Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biomacromolecules ; 23(11): 4629-4644, 2022 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-36288499

RESUMEN

The co-administration of glial cell line-derived neurotrophic factor (GDNF) and mesenchymal stem cells (MSCs) in hydrogels (HGs) has emerged as a powerful strategy to enhance the efficient integration of transplanted cells in Parkinson's disease (PD). This strategy could be improved by controlling the cellular microenvironment and biomolecule release and better mimicking the complex properties of the brain tissue. Here, we develop and characterize a drug delivery system for brain repair where MSCs and GDNF are included in a nanoparticle-modified supramolecular guest-host HA HG. In this system, the nanoparticles act as both carriers for the GDNF and active physical crosslinkers of the HG. The multifunctional HG is mechanically compatible with brain tissue and easily injectable. It also protects GDNF from degradation and achieves its controlled release over time. The cytocompatibility studies show that the developed biomaterial provides a friendly environment for MSCs and presents good compatibility with PC12 cells. Finally, using RNA-sequencing (RNA-seq), we investigated how the three-dimensional (3D) environment, provided by the nanostructured HG, impacted the encapsulated cells. The transcriptome analysis supports the beneficial effect of including MSCs in the nanoreinforced HG. An enhancement in the anti-inflammatory effect of MSCs was observed, as well as a differentiation of the MSCs toward a neuron-like cell type. In summary, the suitable strength, excellent self-healing properties, good biocompatibility, and ability to boost MSC regenerative potential make this nanoreinforced HG a good candidate for drug and cell administration to the brain.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Ratas , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Ingeniería de Tejidos/métodos , Hidrogeles/farmacología , Hidrogeles/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Encéfalo/metabolismo
2.
J Pharmacol Exp Ther ; 370(3): 761-771, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30728248

RESUMEN

Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) are a promising cell source for cardiac repair after myocardial infarction (MI) because they offer several advantages such as potential to remuscularize infarcted tissue, integration in the host myocardium, and paracrine therapeutic effects. However, cell delivery issues have limited their potential application in clinical practice, showing poor survival and engraftment after transplantation. In this work, we hypothesized that the combination of hiPSC-CMs with microparticles (MPs) could enhance long-term cell survival and retention in the heart and consequently improve cardiac repair. CMs were obtained by differentiation of hiPSCs by small-molecule manipulation of the Wnt pathway and adhered to biomimetic poly(lactic-co-glycolic acid) MPs covered with collagen and poly(d-lysine). The potential of the system to support cell survival was analyzed in vitro, demonstrating a 1.70-fold and 1.99-fold increase in cell survival after 1 and 4 days, respectively. The efficacy of the system was tested in a mouse MI model. Interestingly, 2 months after administration, transplanted hiPSC-CMs could be detected in the peri-infarct area. These cells not only maintained the cardiac phenotype but also showed in vivo maturation and signs of electrical coupling. Importantly, cardiac function was significantly improved, which could be attributed to a paracrine effect of cells. These findings suggest that MPs represent an excellent platform for cell delivery in the field of cardiac repair, which could also be translated into an enhancement of the potential of cell-based therapies in other medical applications.


Asunto(s)
Plásticos Biodegradables/uso terapéutico , Cardiopatías/terapia , Células Madre Pluripotentes Inducidas/trasplante , Miocitos Cardíacos/trasplante , Nanopartículas/uso terapéutico , Trasplante de Células Madre/métodos , Animales , Diferenciación Celular , Supervivencia Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Cardiopatías/patología , Pruebas de Función Cardíaca , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Infarto del Miocardio/terapia , Remodelación Ventricular
3.
Adv Drug Deliv Rev ; 208: 115302, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38574952

RESUMEN

Cardiovascular diseases (CVDs) stand as the leading cause of death worldwide, posing a significant global health challenge. Consequently, the development of innovative therapeutic strategies to enhance CVDs treatment is imperative. RNA-based therapies, encompassing non-coding RNAs, mRNA, aptamers, and CRISPR/Cas9 technology, have emerged as promising tools for addressing CVDs. However, inherent challenges associated with RNA, such as poor cellular uptake, susceptibility to RNase degradation, and capture by the reticuloendothelial system, underscore the necessity of combining these therapies with effective drug delivery systems. Various non-viral delivery systems, including extracellular vesicles, lipid-based carriers, polymeric and inorganic nanoparticles, as well as hydrogels, have shown promise in enhancing the efficacy of RNA therapeutics. In this review, we offer an overview of the most relevant RNA-based therapeutic strategies explored for addressing CVDs and emphasize the pivotal role of delivery systems in augmenting their effectiveness. Additionally, we discuss the current status of these therapies and the challenges that hinder their clinical translation.


