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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS One ; 18(1): e0278844, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36701370

RESUMEN

Leiomyosarcoma (LMS) is an aggressive, often poorly differentiated cancer of the smooth muscle (SM) lineage for which the molecular drivers of transformation and progression are poorly understood. In microRNA (miRNA) profiling studies, miR-130b was previously found to be upregulated in LMS vs. normal SM, and down-regulated during the differentiation of mesenchymal stem cells (MSCs) into SM, suggesting a role in LMS tumor progression. In the present study, the effects of miR-130b on human LMS tumorigenesis were investigated. Stable miR-130b overexpression enhanced invasion of LMS cells in vitro, and led to the formation of undifferentiated, pleomorphic tumors in vivo, with increased growth and metastatic potential compared to control LMS cells. TSC1 was identified as a direct miR-130b target in luciferase-3'UTR assays, and shRNA-mediated knockdown of TSC1 replicated miR-130b effects. Loss-of-function and gain-of-function studies showed that miR-130b levels regulate cell morphology and motility. Following miR-130b suppression, LMS cells adopted a rounded morphology, amoeboid mode of cell movement and enhanced invasive capacity that was Rho/ROCK dependent. Conversely, miR-130b-overexpressing LMS cells exhibited Rho-independent invasion, accompanied by down-regulation of Rho-pathway effectors. In mesenchymal stem cells, both miR-130b overexpression and TSC1 silencing independently impaired SM differentiation in vitro. Together, the data reveal miR-130b as a pro-oncogenic miRNA in LMS and support a miR-130b-TSC1 regulatory network that enhances tumor progression via inhibition of SM differentiation.


Asunto(s)
Leiomiosarcoma , MicroARNs , Humanos , Línea Celular Tumoral , Leiomiosarcoma/genética , MicroARNs/genética , ARN Interferente Pequeño , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Proliferación Celular
2.
Front Bioeng Biotechnol ; 10: 901317, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35837555

RESUMEN

In orthopaedics, gene-based treatment approaches are being investigated for an array of common -yet medically challenging- pathologic conditions of the skeletal connective tissues and structures (bone, cartilage, ligament, tendon, joints, intervertebral discs etc.). As the skeletal system protects the vital organs and provides weight-bearing structural support, the various tissues are principally composed of dense extracellular matrix (ECM), often with minimal cellularity and vasculature. Due to their functional roles, composition, and distribution throughout the body the skeletal tissues are prone to traumatic injury, and/or structural failure from chronic inflammation and matrix degradation. Due to a mixture of environment and endogenous factors repair processes are often slow and fail to restore the native quality of the ECM and its function. In other cases, large-scale lesions from severe trauma or tumor surgery, exceed the body's healing and regenerative capacity. Although a wide range of exogenous gene products (proteins and RNAs) have the potential to enhance tissue repair/regeneration and inhibit degenerative disease their clinical use is hindered by the absence of practical methods for safe, effective delivery. Cumulatively, a large body of evidence demonstrates the capacity to transfer coding sequences for biologic agents to cells in the skeletal tissues to achieve prolonged delivery at functional levels to augment local repair or inhibit pathologic processes. With an eye toward clinical translation, we discuss the research progress in the primary injury and disease targets in orthopaedic gene therapy. Technical considerations important to the exploration and pre-clinical development are presented, with an emphasis on vector technologies and delivery strategies whose capacity to generate and sustain functional transgene expression in vivo is well-established.

3.
FASEB J ; 23(1): 79-89, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18780763

RESUMEN

In osteoarthritis (OA) articular chondrocytes undergo phenotypic changes culminating in the progressive loss of cartilage from the joint surface. The molecular mechanisms underlying these changes are poorly understood. Here we report enhanced (approximately 7-fold) expression of F-spondin, a neuronal extracellular matrix glycoprotein, in human OA cartilage (P<0.005). OA-specific up-regulation of F-spondin was also demonstrated in rat knee cartilage following surgical menisectomy. F-spondin treatment of OA cartilage explants caused a 2-fold increase in levels of the active form of TGF-beta1 (P<0.01) and a 10-fold induction of PGE2 (P<0.005) in culture supernatants. PGE2 induction was found to be dependent on TGF-beta and the thrombospondin domain of the F-spondin molecule. F-spondin addition to cartilage explant cultures also caused a 4-fold increase in collagen degradation (P<0.05) and a modest reduction in proteoglycan synthesis (approximately 20%; P<0.05), which were both TGF-beta and PGE2 dependent. F-spondin treatment also led to increased secretion and activation of MMP-13 (P<0.05). Together these studies identify F-spondin as a novel protein in OA cartilage, where it may act in situ at lesional areas to activate latent TGF-beta and induce cartilage degradation via pathways that involve production of PGE2.


Asunto(s)
Cartílago Articular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Osteoartritis/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Cartílago Articular/patología , Células Cultivadas , Condrocitos/metabolismo , Proteínas de la Matriz Extracelular/genética , Humanos , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
4.
Cells ; 9(9)2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32927859

RESUMEN

TSC1 is a tumor suppressor that inhibits cell growth via negative regulation of the mammalian target of rapamycin complex (mTORC1). TSC1 mutations are associated with Tuberous Sclerosis Complex (TSC), characterized by multiple benign tumors of mesenchymal and epithelial origin. TSC1 modulates self-renewal and differentiation in hematopoietic stem cells; however, its effects on mesenchymal stem cells (MSCs) are unknown. We investigated the impact of Tsc1 inactivation in murine bone marrow (BM)-MSCs, using tissue-specific, transgelin (Tagln)-mediated cre-recombination, targeting both BM-MSCs and smooth muscle cells. Tsc1 mutants were viable, but homozygous inactivation led to a dwarfed appearance with TSC-like pathologies in multiple organs and reduced survival. In young (28 day old) mice, Tsc1 deficiency-induced significant cell expansion of non-hematopoietic BM in vivo, and MSC colony-forming potential in vitro, that was normalized upon treatment with the mTOR inhibitor, everolimus. The hyperproliferative BM-MSC phenotype was lost in aged (1.5 yr) mice, and Tsc1 inactivation was also accompanied by elevated ROS and increased senescence. ShRNA-mediated knockdown of Tsc1 in BM-MSCs replicated the hyperproliferative BM-MSC phenotype and led to impaired adipogenic and myogenic differentiation. Our data show that Tsc1 is a negative regulator of BM-MSC proliferation and support a pivotal role for the Tsc1-mTOR axis in the maintenance of the mesenchymal progenitor pool.


Asunto(s)
Proliferación Celular , Células Madre Mesenquimatosas/citología , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Senescencia Celular , Femenino , Ratones , Ratones Noqueados , Serina-Treonina Quinasas TOR/metabolismo , Esclerosis Tuberosa/patología
5.
Biomaterials ; 29(7): 904-16, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18045683

RESUMEN

Ruptures of the anterior cruciate ligament (ACL) are common knee injuries that do not heal, even with surgical repair. Our research is directed towards developing novel, biological approaches that enable suture repair of this ligament. One promising strategy involves the insertion of a collagen hydrogel between the severed ends of the ACL. Cells migrate from the damaged ligament into the hydrogel and produce repair tissue. Here we have investigated the potential for augmenting this process by the transfer of insulin like growth factor (IGF) 1 cDNA to the repair cells using an adenovirus vector. The goal is to achieve direct, in situ gene delivery by loading the hydrogel with vector prior to its insertion into the defect. In a step-wise approach towards evaluating this process, we confirmed that monolayers of ACL fibroblasts were efficiently transduced by adenovirus vectors and continued to express transgenes when subsequently incorporated into the hydrogel; indeed, transgene expression persisted longer within collagen gels than in monolayer culture. Transfer of IGF-1 cDNA increased the cellularity of the gels and led to the synthesis and deposition of increased amounts of types I and III collagen, elastin, tenascin, and vimentin. The cells remained viable, even when subjected to high viral loads. Similar results were obtained when collagen hydrogels were preloaded with adenovirus prior to insertion into an experimental ACL lesion in vitro. These data confirm the promise of using vector-laden hydrogels for the in situ delivery of genes to cells within damaged ligaments and suggest novel possibilities for the biological repair of the ACL.


Asunto(s)
Lesiones del Ligamento Cruzado Anterior , Ligamento Cruzado Anterior/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Transgenes/genética , Adenoviridae/genética , Ligamento Cruzado Anterior/citología , Biomarcadores , Supervivencia Celular , Células Cultivadas , Colágeno , Fibroblastos , Regulación de la Expresión Génica , Humanos , Hidrogeles , Inmunohistoquímica , Factor I del Crecimiento Similar a la Insulina/genética
6.
Tissue Eng ; 13(9): 2227-37, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17561802

RESUMEN

To investigate the use of a scaffold seeded with genetically modified meniscal cells or mesenchymal stem cells (MSCs) for the healing of meniscal lesions, primary meniscus cells and bone marrow-derived MSCs were isolated from bovine calves and transduced with first-generation adenoviral vectors encoding green fluorescent protein, luciferase, or transforming growth factor (TGF)-beta1 complementary deoxyribonucleic acid (cDNA). The genetically modified cells were seeded in type I collagen-glycosaminoglycan (GAG) matrices and transplanted into tears of the avascular zone of bovine menisci. After 3 weeks of in vitro culture, constructs and repair tissues were analyzed histologically, biochemically, and using reverse transcriptase polymerase chain reaction. Recombinant adenovirus readily transduced meniscal cells and MSCs, and transgene expression remained high after the cells were incorporated into collagen-GAG matrices. Transfer of TGF-beta1 cDNA increased cellularitiy and the synthesis of GAG/DNA [microg/microg]. It also led to stronger staining for proteoglycans and type II collagen and enhanced expression of meniscal genes. Transplantation of the TGF-beta1 transduced constructs into meniscal lesions of the avascular zone resulted in filling of the lesions with repair tissue after 3 weeks of in vitro culture. These results indicate that TGF-beta1 cDNA delivery may affect cell-based meniscus repair approaches in vivo.


Asunto(s)
ADN Complementario , Técnicas de Transferencia de Gen , Ingeniería Genética , Meniscos Tibiales , Ingeniería de Tejidos , Factor de Crecimiento Transformador beta1/genética , Animales , Bovinos , Células Cultivadas , Genes Reporteros , Humanos , Meniscos Tibiales/citología , Células Madre Mesenquimatosas , Proteínas Recombinantes/genética
7.
Tissue Eng ; 13(8): 1987-93, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17518747

RESUMEN

Facilitated endogenous repair is a novel approach to tissue engineering that avoids the ex vivo culture of autologous cells and the need for manufactured scaffolds, while minimizing the number and invasiveness of associated clinical procedures. The strategy relies on harnessing the intrinsic regenerative potential of endogenous tissues using molecular stimuli, such as gene transfer, to initiate reparative processes in situ. In the simplest example, direct percutaneous injection of an osteogenic vector is used to stimulate bone healing. If necessary, additional progenitor cells and space-filling scaffolds can be provided by autologous bone marrow, muscle, fat, and perhaps other tissues. These can be harvested, processed, and reimplanted by simple, expedited, intraoperative procedures. Examples of repair of experimental osseous and osteochondral lesions in laboratory animals are described. If successful, these strategies will provide methods for tissue regeneration that are not only effective but also inexpensive, safe, and clinically expeditious. Although orthopaedic examples are given here, the technology should be more generally applicable.


Asunto(s)
Ingeniería de Tejidos/economía , Ingeniería de Tejidos/métodos , Cicatrización de Heridas/fisiología , Animales , Humanos , Ingeniería de Tejidos/tendencias
8.
ACS Biomater Sci Eng ; 2(10): 1787-1795, 2016 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-33440476

RESUMEN

The demands of tissue engineering have driven a tremendous amount of research effort in 3D tissue culture technology and, more recently, in 3D printing. The need to use 3D tissue culture techniques more broadly in all of cell biology is well-recognized, but the transition to 3D has been impeded by the convenience, effectiveness, and ubiquity of 2D culture materials, assays, and protocols, as well as the lack of 3D counterparts of these tools. Interestingly, progress and discoveries in 3D bioprinting research may provide the technical support needed to grow the practice of 3D culture. Here we investigate an integrated approach for 3D printing multicellular structures while using the same platform for 3D cell culture, experimentation, and assay development. We employ a liquid-like solid (LLS) material made from packed granular-scale microgels, which locally and temporarily fluidizes under the focused application of stress and spontaneously solidifies after the applied stress is removed. These rheological properties enable 3D printing of multicellular structures as well as the growth and expansion of cellular structures or dispersed cells. The transport properties of LLS allow molecular diffusion for the delivery of nutrients or small molecules for fluorescence-based assays. Here, we measure viability of 11 different cell types in the LLS medium, we 3D print numerous structures using several of these cell types, and we explore the transport properties in molecular time-release assays.

9.
Methods Mol Med ; 100: 147-64, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15280594

RESUMEN

The major requirement of a successful gene transfer is the efficient delivery of an exogenous therapeutic gene to the appropriate cell type with subsequent high or regulated levels of expression. In this context, viral systems are more efficient than nonviral systems, giving higher levels of gene expression for longer periods. For the application of osteoarthritis (OA), gene products triggering anti-inflammatory or chondroprotective effects are of obvious therapeutic utility. Thus, their cognate genes are candidates for use in the gene therapy of OA. In this chapter, we describe the preparation, the use, and the effect of the transduction of chondrocytes or synovial fibroblasts with an adenoviral vector encoding the cDNA for glutamine: fructose-6-phosphate amidotransferase (GFAT). This is intended to serve as an example of a technology that can be used to evaluate the biological effects of overexpression of other cDNAs.


Asunto(s)
Adenoviridae/genética , Condrocitos/metabolismo , Fibroblastos/metabolismo , Vectores Genéticos/genética , Membrana Sinovial/citología , Transducción Genética/métodos , Adenoviridae/química , Cartílago Articular/química , Cartílago Articular/citología , Cartílago Articular/metabolismo , Condrocitos/química , ADN Recombinante/química , ADN Recombinante/genética , Fibroblastos/química , Terapia Genética/métodos , Vectores Genéticos/química , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/genética , Humanos , Interleucina-1/farmacología , Óxido Nítrico/biosíntesis , Osteoartritis/genética , Osteoartritis/terapia , Plásmidos/química , Plásmidos/genética
10.
Biorheology ; 40(1-3): 61-72, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12454388

RESUMEN

The effects of hypotonic (180 mOsm) and hypertonic (580 mOsm) medium loading on chondrocyte aggrecan gene expression in 2D monolayer and 3D hydrogel culture (agarose or alginate) were studied. Aggrecan promoter activity was monitored using a luciferase reporter gene assay and transient transfection. Osmotic loading was observed to differentially affect promoter activity, with hypotonic loading generally producing at least a 40% elevation in promoter activity, except for the case of alginate where a 50% suppression was observed. Hypertonic loading produced at least a 35% decrease in activity for all cultures. Similar osmolality-induced changes to aggrecan mRNA levels were observed in monolayer cells using qPCR. Deletion of exon 1 blocked the sensitivity of monolayer cells to hypertonic but not hypotonic medium changes. Confocal microscopy measurements suggested that the degree of hypotonic swelling in cells encapsulated in 3D matrix was restricted compared to monolayer cells whereas the degree of hypertonic shrinking was similar under both culture conditions.


Asunto(s)
Cartílago Articular/citología , Condrocitos/metabolismo , Proteínas de la Matriz Extracelular , Proteoglicanos/biosíntesis , Agrecanos , Animales , Bovinos , Técnicas de Cultivo de Célula , Tamaño de la Célula/fisiología , Regulación de la Expresión Génica , Soluciones Hipertónicas , Soluciones Hipotónicas , Lectinas Tipo C , Concentración Osmolar , Proteoglicanos/genética , ARN Mensajero/genética
11.
PLoS One ; 9(5): e98388, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24875054

RESUMEN

F-spondin is a pericellular matrix protein upregulated in developing growth plate cartilage and articular cartilage during osteoarthritis. To address its function in bone and cartilage in vivo, we generated mice that were deficient for the F-spondin gene, Spon1. Spon1-/- mice were viable and developed normally to adulthood with no major skeletal abnormalities. At 6 months, femurs and tibiae of Spon1-/- mice exhibited increased bone mass, evidenced by histological staining and micro CT analyses, which persisted up to 12 months. In contrast, no major abnormalities were observed in articular cartilage at any age group. Immunohistochemical staining of femurs and tibiae revealed increased levels of periostin, alkaline phosphate and tartrate resistant acid phosphatase (TRAP) activity in the growth plate region of Spon1-/- mice, suggesting elevated bone synthesis and turnover. However, there were no differences in serum levels of TRAP, the bone resorption marker, CTX-1, or osteoclast differentiation potential between genotypes. Knockout mice also exhibited reduced levels of TGF-ß1 in serum and cultured costal chondrocytes relative to wild type. This was accompanied by increased levels of the BMP-regulatory SMADs, P-SMAD1/5 in tibiae and chondrocytes. Our findings indicate a previously unrecognized role for Spon1 as a negative regulator of bone mass. We speculate that Spon1 deletion leads to a local and systemic reduction of TGF-ß levels resulting in increased BMP signaling and increased bone deposition in adult mice.


Asunto(s)
Huesos/anatomía & histología , Huesos/metabolismo , Proteínas de la Matriz Extracelular/deficiencia , Fenotipo , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Huesos/citología , Huesos/diagnóstico por imagen , Condrocitos/metabolismo , Orden Génico , Marcación de Gen , Genotipo , Ratones , Ratones Noqueados , Osteoclastos/citología , Osteoclastos/metabolismo , Fosforilación , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Microtomografía por Rayos X
12.
J Orthop Res ; 28(10): 1323-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20839318

RESUMEN

This study examines the role of F-spondin, an extracellular matrix protein of osteoarthritic cartilage, during chondrocyte maturation in embryonic growth plate cartilage. In chick tibia, F-spondin expression localized to the hypertrophic and calcified zones of the growth plate. Functional studies using tibial organ cultures indicated that F-spondin inhibited (∼35%, p = 0.02), and antibodies to F-spondin increased (∼30%, p < 0.1) longitudinal limb growth relative to untreated controls. In cell cultures, induction of chondrocyte maturation, by retinoic acid (RA) or transforming growth factor (TGF)-ß treatment led to a significant upregulation of F-spondin (p < 0.05). F-spondin transfection increased mineral deposition, alkaline phosphatase (AP) and matrix metalloproteinase (MMP)-13 mRNA levels (p < 0.05), and AP activity following RA stimulation, compared to mock transfected controls. Using AP as a differentiation marker we then investigated the mechanism of F-spondin promaturation effects. Blocking endogenous F-spondin via its thrombospondin (TSR) domain inhibited RA induced AP activity 40% compared to controls (p < 0.05). The stimulatory effect of F-spondin on AP expression was also inhibited following depletion of TGF-ß from culture supernatants. Our findings indicate that F-spondin is expressed in embryonic cartilage, where it has the capacity to enhance chondrocyte terminal differentiation and mineralization via interactions in its TSR domain and TGF-ß dependent pathways.


Asunto(s)
Diferenciación Celular/fisiología , Condrocitos/citología , Proteínas de la Matriz Extracelular/fisiología , Osteogénesis/fisiología , Fosfatasa Alcalina/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Condrocitos/efectos de los fármacos , Condrocitos/fisiología , Femenino , Placa de Crecimiento/citología , Placa de Crecimiento/fisiología , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones Endogámicos , Modelos Animales , Embarazo , Factor de Crecimiento Transformador beta/farmacología , Tretinoina/farmacología
14.
Tissue Eng Part A ; 15(5): 1127-39, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18826340

RESUMEN

Because articular cartilage has a poor regeneration capacity, numerous cell-based approaches to therapy are currently being explored. The present study involved the use of gene transfer as a means to provide sustained delivery of chondrogenic proteins to primary mesenchymal stem cells (MSCs). In previous work, we found that adenoviral-mediated gene transfer of transforming growth factor-beta1 (TGF-beta1) and bone morphogenetic protein 2 (BMP-2), but not insulin-like growth factor 1 (IGF-1), could be used to induce chondrogenic differentiation of MSCs in an aggregate culture system. In the present study, we examined the effects on chondrogenesis of these transgenes when delivered in combination. Cultures of bone marrow-derived MSCs were infected with 2.5 x 10(2) or 2.5 x 10(3) viral particles/cell of each adenoviral vector individually, or in combination, seeded into aggregates, and cultured for 3 weeks in a defined serum-free medium. Levels of transgene product in the medium were initially high, approximately 100 ng/mL TGF-beta1, 120 ng/mL BMP-2, and 80 ng/mL IGF-1 at day 3, and declined thereafter. We found that co-expression of IGF-1 and TGF-beta1, BMP-2, or both at low doses resulted in larger aggregates, higher levels of glycosaminoglycan synthesis, stronger staining for proteoglycans and collagen type II and X, and greater expression of cartilage-specific marker genes than with either transgene alone. Gene-induced chondrogenesis of MSCs using multiple genes that act synergistically may enable the administration of reduced viral doses in vivo and could be of considerable benefit for the development of cell-based therapies for cartilage repair.


Asunto(s)
Condrogénesis/genética , Condrogénesis/fisiología , Técnicas de Transferencia de Gen , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Adenoviridae/genética , Animales , Secuencia de Bases , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/fisiología , Bovinos , Agregación Celular , Diferenciación Celular , ADN/biosíntesis , ADN/genética , Cartilla de ADN/genética , Expresión Génica , Vectores Genéticos , Glicosaminoglicanos/biosíntesis , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/fisiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ingeniería de Tejidos/métodos , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/fisiología
15.
Mol Ther ; 15(6): 1114-20, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17440444

RESUMEN

Local gene therapy for chronic joint diseases requires prolonged transgenic expression, but this has not been reliably achieved in animal models. Using normal and immunocompromised animals, we examined the capacity of various cell types in joint tissues to maintain and express exogenous transgenes after direct intra-articular gene delivery. We found that transgenic expression could persist for the lifetime of the animal but required precise immunological compatibility between the vector, transgene product, and host. It was not dependent on vector integration or promoter origin. We identified two phenotypically distinct sub-populations of genetically modified cells within the joint: (i) transient cells, with a half-life of a few weeks, and (ii) stable cells that reside in the joint tissues indefinitely. Contrary to the prevailing assumption, the transient sub-population was composed almost exclusively of synovial fibroblasts, indicating that the synovium is not an appropriate tissue upon which to base a long-term therapy. Instead, fibroblasts in the ligaments, tendons, and capsule emerged as the primary cell types capable of sustained therapeutic transgene expression. This study sheds new light on the cellular dynamics of articular tissues and suggests that cell turnover and immune reactivity are the key determinants in achieving sustained transgenic expression intra-articularly.


Asunto(s)
Terapia Genética/métodos , Artropatías/terapia , Transgenes/genética , Animales , Línea Celular , Enfermedad Crónica , ADN Complementario/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Citometría de Flujo , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteína Antagonista del Receptor de Interleucina 1/genética , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Artropatías/inmunología , Artropatías/patología , Lentivirus/genética , Masculino , Microscopía Fluorescente , Ratas , Ratas Desnudas , Ratas Wistar , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
16.
Mol Ther ; 12(2): 219-28, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16043093

RESUMEN

Adult mesenchymal stem cells (MSCs) have the capacity to differentiate into various connective tissues such as cartilage and bone following stimulation with certain growth factors. However, less is known about the capacity of these cells to undergo chondrogenesis when these proteins are delivered via gene transfer. In this study, we investigated chondrogenesis of primary, bone marrow-derived MSCs in aggregate cultures following genetic modification with adenoviral vectors encoding chondrogenic growth factors. We found that adenoviral-mediated expression of TGF-beta1 and BMP-2, but not IGF-1, induced chondrogenesis of MSCs as evidenced by toluidine blue metachromasia and immunohistochemical detection of type II collagen. Chondrogenesis correlated with the level and duration of expressed protein and was strongest in aggregates expressing 10-100 ng/ml transgene product. Transgene expression in all aggregates was highly transient, showing a marked decrease after 7 days. Chondrogenesis was inhibited in aggregates modified to express >100 ng/ml TGF-beta1 or BMP-2; however, this was found to be partly due to the inhibitory effect of exposure to high adenoviral loads. Our findings indicate that parameters such as these are important functional considerations for adapting gene transfer technologies to induce chondrogenesis of MSCs.


Asunto(s)
Condrogénesis/fisiología , Técnicas de Transferencia de Gen , Terapia Genética , Células Madre Mesenquimatosas/fisiología , Ingeniería de Tejidos/métodos , Adenoviridae , Adulto , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Condrocitos , Condrogénesis/genética , Técnicas de Cultivo , Expresión Génica , Vectores Genéticos , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Osteogénesis , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1 , Transgenes/genética
17.
Mol Ther ; 7(4): 460-6, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12727108

RESUMEN

Clinical translation of gene-based therapies for arthritis could be accelerated by vectors capable of efficient intra-articular gene delivery and long-term transgene expression. Previously, we have shown that lentiviral vectors transduce rat synovium efficiently in vivo. Here, we evaluated the functional capacity of transgene expression provided by lentiviral-mediated gene delivery to the joint. To do this, we measured the ability of a lentiviral vector containing the cDNA for human interleukin-1 receptor antagonist (LV-hIL-1Ra) to suppress intra-articular responses to IL-1beta. Groups of rats were injected in one knee with 5 x 10(7) infectious units of LV-IL-1Ra. After 24 h, a range of doses of fibroblasts (3 x 10(3), 10(4), 3 x 10(4), or 10(5) cells) genetically modified to overexpress IL-1beta was injected into both knees. Intra-articular delivery of LV-hIL-1Ra strongly prevented swelling in all treated knees, even in those receiving the greatest dose of IL-1beta(+) cells. Cellular infiltration, cartilage erosion, and invasiveness of inflamed synovium were effectively prevented in LV-hIL-1Ra-treated knees and were significantly inhibited in contralateral joints. Beneficial effects were also observed systemically in the lentivirus-treated animals. Interestingly, intra-articular expression of the IL-1Ra transgene was found to increase in relation to the number of IL-1beta(+) cells injected. Further experiments using GFP suggest this is due to the proliferation of cells, stably modified by the integrative lentivirus, in response to inflammatory stimulation.


Asunto(s)
Artritis Experimental/terapia , Lentivirus/genética , Sialoglicoproteínas/genética , Membrana Sinovial/metabolismo , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Vectores Genéticos/administración & dosificación , Miembro Posterior/metabolismo , Miembro Posterior/patología , Humanos , Inyecciones Intraarticulares , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-1/genética , Interleucina-1/metabolismo , Articulaciones/metabolismo , Articulaciones/patología , Masculino , Microscopía Fluorescente , Ratas , Ratas Wistar , Sialoglicoproteínas/metabolismo
18.
Mol Ther ; 5(4): 397-404, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11945066

RESUMEN

The delivery of anti-arthritic genes to the synovial lining of joints is being explored as a strategy for the treatment of rheumatoid arthritis. In this study, we have investigated the use of VSV-G pseudotyped, HIV-1-based lentiviral vectors for gene delivery to articular tissues. Recombinant lentivirus containing a beta-galactosidase/neomycin resistance fusion gene under control of the elongation factor (EF) 1alpha promoter efficiently transduced human and rat synoviocytes and chondrocytes in cell culture. When directly injected into the knees of rats, this vector transduced synovial lining cells, but not other articular tissues such as cartilage. We also constructed a lentiviral vector containing the human interleukin-1 receptor antagonist (IL1RA) cDNA and examined transgene expression in vitro and in vivo following injection into the knee joints of rats. In immunocompetent animals, intra-articular IL1RA expression was high and persisted, at a sharply declining rate, for approximately 20 days. In immunocompromised rats, however, lentivirus-mediated intra-articular expression of human IL1RA was found to persist for at least 6 weeks. Extra-articular expression of the transgene was minimal. These results indicate that lentiviral vectors are capable of efficient in vivo gene transfer to synovium and merit further investigation as a means of providing long-term expression for gene-based treatments of arthritis.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/genética , VIH-1/genética , Membrana Sinovial/metabolismo , Animales , Animales Modificados Genéticamente , Antirreumáticos/administración & dosificación , Antirreumáticos/farmacocinética , Artritis Reumatoide/terapia , Resistencia a Medicamentos/genética , Terapia Genética , Humanos , Proteína Antagonista del Receptor de Interleucina 1 , Masculino , Neomicina/farmacología , Especificidad de Órganos , Ratas , Ratas Desnudas , Ratas Wistar , Sialoglicoproteínas/genética , Sialoglicoproteínas/farmacocinética , beta-Galactosidasa/genética
19.
Mol Ther ; 10(2): 327-36, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15294179

RESUMEN

The inability of the ruptured anterior cruciate ligament (ACL) of the knee joint to heal spontaneously presents numerous clinical problems. Here we describe a novel, gene-based approach to augment ACL healing. It is based upon the migration of cells from the ruptured ends of the ligament into a collagen hydrogel laden with recombinant adenovirus. Cells entering the gel become transduced by the vector, which provides a basis for the local synthesis of gene products that aid repair. Monolayers of bovine ACL cells were readily transduced by first-generation, recombinant adenovirus, and transgene expression remained high after the cells were incorporated into collagen hydrogels. Using an in vitro model of ligament repair, cells migrated from the cut ends of the ACL into the hydrogel and were readily transduced by recombinant adenovirus contained within it. The results of experiments in which GFP was used as the transgene suggest highly efficient transduction of ACL cells in this manner. Moreover, during a 21-day period GFP+ cells were observed more than 6 mm from the severed ligament. This distance is ample for the projected clinical application of this technology. In response to TGF-beta1 as the transgene, greater numbers of ACL cells accumulated in the hydrogels, where they deposited larger amounts of type III collagen. These data confirm that it is possible to transduce ACL cells efficiently in situ as they migrate from the ruptured ACL, that transduction does not interfere with the cells' ability to migrate distances necessary for successful repair, and that ACL cells will respond in a suitable manner to the products of the transgenes they express. This permits optimism over a possible clinical use for this technology.


Asunto(s)
Lesiones del Ligamento Cruzado Anterior , Terapia Genética/métodos , Transducción Genética/métodos , Factor de Crecimiento Transformador beta/genética , Cicatrización de Heridas , Adenoviridae/genética , Animales , Ligamento Cruzado Anterior/fisiología , Bovinos , Movimiento Celular , Células Cultivadas , Colágeno Tipo III/análisis , Colágeno Tipo III/metabolismo , Expresión Génica/genética , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Rotura/terapia , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1
20.
Arthritis Res Ther ; 5(5): R301-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12932294

RESUMEN

Anakinra, the recombinant form of IL-1 receptor antagonist (IL-1Ra), has been approved for clinical use in the treatment of rheumatoid arthritis as the drug Kineret trade mark, but it must be administered daily by subcutaneous injection. Gene transfer may offer a more effective means of delivery. In this study, using prostaglandin E2 production as a measure of stimulation, we quantitatively compared the ability of anakinra, as well as that of IL-1Ra delivered by gene transfer, to inhibit the biologic actions of IL-1beta. Human synovial fibroblast cultures were incubated with a range of doses of anakinra or HIG-82 cells genetically modified to constitutively express IL-1Ra. The cultures were then challenged with recombinant human IL-1beta either simultaneously with addition of the source of IL-1Ra or 24 hours later. In a similar manner, the potencies of the two sources of IL-1Ra were compared when human synovial fibroblasts were challenged with IL-1beta produced constitutively by genetically modified cells. No significant difference in inhibitory activity was observed between recombinant protein and IL-1Ra provided by the genetically modified cells, under static culture conditions, even following incubation for 4 days. However, under culture conditions that provided progressive dilution of the culture media, striking differences between these methods of protein delivery became readily apparent. Constitutive synthesis of IL-1Ra by the genetically modified cells provided sustained or increased protection from IL-1 stimulation over time, whereas the recombinant protein became progressively less effective. This was particularly evident under conditions of continuous IL-1beta synthesis.


Asunto(s)
Interleucina-1/antagonistas & inhibidores , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Sialoglicoproteínas/administración & dosificación , Sialoglicoproteínas/farmacología , Animales , Línea Celular , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/virología , Ingeniería Genética/métodos , Vectores Genéticos/genética , Humanos , Proteína Antagonista del Receptor de Interleucina 1 , Osteoartritis/metabolismo , Osteoartritis/patología , Conejos , Ratas , Ratas Wistar , Retroviridae/genética , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/patología , Transducción Genética/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA