Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Eur J Pharm Biopharm ; 169: 268-279, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34748934

RESUMEN

Despite the curative approaches developed against myocardial infarction, cardiac cell death causes dysfunctional heart contractions that depend on the extent of the ischemic area and the reperfusion period. Cardiac regeneration may allow neovascularization and limit the ventricular remodeling caused by the scar tissue. We have previously found that large extracellular vesicles, carrying Sonic Hedgehog (lEVs), displayed proangiogenic and antioxidant properties, and decreased myocardial infarction size when administrated by intravenous injection. We propose to associate lEVs with pharmacology active microcarriers (PAMs) to obtain a combined cardioprotective and regenerative action when administrated by intracardiac injection. PAMs made of poly-D,L-lactic-coglycolic acid-poloxamer 188-poly-D,L-lactic-coglycolic acid and covered by fibronectin/poly-D-lysine provided a biodegradable and biocompatible 3D biomimetic support for the lEVs. When compared with lEVs alone, lEVs-PAMs constructs possessed an enhanced in vitro pro-angiogenic ability. PAMs were designed to continuously release encapsulated hepatocyte growth factor (PAMsHGF) and thus, locally increase the activity of the lEVs by the combined anti-fibrotic properties and regenerative properties. Intracardiac administration of either lEVs alone or lEVs-PAMsHGF improved cardiac function in a similar manner, in a rat model of ischemia-reperfusion. Moreover, lEVs alone or the IEVs-PAMsHGF induced arteriogenesis, but only the latter reduced tissue fibrosis. Taken together, these results highlight a promising approach for lEVs-PAMsHGF in regenerative medicine for myocardial infarction.


Asunto(s)
Portadores de Fármacos/farmacología , Factor de Crecimiento de Hepatocito , Infarto del Miocardio/tratamiento farmacológico , Poloxámero/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología , Regeneración , Animales , Antioxidantes/farmacología , Biomimética/métodos , Cardiotónicos/farmacología , Excipientes/farmacología , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Microesferas , Miocardio/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Regeneración/efectos de los fármacos , Regeneración/fisiología
2.
Mater Sci Eng C Mater Biol Appl ; 121: 111852, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33579486

RESUMEN

Polymeric, biodegradable, microspheres (MS) presenting a biomimetic surface of extracellular matrix (ECM) proteins are currently used for transporting cells and/or encapsulated proteins for regenerative medicine studies. They can be made of (lactic-co-glycolic acid) (PLGA) or of a more hydrophilic PLGA-P188 (Poloxamer188)-PLGA polymer allowing for the complete release of the therapeutic proteins. They promote stem cell adhesion, cell survival and differentiation after transplantation. Although the biological effectiveness of these microcarriers is established, a detailed understanding of the protein and cell interactions with the microcarrier surface remain unclear due to a lack of information of their surface properties. The aim of this study was to characterize the physicochemical properties of two polymeric MS systems and determine the effect of laminin and poly-d-lysine coated microcarriers on stem cell adhesion, survival and neuronal differentiation. The hydrophobicity and topography of PLGA MS promoted protein adsorption and the stem cells quickly adhered and spread on the surface of these microcarriers. In contrast less proteins adsorbed onto PLGA-P188-PLGA MS and although cells adhered to these microcarriers, they remained round and did not spread on their surface. Despite these early-stage differences, our results suggest that the nature of the MS does not strongly influence the long-term cell behavior. The cells exhibit the same cell number, differentiation profile and ability to secrete ECM molecules regardless of the type of microcarrier used. Likely the ECM molecules that form a microenvironment around both of these 3D microcarrier/cell constructs over time play a role in this converging cell behavior. We have thus furthered our understanding of the physicochemical properties of polymeric cell carriers affecting stem cell behavior to help tailor suitable microcarriers for neuroregenerative applications.


Asunto(s)
Células Madre Mesenquimatosas , Adhesión Celular , Moléculas de Adhesión Celular , Células Cultivadas , Ácido Láctico , Microesferas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
3.
Pharmaceutics ; 12(12)2020 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-33317141

RESUMEN

Clinical studies have demonstrated the regenerative potential of stem cells for cardiac repair over the past decades, but their widespread use is limited by the poor tissue integration and survival obtained. Natural or synthetic hydrogels or microcarriers, used as cell carriers, contribute to resolving, in part, the problems encountered by providing mechanical support for the cells allowing cell retention, survival and tissue integration. Moreover, hydrogels alone also possess mechanical protective properties for the ischemic heart. The combined effect of growth factors with cells and an appropriate scaffold allow a therapeutic effect on myocardial repair. Despite this, the effects obtained with cell therapy remain limited and seem to be equivalent to the effects obtained with extracellular vesicles, key actors in intercellular communication. Extracellular vesicles have cardioprotective effects which, when combined proangiogenic properties with antiapoptotic and anti-inflammatory actions, make it possible to act on all the damages caused by ischemia. The evolution of biomaterial engineering allows us to envisage their association with new major players in cardiac therapy, extracellular vesicles, in order to limit undesirable effects and to envisage a transfer to the clinic. This new therapeutic approach could be associated with the release of growth factors to potentialized the beneficial effect obtained.

4.
Biointerphases ; 15(4): 041008, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32752604

RESUMEN

Protein-coated polymer-based microparticles are attractive supports for cell delivery, but the interplay between microparticle properties, protein coating, and cell response is poorly understood. The interest in alternative microparticle formulations increases the need for a better understanding of how functional protein coatings form on different microparticles. In this work, microparticle formulations based on biodegradable polymers [poly (lactic-co-glycolic acid) (PLGA) and the triblock copolymer PLGA-poloxamer-PLGA] were prepared via an emulsion-based process. To explore the impact that the use of a surfactant has on the properties of the microparticles, the emulsion was stabilized by using either a surfactant, poly(vinyl alcohol), or an organic solvent, propylene glycol. Four different types of microparticles were prepared through combinations of the two types of polymers and the two types of stabilizers. The coating of microparticles with proteins/polypeptides such as fibronectin and poly-d-lysine has been demonstrated before and is an integral step for their application as microcarriers, e.g., for cell delivery; however, the impact of the microparticles' surface chemical properties on the formation (prevalence and distribution) of the mixed polypeptide coatings and the influence on subsequent cell attachment remain to be elucidated. Using a colocalization analysis approach on ToF-SIMS images of protein-coated microparticles, we show that the use of propyleneglycol over PVA as well as the substitution of PLGA by the triblock copolymer resulted in enhanced protein adsorption. Furthermore, if propyleneglycol is used, the substitution of PLGA with the triblock copolymer leads to increased stem cell adhesion.


Asunto(s)
Fibronectinas/química , Polilisina/química , Polímeros/química , Adhesión Celular/efectos de los fármacos , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Proliferación Celular/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Polímeros/farmacología , Alcohol Polivinílico/química , Propiedades de Superficie
5.
Int J Pharm ; 587: 119685, 2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32712253

RESUMEN

There is no treatment for spinal cord injury (SCI) that fully repairs the damages. One strategy is to inject mesenchymal stem cells around the lesion to benefit from their immunomodulatory properties and neuroprotective effect. Our hypothesis was that the combination of dental stem cells from the apical papilla (SCAP) with pharmacologically active microcarriers (PAMs) releasing brain-derived neurotrophic factor (BDNF) would improve rat locomotor function by immunomodulation and neuroprotection. BDNF-PAMs were prepared by solid/oil/water emulsion of poly(L-lactide-co-glycolide) and nanoprecipitated BDNF and subsequent coating with fibronectin. SCAP were then seeded on BDNF-PAMs. SCAP expression of neuronal and immunomodulatory factors was evaluated in vitro. SCAP BDNF-PAMs were injected in a rat spinal cord contusion model and their locomotor function was evaluated by Basso, Beattie, and Bresnahan (BBB) scoring. Impact on inflammation and neuroprotection/axonal growth was evaluated by immunofluorescence. Culture on PAMs induced the overexpression of immunomodulatory molecules and neural/neuronal markers. Injection of SCAP BDNF-PAMs at the lesion site improved rat BBB scoring, reduced the expression of inducible nitric oxide synthase and increased the expression of ßIII tubulin, GAP43, and 5-HT. These results confirm the suitability and versatility of PAMs as combined drug and cell delivery system for regenerative medicine applications but also that BDNF-PAMs potentialize the very promising therapeutic potential of SCAP in the scope of SCI.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Células Madre Mesenquimatosas , Fármacos Neuroprotectores , Traumatismos de la Médula Espinal , Animales , Humanos , Neuronas , Ratas , Médula Espinal , Traumatismos de la Médula Espinal/tratamiento farmacológico
6.
J Neuroinflammation ; 17(1): 76, 2020 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-32127025

RESUMEN

BACKGROUND: Tick-borne encephalitis virus (TBEV) is a member of the Flaviviridae family, Flavivirus genus, which includes several important human pathogens. It is responsible for neurological symptoms that may cause permanent disability or death, and, from a medical point of view, is the major arbovirus in Central/Northern Europe and North-Eastern Asia. TBEV tropism is critical for neuropathogenesis, yet little is known about the molecular mechanisms that govern the susceptibility of human brain cells to the virus. In this study, we sought to establish and characterize a new in vitro model of TBEV infection in the human brain and to decipher cell type-specific innate immunity and its relation to TBEV tropism and neuropathogenesis. METHOD: Human neuronal/glial cells were differentiated from neural progenitor cells and infected with the TBEV-Hypr strain. Kinetics of infection, cellular tropism, and cellular responses, including innate immune responses, were characterized by measuring viral genome and viral titer, performing immunofluorescence, enumerating the different cellular types, and determining their rate of infection and by performing PCR array and qRT-PCR. The specific response of neurons and astrocytes was analyzed using the same approaches after enrichment of the neuronal/glial cultures for each cellular subtype. RESULTS: We showed that infection of human neuronal/glial cells mimicked three major hallmarks of TBEV infection in the human brain, namely, preferential neuronal tropism, neuronal death, and astrogliosis. We further showed that these cells conserved their capacity to mount an antiviral response against TBEV. TBEV-infected neuronal/glial cells, therefore, represented a highly relevant pathological model. By enriching the cultures for either neurons or astrocytes, we further demonstrated qualitative and quantitative differential innate immune responses in the two cell types that correlated with their particular susceptibility to TBEV. CONCLUSION: Our results thus reveal that cell type-specific innate immunity is likely to contribute to shaping TBEV tropism for human brain cells. They describe a new in vitro model for in-depth study of TBEV-induced neuropathogenesis and improve our understanding of the mechanisms by which neurotropic viruses target and damage human brain cells.


Asunto(s)
Astrocitos/inmunología , Astrocitos/virología , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/virología , Neuronas/inmunología , Neuronas/virología , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Susceptibilidad a Enfermedades , Virus de la Encefalitis Transmitidos por Garrapatas/fisiología , Humanos , Inmunidad Innata , Tropismo Viral
7.
Pharmaceutics ; 11(10)2019 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-31614758

RESUMEN

For Huntington's disease (HD) cell-based therapy, the transplanted cells are required to be committed to a neuronal cell lineage, survive and maintain this phenotype to ensure their safe transplantation in the brain. We first investigated the role of RE-1 silencing transcription factor (REST) inhibition using siRNA in the GABAergic differentiation of marrow-isolated adult multilineage inducible (MIAMI) cells, a subpopulation of MSCs. We further combined these cells to laminin-coated poly(lactic-co-glycolic acid) PLGA pharmacologically active microcarriers (PAMs) delivering BDNF in a controlled fashion to stimulate the survival and maintain the differentiation of the cells. The PAMs/cells complexes were then transplanted in an ex vivo model of HD. Using Sonic Hedgehog (SHH) and siREST, we obtained GABAergic progenitors/neuronal-like cells, which were able to secrete HGF, SDF1 VEGFa and BDNF, of importance for HD. GABA-like progenitors adhered to PAMs increased their mRNA expression of NGF/VEGFa as well as their secretion of PIGF-1, which can enhance reparative angiogenesis. In our ex vivo model of HD, they were successfully transplanted while attached to PAMs and were able to survive and maintain this GABAergic neuronal phenotype. Together, our results may pave the way for future research that could improve the success of cell-based therapy for HDs.

8.
Emerg Microbes Infect ; 8(1): 1003-1016, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31282298

RESUMEN

Zika virus (ZIKV) is a mosquito-borne Flavivirus that causes Zika disease with particular neurological complications, including Guillain-Barré Syndrome and congenital microcephaly. Although ZIKV has been shown to directly infect human neural progenitor cells (hNPCs), thereby decreasing their viability and growth, it is as yet unknown which of the cellular pathways involved in the disruption of neurogenesis are affected following ZIKV infection. By comparing the effect of two ZIKV strains in vitro on hNPCs, the differentiation process of the latter cells was found to lead to a decreased susceptibility to infection and cell death induced by each of the ZIKV strains, which was associated with an earlier and stronger antiviral innate immune response in infected, differentiated hNPCs, as compared to undifferentiated cells. Moreover, ZIKV modulated, both in hNPCs and in vivo in fetal brain in an experimental mouse model, the expression of the Notch pathway which is involved in cellular proliferation, apoptosis and differentiation during neurogenesis. These results show that the differentiation state of hNPCs is a significant factor contributing to the outcome of ZIKV infection and furthermore suggest that ZIKV infection might initiate early activation of the Notch pathway resulting in an abnormal differentiation process, implicated in ZIKV-induced brain injury.


Asunto(s)
Células-Madre Neurales/virología , Neurogénesis , Receptor Notch1/metabolismo , Infección por el Virus Zika/virología , Virus Zika/fisiología , Animales , Apoptosis , Femenino , Humanos , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Receptor Notch1/genética , Transducción de Señal , Virus Zika/genética , Infección por el Virus Zika/genética , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/fisiopatología
9.
Acta Biomater ; 84: 268-279, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30465922

RESUMEN

Glioblastoma (GB) is a highly infiltrative tumor, recurring, in 90% of cases, within a few centimeters of the surgical resection cavity, even with adjuvant chemo/radiotherapy. Residual GB cells left in the margins or infiltrating the brain parenchyma shelter behind the extremely fragile and sensitive brain tissue and may favor recurrence. Tools for eliminating these cells without damaging the brain microenvironment are urgently required. We propose a strategy involving the implantation, into the tumor bed after resection, of a scaffold to concentrate and trap these cells, to facilitate their destruction by targeted therapies, such as stereotactic radiosurgery. We used bacterial cellulose (BC), an easily synthesized and modifiable random nanofibrous biomaterial, to make the trap. We showed that the structure of BC membranes was ideal for trapping tumor cells and that BC implants were biocompatible with brain parenchyma. We also demonstrated the visibility of BC on magnetic resonance imaging, making it possible to follow its fate in clinical situations and to define the target volume for stereotactic radiosurgery more precisely. Furthermore, BC membranes can be loaded with chemoattractants, which were released and attracted tumor cells in vitro. This is of particular interest for trapping GB cells infiltrating tissues within a few centimeters of the resection cavity. Our data suggest that BC membranes could be a scaffold of choice for implantation after surgical resection to trap residual GB cells. STATEMENT OF SIGNIFICANCE: Glioblastoma is a highly infiltrative tumor, recurring, in 90% of cases, within a few centimeters of the surgical resection cavity, even with adjuvant chemo/radiotherapy. Residual tumor cells left in the margins or infiltrating the brain parenchyma shelter behind the extremely fragile and sensitive brain tissue and contribute to the risk of recurrence. Finding tools to eliminate these cells without damaging the brain microenvironment is a real challenge. We propose a strategy involving the implantation, into the walls of the surgical resection cavity, of a scaffold to concentrate and trap the residual tumor cells, to facilitate their destruction by targeted therapies, such as stereotactic radiosurgery.


Asunto(s)
Materiales Biocompatibles , Neoplasias Encefálicas , Glioblastoma , Imagen por Resonancia Magnética , Membranas Artificiales , Nanofibras , Radiocirugia , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/uso terapéutico , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Celulosa/química , Celulosa/uso terapéutico , Glioblastoma/diagnóstico por imagen , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Humanos , Masculino , Nanofibras/química , Nanofibras/uso terapéutico , Ratas , Ratas Sprague-Dawley , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de la radiación
10.
J Tissue Eng Regen Med ; 12(6): 1363-1373, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29656608

RESUMEN

We propose a regenerative solution in the treatment of critical limb ischaemia (CLI). Poly-lactic/glycolic acid microcarriers were prepared and coated with laminin to be sterilized through γ-irradiation of 25 kGy at low temperature. Stromal vascular fraction (SVF) cells were extracted through enzymatic digestion of adipose tissue. Streptozotocin-induced diabetic mice underwent arteriotomy and received an administration of SVF cells combined or not with biomimetic microcarriers. Functional evaluation of the ischaemic limb was then reported, and tissue reperfusion was evaluated through fluorescence molecular tomography. Microcarriers were stable and functional after γ-irradiation until at least 12 months of storage. Mice that received an injection of SVF cells in the ischaemic limb have 22% of supplementary blood supply within this limb 7 days after surgery compared with vehicle, whereas no difference was observed at Day 14. With the combined therapy, the improvement of blood flow is significantly higher compared with vehicle, of about 31% at Day 7 and of about 11% at Day 14. Injection of SVF cells induces a significant 27% decrease of necrosis compared with vehicle. This effect is more important when SVF cells were mixed with biomimetic microcarriers: -37% compared with control. Although SVF cells injection leads to a non-significant 22% proprioception recovery, the combined therapy induces a significant recovery of about 27% compared with vehicle. We show that the combination of SVF cells from adipose tissue with laminin-coated poly-lactic/glycolic acid microcarriers is efficient for critical limb ischaemia therapy in a diabetic mouse model.


Asunto(s)
Tejido Adiposo/citología , Materiales Biomiméticos/farmacología , Isquemia/terapia , Microesferas , Neovascularización Fisiológica/efectos de los fármacos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología , Adulto , Animales , Adhesión Celular , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/patología , Laminina/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Peso Molecular , Necrosis , Tamaño de la Partícula , Perfusión , Propiocepción , Flujo Sanguíneo Regional , Electricidad Estática , Células del Estroma/efectos de los fármacos
11.
J Exp Clin Cancer Res ; 36(1): 135, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28962658

RESUMEN

BACKGROUND: Glioblastoma (GB) is the most malignant brain tumor in adults. It is characterized by angiogenesis and a high proliferative and invasive capacity. Standard therapy (surgery, radiotherapy and chemotherapy with temozolomide) is of limited efficacy. Innovative anticancer drugs targeting both tumor cells and angiogenesis are urgently required, together with effective systems for their delivery to the brain. We assessed the ability of human mesenchymal stromal cells (MSCs) to uptake the multikinase inhibitor, sorafenib (SFN), and to carry this drug to a brain tumor following intranasal administration. METHOD: MSCs were primed with SFN and drug content and release were quantified by analytical chemistry techniques. The ability of SFN-primed MSCs to inhibit the survival of the human U87MG GB cell line and endothelial cells was assessed in in vitro assays. These cells were then administered intranasally to nude mice bearing intracerebral U87MG xenografts. Their effect on tumor growth and angiogenesis was evaluated by magnetic resonance imaging and immunofluorescence analyses, and was compared with the intranasal administration of unprimed MSCs or SFN alone. RESULTS: MSCs took up about 9 pg SFN per cell, with no effect on viability, and were able to release 60% of the primed drug. The cytostatic activity of the released SFN was entirely conserved, resulting in a significant inhibition of U87MG and endothelial cell survival in vitro. Two intranasal administrations of SFN-primed MSCs in U87MG-bearing mice resulted in lower levels of tumor angiogenesis than the injection of unprimed MSCs or SFN alone, but had no effect on tumor volume. We also observed an increase in the proportion of small intratumoral vessels in animals treated with unprimed MSCs; this effect being abolished if the MSCs were primed with SFN. CONCLUSION: We show the potential of MSCs to carry SFN to brain tumors following an intranasal administration. However, the therapeutic effect is modest probably due to the pro-tumorigenic properties of MSCs, which may limit the action of the released SFN. This calls into question the suitability of MSCs for use in GB therapy and renders it necessary to find methods guaranteeing the safety of this cellular vector after drug delivery.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Administración Intranasal , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Humanos , Células Madre Mesenquimatosas/química , Ratones , Ratones Desnudos , Niacinamida/farmacología , Sorafenib , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Acta Biomater ; 49: 167-180, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27865962

RESUMEN

Stem cells combined with biodegradable injectable scaffolds releasing growth factors hold great promises in regenerative medicine, particularly in the treatment of neurological disorders. We here integrated human marrow-isolated adult multilineage-inducible (MIAMI) stem cells and pharmacologically active microcarriers (PAMs) into an injectable non-toxic silanized-hydroxypropyl methylcellulose (Si-HPMC) hydrogel. The goal is to obtain an injectable non-toxic cell and growth factor delivery device. It should direct the survival and/or neuronal differentiation of the grafted cells, to safely transplant them in the central nervous system, and enhance their tissue repair properties. A model protein was used to optimize the nanoprecipitation conditions of the neuroprotective brain-derived neurotrophic factor (BDNF). BDNF nanoprecipitate was encapsulated in fibronectin-coated (FN) PAMs and the in vitro release profile evaluated. It showed a prolonged, bi-phasic, release of bioactive BDNF, without burst effect. We demonstrated that PAMs and the Si-HPMC hydrogel increased the expression of neural/neuronal differentiation markers of MIAMI cells after 1week. Moreover, the 3D environment (PAMs or hydrogel) increased MIAMI cells secretion of growth factors (b-NGF, SCF, HGF, LIF, PlGF-1, SDF-1α, VEGF-A & D) and chemokines (MIP-1α & ß, RANTES, IL-8). These results show that PAMs delivering BDNF combined with Si-HPMC hydrogel represent a useful novel local delivery tool in the context of neurological disorders. It not only provides neuroprotective BDNF but also bone marrow-derived stem cells that benefit from that environment by displaying neural commitment and an improved neuroprotective/reparative secretome. It provides preliminary evidence of a promising pro-angiogenic, neuroprotective and axonal growth-promoting device for the nervous system. STATEMENT OF SIGNIFICANCE: Combinatorial tissue engineering strategies for the central nervous system are scarce. We developed and characterized a novel injectable non-toxic stem cell and protein delivery system providing regenerative cues for central nervous system disorders. BDNF, a neurotrophic factor with a wide-range effect, was nanoprecipitated to maintain its structure and released in a sustained manner from novel polymeric microcarriers. The combinatorial 3D support, provided by fibronectin-microcarriers and the hydrogel, to the mesenchymal stem cells guided the cells towards a neuronal differentiation and enhanced their tissue repair properties by promoting growth factors and cytokine secretion. The long-term release of physiological doses of bioactive BDNF, combined to the enhanced secretion of tissue repair factors from the stem cells, constitute a promising therapeutic approach.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Diferenciación Celular/efectos de los fármacos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Células Madre Mesenquimatosas/citología , Microesferas , Neuronas/citología , Proteoma/metabolismo , Anciano , Materiales Biocompatibles/farmacología , Forma de la Célula/efectos de los fármacos , Precipitación Química , Liberación de Fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Derivados de la Hipromelosa/química , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/ultraestructura , Nanopartículas/química , Neuronas/efectos de los fármacos , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reología , Silanos/química
13.
Biomaterials ; 88: 60-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26945456

RESUMEN

In the present study, we aimed at evaluating the ability of novel PLGA-P188-PLGA-based microspheres to induce the differentiation of mesenchymal stem/stromal cells (MSC) into chondrocytes. To this aim, we tested microspheres releasing TGFß3 (PAM-T) in vitro and in situ, in a pathological osteoarthritic (OA) environment. We first evaluated the chondrogenic differentiation of human MSCs seeded onto PAM-T in vitro and confirmed the up-regulation of chondrogenic markers while the secretome of the cells was not changed by the 3D environment. We then injected human MSC seeded onto PAM-T in the knee joints of mice with collagenase-induced OA. After 6 weeks, histological analysis revealed that formation of a cartilage-like tissue occurred at the vicinity of PAM-T that was not observed when MSCs were seeded onto PAM. We also noticed that the endogenous articular cartilage was less degraded. The extent of cartilage protection was further analysed by confocal laser microscopy. When MSCs seeded onto PAM-T were injected early after OA induction, protection of cartilage against degradation was evidenced and this effect was associated to a higher survival of MSCs in presence of TGFß3. This study points to the interest of using MSCs seeded onto PAM for cartilage repair and stimulation of endogenous cartilage regeneration.


Asunto(s)
Condrogénesis/efectos de los fármacos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoartritis/terapia , Andamios del Tejido/química , Factor de Crecimiento Transformador beta3/administración & dosificación , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Células Cultivadas , Portadores de Fármacos/química , Humanos , Articulación de la Rodilla/efectos de los fármacos , Articulación de la Rodilla/patología , Ácido Láctico/química , Células Madre Mesenquimatosas/citología , Ratones , Ratones SCID , Microesferas , Osteoartritis/patología , Poloxámero/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Factor de Crecimiento Transformador beta3/farmacología , Factor de Crecimiento Transformador beta3/uso terapéutico
14.
Biomaterials ; 83: 347-62, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26802487

RESUMEN

The potential treatments for neurodegenerative disorders will be revolutionized by the transplantation of stem cells or neuronal progenitors derived from these cells. It is however crucial to better monitor their proliferation, improve their survival and differentiation and hence ameliorate their engraftment after transplantation. To direct stem cell fate, a delicate control of gene expression through RNA interference (RNAi) is emerging as a safe epigenetic approach. The development of novel biomaterials (nano and microcarriers) capable of delivering proteins, nucleic acids and cells, open the possibility to regulate cell fate while achieving neuroprotection and neurorepair and could be applied to Huntington's disease. This review first provides an overview of stem cell therapy for the neurodegenerative disorder Huntington's disease. Within that context, an integrative discussion follows of the control of stem cell behaviour by RNAi delivered by different nanocarriers in vitro prior to their transplantation. Finally, combined in vivo strategies using stem cells, biomaterials and epigenetic cell regulation are reported.


Asunto(s)
Portadores de Fármacos/química , Enfermedad de Huntington/terapia , Nanopartículas/química , Degeneración Nerviosa/terapia , Medicina Regenerativa/métodos , Células Madre/citología , Humanos , Trasplante de Células Madre , Células Madre/metabolismo
15.
Methods Mol Biol ; 1340: 171-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26445838

RESUMEN

In recent years, cell-based therapies using adult stem cells have attracted considerable interest in regenerative medicine. A tissue-engineered construct for cartilage repair should provide a support for the cell and allow sustained in situ delivery of bioactive factors capable of inducing cell differentiation into chondrocytes. Pharmacologically active microcarriers (PAMs), made of biodegradable and biocompatible poly (D,L-lactide-co-glycolide acid) (PLGA), are a unique system which combines these properties in an adaptable and simple microdevice. This device relies on nanoprecipitation of proteins encapsulated in polymeric microspheres with a solid in oil in water emulsion-solvent evaporation process, and their subsequent coating with extracellular matrix protein molecules. Here, we describe their preparation process, and some of their characterization methods for an application in cartilage tissue engineering.


Asunto(s)
Cartílago/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Células Madre Mesenquimatosas/efectos de los fármacos , Polímeros/química , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Cartílago/citología , Cartílago/metabolismo , Cartílago/trasplante , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Química Farmacéutica , Condrocitos/metabolismo , Condrocitos/trasplante , Preparaciones de Acción Retardada , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Nanomedicina , Regeneración/efectos de los fármacos , Factores de Tiempo , Factor de Crecimiento Transformador beta3/administración & dosificación , Factor de Crecimiento Transformador beta3/química
16.
Cardiovasc Res ; 108(1): 39-49, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26187727

RESUMEN

RATIONALE: Engraftment and survival of transplanted stem or stromal cells in the microenvironment of host tissues may be improved by combining such cells with scaffolds to delay apoptosis and enhance regenerative properties. AIMS: We examined whether poly(lactic-co-glycolic acid) pharmacologically active microcarriers (PAMs) releasing vascular endothelial growth factor (VEGF) enhance survival, differentiation, and angiogenesis of adipose tissue-mesenchymal stromal cells (AT-MSCs). We analysed the efficacy of transplanted AT-MSCs conjugated with PAMs in a murine model of acute myocardial infarction (AMI). METHODS AND RESULTS: We used fibronectin-coated (empty) PAMs or VEGF-releasing PAMs covered with murine AT-MSCs. Twelve-month-old C57 mice underwent coronary artery ligation to induce AMI, and were randomized into five treatment groups: AMI control (saline 20 µL, n = 7), AMI followed by intramyocardial injection with AT-MSCs (2.5 × 10(5) cells/20 µL, n = 5), or concentrated medium (CM) from AT-MSCs (20 µL, n = 8), or AT-MSCs (2.5 × 10(5) cells/20 µL) conjugated with empty PAMs (n = 7), or VEGF-releasing PAMs (n = 8). Sham-operated mice (n = 7) were used as controls. VEGF-releasing PAMs increased proliferation and angiogenic potential of AT-MSCs, but did not impact their osteogenic or adipogenic differentiation. AT-MSCs conjugated with VEGF-releasing PAMs inhibited apoptosis, decreased fibrosis, increased arteriogenesis and the number of cardiac-resident Ki-67 positive cells, and improved myocardial fractional shortening compared with AT-MSCs alone when transplanted into the infarcted hearts of C57 mice. With the exception of fractional shortening, all such effects of AT-MSCs conjugated with VEGF-PAMs were paralleled by the injection of CM. CONCLUSIONS: AT-MSCs conjugated with VEGF-releasing PAMs exert paracrine effects that may have therapeutic applications.


Asunto(s)
Tejido Adiposo/citología , Trasplante de Células Madre Mesenquimatosas , Infarto del Miocardio/terapia , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Microesferas
17.
Stem Cells Transl Med ; 4(6): 670-84, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25925835

RESUMEN

UNLABELLED: Stem cell-based regenerative therapies hold great potential for the treatment of degenerative disorders such as Parkinson's disease (PD). We recently reported the repair and functional recovery after treatment with human marrow-isolated adult multilineage inducible (MIAMI) cells adhered to neurotrophin-3 (NT3) releasing pharmacologically active microcarriers (PAMs) in hemiparkinsonian rats. In order to comprehend this effect, the goal of the present work was to elucidate the survival, differentiation, and neuroprotective mechanisms of MIAMI cells and human neural stem cells (NSCs), both adhering to NT3-releasing PAMs in an ex vivo organotypic model of nigrostriatal degeneration made from brain sagittal slices. It was shown that PAMs led to a marked increase in MIAMI cell survival and neuronal differentiation when releasing NT3. A significant neuroprotective effect of MIAMI cells adhering to PAMs was also demonstrated. NSCs barely had a neuroprotective effect and differentiated mostly into dopaminergic neuronal cells when adhering to PAM-NT3. Moreover, those cells were able to release dopamine in a sufficient amount to induce a return to baseline levels. Reverse transcription-quantitative polymerase chain reaction and enzyme-linked immunosorbent assay analyses identified vascular endothelial growth factor (VEGF) and stanniocalcin-1 as potential mediators of the neuroprotective effect of MIAMI cells and NSCs, respectively. It was also shown that VEGF locally stimulated tissue vascularization, which might improve graft survival, without excluding a direct neuroprotective effect of VEGF on dopaminergic neurons. These results indicate a prospective interest of human NSC/PAM and MIAMI cell/PAM complexes in tissue engineering for PD. SIGNIFICANCE: Stem cell-based regenerative therapies hold great potential for the treatment of degenerative disorders such as Parkinson's disease (PD). The present work elucidates and compares the survival, differentiation, and neuroprotective mechanisms of marrow-isolated adult multilineage inducible cells and human neural stem cells both adhered to neurotrophin-3-releasing pharmacologically active microcarriers in an ex vivo organotypic model of PD made from brain sagittal slices.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Portadores de Fármacos/farmacología , Células-Madre Neurales/trasplante , Neurotrofina 3/farmacología , Trastornos Parkinsonianos/terapia , Trasplante de Células Madre , Adulto , Animales , Supervivencia Celular/efectos de los fármacos , Células Inmovilizadas/metabolismo , Células Inmovilizadas/patología , Células Inmovilizadas/trasplante , Preparaciones de Acción Retardada/farmacología , Modelos Animales de Enfermedad , Humanos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Ratas
18.
J Biomed Mater Res A ; 103(9): 3012-25, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25727843

RESUMEN

One of the main cause of ineffective cell therapy in repairing the damaged heart is the poor yield of grafted cells. To overcome this drawback, rats with 4-week-old myocardial infarction (MI) were injected in the border zone with human adipose-derived stem cells (ADSCs) conveyed by poly(lactic-co-glycolic acid) microcarriers (PAMs) releasing hepatocyte growth factor (HGF) and insulin-like growth factor-1 (IGF-1) (GFsPAMs). According to treatments, animals were subdivided into different groups: MI_ADSC, MI_ADSC/PAM, MI_GFsPAM, MI_ADSC/GFsPAM, and untreated MI_V. Two weeks after injection, a 31% increase in ADSC engraftment was observed in MI_ADSC/PAM compared with MI_ADSC (p < 0.05). A further ADSC retention was obtained in MI_ADSC/GFsPAM with respect to MI_ADSC (106%, p < 0.05) and MI_ADSC/PAM (57%, p < 0.05). A 130% higher density of blood vessels of medium size was present in MI_ADSC/GFsPAM compared with MI_ADSC (p < 0.01). MI_ADSC/GFsPAM also improved, albeit slightly, left ventricular remodeling and hemodynamics with respect to the other groups. Notably, ADSCs and/or PAMs, with or without HGF/IGF-1, trended to induce arrhythmias in electrically driven, Langendorff-perfused, hearts of all groups. Thus, PAMs releasing HGF/IGF-1 markedly increase ADSC engraftment 2 weeks after injection and stimulate healing in chronically infarcted myocardium, but attention should be paid to potentially negative electrophysiological consequences.


Asunto(s)
Factor de Crecimiento de Hepatocito/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/terapia , Trasplante de Células Madre/métodos , Tejido Adiposo/citología , Animales , Arritmias Cardíacas/etiología , Materiales Biomiméticos/química , Modelos Animales de Enfermedad , Portadores de Fármacos/administración & dosificación , Humanos , Ácido Láctico , Masculino , Ensayo de Materiales , Microesferas , Infarto del Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Ratas Wistar , Trasplante de Células Madre/efectos adversos , Remodelación Ventricular , Cicatrización de Heridas/efectos de los fármacos
19.
Int J Nanomedicine ; 10: 1259-71, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25709447

RESUMEN

Recently developed drug delivery nanosystems, such as lipid nanocapsules (LNCs), hold great promise for the treatment of glioblastomas (GBs). In this study, we used a subpopulation of human mesenchymal stem cells, "marrow-isolated adult multilineage inducible" (MIAMI) cells, which have endogenous tumor-homing activity, to deliver LNCs containing an organometallic complex (ferrociphenol or Fc-diOH), in the orthotopic U87MG GB model. We determined the optimal dose of Fc-diOH-LNCs that can be carried by MIAMI cells and compared the efficacy of Fc-diOH-LNC-loaded MIAMI cells with that of the free-standing Fc-diOH-LNC system. We showed that MIAMI cells entrapped an optimal dose of about 20 pg Fc-diOH per cell, with no effect on cell viability or migration capacity. The survival of U87MG-bearing mice was longer after the intratumoral injection of Fc-diOH-LNC-loaded MIAMI cells than after the injection of Fc-diOH-LNCs alone. The greater effect of the Fc-diOH-LNC-loaded MIAMI cells may be accounted for by their peritumoral distribution and a longer residence time of the drug within the tumor. These results confirm the potential of combinations of stem cell therapy and nanotechnology to improve the local tissue distribution of anticancer drugs in GB.


Asunto(s)
Antineoplásicos , Compuestos Ferrosos , Glioblastoma/terapia , Lípidos , Trasplante de Células Madre Mesenquimatosas , Nanocápsulas , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Compuestos Ferrosos/administración & dosificación , Compuestos Ferrosos/química , Compuestos Ferrosos/uso terapéutico , Compuestos Ferrosos/toxicidad , Humanos , Lípidos/administración & dosificación , Lípidos/química , Lípidos/uso terapéutico , Lípidos/toxicidad , Ratones , Nanocápsulas/administración & dosificación , Nanocápsulas/química , Nanocápsulas/uso terapéutico , Nanocápsulas/toxicidad , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Acta Biomater ; 15: 77-88, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25556361

RESUMEN

Few effective therapeutic interventions are available to limit brain damage and functional deficits after ischaemic stroke. Within this context, mesenchymal stem cell (MSC) therapy carries minimal risks while remaining efficacious through the secretion of trophic, protective, neurogenic and angiogenic factors. The limited survival rate of MSCs restricts their beneficial effects. The usefulness of a three-dimensional support, such as a pharmacologically active microcarrier (PAM), on the survival of MSCs during hypoxia has been shown in vitro, especially when the PAMs were loaded with vascular endothelial growth factor (VEGF). In the present study, the effect of MSCs attached to laminin-PAMs (LM-PAMs), releasing VEGF or not, was evaluated in vivo in a model of transient stroke. The parameters assessed were infarct volume, functional recovery and endogenous cellular reactions. LM-PAMs induced the expression of neuronal markers by MSCs both in vitro and in vivo. Moreover, the prolonged release of VEGF increased angiogenesis around the site of implantation of the LM-PAMs and facilitated the migration of immature neurons towards the ischaemic tissue. Nonetheless, MSCs/LM-PAMs-VEGF failed to improve sensorimotor functions. The use of LM-PAMs to convey MSCs and to deliver growth factors could be an effective strategy to repair the brain damage caused by a stroke.


Asunto(s)
Isquemia Encefálica/complicaciones , Portadores de Fármacos/química , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/etiología , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Conducta Animal , Vasos Sanguíneos/efectos de los fármacos , Isquemia Encefálica/fisiopatología , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Laminina/farmacología , Imagen por Resonancia Magnética , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/fisiopatología , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...