Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Environ Monit Assess ; 196(3): 293, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38383675

RESUMEN

Utilizing engineered wetlands for the cultivation of vegetables can help to overcome the problems of water and food scarcity. These wetlands are primarily designed for wastewater treatment, and their efficiency and effectiveness can be improved by selecting an appropriate substrate. To investigate the potential for nutrient and microbial removal, the Abelmoschus esculentus, Solanum melongena, and Capsicum annuum L. plants were selected to grow in a hybrid constructed wetland (CW) under natural conditions. The removal efficiency of the A. esculentus, S. melongena, and C. annuum L. in the CW system varied between 59.8 to 68.5% for total phosphorous (TP), 40.3 to 53.1% for ammonium (NH4+), and 33.6 to 45.1% for total nitrogen (TN). The influent sample contained multiple pathogenic bacteria, including Alcaligenes faecalis, Staphylococcus aureus, and Escherichia coli, with Capsicum annuum exhibiting a positive association with 7 of the 11 detected species, whereas microbial removal efficiency was notably higher in the S. melongena bed, potentially attributed to temperature variations and plant-facilitated oxygen release rates. While utilizing constructed wetlands for vegetable cultivation holds promising potential to address the disparity between water and food supply and yield various environmental, economic, and social benefits, it is crucial to note that the wastewater source may contain heavy metals, posing a risk of their transmission to humans through the food chain.


Asunto(s)
Abelmoschus , Capsicum , Solanum melongena , Humanos , Aguas Residuales , Humedales , Monitoreo del Ambiente , Nutrientes , Plantas , Nitrógeno/análisis , Verduras , Agua , Eliminación de Residuos Líquidos
2.
Cell Biol Int ; 45(9): 1926-1934, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34009727

RESUMEN

In the treatment of cardiovascular diseases, vascular scaffold materials play an extremely important role. The appropriate substrate chemistries and 15 dynes/cm2 physiological fluid shear stress (FSS) are both required to ensure normal physiological activity of human umbilical vein endothelial cells (HUVECs). The present study reported the collective influence of substrate chemistries and FSS on HUVECs in the sense of its biological functions. The CH3 , NH2 , and OH functional groups were adopted to offer a variety of substrate chemistries on glass slides by the technology of self-assembled monolayers, whereas FSS was generated by a parallel-plate fluid flow system. Substrate chemistries on its own by no means had noticeable effects on eNOS, ATP, NO, and PGI2 expressions, while FSS stimuli enhanced their production. While substrate chemistries, as well as FSS, were both exerted, the releases of ATP, NO, and PGI2 were dependent on substrate chemistries. Study of F-actin organization and focal adhesions (FAs) formation of HUVECs before FSS exposure proves that F-action organization and FAs formation followed similar chemistry-dependence. Hereby proposed a feasible mechanism, that is, the F-actin organization and FAs formation of HUVECs are controlled by substrate chemistries, further advancing the modulation of FSS-triggered responses of HUVECs.


Asunto(s)
Adhesión Celular , Técnicas de Cultivo de Célula/métodos , Células Endoteliales de la Vena Umbilical Humana , Estrés Mecánico , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos
3.
Immunol Invest ; 49(7): 840-857, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31809611

RESUMEN

Background: Recent advancements in therapeutic strategies have attracted considerable attention to control the acute organs and tissues rejection, which is the main cause of mortality in transplant recipients. The long-term usage of immunosuppressive drugs compromises the body immunity against simple infections and decrease the patients' quality of life. Tolerance of allograft in recipients without harming the rest of host immune system is the basic idea to develop the therapeutic approaches after induction of donor-specific transplant. Methods: Controlled and targeted delivery system by using biomimetic micro and nanoparticles as carriers is an effective strategy to deplete the immune cells in response to allograft in an antigen-specific manner. Polylactic-co-glycolic acid (PLGA) is a biocompatible and biodegradable polymer, which has frequently being used as drug delivery vehicle. Results: This review focuses on the biomedical applications of PLGA based biomimetic micro and nano-sized particles in drug delivery systems to prolong the survival of alloskin graft. Conclusion: We will discuss the mediating factors for rejection of alloskin graft, selective depletion of immune cells, controlled release mechanism, physiochemical properties, size-based body distribution of PLGA particles and their effect on overall host immune system.


Asunto(s)
Portadores de Fármacos , Diseño de Fármacos , Rechazo de Injerto/prevención & control , Factores Inmunológicos/administración & dosificación , Inmunosupresores/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Trasplante de Piel/efectos adversos , Aloinjertos , Animales , Antígenos/administración & dosificación , Antígenos/inmunología , Biomimética/métodos , Fenómenos Químicos , Preparaciones de Acción Retardada , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Desarrollo de Medicamentos , Liberación de Fármacos , Rechazo de Injerto/tratamiento farmacológico , Rechazo de Injerto/inmunología , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Humanos , Inmunomodulación/efectos de los fármacos , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Trasplante de Piel/métodos , Trasplante Homólogo
4.
J Immunol ; 201(4): 1194-1210, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29987163

RESUMEN

In this study, a tolerogenic artificial APC (TaAPC) was developed to directly and selectively modulate myelin-autoreactive CD4+ and CD8+ T cells in the myelin oligodendrocyte glycoprotein (MOG)35-55 peptide-induced experimental autoimmune encephalomyelitis in C57BL/6J mice. Cell-sized polylactic-coglycolic acid microparticles were generated to cocouple target Ags (MOG40-54/H-2Db-Ig dimer, MOG35-55/I-Ab multimer), regulatory molecules (anti-Fas and PD-L1-Fc), and "self-marker" CD47-Fc and encapsulate inhibitory cytokine (TGF-ß1). Four infusions of the TaAPCs markedly and durably inhibited the experimental autoimmune encephalomyelitis progression and reduced the local inflammation in CNS tissue. They circulated throughout vasculature into peripheral lymphoid tissues and various organs, but not into brain, with retention of 36 h and exerted direct effects on T cells in vivo and in vitro. Two infusions of the TaAPCs depleted 65-79% of MOG35-55-specific CD4+ and 46-62% of MOG40-54-specific CD8+ T cells in peripheral blood, spleen, and CNS tissues in an Ag-specific manner and regulatory molecule-dependent fashion; induced robust T cell apoptosis; inhibited the activation and proliferation of MOG peptide-reactive T cells; reduced MOG peptide-reactive Th1, Th17, and Tc17 cells; and expanded regulatory T cells. They also inhibited IFN-γ/IL-17A secretion and elevated IL-10/TGF-ß1 production in splenocytes but not in CNS tissue. More importantly, the TaAPCs treatment did not obviously suppress the overall immune function of host. To our knowledge, this study provides the first experimental evidence for the capability of TaAPCs to directly modulate autoreactive T cells by surface presentation of multiple ligands and paracrine release of cytokine, thus suggesting a novel Ag-specific immunotherapy for the T cell-mediated autoimmune diseases.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Inmunoterapia/métodos , Microesferas , Esclerosis Múltiple/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Apoptosis , Antígeno CD47/química , Antígeno CD47/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Antígeno de Histocompatibilidad H-2D/química , Antígeno de Histocompatibilidad H-2D/inmunología , Antígenos de Histocompatibilidad Clase II/química , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Tolerancia Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/química , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T , Factor de Crecimiento Transformador beta1/química , Factor de Crecimiento Transformador beta1/inmunología
5.
Immunol Invest ; 47(1): 1-17, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28872930

RESUMEN

BACKGROUND: Mouse experimental autoimmune encephalomyelitis (EAE) is widely used model of multiple sclerosis (MS). The role of autoreactive CD4+ and CD8+ T cells in the development of mouse EAE has been demonstrated. However, little information is available about the relation between the frequency and reactivity of myelin antigen-reactive CD4+ and CD8+ T cells in secondary lymphoid organs and their relevance with the inflammation and pathological lesion of CNS during the course of EAE mouse model. METHODS: In this study, an EAE model with a clinical course containing acute onset, peak and chronic remission stages was established in C57BL/6J mice by myelin oligodendrocyte protein (MOG)35-55 peptide immunization, and followed by the monitoring of clinical and pathological parameters and autoreactive T cells at different stages during the course. RESULTS: The dynamic changes of inflammatory infiltration, myelin loss, and astrocyte proliferation in brain and spinal cord were highly consistent with clinical severity observed in EAE course. However, the frequencies of both MOG-specific CD4+ and CD8+ T cells in secondary lymphoid organs presented different dynamic trends from the IFN-γ production by MOG-reactive T cells. Meanwhile, the IL-17 production by MOG-reactive CD4+ T cells was consistent with the proliferation of MOG-specific CD4+ T cells. CONCLUSIONS: Both CD4+ and CD8+ T cells were most sensitive to MOG antigen stimulation for IFN-γ production during the early stage of EAE, but then rapidly lost the function despite their vigorous proliferation at the peak stage and later.


Asunto(s)
Astrocitos/patología , Encéfalo/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Médula Espinal/inmunología , Animales , Autoantígenos/inmunología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología
6.
Cancer Immunol Immunother ; 66(9): 1229-1241, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28501941

RESUMEN

Accumulating evidence indicates that bead-based artificial antigen-presenting cells (aAPCs) are a powerful tool to induce antigen-specific T cell responses in vitro and in vivo. To date, most conventional aAPCs have been generated by coupling an antigen signal (signal 1) and one or two costimulatory signals, such as anti-CD28 with anti-LFA1 or anti-4-1BB (signal 2), onto the surfaces of cell-sized or nanoscale magnetic beads or polyester latex beads. The development of a biodegradable scaffold and the combined use of multiple costimulatory signals as well as third signals for putative clinical applications is the next step in the development of this technology. Here, a novel biodegradable aAPC platform for active immunotherapy was developed by co-encapsulating IL-2 and anti-CTLA-4 inside cell-sized polylactic-co-glycolic acid microparticles (PLGA-MPs) while co-coupling an H-2Kb/TRP2-Ig dimer and anti-CD28 onto the surface. Cytokines (activating signal) and antibodies (anti-inhibition signal) were efficiently co-encapsulated in PLGA-MP-based aAPCs and co-released without interfering with each other. The targeted, sustained co-release of IL-2 and anti-CTLA-4 achieved markedly enhanced, synergistic effects in activating and expanding tumor antigen-specific T cells both in vitro and in vivo, as well as in inhibiting tumor growth in a mouse melanoma model, as compared with conventional two-signal aAPCs and IL-2 or anti-CTLA-4 single-released aAPCs. These data revealed the feasibility and importance of the paracrine release of multiple costimulatory molecules and cytokines from biodegradable aAPCs and thus provide a proof of principle for the future use of polymeric aAPCs for active immunotherapy of tumors and infectious diseases.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Interleucina-2/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Comunicación Paracrina
8.
Immunol Invest ; 44(4): 385-99, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25942349

RESUMEN

BACKGROUND: The strategy of specifically depleting antigen-specific T cells can potentially be used for the treatment of allograft rejection and autoimmunity because it does not suppress the overall immune systems. METHODS: In this study, we generated killer polylactic-co-glycolic acid (PLGA) microspheres by covalently coupling major histocompatibility complex (MHC) class I antigens and apoptosis-inducing anti-Fas monoclonal antibody (mAb) onto PLGA microspheres. A modified double-emulsion method was used for the preparation of cell-sized PLGA microspheres. H-2K(b)/peptide monomers were generated in-house and analyzed through flow cytometry. The killer PLGA microspheres were administered intravenously into BALB/c mice (H-2K(d)) that had previously been grafted with skin squares from C57BL/6 mice (H-2K(b)). Tumor cell challenge and third-party mixed lymphocyte culture were used to assess the general immune functions of host. RESULTS: The alloskin graft survival was prolonged by 4 days. The killer PLGA microspheres could specifically deplete the H-2K(b) alloantigen-reactive CD8(+) T cells that infiltrated into the alloskin graft but not CD4(+) T cells, without impairment of host overall immune function. CONCLUSIONS: Here, we initially report that PLGA microspheres, which have been widely used as medicine-delivering carriers, were used to prepare antigen-specific killer complexes and treat allograft rejection. Our data highlight the therapeutic potential of this biocompatible and biodegradable antigen-specific killer effector for the treatment of allograft rejection and autoimmune disease.


Asunto(s)
Epítopos de Linfocito T/inmunología , Supervivencia de Injerto/inmunología , Ácido Láctico , Microesferas , Ácido Poliglicólico , Trasplante de Piel , Animales , Linfocitos T CD8-positivos/inmunología , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Antígenos H-2/química , Antígenos H-2/inmunología , Masculino , Ratones , Modelos Animales , Péptidos/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Trasplante Homólogo
9.
Front Immunol ; 15: 1429442, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39040099

RESUMEN

Introduction: Allergic rhinitis (AR) is an upper airway inflammatory disease of the nasal mucosa. Conventional treatments such as symptomatic pharmacotherapy and allergen-specific immunotherapy have considerable limitations and drawbacks. As an emerging therapy with regenerative potential and immunomodulatory effect, mesenchymal stem cell-derived exosomes (MSC-Exos) have recently been trialed for the treatment of various inflammatory and autoimmune diseases. Methods: In order to achieve sustained and protected release of MSC-Exos for intranasal administration, we fabricated Poly(lactic-co-glycolic acid) (PLGA) micro and nanoparticles-encapsulated MSC-Exos (PLGA-Exos) using mechanical double emulsion for local treatment of AR. Preclinical in vivo imaging, ELISA, qPCR, flow cytometry, immunohistochemical staining, and multiomics sequencing were used for phenotypic and mechanistic evaluation of the therapeutic effect of PLGA-Exos in vitro and in vivo. Results: The results showed that our PLGA platform could efficiently encapsulate and release the exosomes in a sustained manner. At protein level, PLGA-Exos treatment upregulated IL-2, IL-10 and IFN-γ, and downregulated IL-4, IL-17 and antigen-specific IgE in ovalbumin (OVA)-induced AR mice. At cellular level, exosomes treatment reduced Th2 cells, increased Tregs, and reestablished Th1/Th2 balance. At tissue level, PLGA-Exos significantly attenuated the infiltration of immune cells (e.g., eosinophils and goblet cells) in nasal mucosa. Finally, multiomics analysis discovered several signaling cascades, e.g., peroxisome proliferator-activated receptor (PPAR) pathway and glycolysis pathway, that might mechanistically support the immunomodulatory effect of PLGA-Exos. Discussion: For the first time, we present a biomaterial-facilitated local delivery system for stem cell-derived exosomes as a novel and promising strategy for AR treatment.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Rinitis Alérgica , Exosomas/inmunología , Exosomas/metabolismo , Animales , Rinitis Alérgica/terapia , Rinitis Alérgica/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Inmunomodulación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Administración Intranasal
10.
ACS Appl Mater Interfaces ; 16(29): 37497-37512, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-38980910

RESUMEN

Traumatic brain injury poses serious physical, psychosocial, and economic threats. Although systemic administration of stem cell-derived exosomes has recently been proven to be a promising modality for traumatic brain injury treatment, they come with distinct drawbacks. Luckily, various biomaterials have been developed to assist local delivery of exosomes to improve the targeting of organs, minimize nonspecific accumulation in vital organs, and ensure the protection and release of exosomes. In this study, we developed an electrospun nanofibrous scaffold to provide sustained delivery of dual exosomes derived from mesenchymal stem cells and neural stem cells for traumatic brain injury treatment. The electrospun nanofibrous scaffold employed a functionalized layer of polydopamine on electrospun poly(ε-caprolactone) nanofibers, thereby enhancing the efficient incorporation of exosomes through a synergistic interplay of adhesive forces, hydrogen bonding, and electrostatic interactions. First, the mesenchymal stem cell-derived exosomes and the neural stem cell-derived exosomes were found to modulate microglial polarization toward M2 phenotype, play an important role in the modulation of inflammatory responses, and augment axonal outgrowth and neural repair in PC12 cells. Second, the nanofibrous scaffold loaded with dual stem cell-derived exosomes (Duo-Exo@NF) accelerated functional recovery in a murine traumatic brain injury model, as it mitigated the presence of reactive astrocytes and microglia while elevating the levels of growth associated protein-43 and doublecortin. Additionally, multiomics analysis provided mechanistic insights into how dual stem cell-derived exosomes exerted its therapeutic effects. These findings collectively suggest that our novel Duo-Exo@NF system could function as an effective treatment modality for traumatic brain injury using sustained local delivery of dual exosomes from stem cells.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Exosomas , Células Madre Mesenquimatosas , Nanofibras , Células-Madre Neurales , Exosomas/metabolismo , Exosomas/química , Animales , Lesiones Traumáticas del Encéfalo/terapia , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Nanofibras/química , Ratas , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Células PC12 , Ratones , Andamios del Tejido/química , Poliésteres/química , Proteína Doblecortina , Polímeros/química , Masculino , Indoles/química
11.
Biomolecules ; 14(1)2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38254695

RESUMEN

Recent advances in cochlear implantology are exemplified by novel functional strategies such as bimodal electroacoustic stimulation, in which the patient has intact low-frequency hearing and profound high-frequency hearing pre-operatively. Therefore, the synergistic restoration of dysfunctional cochlear hair cells and the protection of hair cells from ototoxic insults have become a persistent target pursued for this hybrid system. In this study, we developed a composite GelMA/PEDOT:PSS conductive hydrogel that is suitable as a coating for the cochlear implant electrode for the potential local delivery of otoregenerative and otoprotective drugs. Various material characterization methods (e.g., 1H NMR spectroscopy, FT-IR, EIS, and SEM), experimental models (e.g., murine cochlear organoid and aminoglycoside-induced ototoxic HEI-OC1 cellular model), and biological analyses (e.g., confocal laser scanning microscopy, real time qPCR, flow cytometry, and bioinformatic sequencing) were used. The results demonstrated decent material properties of the hydrogel, such as mechanical (e.g., high tensile stress and Young's modulus), electrochemical (e.g., low impedance and high conductivity), biocompatibility (e.g., satisfactory cochlear cell interaction and free of systemic toxicity), and biosafety (e.g., minimal hemolysis and cell death) features. In addition, the CDR medicinal cocktail sustainably released by the hydrogel not only promoted the expansion of the cochlear stem cells but also boosted the trans-differentiation from cochlear supporting cells into hair cells. Furthermore, hydrogel-based drug delivery protected the hair cells from oxidative stress and various forms of programmed cell death (e.g., apoptosis and ferroptosis). Finally, using large-scale sequencing, we enriched a complex network of signaling pathways that are potentially downstream to various metabolic processes and abundant metabolites. In conclusion, we present a conductive hydrogel-based local delivery of bifunctional drug cocktails, thereby serving as a potential solution to intracochlear therapy of bimodal auditory rehabilitation and diseases beyond.


Asunto(s)
Células Ciliadas Auditivas , Hidrogeles , Humanos , Animales , Ratones , Hidrogeles/farmacología , Espectroscopía Infrarroja por Transformada de Fourier , Comunicación Celular , Transducción de Señal
12.
Biomed Mater ; 19(2)2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38181444

RESUMEN

Glioblastoma (GBM) remains a challenging malignancy due to its aggressive nature and the lack of efficacious therapeutic interventions. Nanotechnology-based approaches exhibit promise in GBM treatment; however, the successful translation of these strategies from preclinical models to clinical settings is hindered by inefficient nanoparticle clearance from vital organs. Addressing this concern, we investigated the therapeutic potential of amrubicin (AMR) encapsulated within poly (lactic-co-glycolic acid) nanoparticles (AMR-PLGA-NPs) in combating temozolomide (TMZ) resistant GBM. The study demonstrated that AMR-PLGA-NPs exerted a pronounced inhibitory effect on the cellular viability and migratory capacity of TMZ-resistant GBM cells. Furthermore, these nanoparticles exhibited considerable efficacy in downregulating the PI3K/AKT signaling pathway, thereby inducing apoptosis specifically in TMZ-resistant glioma cells and glioma stem-like cells through the activation of PTEN. Notably,in vivoexperimentation revealed the ability of AMR-PLGA-NPs to traverse biological barriers within murine models. Collectively, these findings underscore the potential therapeutic utility of AMR-PLGA-NPs as a versatile nanoplatform for addressing the formidable challenges posed by GBM, particularly in mitigating drug resistance mechanisms. The study substantiates the stability and safety profile of AMR-PLGA-NPs, positioning them as a promising avenue for combating drug resistance in GBM therapeutics.


Asunto(s)
Antraciclinas , Neoplasias Encefálicas , Glioblastoma , Glioma , Animales , Ratones , Antraciclinas/farmacología , Apoptosis , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Glioma/tratamiento farmacológico , Glioma/metabolismo , Glioma/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Temozolomida/farmacología
13.
Artículo en Inglés | MEDLINE | ID: mdl-34952030

RESUMEN

Long noncoding RNA (lncRNA) has been recently revealed as a main regulatory molecule, which implicates many cellular functions. Studies showed that lncRNA abnormally expressed and involved in the progression and tumorigenesis of glioma. Present study identified a novel lncRNA associated with glioma, glioma stem-like cells (GSCs), and then revealed their potential functions. During the screening of lncRNAs, we investigated overexpression of lncRNA RP5-821D11.7 (lncRNA-RP5) in GSCs compared to glioma cells. Lentivirus-mediated shRNA for lncRNA-RP5 was constructed and transfected into glioma cells. Transfected stable glioma cells were transplanted into nude mice and tumor growth was observed. Knockdown of lncRNA-RP5 significantly inhibits proliferation, colony formation, migration and reduces epithelial-mesenchymal transition (EMT) by activating the Wnt/ß-catenin pathway. Additionally, the results showed that lncRNA RP5 knockdown enhances cell apoptosis through endoplasmic reticulum stress. Therefore, this study may provide a better understanding about lncRNA-RP5 which revealed that it might be a potential therapeutic target in case of glioma progression and recurrence.

14.
Cancer Immunol Res ; 7(7): 1188-1201, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31113806

RESUMEN

Antigen-presenting cells expand antigen-specific T cells ex vivo and in vivo for tumor immunotherapy, but are time-consuming to generate and, as live cells, raise biosafety concerns. An alternative is found in cell-free artificial antigen-presenting cells (aAPC), but these only present two or three kinds of immune molecules. Here, we describe a multipotent artificial antigen-presenting cell (MaAPC) that delivered 11 kinds of immune moleclues. This MaAPC simulated natural APCs through the concurent coupling of target antigens (H-2Kb/TRP2180-188-Ig dimers and H-2Db/gp10025-33-Ig dimers), costimulatory molecules (anti-CD28, anti-4-1BB, and anti-CD2), and "self-marker" CD47-Fc onto surface-modified polylactic-co-glycolic acid microparticles (PLGA-MP). These PLGA-MPs also encapsulated cytokines (IL2 and IL15), a chemokine (CCL21), and checkpoint inhibitors (anti-CTLA-4 and anti-PD-1). Culture of MaAPCs with naïve T cells for 1 week elevated the frequencies of TRP2180-188-specific and gp10025-33-specific CTLs to 51.0% and 43.3%, respectively, with enhanced cytotoxicity. Three infusions of MaAPCs inhibited subcutaneous melanoma growth in a mouse model and expanded TRP2180-188 and gp10025-33-specific CTLs 59-86-fold in peripheral blood, 76-77-fold in spleen, and 205-212-fold in tumor tissue, in an antigen-specific manner. Compared with conventional aAPCs carrying two or three immune molecules, the 11-signal MaAPCs exerted greater impact on T cells, including activation, proliferation, cytotoxicity, differentiation to memory CTLs or regulatory T cells and cytokines profiles, without detected side effects. Such MaAPCs could be used to individualize tumor immunotherapy.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Oxidorreductasas Intramoleculares/inmunología , Melanoma Experimental/inmunología , Linfocitos T Citotóxicos/inmunología , Antígeno gp100 del Melanoma/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Apoptosis , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Proliferación Celular , Quimiocinas/inmunología , Citocinas/inmunología , Femenino , Inmunoterapia , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Células Tumorales Cultivadas
15.
Int J Nanomedicine ; 13: 3731-3750, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29983566

RESUMEN

PURPOSE: Numerous nanomaterials have been reported in the treatment of multiple sclerosis or experimental autoimmune encephalomyelitis (EAE). But most of these nanoscale therapeutics deliver myelin antigens together with toxins or cytokines and underlay the cellular uptake and induction of tolerogenic antigen-presenting cells by which they indirectly induce T cell tolerance. This study focuses on the on-target and direct modulation of myelin-autoreactive T cells and combined use of multiple regulatory molecules by generating a tolerogenic nanoparticle. MATERIALS AND METHODS: Poly(lactic-co-glycolic acid) nanoparticles (PLGA-NPs) were fabricated by co-coupling MOG40-54/H-2Db-Ig dimer, MOG35-55/I-Ab multimer, anti-Fas, PD-L1-Fc and CD47-Fc and encapsulating transforming growth factor-ß1. The resulting 217 nm tolerogenic nanoparticles (tNPs) were administered intravenously into MOG35-55 peptide-induced EAE mice, which was followed by the investigation of therapeutic outcomes and the in vivo mechanism. RESULTS: Four infusions of the tNPs durably ameliorated EAE with a marked reduction of clinical score, neuroinflammation and demyelination. They were distributed in secondary lymphoid tissues, various organs and brain after intravenous injection, with retention over 36 h, and made contacts with CD4+ and CD8+ T cells. Two injections of the tNPs markedly decreased the MOG35-55-reactive Th1 and Th17 cells and MOG40-55-reactive Tc1 and Tc17 cells, increased regulatory T cells, inhibited T cell proliferation and elevated T cell apoptosis in spleen. Transforming growth factor-ß1 and interleukin-10 were upregulated in the homogenates of central nervous system and supernatant of spleen cells. CONCLUSION: Our data suggest a novel therapeutic nanoparticle to directly modulate autoreactive T cells by surface presentation of multiple ligands and paracrine release of cytokine in the antigen-specific combination immunotherapy for T cell-mediated autoimmune diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Antígeno CD47/metabolismo , Linfocitos T CD8-positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Tolerancia Inmunológica , Complejo Mayor de Histocompatibilidad , Glicoproteína Mielina-Oligodendrócito/inmunología , Nanopartículas/química , Animales , Apoptosis , Proliferación Celular , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ácido Láctico/química , Activación de Linfocitos , Ratones Endogámicos C57BL , Nanopartículas/ultraestructura , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Linfocitos T Reguladores/inmunología , Distribución Tisular
16.
Drug Deliv ; 25(1): 703-715, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29508634

RESUMEN

Biomimetic nanoparticles have been reported as immune modulators in autoimmune diseases and allograft rejections by numerous researchers. However, most of the therapeutics carrying antigens, toxins or cytokines underlay the mechanism of antigen presentation by cellular uptake of NPs through pinocytosis and phagocytosis. Few researches focus on the direct and antigen-specific modulation on T cells by NPs and combined use of multiple regulatory molecules. Here, polylactic-co-glycolic acid nanoparticles (PLGA-NPs) were fabricated as scaffold to cocoupling H-2Kb-Ig dimer, anti-Fas mAb, PD-L1-Fc, TGF-ß and CD47-Fc for the generation of alloantigen-presenting and tolerance-inducing NPs, termed killer NPs and followed by i.v. injection into a single MHC-mismatched murine model of alloskin transplantation. Three infusions prolonged alloskin graft survival for 45 days; depleted most of H-2Kb alloreactive CD8+ T cells in peripheral blood, spleen and local graft, in an antigen-specific manner. The killer NPs circulated throughout vasculature into various organs and local allograft, with a retention time up to 30 h. They made contacts with CD8+ T cells to facilitate vigorous apoptosis, inhibit the activation and proliferation of alloreactive CD8+ T cells and induce regulatory T cells in secondary lymphoid organs, with the greatly minimized uptake by phagocytes. More importantly, the impairment of host overall immune function and visible organ toxicity were not found. Our results provide the first experimental evidence for the direct and on-target modulation on alloreactive T cells by the biodegradable 200-nm killer NPs via co-presentation of alloantigen and multiple regulatory molecules, thus suggest a novel antigen-specific immune modulator for allograft rejections.


Asunto(s)
Antígeno CD47/administración & dosificación , Isoantígenos/administración & dosificación , Complejo Mayor de Histocompatibilidad , Nanopartículas/administración & dosificación , Trasplante de Piel/métodos , Linfocitos T/metabolismo , Animales , Antígeno CD47/inmunología , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Isoantígenos/inmunología , Masculino , Melanoma Experimental , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Trasplante Homólogo
17.
Front Immunol ; 8: 657, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28649247

RESUMEN

Selectively depleting the pathogenic T cells is a fundamental strategy for the treatment of allograft rejection and autoimmune disease since it retains the overall immune function of host. The concept of killer artificial antigen-presenting cells (KaAPCs) has been developed by co-coupling peptide-major histocompatibility complex (pMHC) multimer and anti-Fas monoclonal antibody (mAb) onto the polymeric microparticles (MPs) to induce the apoptosis of antigen-specific T cells. But little information is available about its in vivo therapeutic potential and mechanism. In this study, polyethylenimine (PEI)-coated poly lactic-co-glycolic acid microparticle (PLGA MP) was fabricated as a cell-sized scaffold to covalently co-couple H-2Kb-Ig dimer and anti-Fas mAb for the generation of alloantigen-presenting and apoptosis-inducing MPs. Intravenous infusions of the biodegradable KaAPCs prolonged the alloskin graft survival for 43 days in a single MHC-mismatched murine model, depleted the most of H-2Kb-alloreactive CD8+ T cells in peripheral blood, spleen, and alloskin graft in an antigen-specific manner and anti-Fas-dependent fashion. The cell-sized KaAPCs circulated throughout vasculature into liver, kidney, spleen, lymph nodes, lung, and heart, but few ones into local allograft at early stage, with a retention time up to 36 h in vivo. They colocalized with CD8+ T cells in secondary lymphoid organs while few ones contacted with CD4+ T cells, B cells, macrophage, and dendritic cells, or internalized by phagocytes. Importantly, the KaAPC treatment did not significantly impair the native T cell repertoire or non-pathogenic immune cells, did not obviously suppress the overall immune function of host, and did not lead to visible organ toxicity. Our results strongly document the high potential of PLGA MP-based KaAPCs as a novel antigen-specific immunotherapy for allograft rejection and autoimmune disorder. The in vivo mechanism of alloinhibition, tissue distribution, and biosafety were also initially characterized, which will facilitate its translational studies from bench to bedside.

18.
Sci Rep ; 7(1): 16400, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-29180767

RESUMEN

Conventional peptide-major histocompatibility complex (pMHC) multimer staining, intracellular cytokine staining, and enzyme-linked immunospot (ELISPOT) assay cannot concurrently determine the frequency and reactivity of antigen-specific T cells (AST) in a single assay. In this report, pMHC multimer, magnetic-activated cell sorting (MACS), and ELISPOT techniques have been integrated into a micro well by coupling pMHC multimers onto cell-sized magnetic beads to characterize AST cell populations in a 96-well microplate which pre-coated with cytokine-capture antibodies. This method, termed AAPC-microplate, allows the enumeration and local cytokine production of AST cells in a single assay without using flow cytometry or fluorescence intensity scanning, thus will be widely applicable. Here, ovalbumin257-264-specific CD8+ T cells from OT-1 T cell receptor (TCR) transgenic mice were measured. The methodological accuracy, specificity, reproducibility, and sensitivity in enumerating AST cells compared well with conventional pMHC multimer staining. Furthermore, the AAPC-microplate was applied to detect the frequency and reactivity of Hepatitis B virus (HBV) core antigen18-27- and surface antigen183-191-specific CD8+ T cells for the patients, and was compared with conventional method. This method without the need of high-end instruments may facilitate the routine analysis of patient-specific cellular immune response pattern to a given antigen in translational studies.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Antígenos/inmunología , Recuento de Linfocitos , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Células Presentadoras de Antígenos/metabolismo , Biomarcadores , Ensayo de Immunospot Ligado a Enzimas/métodos , Ensayo de Immunospot Ligado a Enzimas/normas , Citometría de Flujo/métodos , Citometría de Flujo/normas , Hepatitis B/inmunología , Hepatitis B/virología , Virus de la Hepatitis B/inmunología , Humanos , Separación Inmunomagnética , Ratones , Fenotipo , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
19.
J Microbiol Biotechnol ; 26(12): 2184-2191, 2016 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-27586530

RESUMEN

The time-consuming and high-cost preparation of soluble peptide-major histocompatibility complexes (pMHC) currently limits their wide uses in monitoring antigen-specific T cells. The single-chain trimer (SCT) of peptide-ß2m-MHC class I heavy chain was developed as an alternative strategy, but its gene fusion is hindered in many cases owing to the incompatibility between the multiple restriction enzymes and the restriction endonuclease sites of plasmid vectors. In this study, overlap extension PCR and one-step cloning were adopted to overcome this restriction. The SCT gene of the OVA257₋264 peptide-(GS4)3-ß2m-(GS4)4-H-2Kb heavy chain was constructed and inserted into plasmid pET28a by overlap extension PCR and one-step cloning, without the requirement of restriction enzymes. The SCT protein was expressed in Escherichia coli, and then purified and refolded. The resulting H-2Kb/OVA257₋264 complex showed the correct structural conformation and capability to bind with OVA257₋264-specific T-cell receptor. The overlap extension PCR and one-step cloning ensure the construction of single-chain MHC class I molecules associated with random epitopes, and will facilitate the preparation of soluble pMHC multimers.


Asunto(s)
Clonación Molecular/métodos , Epítopos de Linfocito T/genética , Antígenos de Histocompatibilidad Clase I/genética , Péptidos/genética , Reacción en Cadena de la Polimerasa/métodos , Animales , Epítopos de Linfocito T/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Ratones , Péptidos/metabolismo , Plásmidos/genética , Plásmidos/metabolismo
20.
Oncotarget ; 7(11): 12176-90, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26910923

RESUMEN

The specific eradication of pathogenic T cells for the treatment of allograft rejections and autoimmune disorders without impairment of overall immune function is a fundamental goal. Here, cell-sized poly(lactic-co-glycolic acid) microparticles (PLGA MPs) were prepared as a scaffold to co-display the peptide/major histocompatibility complex (pMHC, target antigen) and anti-Fas monoclonal antibody (apoptosis-inducing molecule) for the generation of biodegradable killer MPs. Ovalbumin (OVA) antigen-targeted killer MPs significantly depleted OVA-specific CD8+ T cells in an antigen-specific manner, both in vitro and in OT-1 mice. After intravenous administration, the killer MPs predominantly accumulated in the liver, lungs, and gut of OT-1 mice with a retention time of up to 48 hours. The killing effects exerted by killer MPs persisted for 4 days after two injections. Moreover, the H-2Kb alloantigen-targeted killer MPs were able to eliminate low-frequency alloreactive T cells and prolong alloskin graft survival for 41.5 days in bm1 mice. Our data indicate that PLGA-based killer MPs are capable of specifically depleting pathogenic T cells, which highlights their therapeutic potential for treating allograft rejection and autoimmune disorders.


Asunto(s)
Epítopos de Linfocito T/administración & dosificación , Rechazo de Injerto/inmunología , Ácido Láctico/administración & dosificación , Depleción Linfocítica/métodos , Ácido Poliglicólico/administración & dosificación , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales , Anticuerpos Monoclonales de Origen Murino , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/inmunología , Ácido Láctico/química , Ácido Láctico/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Microesferas , Ovalbúmina/administración & dosificación , Ovalbúmina/química , Ovalbúmina/inmunología , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Linfocitos T/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA