Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
PLoS One ; 16(12): e0261599, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34905584

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0233263.].

2.
PLoS One ; 16(5): e0251601, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34038436

RESUMEN

Traumatic brain injury (TBI) causes both physical disruption of the blood brain barrier (BBB) and altered immune responses that can lead to significant secondary brain injury and chronic inflammation within the central nervous system (CNS). Cell therapies, including mesenchymal stromal cells (MSC), have been shown to restore BBB integrity and augment endogenous splenic regulatory T cells (Treg), a subset of CD4+ T cells that function to regulate immune responses and prevent autoimmunity. We have recently shown that infusion of human cord blood-derived Treg decreased neuroinflammation after TBI in vivo and in vitro. However, while both cells have demonstrated anti-inflammatory and regenerative potential, they likely utilize differing, although potentially overlapping, mechanisms. Furthermore, studies investigating these two cell types together, as a combination therapy, are lacking. In this study, we compared the ability of Treg+MSC combination therapy, as well as MSC and Treg monotherapies, to improve BBB permeability in vivo and suppress inflammation in vitro. While Treg+MSC combination did not significantly augment potency in vivo, our in vitro data demonstrates that combination therapy may augment therapeutic potency and immunosuppressive potential compared to Treg or MSC monotherapy.


Asunto(s)
Barrera Hematoencefálica/inmunología , Lesiones Traumáticas del Encéfalo , Tolerancia Inmunológica , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Linfocitos T Reguladores , Animales , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/terapia , Modelos Animales de Enfermedad , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante
3.
Stem Cells ; 39(3): 358-370, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33368792

RESUMEN

The inflammatory response after traumatic brain injury (TBI) can lead to significant secondary brain injury and chronic inflammation within the central nervous system. Cell therapies, including mesenchymal stromal cells (MSC), have led to improvements in animal models of TBI and are under investigation in human trials. One potential mechanism for the therapeutic potential of MSC is their ability to augment the endogenous response of immune suppressive regulatory T cells (Treg). We have recently shown that infusion of human cord blood Treg decreased chronic microgliosis after TBI and altered the systemic immune response in a rodent model. These cells likely use both overlapping and distinct mechanisms to modulate the immune system; therefore, combining Treg and MSC as a combination therapy may confer therapeutic benefit over either monotherapy. However, investigation of Treg + MSC combination therapy in TBI is lacking. In this study, we compared the ability MSC + Treg combination therapy, as well as MSC and Treg monotherapies, to inhibit the neuroinflammatory response to TBI in vivo and in vitro. Treg + MSC combination therapy demonstrated increased potency to reduce the neuro- and peripheral inflammatory response compared to monotherapy; furthermore, the timing of infusion proved to be a significant variable in the efficacy of both MSC monotherapy and Treg + MSC combination therapy in vivo and in vitro.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Linfocitos T Reguladores/inmunología , Animales , Lesiones Traumáticas del Encéfalo/inmunología , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Inmunidad , Inflamación/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Ratas Sprague-Dawley
4.
Sci Rep ; 10(1): 10729, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32612177

RESUMEN

The immune system plays critical roles in promoting tissue repair during recovery from neurotrauma but is also responsible for unchecked inflammation that causes neuronal cell death, systemic stress, and lethal immunodepression. Understanding the immune response to neurotrauma is an urgent priority, yet current models of traumatic brain injury (TBI) inadequately recapitulate the human immune response. Here, we report the first description of a humanized model of TBI and show that TBI places significant stress on the bone marrow. Hematopoietic cells of the marrow are regionally decimated, with evidence pointing to exacerbation of underlying graft-versus-host disease (GVHD) linked to presence of human T cells in the marrow. Despite complexities of the humanized mouse, marrow aplasia caused by TBI could be alleviated by cell therapy with human bone marrow mesenchymal stromal cells (MSCs). We conclude that MSCs could be used to ameliorate syndromes triggered by hypercytokinemia in settings of secondary inflammatory stimulus that upset marrow homeostasis such as TBI. More broadly, this study highlights the importance of understanding how underlying immune disorders including immunodepression, autoimmunity, and GVHD might be intensified by injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/fisiopatología , Enfermedad Injerto contra Huésped/etiología , Tolerancia Inmunológica/inmunología , Células Madre Mesenquimatosas/citología , Linfocitos T/inmunología , Animales , Femenino , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/terapia , Masculino , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos NOD , Ratones SCID
5.
Stem Cells Transl Med ; 9(8): 903-916, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32383348

RESUMEN

Traumatic brain injury (TBI) causes a profound inflammatory response within the central nervous system and peripheral immune system, which contributes to secondary brain injury and further morbidity and mortality. Preclinical investigations have demonstrated that treatments that downregulate microglia activation and polarize them toward a reparative/anti-inflammatory phenotype have improved outcomes in preclinical models. However, no therapy to date has translated into proven benefits in human patients. Regulatory T cells (Treg) have been shown to downregulate pathologic immune responses of the innate and adaptive immune system across a variety of pathologies. Furthermore, cellular therapy has been shown to augment host Treg responses in preclinical models; yet, studies investigating the use of Treg as a therapeutic for TBI are lacking. In a rodent TBI model, we demonstrate that human umbilical cord blood Treg modulate the central and peripheral immune response after injury in vitro and in vivo.


Asunto(s)
Lesiones Traumáticas del Encéfalo/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Inmunidad/inmunología , Inmunofenotipificación/métodos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Animales , Lesiones Traumáticas del Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Ratas , Ratas Sprague-Dawley
6.
PLoS One ; 15(5): e0233263, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32453741

RESUMEN

In the U.S., approximately 1.7 million people suffer traumatic brain injury each year, with many enduring long-term consequences and significant medical and rehabilitation costs. The primary injury causes physical damage to neurons, glia, fiber tracts and microvasculature, which is then followed by secondary injury, consisting of pathophysiological mechanisms including an immune response, inflammation, edema, excitotoxicity, oxidative damage, and cell death. Most attempts at intervention focus on protection, repair or regeneration, with regenerative medicine becoming an intensively studied area over the past decade. The use of stem cells has been studied in many disease and injury models, using stem cells from a variety of sources and applications. In this study, human adipose-derived mesenchymal stromal cells (MSCs) were administered at early (3 days) and delayed (14 days) time points after controlled cortical impact (CCI) injury in rats. Animals were routinely assessed for neurological and vestibulomotor deficits, and at 32 days post-injury, brain tissue was processed by flow cytometry and immunohistochemistry to analyze neuroinflammation. Treatment with HB-adMSC at either 3d or 14d after injury resulted in significant improvements in neurocognitive outcome and a change in neuroinflammation one month after injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Trasplante de Células Madre Mesenquimatosas , Tejido Adiposo/citología , Animales , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/psicología , Modelos Animales de Enfermedad , Humanos , Inflamación/patología , Masculino , Aprendizaje por Laberinto , Células Madre Mesenquimatosas/citología , Destreza Motora , Neurogénesis , Ratas Sprague-Dawley , Factores de Tiempo
7.
Stem Cells Transl Med ; 9(4): 491-498, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31903737

RESUMEN

Clinical trials in trauma populations are exploring the use of clinical cellular therapeutics (CCTs) like human mesenchymal stromal cells (MSC) and mononuclear cells (MNC). Recent studies demonstrate a procoagulant effect of these CCTs related to their expression of tissue factor (TF). We sought to examine this relationship in blood from severely injured trauma patients and identify methods to reverse this procoagulant effect. Human MSCs from bone marrow, adipose, and amniotic tissues and freshly isolated bone marrow MNC samples were tested. TF expression and phenotype were quantified using flow cytometry. CCTs were mixed individually with trauma patients' whole blood, assayed with thromboelastography (TEG), and compared with healthy subjects mixed with the same cell sources. Heparin was added to samples at increasing concentrations until TEG parameters normalized. Clotting time or R time in TEG decreased relative to the TF expression of the CCT treatment in a logarithmic fashion for trauma patients and healthy subjects. Nonlinear regression curves were significantly different with healthy subjects demonstrating greater relative decreases in TEG clotting time. In vitro coadministration of heparin normalized the procoagulant effect and required dose escalation based on TF expression. TF expression in human MSC and MNC has a procoagulant effect in blood from trauma patients and healthy subjects. The procoagulant effect is lower in trauma patients possibly because their clotting time is already accelerated. The procoagulant effect due to MSC/MNC TF expression could be useful in the bleeding trauma patient; however, it may emerge as a safety release criterion due to thrombotic risk. The TF procoagulant effect is reversible with heparin.


Asunto(s)
Coagulación Sanguínea , Heridas y Lesiones/sangre , Adulto , Biomarcadores/metabolismo , Coagulación Sanguínea/efectos de los fármacos , Estudios de Casos y Controles , Femenino , Heparina/farmacología , Humanos , Masculino , Tromboelastografía
8.
Front Immunol ; 10: 1645, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31417542

RESUMEN

For several decades, multipotent mesenchymal stromal cells (MSCs) have been extensively studied for their therapeutic potential across a wide range of diseases. In the preclinical setting, MSCs demonstrate consistent ability to promote tissue healing, down-regulate excessive inflammation and improve outcomes in animal models. Several proposed mechanisms of action have been posited and demonstrated across an array of in vitro models. However, translation into clinical practice has proven considerably more difficult. A number of prominent well-funded late-phase clinical trials have failed, thus calling out for new efforts to optimize product delivery in the clinical setting. In this review, we discuss novel topics critical to the successful translation of MSCs from pre-clinical to clinical applications. In particular, we focus on the major routes of cell delivery, aspects related to hemocompatibility, and potential safety concerns associated with MSC therapy in the different settings.


Asunto(s)
Diferenciación Celular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Animales , Modelos Animales de Enfermedad , Humanos
9.
Stem Cells Transl Med ; 7(10): 731-739, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30070065

RESUMEN

Clinical cellular therapeutics (CCTs) have shown preliminary efficacy in reducing inflammation after trauma, preserving cardiac function after myocardial infarction, and improving functional recovery after stroke. However, most clinically available cell lines express tissue factor (TF) which stimulates coagulation. We sought to define the degree of procoagulant activity of CCTs as related to TF expression. CCT samples from bone marrow, adipose, amniotic fluid, umbilical cord, multi-potent adult progenitor cell donors, and bone marrow mononuclear cells were tested. TF expression and phenotype were quantified using flow cytometry. Procoagulant activity of the CCTs was measured in vitro with thromboelastography and calibrated thrombogram. Fluorescence-activated cell sorting (FACS) separated samples into high- and low-TF expressing populations to isolate the contribution of TF to coagulation. A TF neutralizing antibody was incubated with samples to demonstrate loss of procoagulant function. All CCTs tested expressed procoagulant activity that correlated with expression of tissue factor. Time to clot and thrombin formation decreased with increasing TF expression. High-TF expressing cells decreased clotting time more than low-TF expressing cells when isolated from a single donor using FACS. A TF neutralizing antibody restored clotting time to control values in some, but not all, CCT samples. CCTs demonstrate wide variability in procoagulant activity related to TF expression. Time to clot and thrombin formation decreases as TF load increases and this procoagulant effect is neutralized by a TF blocking antibody. Clinical trials using CCTs are in progress and TF expression may emerge as a safety release criterion. Stem Cells Translational Medicine 2018;7:731-739.


Asunto(s)
Coagulación Sanguínea , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Tromboplastina/metabolismo , Tejido Adiposo/citología , Líquido Amniótico/citología , Células de la Médula Ósea/citología , Sangre Fetal/citología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Tromboelastografía , Trombina/metabolismo , Tromboplastina/genética
10.
Sci Rep ; 8(1): 480, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29323194

RESUMEN

No current clinical intervention can alter the course of acute spinal cord injury (SCI), or appreciably improve neurological outcome. Mesenchymal stromal cells (MSCs) have been shown to modulate the injury sequelae of SCI largely via paracrine effects, although the mechanisms remain incompletely understood. One potential modality is through secretion of extracellular vesicles (EVs). In this study, we investigate whether systemic administration of EVs isolated from human MSCs (MSCEv) has the potential to be efficacious as an alternative to cell-based therapy for SCI. Additionally, we investigate whether EVs isolated from human MSCs stimulated with pro-inflammatory cytokines have enhanced anti-inflammatory effects when administered after SCI. Immunohistochemistry supported the quantitative analysis, demonstrating a diminished inflammatory response with apparent astrocyte and microglia disorganization in cord tissue up to 10 mm caudal to the injury site. Locomotor recovery scores showed significant improvement among animals treated with MSCEv. Significant increases in mechanical sensitivity threshold were observed in animals treated with EVs from either naïve MSC (MSCEvwt) or stimulated MSC (MSCEv+), with a statistically significant increase in threshold for MSCEv+-treated animals when compared to those that received MSCEvwt. In conclusion, these data show that treatment of acute SCI with extracellular vesicles derived from human MSCs attenuates neuroinflammation and improves functional recovery.


Asunto(s)
Vesículas Extracelulares/trasplante , Microglía/metabolismo , Traumatismos de la Médula Espinal/terapia , Animales , Astrocitos/citología , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Vesículas Extracelulares/metabolismo , Humanos , Inflamación , Locomoción/fisiología , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Microglía/citología , Neutrófilos/citología , Neutrófilos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de IgG/metabolismo , Recuperación de la Función , Bazo/citología , Bazo/inmunología , Resultado del Tratamiento
11.
Stem Cells ; 36(1): 79-90, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29076623

RESUMEN

Extracellular vesicles (EVs) secreted by mesenchymal stromal cells (MSCs) have been proposed to be a key mechanistic link in the therapeutic efficacy of cells in response to cellular injuries through paracrine effects. We hypothesize that inflammatory stimulation of MSCs results in the release of EVs that have greater anti-inflammatory effects. The present study evaluates the immunomodulatory abilities of EVs derived from inflammation-stimulated and naive MSCs (MSCEv+ and MSCEv, respectively) isolated using a current Good Manufacturing Practice-compliant tangential flow filtration system. Detailed characterization of both EVs revealed differences in protein composition, cytokine profiles, and RNA content, despite similarities in size and expression of common surface markers. MSCEv+ further attenuated release of pro-inflammatory cytokines in vitro when compared to MSCEv, with a distinctly different pattern of EV-uptake by activated primary leukocyte subpopulations. The efficacy of EVs was partially attributed to COX2/PGE2 expression. The present study demonstrates that inflammatory stimulation of MSCs renders release of EVs that have enhanced anti-inflammatory properties partially due to COX2/PGE2 pathway alteration. Stem Cells 2018;36:79-90.


Asunto(s)
Vesículas Extracelulares/metabolismo , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Microscopía Electrónica de Transmisión/métodos , Humanos
13.
Nanoscale ; 9(38): 14581-14591, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28932838

RESUMEN

Inflammatory bowel disease (IBD), which includes Crohn's disease and ulcerative colitis, is a chronic inflammatory condition of the gastrointestinal (GI) tract. Currently, it is treated with immunosuppressant or biologics that often induce severe adverse effects. Thus, there is an urgent clinical need for more specific treatments. To provide a valid therapeutic tool for IBD therapy, in this work we developed biomimetic nanovesicles by manipulating leukocyte membranes to exploit mechanisms of T-cell recruitment during inflammation. A subset of T-lymphocytes participates in homing to inflamed tissue in the gastrointestinal tract by overexpressing the α4ß7 integrin, which is responsible for binding to its receptor on the endothelial membrane, the mucosal addressin cell adhesion molecule 1. Based on this principle, we engineered biomimetic vesicles, referred to as specialized leukosomes (SLKs), which are leukocyte-like carriers 'doped' with the α4ß7 integrin over-induced in purified immune cells. We tested SLKs in an in vivo murine model of IBD induced by treatment with dextran sulfate sodium. Notably, treatment of IBD mice with SLKs allowed us to observe a reduction of inflammation (favorable modulation of both pro- and anti-inflammatory genes, as well as reduction of immune cells infiltration into the colon tissue), and a consequent enhanced intestinal repair (low epithelial damage). In this study, we demonstrate that biological-derived nanoparticles can be used not only as naturally targeted drug delivery systems, but also as nano-therapeutics endowed with intrinsic anti-inflammatory properties.


Asunto(s)
Antiinflamatorios/farmacología , Materiales Biomiméticos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Nanopartículas , Animales , Sulfato de Dextran , Enfermedades Inflamatorias del Intestino/inducido químicamente , Integrinas , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología
14.
ACS Nano ; 11(3): 3262-3273, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28264157

RESUMEN

Understanding interactions occurring at the interface between nanoparticles and biological components is an urgent challenge in nanomedicine due to their effect on the biological fate of nanoparticles. After the systemic injection of nanoparticles, a protein corona constructed by blood components surrounds the carrier's surface and modulates its pharmacokinetics and biodistribution. Biomimicry-based approaches in nanotechnology attempt to imitate what happens in nature in order to transfer specific natural functionalities to synthetic nanoparticles. Several biomimetic formulations have been developed, showing superior in vivo features as a result of their cell-like identity. We have recently designed biomimetic liposomes, called leukosomes, which recapitulate the ability of leukocytes to target inflamed endothelium and escape clearance by the immune system. To gain insight into the properties of leukosomes, we decided to investigate their protein corona in vivo. So far, most information about the protein corona has been obtained using in vitro experiments, which have been shown to minimally reproduce in vivo phenomena. Here we directly show a time-dependent quantitative and qualitative analysis of the protein corona adsorbed in vivo on leukosomes and control liposomes. We observed that leukosomes absorb fewer proteins than liposomes, and we identified a group of proteins specifically adsorbed on leukosomes. Moreover, we hypothesize that the presence of macrophage receptors on leukosomes' surface neutralizes their protein corona-meditated uptake by immune cells. This work unveils the protein corona of a biomimetic carrier and is one of the few studies on the corona performed in vivo.


Asunto(s)
Nanopartículas/química , Corona de Proteínas/química , Adsorción , Animales , Línea Celular , Microscopía por Crioelectrón , Electroforesis Capilar , Liposomas/sangre , Liposomas/química , Espectrometría de Masas , Ratones , Microscopía Confocal
15.
Methodist Debakey Cardiovasc J ; 12(3): 141-145, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27826367

RESUMEN

Atherosclerosis is a complex process responsible for a major burden of cardiovascular morbidity and mortality. Macrophages and smooth muscle cells (SMCs) are abundant within atherosclerotic plaques. This review discusses the role of macrophages and SMCs in plaque progression and provides an overview of nanoparticle-based approaches and other current methods for local targeting of atherosclerotic plaques.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Macrófagos/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Placa Aterosclerótica/prevención & control , Animales , Proliferación Celular , Progresión de la Enfermedad , Humanos , Macrófagos/patología , Músculo Liso Vascular/patología , Placa Aterosclerótica/patología
16.
Methodist Debakey Cardiovasc J ; 12(3): 169-174, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27826372

RESUMEN

Despite an improved understanding of its pathophysiology and a wide range of new treatments, cardiovascular disease (CVD) remains a serious public health issue and the number one cause of mortality in the United States. Conditions that promote chronic systemic inflammation, such as obesity, cancer, and autoimmune and infectious diseases, are now known to play an important role in promoting CVD by inducing the expression of endothelial adhesion molecules and chemokines; these in turn promote leukocyte adherence and infiltration, which initiates and spurs the progression of CVD. In response to this new understanding, researchers are evaluating the potential cardiovascular benefits of new-generation therapies based on endogenous molecules with anti-inflammatory properties. Similarly, targeted approaches that leverage the phenotypic differences between non-inflamed and inflamed endothelia have the potential to selectively deliver therapeutics and decrease the morbidity and mortality of CVD patients. In this review, we discuss the role of inflammation in CVD and explore the therapeutic potential of targeting inflamed vasculature through conventional and biomimetic approaches.


Asunto(s)
Factores Biológicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Nanomedicina/métodos , Nanopartículas , Vasculitis/tratamiento farmacológico , Humanos
17.
Int J Nanomedicine ; 11: 3049-63, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27445473

RESUMEN

A thorough understanding of interactions occurring at the interface between nanocarriers and biological systems is crucial to predict and interpret their biodistribution, targeting, and efficacy, and thus design more effective drug delivery systems. Upon intravenous injection, nanoparticles are coated by a protein corona (PC). This confers a new biological identity on the particles that largely determines their biological fate. Liposomes have great pharmaceutical versatility, so, as proof of concept, their PC has recently been implicated in the mechanism and efficiency of their internalization into the cell. In an attempt to better understand the interactions between nanocarriers and biological systems, we analyzed the plasma proteins adsorbed on the surface of multicomponent liposomes. Specifically, we analyzed the physical properties and ultrastructure of liposome/PC complexes and the aggregation process that occurs when liposomes are dispersed in plasma. The results of combined confocal microscopy and flow cytometry experiments demonstrated that the PC favors liposome internalization by both macrophages and tumor cells. This work provides insights into the effects of the PC on liposomes' physical properties and, consequently, liposome-liposome and liposome-cell interactions.


Asunto(s)
Comunicación Celular , Liposomas/química , Corona de Proteínas/química , Adsorción , Animales , Proteínas Sanguíneas/metabolismo , Línea Celular Tumoral , Microscopía por Crioelectrón , Dispersión Dinámica de Luz , Endocitosis , Citometría de Flujo , Humanos , Ratones Endogámicos BALB C , Microscopía de Fuerza Atómica , Nanopartículas/química , Proteómica
18.
Nanomedicine (Lond) ; 11(1): 81-100, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26653875

RESUMEN

In a perfect sequence of events, nanoparticles (NPs) are injected into the bloodstream where they circulate until they reach the target tissue. The ligand on the NP surface recognizes its specific receptor expressed on the target tissue and the drug is released in a controlled manner. However, once injected in a physiological environment, NPs interact with biological components and are surrounded by a protein corona (PC). This can trigger an immune response and affect NP toxicity and targeting capabilities. In this review, we provide a survey of recent findings on the NP-PC interactions and discuss how the PC can be used to modulate both cytotoxicity and the immune response as well as to improve the efficacy of targeted delivery of nanocarriers.


Asunto(s)
Nanopartículas/toxicidad , Animales , Activación de Complemento , Sistemas de Liberación de Medicamentos , Humanos , Nanopartículas/administración & dosificación
19.
Nano Lett ; 13(7): 3248-55, 2013 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-23786263

RESUMEN

The ultimate goal in cancer therapy is achieving selective targeting of cancer cells. We report a novel delivery platform, based on nanoghosts (NGs) produced from the membranes of mesenchymal stem cells (MSCs). Encompassing MSC surface molecules, the MSC-NGs retained MSC-specific in vitro and in vivo tumor targeting capabilities and were cleared from blood-filtering organs. MSC-NGs were found to be biocompatible. Systemic administration of drug loaded MSC-NGs demonstrated 80% inhibition of human prostate cancer.


Asunto(s)
Membrana Celular/química , Células Madre Mesenquimatosas/química , Nanocápsulas/química , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/farmacocinética , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/farmacocinética , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Nanocápsulas/administración & dosificación , Nanocápsulas/ultraestructura , Especificidad de Órganos , Tamaño de la Partícula , Distribución Tisular , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...