Asunto(s)
Enfermedades Cardiovasculares , Nanopartículas , Humanos , ARN , Enfermedades Cardiovasculares/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , ARN Mensajero/genética
4.
Eur J Pharm Sci ; 185: 106439, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37003408

RESUMEN

Myocardial infarction is one of the major causes of morbidity and mortality worldwide. Current treatments can relieve the symptoms of myocardial ischemia but cannot repair the necrotic myocardial tissue. Novel therapeutic strategies based on cellular therapy, extracellular vesicles, non-coding RNAs and growth factors have been designed to restore cardiac function while inducing cardiomyocyte cycle re-entry, ensuring angiogenesis and cardioprotection, and preventing ventricular remodeling. However, they face low stability, cell engraftment issues or enzymatic degradation in vivo, and it is thus essential to combine them with biomaterial-based delivery systems. Microcarriers, nanocarriers, cardiac patches and injectable hydrogels have yielded promising results in preclinical studies, some of which are currently being tested in clinical trials. In this review, we cover the recent advances made in cellular and acellular therapies used for cardiac repair after MI. We present current trends in cardiac tissue engineering related to the use of microcarriers, nanocarriers, cardiac patches and injectable hydrogels as biomaterial-based delivery systems for biologics. Finally, we discuss some of the most crucial aspects that should be addressed in order to advance towards the clinical translation of cardiac tissue engineering approaches.


Asunto(s)
Infarto del Miocardio , Ingeniería de Tejidos , Humanos , Ingeniería de Tejidos/métodos , Infarto del Miocardio/terapia , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos , Materiales Biocompatibles , Hidrogeles
5.
J Control Release ; 361: 130-146, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37532145

RESUMEN

RNA-based therapies, and siRNAs in particular, have attractive therapeutic potential for cancer treatment due to their ability to silence genes that are imperative for tumor progression. To be effective and solve issues related to their poor half-life and poor pharmacokinetic properties, siRNAs require adequate drug delivery systems that protect them from degradation and allow intracellular delivery. Among the various delivery vehicles available, lipid nanoparticles have emerged as the leading choice. These nanoparticles consist of cholesterol, phospholipids, PEG-lipids and most importantly ionizable cationic lipids. These ionizable lipids enable the binding of negatively charged siRNA, resulting in the formation of stable and neutral lipid nanoparticles with exceptionally high encapsulation efficiency. Lipid nanoparticles have demonstrated their effectiveness and versatility in delivering not only siRNAs but also multiple RNA molecules, contributing to their remarkable success. Furthermore, the advancement of efficient manufacturing techniques such as microfluidics, enables the rapid mixing of two miscible solvents without the need for shear forces. This facilitates the reproducible production of lipid nanoparticles and holds enormous potential for scalability. This is shown by the increasing number of preclinical and clinical trials evaluating the potential use of siRNA-LNPs for the treatment of solid and hematological tumors as well as in cancer immunotherapy. In this review, we provide an overview of the progress made on siRNA-LNP development for cancer treatment and outline the current preclinical and clinical landscape in this area. Finally, the translational challenges required to bring siRNA-LNPs further into the clinic are also discussed.


Asunto(s)
Nanopartículas , Neoplasias , ARN Interferente Pequeño , Liposomas , Nanopartículas/química , Fosfolípidos , Neoplasias/genética , Neoplasias/terapia
6.
Expert Opin Drug Deliv ; 19(11): 1521-1537, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36240170

RESUMEN

INTRODUCTION: Parkinson's disease is the second most common neurodegenerative disease. Currently, there are no curative therapies, with only symptomatic treatment available. One of the principal reasons for the lack of treatments is the problem of delivering drugs to the brain, mainly due to the blood-brain barrier. Hydrogels are presented as ideal platforms for delivering treatments to the brain ranging from small molecules to cell replacement therapies. AREAS COVERED: The potential application of hydrogel-based therapies for Parkinson's disease is addressed. The desirable composition and mechanical properties of these therapies for brain application are discussed, alongside the preclinical research available with hydrogels in Parkinson's disease. Lastly, translational and manufacturing challenges are presented. EXPERT OPINION: Parkinson's disease urgently needs novel therapies to delay its progression and for advanced stages, at which conventional therapies fail to control motor symptoms. Neurotrophic factor-loaded hydrogels with stem cells offer one of the most promising therapies. This approach may increase the striatal dopamine content while protecting and promoting the differentiation of stem cells although the generation of synapses between engrafted and host cells remains an issue to overcome. Other challenges to consider are related to the route of administration of hydrogels and their large-scale production, required to accelerate their translation toward the clinic.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Hidrogeles , Enfermedades Neurodegenerativas/tratamiento farmacológico , Encéfalo , Dopamina/uso terapéutico
7.
J Control Release ; 348: 553-571, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35705114

RESUMEN

Embryonal tumors of the nervous system are neoplasms predominantly affecting the pediatric population. Among the most common and aggressive ones are neuroblastoma (NB) and medulloblastoma (MB). NB is a sympathetic nervous system tumor, which is the most frequent extracranial solid pediatric cancer, usually detected in children under two. MB originates in the cerebellum and is one of the most lethal brain tumors in early childhood. Their tumorigenesis presents some similarities and both tumors often have treatment resistances and poor prognosis. High-risk (HR) patients require high dose chemotherapy cocktails associated with acute and long-term toxicities. Nanomedicine and cell therapy arise as potential solutions to improve the prognosis and quality of life of children suffering from these tumors. Indeed, nanomedicines have been demonstrated to efficiently reduce drug toxicity and improve drug efficacy. Moreover, these systems have been extensively studied in cancer research over the last few decades and an increasing number of anticancer nanocarriers for adult cancer treatment has reached the clinic. Among cell-based strategies, the clinically most advanced approach is chimeric-antigen receptor (CAR) T therapy for both pathologies, which is currently under investigation in phase I/II clinical trials. However, pediatric drug research is especially hampered due not only to ethical issues but also to the lack of efficient pre-clinical models and the inadequate design of clinical trials. This review provides an update on progress in the treatment of the main embryonal tumors of the nervous system using nanotechnology and cell-based therapies and discusses key issues behind the gap between preclinical studies and clinical trials in this specific area. Some directions to improve their translation into clinical practice and foster their development are also provided.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Neoplasias de Células Germinales y Embrionarias , Neuroblastoma , Adulto , Tratamiento Basado en Trasplante de Células y Tejidos , Niño , Preescolar , Humanos , Meduloblastoma/tratamiento farmacológico , Nanomedicina , Neoplasias de Células Germinales y Embrionarias/tratamiento farmacológico , Calidad de Vida
8.
Eur J Pharm Biopharm ; 170: 187-196, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34968647

RESUMEN

Since the discovery of the beneficial therapeutical effects of extracellular vesicles (EVs), these agents have been attracting great interest as next-generation therapies. EVs are nanosized membrane bodies secreted by all types of cells that mediate cell-cell communication. Although the classification of different subpopulations of EVs can be complex, they are broadly divided into microvesicles and exosomes based on their biogenesis and in large and small EVs based on their size. As this is an emerging field, current investigations are focused on basic aspects such as the more convenient method for EV isolation. In the present paper, we used cardiac progenitor cells (CPCs) to study and compare different cell culture conditions for EV isolation as well as two of the most commonly employed purification methods: ultracentrifugation (UC) and size-exclusion chromatography (SEC). Large and small EVs were separately analysed. We found that serum starvation of cells during the EV collecting period led to a dramatic decrease in EV secretion and major cell death. Regarding the isolation method, our findings suggest that UC and SEC gave similar EV recovery rates. Separation of large and small EV-enriched subpopulations was efficiently achieved with both purification protocols although certain difference in sample heterogeneity was observed. Noteworthy, while calnexin was abundant in large EVs, ALIX and CD63 were mainly found in small EVs. Finally, when the functionality of EVs was assessed on primary culture of adult murine cardiac fibroblasts, we found that EVs were taken up by these cells, which resulted in a pronounced reduction in the proliferative and migratory capacity of the cells. Specifically, a tendency towards a larger effect of SEC-related EVs was observed. No differences could be found between large and small EVs. Altogether, these results contribute to establish the basis for the use of EVs as therapeutic platforms, in particular in regenerative fields.


Asunto(s)
Vesículas Extracelulares , Miocardio/citología , Miofibroblastos/metabolismo , Células Madre/citología , Animales , Proteínas de Unión al Calcio/metabolismo , Calnexina/metabolismo , Células Cultivadas , Masculino , Ratones , Ratas Wistar , Tetraspanina 30/metabolismo
9.
Int J Pharm ; 629: 122356, 2022 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36332831

RESUMEN

Extracellular vesicles (EVs) are nanosized particles with attractive therapeutic potential for cardiac repair. However, low retention and stability after systemic administration limit their clinical translation. As an alternative, the combination of EVs with biomaterial-based hydrogels (HGs) is being investigated to increase their exposure in the myocardium and achieve an optimal therapeutic effect. In this study, we developed and characterized a novel injectable in-situ forming HG based on alginate and collagen as a cardiac delivery vehicle for EVs. Different concentrations of alginate and collagen crosslinked with calcium gluconate were tested. Based on injectability studies, 1% alginate, 0.5 mg/mL collagen and 0.25% calcium gluconate HG was selected as the idoneous combination for cardiac administration using catheter-based systems. Rheological examination revealed that the HG possessed an internal gel structure, weak mechanical properties and low viscosity, facilitating an easy administration. In addition, EVs were successfully incorporated and homogeneously distributed in the HG. After administration in a rat model of myocardial infarction, the HG showed long-term retention in the heart and allowed for a sustained release of EVs for at least 7 days. Thus, the combination of HGs and EVs represents a promising therapeutic strategy for myocardial repair. Besides EVs delivery, the developed HG could represent a useful platform for cardiac delivery of multiple therapeutic agents.


Asunto(s)
Vesículas Extracelulares , Hidrogeles , Ratas , Animales , Hidrogeles/química , Alginatos/química , Gluconato de Calcio , Colágeno
10.
J Neurochem ; 119(5): 972-88, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21496021

RESUMEN

Cell-based therapies for global cerebral ischemia represent promising approaches for neuronal damage prevention and tissue repair promotion. We examined the potential of marrow-isolated adult multilineage-inducible (MIAMI) cells, a homogeneous subpopulation of immature human mesenchymal stromal cell, injected into the hippocampus to prevent neuronal damage induced by global ischemia using rat organotypic hippocampal slices exposed to oxygen-glucose deprivation and rats subjected to asphyxial cardiac arrest. We next examined the value of combining fibronectin-coated biomimetic microcarriers (FN-BMMs) with epidermal growth factor (EGF)/basic fibroblast growth factor (bFGF) pre-treated MIAMI compared to EGF/bFGF pre-treated MIAMI cells alone, for their in vitro and in vivo neuroprotective capacity. Naïve and EGF/bFGF pre-treated MIAMI cells significantly protected the Cornu Ammonis layer 1 (CA1) against ischemic death in hippocampal slices and increased CA1 survival in rats. MIAMI cells therapeutic value was significantly increased when delivering the cells complexed with FN-BMMs, probably by increasing stem cell survival and paracrine secretion of pro-survival and/or anti-inflammatory molecules as concluded from survival, differentiation and gene expression analysis. Four days after oxygen and glucose deprivation and asphyxial cardiac arrest, few transplanted cells administered alone survived in the brain whereas stem cell survival improved when injected complexed with FN-BMMs. Interestingly, a large fraction of the transplanted cells administered alone or in complexes expressed ßIII-tubulin suggesting that partial neuronal transdifferentiation may be a contributing factor to the neuroprotective mechanism of MIAMI cells.


Asunto(s)
Materiales Biomiméticos/farmacología , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Diferenciación Celular/fisiología , Hipocampo/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Multipotentes/citología , Animales , Células de la Médula Ósea/citología , Células Cultivadas , Humanos , Ácido Láctico/farmacología , Masculino , Células Madre Mesenquimatosas/citología , Neuronas/patología , Técnicas de Cultivo de Órganos , Ácido Poliglicólico/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Trasplante Heterólogo/métodos , Adulto Joven
11.
Mov Disord ; 26(10): 1943-7, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21661048

RESUMEN

BACKGROUND: Glial cell-derived neurotrophic factor is a survival factor for dopaminergic neurons and a promising candidate for the treatment of Parkinson's disease. However, the delivery issue of the protein to the brain still remains unsolved. Our aim was to investigate the effect of long-term delivery of encapsulated glial cell-derived neurotrophic factor within microspheres. METHODS: A single dose of microspheres containing 2.5 µg of glial cell-derived neurotrophic factor was implanted intrastriatally in animals 2 weeks after a 6-hydroxydopamine lesion. RESULTS: The amphetamine test showed a complete behavioral recovery after 16 weeks of treatment, which was maintained until the end of the study (week 30). This effect was accompanied by an increase in dopaminergic striatal terminals and neuroprotection of dopaminergic neurons. CONCLUSIONS: The main achievement was the long-term neurorestoration in parkinsonian animals induced by encapsulated glial cell-derived neurotrophic factor, suggesting that microspheres may be considered as a means to deliver glial cell-derived neurotrophic factor for Parkinson's disease treatment.


Asunto(s)
Factores Neurotróficos Derivados de la Línea Celular Glial/administración & dosificación , Microesferas , Fármacos Neuroprotectores/administración & dosificación , Trastornos Parkinsonianos/tratamiento farmacológico , Implantes Absorbibles , Adrenérgicos/toxicidad , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Femenino , Lateralidad Funcional , Actividad Motora/efectos de los fármacos , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/patología , Trastornos Parkinsonianos/fisiopatología , Ratas , Ratas Sprague-Dawley , Estadísticas no Paramétricas , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Factores de Tiempo , Tirosina 3-Monooxigenasa
12.
Eur J Pharm Sci ; 159: 105726, 2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33482318

RESUMEN

Human glial cell line-derived neurotrophic factor (hGDNF) is the most potent dopaminergic factor described so far, and it is therefore considered a promising drug for Parkinson's disease (PD) treatment. However, the production of therapeutic proteins with a high degree of purity and a specific glycosylation pattern is a major challenge that hinders its commercialization. Although a variety of systems can be used for protein production, only a small number of them are suitable to produce clinical-grade proteins. Specifically, the baby hamster kidney cell line (BHK-21) has shown to be an effective system for the expression of high levels of hGDNF, with appropriate post-translational modifications and protein folding. This system, which is based on the electroporation of BHK-21 cells using a Semliki Forest virus (SFV) as expression vector, induces a strong shut-off of host cell protein synthesis that simplify the purification process. However, SFV vector exhibits a temperature-dependent cytopathic effect on host cells, which could limit hGDNF expression. The aim of this study was to improve the expression and purification of hGDNF using a biphasic temperature cultivation protocol that would decrease the cytopathic effect induced by SFV. Here we show that an increase in the temperature from 33°C to 37°C during the "shut-off period", produced a significant improvement in cell survival and hGDNF expression. In consonance, this protocol led to the production of almost 3-fold more hGDNF when compared to the previously described methods. Therefore, a "recovery period" at 37°C before cells are exposed at 33°C is crucial to maintain cell viability and increase hGDNF expression. The protocol described constitutes an efficient and highly scalable method to produce highly pure hGDNF.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial , Virus de los Bosques Semliki , Animales , Línea Celular , Cricetinae , Dopamina , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos , Virus de los Bosques Semliki/genética
13.
Nanomaterials (Basel) ; 11(3)2021 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-33668836

RESUMEN

Extracellular vesicles (EVs) are constituted by a group of heterogeneous membrane vesicles secreted by most cell types that play a crucial role in cell-cell communication. In recent years, EVs have been postulated as a relevant novel therapeutic option for cardiovascular diseases, including myocardial infarction (MI), partially outperforming cell therapy. EVs may present several desirable features, such as no tumorigenicity, low immunogenic potential, high stability, and fine cardiac reparative efficacy. Furthermore, the natural origin of EVs makes them exceptional vehicles for drug delivery. EVs may overcome many of the limitations associated with current drug delivery systems (DDS), as they can travel long distances in body fluids, cross biological barriers, and deliver their cargo to recipient cells, among others. Here, we provide an overview of the most recent discoveries regarding the therapeutic potential of EVs for addressing cardiac damage after MI. In addition, we review the use of bioengineered EVs for targeted cardiac delivery and present some recent advances for exploiting EVs as DDS. Finally, we also discuss some of the most crucial aspects that should be addressed before a widespread translation to the clinical arena.

14.
BMC Mol Biol ; 11: 61, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20716364

RESUMEN

BACKGROUND: RT-qPCR analysis is a widely used method for the analysis of mRNA expression throughout the field of mesenchymal stromal cell (MSC) research. Comparison between MSC studies, both in vitro and in vivo, are challenging due to the varied methods of RT-qPCR data normalization and analysis. Therefore, this study focuses on putative housekeeping genes for the normalization of RT-qPCR data between heterogeneous commercially available human MSC, compared with more homogeneous populations of MSC such as MIAMI and RS-1 cells. RESULTS: Eight genes including; ACTB, B2M, EF1alpha, GAPDH, RPL13a, YWHAZ, UBC and HPRT1 were tested as possible housekeeping genes based on their expression level and variability. EF1alpha and RPL13a were validated for RT-qPCR analysis of MIAMI cells during expansion in varied oxygen tensions, endothelial differentiation, neural precursor enrichment, and during the comparison with RS-1 cells and commercially available MSC. RPL13a and YWHAZ were validated as normalization genes for the cross-species analysis of MIAMI cells in an animal model of focal ischemia. GAPDH, which is one of the most common housekeeping genes used for the normalization of RT-qPCR data in the field of MSC research, was found to have the highest variability and deemed not suitable for normalization of RT-qPCR data. CONCLUSIONS: In order to make comparisons between heterogeneous MSC populations, as well as adult stem cell like MSC which are used in different laboratories throughout the world, it is important to have a standardized, reproducible set of housekeeping genes for RT-qPCR analysis. In this study we demonstrate that EF1alpha, RPL13a and YWHAZ are suitable genes for the RT-qPCR analysis and comparison of several sources of human MSC during in vitro characterization and differentiation as well as in an ex vivo animal model of global cerebral ischemia. This will allow for the comparative RT-qPCR analysis of multiple MSC populations with the goal of future use in animal models of disease as well as tissue repair.


Asunto(s)
Células de la Médula Ósea/fisiología , Perfilación de la Expresión Génica/normas , Células Madre Mesenquimatosas/fisiología , Proteínas de Neoplasias/genética , Factor 1 de Elongación Peptídica/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/normas , Proteínas Ribosómicas/genética , Proteínas 14-3-3/genética , Animales , Isquemia Encefálica/genética , Factor de Crecimiento Epidérmico/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Humanos , Masculino , Chaperonas Moleculares/genética , Células-Madre Neurales/fisiología , Oxígeno/metabolismo , Ratas , Estándares de Referencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Adulto Joven
15.
Artículo en Inglés | MEDLINE | ID: mdl-32351045

RESUMEN

Nanomedicine and drug delivery technologies play a prominent role in modern medicine, facilitating better treatments than conventional drugs. Nanomedicine is being increasingly used to develop new methods of cancer diagnosis and treatment, since this technology can modulate the biodistribution and the target site accumulation of chemotherapeutic drugs, thereby reducing their toxicity. Regenerative medicine provides another area where innovative drug delivery technology is being studied for improved tissue regeneration. Drug delivery systems can protect therapeutic proteins and peptides against degradation in biological environments and deliver them in a controlled manner. Similarly, the combination of drug delivery systems and stem cells can improve their survival, differentiation, and engraftment. The present review summarizes the most important steps carried-out by the group of Prof Blanco-Prieto in nanomedicine and drug delivery technologies. Throughout her scientific career, she has contributed to the area of nanomedicine to improve anticancer therapy. In particular, nanoparticles loaded with edelfosine, doxorubicin, or methotrexate have demonstrated great anticancer activity in preclinical settings of lymphoma, glioma, and pediatric osteosarcoma. In regenerative medicine, a major focus has been the development of drug delivery systems for brain and cardiac repair. In this context, several microparticle-based technologies loaded with biologics have demonstrated efficacy in clinically relevant animal models such as monkeys and pigs. The latest research by this group has shown that drug delivery systems combined with cell therapy can achieve a more complete and potent regenerative response. Cutting-edge areas such as noninvasive intravenous delivery of cardioprotective nanomedicines or extracellular vesicle-based therapies are also being explored. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanomedicina , Nanoestructuras , Neoplasias/tratamiento farmacológico , Medicina Regenerativa , Animales , Humanos
16.
J Control Release ; 295: 201-213, 2019 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-30579984

RESUMEN

Current therapies for Parkinson's disease are symptomatic and unable to regenerate the brain tissue. In recent years, the therapeutic potential of a wide variety of neuroprotective and neuroregenerative molecules such as neurotrophic factors, antioxidants and RNA-based therapeutics has been explored. However, drug delivery to the brain is still a challenge and the therapeutic efficacy of many drugs is limited. In the last decade, micro- and nanoparticles have proved to be powerful tools for the administration of these molecules to the brain, enabling the development of new strategies against Parkinson's disease. The list of encapsulated drugs and the nature of the particles used is long, and numerous studies have been carried out supporting their efficacy in treating this pathology. This review aims to give an overview of the latest advances and emerging frontiers in micro- and nanomedical approaches for repairing dopaminergic neurons. Special emphasis will be placed on offering a new perspective to link these advances with the most relevant clinical trials and with the real possibility of transferring micro- and nanoformulations to industrial scale-up processes. This review is intended as a contribution towards facing the challenges that still exist in the clinical translation of micro- and nanotechnologies to administer therapeutic agents in Parkinson's disease.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanomedicina/métodos , Factores de Crecimiento Nervioso/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Antiparkinsonianos/administración & dosificación , Antiparkinsonianos/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Portadores de Fármacos/química , Terapia Genética/métodos , Humanos , Nanopartículas/química , Nanotecnología/métodos , Factores de Crecimiento Nervioso/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia
17.
Maturitas ; 110: 1-9, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29563026

RESUMEN

The capacity of the heart to heal after a myocardial infarction is not enough to restore normal cardiac function. Fortunately, delivery of therapeutics such as stem cells, growth factors, exosomes and small interfering ribonucleic acid (siRNA), among other bioactive molecules, has been shown to enhance heart repair and improve cardiac function. Furthermore, new delivery systems for these therapeutic agents have enhanced their regenerative and cardioprotective potential. In particular, nano- and microparticles (NPs and MPs) are promising. These systems may be administered directly in the infarcted myocardium or reach the heart after intravenous injection due to the enhanced permeability and retention effect or active targeting. Thus, NPs and MPs have made it possible to administer a wide range of potential drugs, including therapeutic molecules and/or stem cells, and evidence in favor of their use has been reported in several preclinical studies. Here, we review the studies done over the last 5 years using NPs and MPs loaded with therapeutics for repairing cardiac tissue after a myocardial infarction, and discuss some of the advances, challenges and future prospects in this field. In addition, we address the application of NPs and MPs for cardioprotective purposes.


Asunto(s)
Sistemas de Liberación de Medicamentos , Infarto del Miocardio/tratamiento farmacológico , Animales , Humanos , Nanopartículas/administración & dosificación , Nanopartículas/uso terapéutico
18.
Front Neuroanat ; 12: 113, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30618654

RESUMEN

When James Parkinson described the classical symptoms of the disease he could hardly foresee the evolution of our understanding over the next two hundred years. Nowadays, Parkinson's disease is considered a complex multifactorial disease in which genetic factors, either causative or susceptibility variants, unknown environmental cues, and the potential interaction of both could ultimately trigger the pathology. Noteworthy advances have been made in different fields from the clinical phenotype to the decoding of some potential neuropathological features, among which are the fields of genetics, drug discovery or biomaterials for drug delivery, which, though recent in origin, have evolved swiftly to become the basis of research into the disease today. In this review, we highlight some of the key advances in the field over the past two centuries and discuss the current challenges focusing on exciting new research developments likely to come in the next few years. Also, the importance of pre-motor symptoms and early diagnosis in the search for more effective therapeutic options is discussed.

20.
Int J Pharm ; 523(2): 454-475, 2017 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-27989830

RESUMEN

Heart failure still represents the leading cause of death worldwide. Novel strategies using stem cells and growth factors have been investigated for effective cardiac tissue regeneration and heart function recovery. However, some major challenges limit their translation to the clinic. Recently, biomaterials have emerged as a promising approach to improve delivery and viability of therapeutic cells and proteins for the regeneration of the damaged heart. In particular, hydrogels are considered one of the most promising vehicles. They can be administered through minimally invasive techniques while maintaining all the desirable characteristics of drug delivery systems. This review discusses recent advances made in the field of hydrogels for cardiac tissue regeneration in detail, focusing on the type of hydrogel (conventional, injectable, smart or nano- and micro-gel), the biomaterials used for its manufacture (natural, synthetic or hybrid) and the therapeutic agent encapsulated (stem cells or proteins). We expect that these novel hydrogel-based approaches will open up new possibilities in drug delivery and cell therapies.


Asunto(s)
Hidrogeles/química , Miocardio , Ingeniería de Tejidos , Corazón , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA