Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38953708

RESUMEN

The coronavirus (COVID-19) pandemic has underscored the critical role of mRNA-based vaccines as powerful, adaptable, readily manufacturable, and safe methodologies for prophylaxis. mRNA-based treatments are emerging as a hopeful avenue for a plethora of conditions, encompassing infectious diseases, cancer, autoimmune diseases, genetic diseases, and rare disorders. Nonetheless, the in vivo delivery of mRNA faces challenges due to its instability, suboptimal delivery, and potential for triggering undesired immune reactions. In this context, the development of effective drug delivery systems, particularly nanoparticles (NPs), is paramount. Tailored with biophysical and chemical properties and susceptible to surface customization, these NPs have demonstrated enhanced mRNA delivery in vivo and led to the approval of several NPs-based formulations for clinical use. Despite these advancements, the necessity for developing a refined, targeted NP delivery system remains imperative. This review comprehensively surveys the biological, translational, and clinical progress in NPs-mediated mRNA therapeutics for both the prevention and treatment of diverse diseases. By addressing critical factors for enhancing existing methodologies, it aims to inform the future development of precise and efficacious mRNA-based therapeutic interventions.

2.
Am J Transplant ; 22(9): 2246-2253, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35373451

RESUMEN

Thrombospondin-1 (TSP-1) is a key mediator of renal ischemia-reperfusion injury (IRI), a major cause of kidney dysfunction under various disease conditions and a risk factor of renal allograft rejection. In this study, we developed a nanotechnology-based therapy targeting TSP-1 to prevent renal IRI. A biocompatible nanoparticle (NP) capable of specific binding to TSP-1 was prepared by conjugating NPs with TSP-1-binding (LSKL) peptides. LSKL/NPs not only effectively adsorbed recombinant TSP-1 proteins in vitro, but also efficiently neutralized TSP-1 in mice undergoing renal IRI. IRI-induced elevation of TSP-1 in the kidney was significantly inhibited by post-IR treatment with LSKL/NPs, but not free LSKL or NPs. Furthermore, TSP-1 proteins adsorbed on LSKL/NPs were functionally inactive and unable to induce apoptosis in renal tubular epithelial cells. Importantly, LSKL/NPs induced strong protection against renal IRI, as shown by markedly diminished serum creatinine levels and improved histological lesions of the kidney. Thus, LSKL/NPs provide a useful means of depleting and inactivating TSP-1 and a potential therapy for renal IRI.


Asunto(s)
Trasplante de Riñón , Nanopartículas , Daño por Reperfusión , Animales , Apoptosis , Riñón/patología , Trasplante de Riñón/efectos adversos , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Trombospondina 1/antagonistas & inhibidores , Trombospondina 1/metabolismo , Trombospondina 1/farmacología
3.
Angew Chem Int Ed Engl ; 61(20): e202117158, 2022 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-35102683

RESUMEN

In photoswitches that undergo fluorescence switching upon ultraviolet irradiation, photoluminescence and photoisomerization often occur simultaneously, leading to unstable fluorescence properties. Here, we successfully demonstrated reversible solid-state triple fluorescence switching through "Pump-Trigger" multiphoton manipulation. A novel fluorescence photoswitch, BOSA-SP, achieved green, yellow, and red fluorescence under excitation by pump light and isomerization induced by trigger light. The energy ranges of photoexcitation and photoisomerization did not overlap, enabling appropriate selection of the multiphoton light for "pump" and "trigger" photoswitching, respectively. Additionally, the large free volume of the spiropyran (SP) moiety in the solid state promoted reversible photoisomerization. Switching between "pump" and "trigger" light is useful for three-color tunable switching cell imaging, which can be exploited in programmable fluorescence switching. Furthermore, we exploited reversible dual-fluorescence switching in a single molecular system to successfully achieve two-color super-resolution imaging.


Asunto(s)
Rayos Ultravioleta , Microscopía Fluorescente
4.
Nano Lett ; 20(8): 6191-6198, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32697585

RESUMEN

Vascular disrupting agents (VDAs) are emerging anticancer agents, which show rising demand for combination with cytostatic drugs (CSDs), owing to inadequate tumor inhibition when applied singly. Nevertheless, the combination remains a challenge due to the different working sites of VDAs and CSDs and hypoxia-induced drug resistance after disrupting neovasculature by VDAs. Herein, we developed a shell-stacked nanoparticle (SNP) for coencapsulation of a VDA combretastatin A-4 phosphate (CA4P) and a proteasome inhibitor bortezomib (BTZ). The SNP could spatiotemporally deliver CA4P to tumor neovasculature and BTZ to tumor cells mediated by the site-specific stimuli-activated drug release. Moreover, the SNP also reversed the drug resistance caused by the overexpressed ABCG2 under CA4P-induced hypoxic conditions. The spatiotemporally targeted combination therapy significantly inhibited the growth of both the human A549 pulmonary adenocarcinoma xenograft model and patient-derived xenograft (PDX) model of colon cancer in mice, providing a promising strategy for treating advanced cancers.


Asunto(s)
Antineoplásicos , Estilbenos , Animales , Antineoplásicos/farmacología , Resistencia a Medicamentos , Hipoxia , Ratones , Nanomedicina
5.
Biochem Biophys Res Commun ; 510(4): 551-557, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30739781

RESUMEN

Currently, SAMHD1 is the only known dNTPase in human cells. It also suppresses the replication of both retroviruses and retroelements. SAMHD1 contains a classic nuclear localization sequence (NLS) and resides in the nucleus in live or fixed cells. It has been reported that alteration or removal of NLS does not affect the dNTPase or the antiviral activity of SAMHD1. However, it was unclear whether the nuclear localization was involved in SAMHD1-mediated suppression against retroelements such as long interspersed element type 1 (LINE-1 or L1). In this study, we reported that SAMHD1 is a nucleocytoplasmic shuttling protein. Digitonin-based cytoplasm/nucleus fractionation tests suggested that SAMHD1 is capable of being exported from the nucleus, which was confirmed by introducing exogenous exportin Xpo1 in live cells. Interestingly, altering the protein's subcellular localization by mutating or removing NLS significantly enhances SAMHD1's potency in L1 suppression. Further tests with SAMHD1 mutants indicated that nucleocytoplasmic shuttling is important for SAMHD1-mediated L1 suppression. Finally, we demonstrated that the cytoplasmic distribution of SAMHD1 leads to an enhanced depletion of L1 ORF2p. Taken together, our data have revealed SAMHD1 as a nucleocytoplasmic shuttling protein, and associated such a new feature of SAMHD1 with its potency against L1 retrotransposition, which provides more insights to the understanding of SAMHD1 and its role in L1 suppression.


Asunto(s)
Elementos de Nucleótido Esparcido Largo , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HEK293 , Humanos , Señales de Localización Nuclear/metabolismo
6.
Biochem Biophys Res Commun ; 511(4): 910-915, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30851937

RESUMEN

Interaction between HIV-1 Vif and host factor CBFß leads to the assembly of the Vif-Cul5-EloB/C ubiquitin ligase (E3 complex). By inducing the formation of E3 complex, Vif depletes host APOBEC3 restriction factors and promotes HIV-1 infection. In addition, Vif is known to arrest host cells at G2/M phase (G2 arrest), benefiting HIV-1 replication and contributing to the depletion of CD4+ T cells. However, whether CBFß is also involved in Vif-induced cell cycle arrest remains unclear. In the present study, we report that CBFß is an essential factor for Vif-induced G2 arrest. Reducing endogenous CBFß expression significantly compromised Vif's potency in cell cycle regulation. In addition, tests with CBFß and Vif mutants indicated that Vif-CBFß interaction is crucial for Vif to induce G2 arrest. Furthermore, suppressors against Vif-hijacked E3 complex or proteasome-mediated proteolysis also abolished Vif's ability to cause G2 arrest. In general, our data indicated that Vif induces G2 arrest through depletion of a yet-unknown cellular factor, where the involvement of CBFß is essential. On the other hand, our data also suggested that, antiviral drugs targeting the Vif-CBFß interaction have the potential to abolish Vif's ability to cause APOBEC3 degradation as well as G2 arrest in host cells, thus reducing both HIV-1 replication and Vif-induced CD4+ T-cell depletion.


Asunto(s)
Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Infecciones por VIH/metabolismo , VIH-1/fisiología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Células HEK293 , Infecciones por VIH/patología , Interacciones Huésped-Patógeno , Humanos , Mapas de Interacción de Proteínas
7.
Nano Lett ; 17(7): 4526-4533, 2017 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-28644032

RESUMEN

Targeted delivery of chemotherapeutic drugs to the desired lesion sites is the main objective in malignancy treatment, especially in highly metastatic malignancies. However, extensive studies around the world on traditional targeting strategies of recognizing either overexpressed receptors or microenvironments in tumors show great limitations, owing to the off-target effect and tumor homogeneity. Integration of both receptor-mediated targeting (RMT) and environment-mediated targeting (EMT) enhances the tumor accumulation and subsequent cell uptake at the same time, which may avoid these limitations. Herein, a dual targeting nanogel of PMNG engineered with both phenylboronic acid (PBA) and morpholine (MP) was reported for not only RMT via specific recognition of sialyl (SA) epitopes but also EMT toward extracellular acidity. Further engineering the nanoparticles via loading doxorubicin (DOX) brought a novel dual targeting system, that is, PMNG/DOX. PMNG/DOX demonstrated a greater targeting effect to both primary and metastatic B16F10 melanoma than the single PBA-modified nanogel (PNG) with only RMT in vitro and in vivo. Moreover, PMNG/DOX was also proved to be highly potent on inhibiting primary tumor growth as well as tumor metastasis on B16F10 melanoma-grafted mouse model. The results demonstrated the dual targeting design as a translational approach for drug delivery to highly metastatic tumor.


Asunto(s)
Antineoplásicos/administración & dosificación , Nanopartículas/química , Microambiente Tumoral/fisiología , Animales , Antineoplásicos/química , Ácidos Borónicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Portadores de Fármacos/química , Liberación de Fármacos , Geles , Humanos , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Ratones , Morfolinas/química , Metástasis de la Neoplasia , Tamaño de la Partícula , Péptidos/química , Propiedades de Superficie , Distribución Tisular
8.
Angew Chem Int Ed Engl ; 53(46): 12320-64, 2014 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-25294565

RESUMEN

In medicine, nanotechnology has sparked a rapidly growing interest as it promises to solve a number of issues associated with conventional therapeutic agents, including their poor water solubility (at least, for most anticancer drugs), lack of targeting capability, nonspecific distribution, systemic toxicity, and low therapeutic index. Over the past several decades, remarkable progress has been made in the development and application of engineered nanoparticles to treat cancer more effectively. For example, therapeutic agents have been integrated with nanoparticles engineered with optimal sizes, shapes, and surface properties to increase their solubility, prolong their circulation half-life, improve their biodistribution, and reduce their immunogenicity. Nanoparticles and their payloads have also been favorably delivered into tumors by taking advantage of the pathophysiological conditions, such as the enhanced permeability and retention effect, and the spatial variations in the pH value. Additionally, targeting ligands (e.g., small organic molecules, peptides, antibodies, and nucleic acids) have been added to the surface of nanoparticles to specifically target cancerous cells through selective binding to the receptors overexpressed on their surface. Furthermore, it has been demonstrated that multiple types of therapeutic drugs and/or diagnostic agents (e.g., contrast agents) could be delivered through the same carrier to enable combination therapy with a potential to overcome multidrug resistance, and real-time readout on the treatment efficacy. It is anticipated that precisely engineered nanoparticles will emerge as the next-generation platform for cancer therapy and many other biomedical applications.


Asunto(s)
Antineoplásicos/administración & dosificación , Preparaciones de Acción Retardada/química , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Animales , Preparaciones de Acción Retardada/metabolismo , Sistemas de Liberación de Medicamentos , Humanos , Nanomedicina/métodos , Nanopartículas/metabolismo , Nanotecnología/métodos
9.
Biomed Pharmacother ; 175: 116702, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38729052

RESUMEN

In recent years, nanoparticles have been broadly utilized in various drugs delivery formulations. Nanodelivery systems have shown promise in solving problems associated with the distribution of hydrophobic drugs and have promoted the accumulation of nanomedicines in the circulation or in organs. However, the injection dose of nanoparticles (NPs) is much greater than that needed by diseased tissues or organs. In other words, most of the NPs are localized off-target and do not reach the desired tissue or organs. With the rapid development of biodegradable and biosafety nanomaterials, the nanovectors represent assurance of safety. However, the off-target effects also induce concerns about the application of NPs, especially in the delivery of gene editing tools. Therefore, a complete understanding of the biological responses to NPs in the body will clearly guide the design of targeted delivery of NPs. The different properties of various nanodelivery systems may induce diverse interactions between carriers and organs. In this review, we describe the relationship between the liver, the most influenced organ of systemic administration of NPs, and targeted delivery nanoplatforms. Various transport vehicles have adopted multiple delivery strategies for the targeted delivery to the cells in the homeostasis liver and in diseased liver. Additionally, nanodelivery systems provide a novel strategy for treating incurable diseases. The appearance of a targeted delivery has profoundly improved the application of NPs to liver diseases.


Asunto(s)
Sistemas de Liberación de Medicamentos , Hepatopatías , Nanopartículas , Humanos , Hepatopatías/tratamiento farmacológico , Hepatopatías/metabolismo , Animales , Sistemas de Liberación de Medicamentos/métodos , Sistema de Administración de Fármacos con Nanopartículas/química , Hígado/metabolismo , Hígado/efectos de los fármacos , Portadores de Fármacos/química , Nanomedicina/métodos
10.
Int J Biol Macromol ; 268(Pt 1): 131679, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38641274

RESUMEN

Over the past few decades, significant strides have been made in understanding the pivotal roles that chemokine networks play in tumor biology. These networks, comprising chemokines and their receptors, wield substantial influence over cancer immune regulation and therapeutic outcomes. As a result, targeting these chemokine systems has emerged as a promising avenue for cancer immunotherapy. However, therapies targeting chemokines face significant challenges in solid tumor treatment, due to the complex and fragile of the chemokine networks. A nuanced comprehension of the complicacy and functions of chemokine networks, and their impact on the tumor microenvironment, is essential for optimizing their therapeutic utility in oncology. This review elucidates the ways in which chemokine networks interact with cancer immunity and tumorigenesis. We particularly elaborate on recent innovations in manipulating these networks for cancer treatment. The review also highlights future challenges and explores potential biomaterial strategies for clinical applications.


Asunto(s)
Quimiocinas , Neoplasias , Microambiente Tumoral , Humanos , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Quimiocinas/metabolismo , Animales , Inmunoterapia/métodos , Portadores de Fármacos/química , Nanopartículas/química , Nanopartículas/uso terapéutico , Receptores de Quimiocina/metabolismo
11.
Mater Today Bio ; 27: 101131, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39050986

RESUMEN

The limitations of platinum in ovarian cancer therapy, such as poor solubility and significant side effects, often lead to suboptimal therapeutic outcome and mortality. In this study, we have developed a novel approach utilizing biodegradable polymeric nanoparticles as a drug delivery system (NDDS), loaded with advanced platinum (IV) (Pt(IV)) prodrugs. A key feature of our approach is the enhancement of nanoparticles with maleimide, a modification hypothesized to significantly boost tumor tissue accumulation. When tested in mouse models of orthotopic and peritoneal metastasis ovarian cancer, these maleimide-modified nanoparticles are anticipated to show preferential accumulation in tumor tissues, enhancing therapeutic efficiency and minimizing systemic drug exposure. Our findings demonstrate that the maleimide-modified Pt(IV)-loaded NDDSs significantly reduce tumor burden in comparison to traditional cisplatin therapy, while simultaneously reducing adverse side effects. This leads to markedly improved survival rates in models of peritoneal metastasis ovarian cancer, offering a promising new direction in the treatment of this challenging disease.

12.
J Control Release ; 366: 425-447, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38154540

RESUMEN

The development of autoimmune diseases and the rejection of transplanted organs are primarily caused by an exaggerated immune response to autoantigens or graft antigens. Achieving immune tolerance is crucial for the effective treatment of these conditions. However, traditional therapies often have limited therapeutic efficacy and can result in systemic toxic effects. The emergence of nanomedicine offers a promising avenue for addressing immune-related diseases. Among the various nanoparticle formulations, cationic nanoparticles have demonstrated significant potential in inducing immune tolerance. In this review, we provide an overview of the underlying mechanism of autoimmune disease and organ transplantation rejection. We then highlight the recent advancements and advantages of utilizing cationic nanoparticles for inducing immune tolerance in the treatment of autoimmune diseases and the prevention of transplant rejection.


Asunto(s)
Enfermedades Autoinmunes , Nanopartículas , Humanos , Enfermedades Autoinmunes/terapia , Autoantígenos , Cationes , Tolerancia Inmunológica
13.
Biomater Sci ; 12(9): 2381-2393, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38500446

RESUMEN

The inability to systemic administration of nanoparticles, particularly cationic nanoparticles, has been a significant barrier to their clinical translation due to toxicity concerns. Understanding the in vivo behavior of cationic lipids is crucial, given their potential impact on critical biological components such as immune cells and hematopoietic stem cells (HSC). These cells are essential for maintaining the body's homeostasis, and their interaction with cationic lipids is a key factor in determining the safety and efficacy of these nanoparticles. In this study, we focused on the cytotoxic effects of cationic lipid/DNA complexes (CLN/DNA). Significantly, we observed that the most substantial cytotoxic effects, including a marked increase in numbers of long-term hematopoietic stem cells (LT-HSC), occurred 24 h post-CLN/DNA treatment in mice. Furthermore, we found that CLN/DNA-induced HSC expansion in bone marrow (BM) led to a notable decrease in the ability to reestablish blood cell production. Our study provides crucial insights into the interaction between cationic lipids and vital cellular components of the immune and hematopoietic systems.


Asunto(s)
Cationes , ADN , Células Madre Hematopoyéticas , Lípidos , Animales , ADN/química , ADN/administración & dosificación , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Cationes/química , Lípidos/química , Nanopartículas/química , Nanopartículas/administración & dosificación , Ratones Endogámicos C57BL
14.
Sci Bull (Beijing) ; 69(7): 922-932, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38331707

RESUMEN

Neoantigen cancer vaccines have been envisioned as one of the most promising means for cancer therapies. However, identifying neoantigens for tumor types with low tumor mutation burdens continues to limit the effectiveness of neoantigen vaccines. Herein, we proposed a "hit-and-run" vaccine strategy which primes T cells to attack tumor cells decorated with exogenous "neo-antigens". This vaccine strategy utilizes a peptide nanovaccine to elicit antigen-specific T cell responses after tumor-specific decoration with a nanocarrier containing the same peptide antigens. We demonstrated that a poly(2-oxazoline)s (POx) conjugated with OVA257-264 peptide through a matrix metalloprotease 2 (MMP-2) sensitive linker could efficiently and selectively decorate tumor cells with OVA peptides in vivo. Then, a POx-based nanovaccine containing OVA257-264 peptides to elicit OVA-specific T cell responses was designed. In combination with this hit-and-run vaccine system, an effective vaccine therapy was demonstrated across tumor types even without OVA antigen expression. This approach provides a promising and uniform vaccine strategy against tumors with a low tumor mutation burden.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Humanos , Epítopos , Antígenos de Neoplasias , Neoplasias/terapia , Péptidos
15.
Biomed Pharmacother ; 177: 117011, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38917758

RESUMEN

Microglia are essential for maintaining homeostasis and responding to pathological events in the central nervous system (CNS). Their dynamic and multidimensional states in different environments are pivotal factors in various CNS disorders. However, therapeutic modulation of microglial states is challenging due to the intricate balance these cells maintain in the CNS environment and the blood-brain barrier's restriction of drug delivery. Nanomedicine presents a promising avenue for addressing these challenges, offering a method for the targeted and efficient modulation of microglial states. This review covers the challenges faced in microglial therapeutic modulation and potential use of nanoparticle-based drug delivery systems. We provide an in-depth examination of nanoparticle applications for modulating microglial states in a range of CNS disorders, encompassing neurodegenerative and autoimmune diseases, infections, traumatic injuries, stroke, tumors, chronic pain, and psychiatric conditions. This review highlights the recent advancements and future prospects in nanomedicine for microglial modulation, paving the way for future research and clinical applications of therapeutic interventions in CNS disorders.

17.
Open Life Sci ; 18(1): 20220595, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37070078

RESUMEN

The cytoskeleton plays an important role in epilepsy; however, the mechanism is unknown. Therefore, this study aimed to reveal the mechanism of cytoskeletal proteins in epilepsy by investigating the expression of cytoskeletal proteins and synaptophysin (SYP) in mice at 0, 3, 6, and 24 h, 3 days, and 7 days in a kainic acid (KA)-induced epileptic model. Our results demonstrated that the expression of F-actin decreased significantly between 3 and 6 h, 6 and 24 h, and 24 h and 3 days (P < 0.05). Meanwhile, the expression of the neurofilament light chain, neurofilament medium chain, and neurofilament heavy chain subunits was significantly decreased (P < 0.001) at 3 h after the KA injection compared to the KA 0 h group, followed by an elevation at 6 h and a further decrease at 24 h compared to at 6 h. SYP expression was significantly decreased between 0 and 3 h as well as between 3 and 6 h (P < 0.05). At 24 h, the level was increased compared to at 6 h and continued to increase at 3 days after the KA injection. Thus, we propose that cytoskeletal proteins may be involved in the pathogenesis of epilepsy.

18.
Front Bioeng Biotechnol ; 11: 1242126, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37877041

RESUMEN

Dendritic cells (DCs) are the major specialized antigen-presenting cells (APCs), play a key role in initiating the body's immune response, maintain the balance of immunity. DCs can also induce immune tolerance by rendering effector T cells absent and anergy, and promoting the expansion of regulatory T cells. Induction of tolerogenic DCs has been proved to be a promising strategy for the treatment of autoimmune diseases, organ transplantation, and allergic diseases by various laboratory researches and clinical trials. The development of nano-delivery systems has led to advances in situ modulation of the tolerance phenotype of DCs. By changing the material composition, particle size, zeta-potential, and surface modification of nanoparticles, nanoparticles can be used for the therapeutic payloads targeted delivery to DCs, endowing them with great potential in the induction of immune tolerance. This paper reviews how nano-delivery systems can be modulated for targeted delivery to DCs and induce immune tolerance and reviews their potential in the treatment of autoimmune diseases, organ transplantation, and allergic diseases.

19.
Adv Mater ; 35(47): e2309667, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37807931

RESUMEN

T cells play a basic and key role in immunotherapy against solid tumors, and efficiently recruiting them into neoplastic foci and sustaining long-term effector function are consistent goals that remain a critical challenge. Here, an injectable alginate-based hydrogel with abundant ß-cyclodextrin (ALG-ßCD) sites is developed and intratumorally injected to recruit CCR9+ CD8+ T cells (a subset of T cells with robust antitumor activity) via the trapped chemokine CCL25. In the meantime, an intravenously injected adamantane-decorated anti-PD1 antibody (Ad-aPD1) would hitchhike on recruited CCR9+ CD8+ T cells to achieve the improved intratumoral accumulation of Ad-aPD1. Moreover, the Ad-PD1 and Ad-PDL1 antibodies are immobilized in the ALG-ßCD hydrogel through supramolecular host-guest interactions of Ad and ßCD, which facilitate engagement between CD8+ T cells and tumor cells and reinvigorate CD8+ T cells to avoid exhaustion. Based on this treatment strategy, T cell-mediated anticancer activity is promoted at multiple levels, eventually achieving superior antitumor efficacy in both orthotopic and postsurgical B16-F10 tumor models.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Hidrogeles/metabolismo , Inmunoterapia , Neoplasias/terapia , Neoplasias/metabolismo
20.
Biomater Sci ; 11(17): 5931-5941, 2023 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-37470222

RESUMEN

The imbalance of the immune system can lead to the occurrence of autoimmune diseases. Controlling and regulating the proliferation and function of effector T (Teff) cells and regulatory T (Treg) cells becomes the key to treating these diseases. Dendritic cells (DCs), as dedicated antigen-presenting cells, play a key role in inducing the differentiation of naive CD4+ T cells. In this study, we designed a cationic lipid-assisted PEG-PLGA nanoparticle (NPs/VD3/siLkb1) to deliver 1,25-dihydroxyvitamin D3 (VD3) and small interfering RNA (siRNA) to DC cells in the draining lymph nodes. By modulating the phenotypic changes of DC cells, this approach expands Treg cells and reduces the occurrence of autoimmune diseases. Thus, this study provides a novel approach to alleviating the occurrence and development of autoimmune diseases while also minimizing the risk of unwanted complications.


Asunto(s)
Enfermedades Autoinmunes , Nanopartículas , Humanos , Colecalciferol/farmacología , Células Dendríticas , ARN Interferente Pequeño/genética , Enfermedades Autoinmunes/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA