Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Adv Mater ; 32(39): e2003368, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32812291

RESUMEN

Cancer immunotherapies, including adoptive T cell transfer and immune checkpoint blockades, have recently shown considerable success in cancer treatment. Nevertheless, transferred T cells often become exhausted because of the immunosuppressive tumor microenvironment. Immune checkpoint blockades, in contrast, can reinvigorate the exhausted T cells; however, the therapeutic efficacy is modest in 70-80% of patients. To address some of the challenges faced by the current cancer treatments, here T-cell-membrane-coated nanoparticles (TCMNPs) are developed for cancer immunotherapy. Similar to cytotoxic T cells, TCMNPs can be targeted at tumors via T-cell-membrane-originated proteins and kill cancer cells by releasing anticancer molecules and inducing Fas-ligand-mediated apoptosis. Unlike cytotoxic T cells, TCMNPs are resistant to immunosuppressive molecules (e.g., transforming growth factor-ß1 (TGF-ß1)) and programmed death-ligand 1 (PD-L1) of cancer cells by scavenging TGF-ß1 and PD-L1. Indeed, TCMNPs exhibit higher therapeutic efficacy than an immune checkpoint blockade in melanoma treatment. Furthermore, the anti-tumoral actions of TCMNPs are also demonstrated in the treatment of lung cancer in an antigen-nonspecific manner. Taken together, TCMNPs have a potential to improve the current cancer immunotherapy.


Asunto(s)
Materiales Biomiméticos/química , Materiales Biomiméticos/uso terapéutico , Inmunoterapia/métodos , Nanopartículas/uso terapéutico , Linfocitos T/inmunología , Línea Celular Tumoral , Humanos , Nanomedicina
2.
ACS Nano ; 12(9): 8977-8993, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30133260

RESUMEN

Cancer immunotherapy modulates immune cells to induce antitumor immune responses. Tumors employ immune checkpoints to evade immune cell attacks. Immune checkpoint inhibitors such as anti-PD-L1 antibody (aPD-L1), which is being used clinically for cancer treatments, can block immune checkpoints so that the immune system can attack tumors. However, immune checkpoint inhibitor therapy may be hampered by polarization of macrophages within the tumor microenvironment (TME) into M2 tumor-associated macrophages (TAMs), which suppress antitumor immune responses and promote tumor growth by releasing anti-inflammatory cytokines and angiogenic factors. In this study, we used exosome-mimetic nanovesicles derived from M1 macrophages (M1NVs) to repolarize M2 TAMs to M1 macrophages that release pro-inflammatory cytokines and induce antitumor immune responses and investigated whether the macrophage repolarization can potentiate the anticancer efficacy of aPD-L1. M1NV treatment induced successful polarization of M2 macrophages to M1 macrophages in vitro and in vivo. Intravenous injection of M1NVs into tumor-bearing mice suppressed tumor growth. Importantly, injection of a combination of M1NVs and aPD-L1 further reduced the tumor size, compared to the injection of either M1NVs or aPD-L1 alone. Thus, our study indicates that M1NV injection can repolarize M2 TAMs to M1 macrophages and potentiate antitumor efficacy of the checkpoint inhibitor therapy.


Asunto(s)
Anticuerpos/inmunología , Antineoplásicos/farmacología , Inmunoterapia , Macrófagos/química , Nanoestructuras/química , Neoplasias/terapia , Animales , Reacciones Antígeno-Anticuerpo , Células Cultivadas , Femenino , Humanos , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Neoplasias/inmunología , Células RAW 264.7 , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
3.
Tissue Eng Part A ; 22(7-8): 654-64, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26976076

RESUMEN

The rapid recruitment of osteoblasts in bone defects is an essential prerequisite for efficient bone repair. Conventionally, osteoblast recruitment to bone defects and subsequent bone repair has been achieved using growth factors. Here, we present a methodology that can guide the recruitment of osteoblasts to bone defects with topographically defined implants (TIs) for efficient in vivo bone repair. We compared circular TIs that had microgrooves in parallel or radial arrangements with nonpatterned implants for osteoblast migration and in vivo bone formation. In vitro, the microgrooves in the TIs enhanced both the migration and proliferation of osteoblasts. Especially, the microgrooves with radial arrangement demonstrated a much higher efficiency of osteoblast recruitment to the implants than did the other types of implants, which may be due to the efficient guidance of cell migration toward the cell-free area of the implants. The expression of the intracellular signaling molecules responsible for the cell migration was also upregulated in osteoblasts on the microgrooved TIs. In vivo, the TI with radially defined topography demonstrated much greater bone repair in mouse calvarial defect models than in the other types of implants. Taken together, these results indicate that implants with physical guidance can enhance tissue repair by rapid cell recruitment.


Asunto(s)
Huesos/patología , Osteoblastos/citología , Prótesis e Implantes , Cicatrización de Heridas , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Materiales Biocompatibles Revestidos/farmacología , Espacio Intracelular/metabolismo , Ratones Endogámicos ICR , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
4.
ACS Nano ; 9(10): 9678-90, 2015 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-26348606

RESUMEN

Gold nanoparticles (AuNPs) have been extensively studied for photothermal cancer therapy because AuNPs can generate heat upon near-infrared irradiation. However, improving their tumor-targeting efficiency and optimizing the nanoparticle size for maximizing the photothermal effect remain challenging. We demonstrate that mesenchymal stem cells (MSCs) can aggregate pH-sensitive gold nanoparticles (PSAuNPs) in mildly acidic endosomes, target tumors, and be used for photothermal therapy. These aggregated structures had a higher cellular retention in comparison to pH-insensitive, control AuNPs (cAuNPs), which is important for the cell-based delivery process. PSAuNP-laden MSCs (MSC-PSAuNPs) injected intravenously to tumor-bearing mice show a 37-fold higher tumor-targeting efficiency (5.6% of the injected dose) and 8.3 °C higher heat generation compared to injections of cAuNPs after irradiation, which results in a significantly enhanced anticancer effect.


Asunto(s)
Agregación Celular , Sistemas de Liberación de Medicamentos , Oro/uso terapéutico , Células Madre Mesenquimatosas/citología , Nanopartículas del Metal/uso terapéutico , Neoplasias/terapia , Animales , Femenino , Oro/administración & dosificación , Oro/farmacocinética , Hipertermia Inducida/métodos , Nanopartículas del Metal/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fototerapia/métodos
5.
Tissue Eng Part A ; 21(7-8): 1275-87, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25517212

RESUMEN

Implantation of ex vivo expanded and osteogenically differentiated mesenchymal stem cells (MSCs) for bone regeneration has drawbacks for clinical applications, such as poor survival of implanted cells and increased treatment expenses. As a new approach for bone regeneration that can circumvent these limitations, we propose dual delivery of substance P (SP) and bone morphogenetic protein-2 (BMP-2) to facilitate endogenous stem cell recruitment to bone defects by SP and subsequent in situ osteogenic differentiation of those cells by BMP-2. A heparin-conjugated fibrin (HCF) gel enabled dual delivery with fast release of SP and slow release of BMP-2, which would be ideal for prompt recruitment of endogenous stem cells in the first stage and time-consuming osteogenic differentiation of the recruited stem cells in the second stage. The HCF gels with SP and/or BMP-2 were implanted into mouse calvarial defects for 8 weeks. Local delivery of SP to the calvarial defects using HCF gel was more effective in recruiting MSCs to the calvarial defects than intraperitoneal or intravenous administration of SP. Many of the cells recruited by SP underwent osteogenic differentiation through local delivery of BMP-2. The efficacy of in vivo bone regeneration was significantly higher in the SP/BMP-2 dual delivery group. The dual delivery of SP and BMP-2 using the HCF gel therefore has potential as an effective bone regeneration strategy.


Asunto(s)
Proteína Morfogenética Ósea 2/farmacología , Regeneración Ósea/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Sustancia P/farmacología , Animales , Bovinos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Fibrina/farmacología , Citometría de Flujo , Heparina/farmacología , Humanos , Inflamación/patología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Cráneo/efectos de los fármacos , Cráneo/patología , Andamios del Tejido/química
6.
Tissue Eng Part A ; 21(1-2): 374-81, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25065511

RESUMEN

Human embryonic stem cells (hESCs) are a useful cell source for cardiac regeneration by stem cell therapy. In this study, we show that incorporation of gold-coated microspheres into hESC-derived embryoid bodies (EBs) enhances the cardiomyogenic differentiation process of pluripotent embryonic stem cells. A polycaprolactone (PCL) microsphere surface was coated with gold. Either gold-coated PCL microspheres (AuMS) or PCL microspheres (MS) were incorporated into hESC-derived EBs. AuMS and MS were not cytotoxic. AuMS promoted the expression of genes for mesodermal and cardiac mesodermal lineage cells, both of which are intermediates in the process of cardiac differentiation of hESCs on day 4 and the expression of cardiomyogenic differentiation markers on day 14 compared to MS. AuMS also enhanced gene expression of cardiac-specific extracellular matrices. Incorporation of gold-coated MS into hESC-derived EBs may provide a new platform for inducing cardiomyogenic differentiation of pluripotent embryonic stem cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Materiales Biocompatibles Revestidos/farmacología , Cuerpos Embrioides/citología , Oro/farmacología , Microesferas , Miocitos Cardíacos/citología , Apoptosis/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cuerpos Embrioides/efectos de los fármacos , Endodermo/citología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos , Mesodermo/citología , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Especificidad de Órganos/efectos de los fármacos , Poliésteres/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo
7.
Angew Chem Int Ed Engl ; 53(35): 9213-7, 2014 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-25044682

RESUMEN

Carbon-based materials have been extensively studied for stem cell culture. However, difficulties associated with engineering pure carbon materials into 3D scaffolds have hampered applications in tissue engineering and regenerative medicine. Carbonized polyacrylonitrile (cPAN) could be a promising alternative, as cPAN is a highly ordered carbon isomorph that resembles the graphitic structure and can be easily processed into 3D scaffolds. Despite the notable features of cPAN, application of cPAN in tissue engineering and regenerative medicine have not been explored. This study, for the first time, demonstrates the fabrication of microporous 3D scaffolds of cPAN and excellent osteoinductivity of cPAN, suggesting utility of 3D cPAN scaffolds as synthetic bone graft materials. The combination of excellent processability and unique bioactive properties of cPAN may lead to future applications in orthopedic regenerative medicine.


Asunto(s)
Resinas Acrílicas/química , Regeneración Ósea , Huesos/fisiología , Ingeniería de Tejidos , Andamios del Tejido/química
8.
Tissue Eng Part A ; 20(7-8): 1306-13, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24224833

RESUMEN

Human mesenchymal stem cells (hMSCs) have the ability to differentiate into mesenchymal lineages. In this study, we hypothesized that treatment of embryoid bodies (EBs) composed of either human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs) with a hMSC-conditioned medium (CM) can stimulate the induction of the mesodermal lineage and subsequent differentiation toward the osteogenic and chondrogenic lineage. Quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) analysis indicated that the hMSC-CM treatment increased gene expression related to the mesodermal lineage and decreased gene expression related to the endodermal and ectodermal lineage in EBs. Fourteen days after culturing the mesodermal lineage-induced EBs in the osteogenic or chondrogenic differentiation medium, we observed enhanced osteogenic and chondrogenic differentiation compared with untreated EBs, as evaluated using qRT-PCR, cytochemistry, immunocytochemistry, and flow cytometry. This method may be useful for enhancing the osteogenic or chondrogenic differentiation of hESCs or hiPSCs.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Células Madre Mesenquimatosas/citología , Mesodermo/citología , Línea Celular , Linaje de la Célula/efectos de los fármacos , Cuerpos Embrioides/citología , Citometría de Flujo , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Osteocalcina/metabolismo , Osteogénesis/efectos de los fármacos
9.
Biomaterials ; 34(33): 8258-68, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23932503

RESUMEN

Peripheral arterial disease (PAD) is characterized by the altered structure and function of arteries caused by accumulated plaque. There have been many studies on treating this disease by the direct injection of various types of therapeutic cells, however, the low cell engraftment efficiency and diffusion of the transplanted cells have been major problems. In this study, we developed an approach (transfer printing) to deliver monolayer of cells to the hindlimb ischemic tissue using thermosensitive hydrogels, and investigated its efficacy in long term retention upon transplantation and therapeutic angiogenesis. We first investigated the in vitro maintenance of robust cell-cell contacts and stable expression of the ECM proteins in myoblast layer following transfer printing process. In order to confirm the therapeutic effect of the myoblasts in vivo, we cultured a monolayer of C2C12 myoblasts on thermosensitive hydrogels, which was then transferred to the hindlimb ischemia tissue of athymic mice directly from the hydrogel by conformal contact. The transferred myoblast layer was retained for a longer period of time than an intramuscularly injected cell suspension. In addition, the morphology of the mice and laser Doppler perfusion (28 days after treatment) supported that the myoblast layer enhanced the therapeutic effects on the ischemic tissue. In summary, the transplantation of the C2C12 myoblast layer using a tissue transfer printing method could represent a new approach for the treatment of PAD by therapeutic angiogenesis.


Asunto(s)
Hidrogeles/química , Mioblastos/citología , Animales , Western Blotting , Línea Celular , Femenino , Miembro Posterior/patología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Isquemia/terapia , Ratones , Ratones Desnudos , Mioblastos/trasplante
10.
Biomaterials ; 34(28): 6871-81, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23773822

RESUMEN

Matrix metalloproteinase (MMP)-2 and MMP-9 have been known to play the role of essential mediators in angiogenesis. Non-invasive in vivo imaging approach using imaging probes is a potential method of detecting MMP activity in living animals, wherein imaging probes must include the characteristics of non-toxicity, specific targetability, and reasonable signal intensity. Here, we developed MMP-specific and self-quenched human serum albumin (HSA)-based (MMP-HSA) nanoprobes for non-invasive optical imaging of MMP activity during angiogenesis in the mouse hindlimb ischemia model. MMP-specific fluorogenic peptide probes, which were self-quenched with a near-infrared fluorophore and a quencher, were covalently conjugated to HSA (MMP-HSA nanoprobes). MMP-HSA nanoprobes formed stable nanoparticle structures of approximately 36 nm in diameter. Strongly self-quenched MMP-HSA nanoprobes boosted intense fluorescence signals in the presence of MMP-2 and MMP-9. Furthermore, MMP-HSA nanoprobes showed no cytotoxicity in cell culture. Importantly, intravenous injection of MMP-HSA nanoprobes provided longer blood half-life and successful non-invasive optical imaging of MMP activity during angiogenesis in the mouse hindlimb ischemia model. In addition, the MMP activity visualized by MMP-HSA nanoprobes was consistent with the results of zymography, Western blot, and immunohistochemistry. MMP-HSA nanoprobes may be useful for monitoring of the initial process of angiogenesis through non-invasive MMP imaging.


Asunto(s)
Miembro Posterior/metabolismo , Isquemia/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Imagen Óptica/métodos , Albúmina Sérica/química , Animales , Western Blotting , Femenino , Miembro Posterior/patología , Humanos , Inmunohistoquímica , Ratones
11.
Biochem Biophys Res Commun ; 430(2): 793-7, 2013 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23206696

RESUMEN

Recently, it was reported that bone morphogenetic protein 4 (BMP4) alone or BMP4 combined with fibroblast growth factor 2 (FGF2) treatment enhanced mesodermal differentiation of human embryonic stem cells (hESCs) that were cultured feeder-free on Matrigel. In this study, we show that mesodermal lineage-induced embryoid bodies (EBs) generate greater numbers of osteogenic and chondrogenic lineage cells. To induce the mesodermal lineage, hESCs were treated with BMP4 and FGF2 during the EB state. Quantitative real-time reverse transcription-polymerase chain reaction analysis showed that the treatment decreased endodermal and ectodermal lineage gene expression and increased mesodermal lineage gene expression. Importantly, the mesodermal lineage-induced EBs underwent enhanced osteogenic and chondrogenic differentiation after differentiation induction. This method could be useful to enhance the osteogenic or chondrogenic differentiation of hESCs.


Asunto(s)
Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Células Madre Embrionarias/citología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Mesodermo/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Células Cultivadas , Cuerpos Embrioides/efectos de los fármacos , Humanos , Mesodermo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA