outlines the approaches toward effective use of iSyTE to gain insights into normal gene expression in the mouse lens, enriched expression in the lens, and differential gene expression in select mouse gene-perturbation cataract/lens defects models, which in turn can be used to evaluate expression of key lens-relevant genes in lens organoids by transcriptomics (e.g., RNA-sequencing (RNA-seq), microarrays, etc.) or other downstream methods (e.g., RT-qPCR, etc.).


Asunto(s)
Cristalino , Organoides , Regeneración , Cristalino/citología , Cristalino/metabolismo , Organoides/metabolismo , Organoides/citología , Animales , Ratones , Regeneración/genética , Perfilación de la Expresión Génica/métodos , Biología Computacional/métodos , Simulación por Computador , Humanos , Catarata/genética , Catarata/patología , Catarata/metabolismo , Transcriptoma , Bases de Datos Genéticas
3.
Methods Mol Biol ; 2848: 117-134, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39240520

RESUMEN

Retinal degenerative diseases including age-related macular degeneration and glaucoma are estimated to currently affect more than 14 million people in the United States, with an increased prevalence of retinal degenerations in aged individuals. An expanding aged population who are living longer forecasts an increased prevalence and economic burden of visual impairments. Improvements to visual health and treatment paradigms for progressive retinal degenerations slow vision loss. However, current treatments fail to remedy the root cause of visual impairments caused by retinal degenerations-loss of retinal neurons. Stimulation of retinal regeneration from endogenous cellular sources presents an exciting treatment avenue for replacement of lost retinal cells. In multiple species including zebrafish and Xenopus, Müller glial cells maintain a highly efficient regenerative ability to reconstitute lost cells throughout the organism's lifespan, highlighting potential therapeutic avenues for stimulation of retinal regeneration in humans. Here, we describe how the application of single-cell RNA-sequencing (scRNA-seq) has enhanced our understanding of Müller glial cell-derived retinal regeneration, including the characterization of gene regulatory networks that facilitate/inhibit regenerative responses. Additionally, we provide a validated experimental framework for cellular preparation of mouse retinal cells as input into scRNA-seq experiments, including insights into experimental design and analyses of resulting data.


Asunto(s)
Células Ependimogliales , Retina , Análisis de la Célula Individual , Animales , Ratones , Análisis de la Célula Individual/métodos , Retina/metabolismo , Células Ependimogliales/metabolismo , Regeneración/genética , Análisis de Secuencia de ARN/métodos , Degeneración Retiniana/genética , Degeneración Retiniana/terapia , RNA-Seq/métodos , Modelos Animales de Enfermedad
4.
Methods Mol Biol ; 2848: 59-71, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39240516

RESUMEN

Glaucoma is one of the leading causes of irreversible blindness. Stem cell therapy has shown promise in the treatment of primary open-angle glaucoma in animal models. Stem cell-free therapy using stem cell-derived trophic factors might be in demand in patients with high-risk conditions or religious restrictions. In this chapter, we describe methods for trabecular meshwork stem cell (TMSC) cultivation, secretome harvesting, and protein isolation, as well as assays to ensure the health of TMSC post-secretome harvesting and for secretome periocular injection into mice for therapeutic purposes.


Asunto(s)
Células Madre , Malla Trabecular , Malla Trabecular/metabolismo , Malla Trabecular/citología , Animales , Ratones , Humanos , Células Madre/citología , Células Madre/metabolismo , Regeneración , Glaucoma/terapia , Trasplante de Células Madre/métodos , Secretoma , Modelos Animales de Enfermedad , Glaucoma de Ángulo Abierto/terapia , Células Cultivadas , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Técnicas de Cultivo de Célula/métodos
5.
Methods Mol Biol ; 2848: 151-167, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39240522

RESUMEN

High-quality imaging of the retina is crucial to the diagnosis and monitoring of disease, as well as for evaluating the success of therapeutics in human patients and in preclinical animal models. Here, we describe the basic principles and methods for in vivo retinal imaging in rodents, including fundus imaging, fluorescein angiography, optical coherence tomography, fundus autofluorescence, and infrared imaging. After providing a concise overview of each method and detailing the retinal diseases and conditions that can be visualized through them, we will proceed to discuss the advantages and disadvantages of each approach. These protocols will facilitate the acquisition of optimal images for subsequent quantification and analysis. Additionally, a brief explanation will be given regarding the potential results and the clinical significance of the detected abnormalities.


Asunto(s)
Modelos Animales de Enfermedad , Angiografía con Fluoresceína , Retina , Enfermedades de la Retina , Tomografía de Coherencia Óptica , Animales , Tomografía de Coherencia Óptica/métodos , Enfermedades de la Retina/diagnóstico por imagen , Enfermedades de la Retina/patología , Enfermedades de la Retina/diagnóstico , Retina/diagnóstico por imagen , Retina/patología , Angiografía con Fluoresceína/métodos , Ratones , Ratas , Roedores , Imagen Óptica/métodos , Humanos , Fondo de Ojo
6.
Methods Mol Biol ; 2848: 169-186, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39240523

RESUMEN

The retinal explant culture system is a valuable tool for studying the pharmacological, toxicological, and developmental aspects of the retina. It is also used for translational studies such as gene therapy. While no photoreceptor-like cell lines are available for in vitro studies of photoreceptor cell biology, the retinal explant culture maintains the laminated retinal structure ex vivo for as long as a month. Human and nonhuman primate (NHP) postmortem retinal explants cut into small pieces offer the possibility of testing multiple conditions for safety and adeno-associated viral (AAV) vector optimization. In addition, the cone-enriched foveal area can be studied using the retinal explants. Here, we present a detailed working protocol for retinal explant isolation and culture from mouse, human, and NHP for testing drug efficacy and AAV transduction. Future applications of this protocol include combining live imaging and multiwell retinal explant culture for high-throughput drug screening systems in rodent and human retinal explants to identify new drugs against retinal degeneration.


Asunto(s)
Dependovirus , Retina , Animales , Humanos , Ratones , Retina/citología , Dependovirus/genética , Primates , Vectores Genéticos/genética , Técnicas de Cultivo de Tejidos/métodos , Transducción Genética
7.
Methods Mol Biol ; 2848: 135-150, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39240521

RESUMEN

Mammals do not possess the ability to spontaneously repair or regenerate damaged retinal tissue. In contrast to teleost fish which are capable of retina regeneration through the action of Müller glia, mammals undergo a process of reactive gliosis and scarring that inhibits replacement of lost neurons. Thus, it is important to discover novel methods for stimulating mammalian Müller glia to dedifferentiate and produce progenitor cells that can replace lost retinal neurons. Inducing an endogenous regenerative pathway mediated by Müller glia would provide an attractive alternative to stem cell injections or gene therapy approaches. Extracellular vesicles (EVs) are now recognized to serve as a novel form of cell-cell communication through the transfer of cargo from donor to recipient cells or by the activation of signaling cascades in recipient cells. EVs have been shown to promote proliferation and regeneration raising the possibility that delivery of EVs could be a viable treatment for visual disorders. Here, we provide protocols to isolate EVs for use in retina regeneration experiments.


Asunto(s)
Vesículas Extracelulares , Regeneración , Retina , Animales , Vesículas Extracelulares/metabolismo , Retina/metabolismo , Retina/citología , Retina/fisiología , Células Ependimogliales/metabolismo , Células Ependimogliales/citología , Ratones , Comunicación Celular , Proliferación Celular , Regeneración Nerviosa/fisiología
8.
Biomaterials ; 312: 122732, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39088913

RESUMEN

Fully restoring the lost population of cardiomyocytes and heart function remains the greatest challenge in cardiac repair post myocardial infarction. In this study, a pioneered highly ROS-eliminating hydrogel was designed to enhance miR-19a/b induced cardiomyocyte proliferation by lowering the oxidative stress and continuously releasing miR-19a/b in infarcted myocardium in situ. In vivo lineage tracing revealed that ∼20.47 % of adult cardiomyocytes at the injected sites underwent cell division in MI mice. In MI pig the infarcted size was significantly reduced from 40 % to 18 %, and thereby marked improvement of cardiac function and increased muscle mass. Most importantly, our treatment solved the challenge of animal death--all the treated pigs managed to live until their hearts were harvested at day 50. Therefore, our strategy provides clinical conversion advantages and safety for healing damaged hearts and restoring heart function post MI, which will be a powerful tool to battle cardiovascular diseases in patients.


Asunto(s)
Proliferación Celular , MicroARNs , Infarto del Miocardio , Miocitos Cardíacos , Estrés Oxidativo , Animales , MicroARNs/metabolismo , MicroARNs/genética , Miocitos Cardíacos/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Ratones , Porcinos , Hidrogeles/química , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
9.
Biomaterials ; 312: 122739, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39096840

RESUMEN

The biofilm-induced "relatively immune-compromised zone" creates an immunosuppressive microenvironment that is a significant contributor to refractory infections in orthopedic endophytes. Consequently, the manipulation of immune cells to co-inhibit or co-activate signaling represents a crucial strategy for the management of biofilm. This study reports the incorporation of Mn2+ into mesoporous dopamine nanoparticles (Mnp) containing the stimulator of interferon genes (STING) pathway activator cGAMP (Mncp), and outer wrapping by M1-like macrophage cell membrane (m-Mncp). The cell membrane enhances the material's targeting ability for biofilm, allowing it to accumulate locally at the infectious focus. Furthermore, m-Mncp mechanically disrupts the biofilm through photothermal therapy and induces antigen exposure through photodynamic therapy-generated reactive oxygen species (ROS). Importantly, the modulation of immunosuppression and immune activation results in the augmentation of antigen-presenting cells (APCs) and the commencement of antigen presentation, thereby inducing biofilm-specific humoral immunity and memory responses. Additionally, this approach effectively suppresses the activation of myeloid-derived suppressor cells (MDSCs) while simultaneously boosting the activity of T cells. Our study showcases the efficacy of utilizing m-Mncp immunotherapy in conjunction with photothermal and photodynamic therapy to effectively mitigate residual and recurrent infections following the extraction of infected implants. As such, this research presents a viable alternative to traditional antibiotic treatments for biofilm that are challenging to manage.


Asunto(s)
Biopelículas , Indoles , Proteínas de la Membrana , Polímeros , Biopelículas/efectos de los fármacos , Polímeros/química , Animales , Indoles/química , Indoles/farmacología , Ratones , Proteínas de la Membrana/metabolismo , Nanopartículas/química , Fotoquimioterapia/métodos , Porosidad , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Femenino , Transducción de Señal/efectos de los fármacos , Terapia Fototérmica , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/efectos de los fármacos , Ratones Endogámicos C57BL
10.
Biomaterials ; 312: 122733, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39106819

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) demonstrates unique characteristics in anticancer therapies as it selectively induces apoptosis in cancer cells. However, most cancer cells are TRAIL-resistant. Odanacatib (ODN), a cathepsin K inhibitor, is considered a novel sensitizer for cancer treatment. Combination therapy between TRAIL and sensitizers is considered a potent platform that improves TRAIL-based anticancer therapies beyond TRAIL monotherapy. Herein, we developed ODN loaded poly(lactic-co-glycolic) nanoparticles conjugated to GST-TRAIL (TRAIL-ODN-PLGA-NPs) to target and treat TRAIL-resistant cancer. TRAIL-ODN-PLGA-NPs demonstrated a significant increase in cellular uptake via death receptors (DR5 and DR4) on surface of cancer cells. TRAIL-ODN-PLGA-NPs exposure destroyed more TRAIL-resistant cells compared to a single treatment with free drugs. The released ODN decreased the Raptor protein, thereby increasing damage to mitochondria by elevating reactive oxygen species (ROS) generation. Additionally, Bim protein stabilization improved TRAIL-resistant cell sensitization to TRAIL-induced apoptosis. The in vivo biodistribution study revealed that TRAIL-ODN-PLGA-NPs demonstrated high location and retention in tumor sites via the intravenous route. Furthermore, TRAIL-ODN-PLGA-NPs significantly inhibited xenograft tumor models of TRAIL-resistant Caki-1 and TRAIL-sensitive MDA-MB-231 cells.The inhibition was associated with apoptosis activation, Raptor protein stabilizing Bim protein downregulation, Bax accumulation, and mitochondrial ROS generation elevation. Additionally, TRAIL-ODN-PLGA-NPs affected the tumor microenvironment by increasing tumor necrosis factor-α and reducing interleukin-6. In conclusion, we evealed that our formulation demonstrated synergistic effects against TRAIL compared with the combination of free drug in vitro and in vivo models. Therefore, TRAIL-ODN-PLGA-NPs may be a novel candidate for TRAIL-induced apoptosis in cancer treatment.


Asunto(s)
Antineoplásicos , Compuestos de Bifenilo , Resistencia a Antineoplásicos , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ligando Inductor de Apoptosis Relacionado con TNF , Animales , Femenino , Humanos , Ratones , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/uso terapéutico , Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/química , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Especies Reactivas de Oxígeno/metabolismo , Distribución Tisular , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
11.
Biomaterials ; 312: 122707, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39121729

RESUMEN

Polypyrimidine tract-binding protein 1 (PTBP1) regulates numerous alternative splicing events during tumor progression and neurogenesis. Previously, PTBP1 downregulation was reported to convert astrocytes into functional neurons; however, how PTBP1 regulates astrocytic physiology remains unclear. In this study, we revealed that PTBP1 modulated glutamate uptake via ATP1a2, a member of Na+/K+-ATPases, and glutamate transporters in astrocytes. Ptbp1 knockdown altered mitochondrial function and energy metabolism, which involved PTBP1 regulating mitochondrial redox homeostasis via the succinate dehydrogenase (SDH)/Nrf2 pathway. The malfunction of glutamate transporters following Ptbp1 knockdown resulted in enhanced excitatory synaptic transmission in the cortex. Notably, we developed a biomimetic cationic triblock polypeptide system, i.e., polyethylene glycol44-polylysine30-polyleucine10 (PEG44-PLL30-PLLeu10) with astrocytic membrane coating to deliver Ptbp1 siRNA in vitro and in vivo, which approach allowed Ptbp1 siRNA to efficiently cross the blood-brain barrier and target astrocytes in the brain. Collectively, our findings suggest a framework whereby PTBP1 serves as a modulator in glutamate transport machinery, and indicate that biomimetic methodology is a promising route for in vivo siRNA delivery.


Asunto(s)
Astrocitos , Ácido Glutámico , Ribonucleoproteínas Nucleares Heterogéneas , Homeostasis , Factor 2 Relacionado con NF-E2 , Proteína de Unión al Tracto de Polipirimidina , ARN Interferente Pequeño , Animales , Astrocitos/metabolismo , Ácido Glutámico/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Factor 2 Relacionado con NF-E2/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ratones , Transducción de Señal , Membrana Celular/metabolismo , Ratones Endogámicos C57BL , Masculino , Humanos , Mitocondrias/metabolismo
12.
Biomaterials ; 312: 122731, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39153324

RESUMEN

Tumor-associated inflammation drives cancer progression and therapy resistance, often linked to the infiltration of monocyte-derived tumor-associated macrophages (TAMs), which are associated with poor prognosis in various cancers. To advance immunotherapies, testing on immunocompetent pre-clinical models of human tissue is crucial. We have developed an in vitro model of microvascular networks with tumor spheroids or patient tissues to assess monocyte trafficking into tumors and evaluate immunotherapies targeting the human tumor microenvironment. Our findings demonstrate that macrophages in vascularized breast and lung tumor models can enhance monocyte recruitment via CCL7 and CCL2, mediated by CSF-1R. Additionally, a multispecific antibody targeting CSF-1R, CCR2, and neutralizing TGF-ß (CSF1R/CCR2/TGF-ß Ab) repolarizes TAMs towards an anti-tumoral M1-like phenotype, reduces monocyte chemoattractant protein secretion, and blocks monocyte migration. This antibody also inhibits monocyte recruitment in patient-specific vascularized tumor models. In summary, this vascularized tumor model recapitulates the monocyte recruitment cascade, enabling functional testing of innovative therapeutic antibodies targeting TAMs in the tumor microenvironment.


Asunto(s)
Monocitos , Receptor de Factor Estimulante de Colonias de Macrófagos , Receptores CCR2 , Microambiente Tumoral , Humanos , Receptores CCR2/metabolismo , Receptores CCR2/antagonistas & inhibidores , Monocitos/metabolismo , Monocitos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral , Femenino , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Ratones , Movimiento Celular/efectos de los fármacos , Neoplasias/inmunología , Neoplasias/patología
13.
Biomaterials ; 312: 122746, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39106816

RESUMEN

Postoperative radiotherapy remains the gold standard for malignant glioma treatment. Clinical limitations, including tumor growth between surgery and radiotherapy and the emergence of radioresistance, reduce treatment effectiveness and result in local disease progression. This study aimed to develop a local drug delivery system to inhibit tumor growth before radiotherapy and enhance the subsequent anticancer effects of limited-dose radiotherapy. We developed a compound of carboplatin-loaded hydrogel (CPH) incorporated with carboplatin-loaded calcium carbonate (CPCC) to enable two-stage (peritumoral and intracellular) release of carboplatin to initially inhibit tumor growth and to synergize with limited-dose radiation (10 Gy in a single fraction) to eliminate malignant glioma (ALTS1C1 cells) in a C57BL/6 mouse subcutaneous tumor model. The doses of carboplatin in CPH and CPCC treatments were 150 µL (carboplatin concentration of 5 mg/mL) and 15 mg (carboplatin concentration of 4.1 µg/mg), respectively. Mice receiving the combination of CPH-CPCC treatment and limited-dose radiation exhibited significantly reduced tumor growth volume compared to those receiving double-dose radiation alone. Furthermore, combining CPH-CPCC treatment with limited-dose radiation resulted in significantly longer progression-free survival than combining CPH treatment with limited-dose radiation. Local CPH-CPCC delivery synergized effectively with limited-dose radiation to eliminate mouse glioma, offering a promising solution for overcoming clinical limitations.


Asunto(s)
Carbonato de Calcio , Carboplatino , Glioma , Hidrogeles , Ratones Endogámicos C57BL , Animales , Glioma/patología , Glioma/tratamiento farmacológico , Glioma/radioterapia , Carboplatino/administración & dosificación , Carboplatino/uso terapéutico , Carboplatino/farmacología , Hidrogeles/química , Línea Celular Tumoral , Carbonato de Calcio/química , Ratones , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia
14.
Biomaterials ; 312: 122744, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39106820

RESUMEN

Inflammation within the central nervous system (CNS), which may be triggered by surgical trauma, has been implicated as a significant factor contributing to postoperative cognitive dysfunction (POCD). The relationship between mitigating inflammation at peripheral surgical sites and its potential to attenuate the CNS inflammatory response, thereby easing POCD symptoms, remains uncertain. Notably, carbon monoxide (CO), a gasotransmitter, exhibits pronounced anti-inflammatory effects. Herein, we have developed carbon monoxide-releasing micelles (CORMs), a nanoparticle that safely and locally liberates CO upon exposure to 650 nm light irradiation. In a POCD mouse model, treatment with CORMs activated by light (CORMs + hv) markedly reduced the concentrations of interleukin (IL)-6, IL-1ß, and tumor necrosis factor-alpha (TNF-α) in both the peripheral blood and the hippocampus, alongside a decrease in ionized calcium-binding adapter molecule 1 in the hippocampal CA1 region. Furthermore, CORMs + hv treatment diminished Evans blue extravasation, augmented the expression of tight junction proteins zonula occludens-1 and occludin, enhanced neurocognitive functions, and fostered fracture healing. Bioinformatics analysis and experimental validation has identified Htr1b and Trhr as potential key regulators in the neuroactive ligand-receptor interaction signaling pathway implicated in POCD. This work offers new perspectives on the mechanisms driving POCD and avenues for therapeutic intervention.


Asunto(s)
Monóxido de Carbono , Luz , Complicaciones Cognitivas Postoperatorias , Animales , Complicaciones Cognitivas Postoperatorias/etiología , Complicaciones Cognitivas Postoperatorias/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Micelas , Luz Roja
15.
Biomaterials ; 312: 122721, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39106817

RESUMEN

Silver nanoparticles (AgNPs) are a potential antiviral agent due to their ability to disrupt the viral particle or alter the virus metabolism inside the host cell. In vitro, AgNPs exhibit antiviral activity against the most common human respiratory viruses. However, their capacity to modulate immune responses during respiratory viral infections has yet to be explored. This study demonstrates that administering AgNPs directly into the lungs prior to infection can reduce viral loads and therefore virus-induced cytokines in mice infected with influenza virus or murine pneumonia virus. The prophylactic effect was diminished in mice with depleted lymphoid cells. We showed that AgNPs-treatment resulted in the recruitment and activation of lymphocytes in the lungs, particularly natural killer (NK) cells. Mechanistically, AgNPs enhanced the ability of alveolar macrophages to promote both NK cell migration and IFN-γ production. By contrast, following infection, in mice treated with AgNPs, NK cells exhibited decreased activation, indicating that these nanoparticles can regulate the potentially deleterious activation of these cells. Overall, the data suggest that AgNPs may possess prophylactic antiviral properties by recruiting and controlling the activation of lymphoid cells through interaction with alveolar macrophages.


Asunto(s)
Células Asesinas Naturales , Pulmón , Nanopartículas del Metal , Infecciones por Orthomyxoviridae , Plata , Animales , Plata/química , Plata/farmacología , Nanopartículas del Metal/química , Pulmón/virología , Pulmón/patología , Pulmón/efectos de los fármacos , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Ratones , Células Asesinas Naturales/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Ratones Endogámicos C57BL , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , Femenino , Activación de Linfocitos/efectos de los fármacos
16.
Biomaterials ; 312: 122724, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39106818

RESUMEN

The residual bone tumor and defects which is caused by surgical therapy of bone tumor is a major and important problem in clinicals. And the sequential treatment for irradiating residual tumor and repairing bone defects has wildly prospects. In this study, we developed a general modification strategy by gallic acid (GA)-assisted coordination chemistry to prepare black calcium-based materials, which combines the sequential photothermal therapy of bone tumor and bone defects. The GA modification endows the materials remarkable photothermal properties. Under the near-infrared (NIR) irradiation with different power densities, the black GA-modified bone matrix (GBM) did not merely display an excellent performance in eliminating bone tumor with high temperature, but showed a facile effect of the mild-heat stimulation to accelerate bone regeneration. GBM can efficiently regulate the microenvironments of bone regeneration in a spatial-temporal manner, including inflammation/immune response, vascularization and osteogenic differentiation. Meanwhile, the integrin/PI3K/Akt signaling pathway of bone marrow mesenchymal stem cells (BMSCs) was revealed to be involved in the effect of osteogenesis induced by the mild-heat stimulation. The outcome of this study not only provides a serial of new multifunctional biomaterials, but also demonstrates a general strategy for designing novel blacked calcium-based biomaterials with great potential for clinical use.


Asunto(s)
Neoplasias Óseas , Regeneración Ósea , Calcio , Ácido Gálico , Células Madre Mesenquimatosas , Ácido Gálico/química , Regeneración Ósea/efectos de los fármacos , Animales , Calcio/metabolismo , Neoplasias Óseas/terapia , Neoplasias Óseas/tratamiento farmacológico , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Terapia Fototérmica/métodos , Osteogénesis/efectos de los fármacos , Ratones , Humanos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Línea Celular Tumoral
17.
Biomaterials ; 312: 122742, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39106821

RESUMEN

Hypertrophic scar (HS) tends to raised above skin level with high inflammatory microenvironment and excessive proliferation of myofibroblasts. The HS therapy remains challenging due to dense scar tissue which makes it hard to penetrate, and the side effects resulting from intralesional corticosteroid injection which is the mainstay treatment in clinic. Herein, bilayer microneedle patches combined with dexamethasone and colchicine (DC-MNs) with differential dual-release pattern is designed. Two drugs loaded in commercially available materials HA and PLGA, respectively. Specifically, after administration, outer layer rapidly releases the anti-inflammatory drug dexamethasone, which inhibits macrophage polarization to pro-inflammatory phenotype in scar tissue. Subsequently, inner layer degrades sustainedly, releasing antimicrotubular agent colchicine, which suppresses the overproliferation of myofibroblasts with extremely narrow therapeutic window, and inhibits the overexpression of collagen, as well as promotes the regular arrangement of collagen. Only applied once, DC-MNs directly delivered drugs to the scar tissue. Compared to traditional treatment regimen, DC-MNs significantly suppressed HS at lower dosage and frequency by differential dual-release design. Therefore, this study put forward the idea of integrated DC-MNs accompany the development of HS, providing a non-invasive, self-applicable, more efficient and secure strategy for treatment of HS.


Asunto(s)
Antiinflamatorios , Cicatriz Hipertrófica , Colchicina , Dexametasona , Miofibroblastos , Agujas , Cicatriz Hipertrófica/tratamiento farmacológico , Cicatriz Hipertrófica/patología , Animales , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Dexametasona/farmacología , Dexametasona/administración & dosificación , Dexametasona/uso terapéutico , Antiinflamatorios/uso terapéutico , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Colchicina/farmacología , Colchicina/administración & dosificación , Ratones , Sistemas de Liberación de Medicamentos , Humanos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química
18.
Biomaterials ; 312: 122743, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39111233

RESUMEN

Photodynamic therapy (PDT) is an appealing modality for cancer treatments. However, the limited tissue penetration depth of external-excitation light makes PDT impossible in treating deep-seated tumors. Meanwhile, tumor hypoxia and intracellular reductive microenvironment restrain the generation of reactive oxygen species (ROS). To overcome these limitations, a tumor-targeted self-illuminating supramolecular nanoparticle T-NPCe6-L-N is proposed by integrating photosensitizer Ce6 with luminol and nitric oxide (NO) for chemiluminescence resonance energy transfer (CRET)-activated PDT. The high H2O2 level in tumor can trigger chemiluminescence of luminol to realize CRET-activated PDT without exposure of external light. Meanwhile, the released NO significantly relieves tumor hypoxia via vascular normalization and reduces intracellular reductive GSH level, further enhancing ROS abundance. Importantly, due to the different ROS levels between cancer cells and normal cells, T-NPCe6-L-N can selectively trigger PDT in cancer cells while sparing normal cells, which ensured low side effect. The combination of CRET-based photosensitizer-activation and tumor microenvironment modulation overcomes the innate challenges of conventional PDT, demonstrating efficient inhibition of orthotopic and metastatic tumors on mice. It also provoked potent immunogenic cell death to ensure long-term suppression effects. The proof-of-concept research proved as a new strategy to solve the dilemma of PDT in treatment of deep-seated tumors.


Asunto(s)
Nanopartículas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Microambiente Tumoral , Fotoquimioterapia/métodos , Microambiente Tumoral/efectos de los fármacos , Animales , Nanopartículas/química , Fármacos Fotosensibilizantes/uso terapéutico , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Humanos , Ratones , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Transferencia de Energía , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Ratones Endogámicos BALB C , Luz , Ratones Desnudos , Óxido Nítrico/metabolismo
19.
Biomaterials ; 312: 122749, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39121725

RESUMEN

The prevalence of Alzheimer's disease (AD) is increasing globally due to population aging. However, effective clinical treatment strategies for AD still remain elusive. The mechanisms underlying AD onset and the interplay between its pathological factors have so far been unclear. Evidence indicates that AD progression is ultimately driven by neuronal loss, which in turn is caused by neuroapoptosis and neuroinflammation. Therefore, the inhibition of neuroapoptosis and neuroinflammation could be a useful anti-AD strategy. Nonetheless, the delivery of active drug agents into the brain parenchyma is hindered by the blood-brain barrier (BBB). To address this challenge, we fabricated a black phosphorus nanosheet (BP)-based methylene blue (MB) delivery system (BP-MB) for AD therapy. After confirming the successful preparation of BP-MB, we proved that its BBB-crossing ability was enhanced under near-infrared light irradiation. In vitro pharmacodynamics analysis revealed that BP and MB could synergistically scavenge excessive reactive oxygen species (ROS) in okadaic acid (OA)-treated PC12 cells and lipopolysaccharide (LPS)-treated BV2 cells, thus efficiently reversing neuroapoptosis and neuroinflammation. To study in vivo pharmacodynamics, we established a mouse model of AD mice, and behavioral tests confirmed that BP-MB treatment could successfully improve cognitive function in these animals. Notably, the results of pathological evaluation were consistent with those of the in vitro assays. The findings demonstrated that BP-MB could scavenge excessive ROS and inhibit Tau hyperphosphorylation, thereby alleviating downstream neuroapoptosis and regulating the polarization of microglia from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. Overall, this study highlights the therapeutic potential of a smart nanomedicine with the capability of reversing neuroapoptosis and neuroinflammation for AD treatment.


Asunto(s)
Enfermedad de Alzheimer , Apoptosis , Barrera Hematoencefálica , Azul de Metileno , Nanomedicina , Enfermedades Neuroinflamatorias , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Apoptosis/efectos de los fármacos , Células PC12 , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Ratas , Ratones , Nanomedicina/métodos , Azul de Metileno/farmacología , Azul de Metileno/uso terapéutico , Masculino , Especies Reactivas de Oxígeno/metabolismo , Ratones Endogámicos C57BL
20.
Biomaterials ; 312: 122751, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39121726

RESUMEN

Tumor immunotherapies have emerged as a promising frontier in the realm of cancer treatment. However, challenges persist in achieving localized, durable immunostimulation while counteracting the tumor's immunosuppressive environment. Here, we develop a natural mussel foot protein-based nanomedicine with spatiotemporal control for tumor immunotherapy. In this nanomedicine, an immunoadjuvant prodrug and a photosensitizer are integrated, which is driven by their dynamic bonding and non-covalent assembling with the protein carrier. Harnessing the protein carrier's bioadhesion, this nanomedicine achieves a drug co-delivery with spatiotemporal precision, by which it not only promotes tumor photothermal ablation but also broadens tumor antigen repertoire, facilitating in situ immunotherapy with durability and maintenance. This nanomedicine also modulates the tumor microenvironment to overcome immunosuppression, thereby amplifying antitumor responses against tumor progression. Our strategy underscores a mussel foot protein-derived design philosophy of drug delivery aimed at refining combinatorial immunotherapy, offering insights into leveraging natural proteins for cancer treatment.


Asunto(s)
Inmunoterapia , Nanomedicina , Animales , Inmunoterapia/métodos , Nanomedicina/métodos , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/uso terapéutico , Fármacos Fotosensibilizantes/farmacología , Terapia Fototérmica/métodos , Ratones , Humanos , Microambiente Tumoral/efectos de los fármacos , Línea Celular Tumoral , Proteínas/química , Femenino , Neoplasias/terapia , Neoplasias/inmunología , Adhesivos/química , Ratones Endogámicos C57BL , Adyuvantes Inmunológicos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA