Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cells Transl Med ; 4(6): 643-54, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25848122

RESUMEN

UNLABELLED: Cultured epithelial autografts (CEAs) produced from a small, healthy skin biopsy represent a lifesaving surgical technique in cases of full-thickness skin burn covering >50% of total body surface area. CEAs also present numerous drawbacks, among them the use of animal proteins and cells, the high fragility of keratinocyte sheets, and the immaturity of the dermal-epidermal junction, leading to heavy cosmetic and functional sequelae. To overcome these weaknesses, we developed a human plasma-based epidermal substitute (hPBES) for epidermal coverage in cases of massive burn, as an alternative to traditional CEA, and set up critical quality controls for preclinical and clinical studies. In this study, phenotypical analyses in conjunction with functional assays (clonal analysis, long-term culture, or in vivo graft) showed that our new substitute fulfills the biological requirements for epidermal regeneration. hPBES keratinocytes showed high potential for cell proliferation and subsequent differentiation similar to healthy skin compared with a well-known reference material, as ascertained by a combination of quality controls. This work highlights the importance of integrating relevant multiparameter quality controls into the bioengineering of new skin substitutes before they reach clinical development. SIGNIFICANCE: This work involves the development of a new bioengineered epidermal substitute with pertinent functional quality controls. The novelty of this work is based on this quality approach.


Asunto(s)
Quemaduras/terapia , Proliferación Celular , Queratinocitos/metabolismo , Queratinocitos/trasplante , Plasma , Piel Artificial , Animales , Quemaduras/metabolismo , Quemaduras/patología , Dermis/metabolismo , Dermis/patología , Epidermis/metabolismo , Epidermis/patología , Femenino , Xenoinjertos , Humanos , Queratinocitos/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID
2.
PLoS One ; 10(3): e0118709, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25789482

RESUMEN

BACKGROUND: Radiation overexposure accidents are rare but can have severe long-term health consequences. Although underreporting can be an issue, some extensive literature reviews of reported radiation overexposures have been performed and constitute a sound basis for conclusions on general trends. Building further on this work, we performed a systematic review that completes previous reviews and provides new information on characteristics and trends of reported radiation accidents. METHODS: We searched publications and reports from MEDLINE, EMBASE, the International Atomic Energy Agency, the International Radiation Protection Association, the United Nations Scientific Committee on the Effects of Atomic Radiation, the United States Nuclear Regulatory Commission, and the Radiation Emergency Assistance Center/Training Site radiation accident registry over 1980-2013. We retrieved the reported overexposure cases, systematically extracted selected information, and performed a descriptive analysis. RESULTS: 297 out of 5189 publications and reports and 194 records from the REAC/TS registry met our eligibility criteria. From these, 634 reported radiation accidents were retrieved, involving 2390 overexposed people, of whom 190 died from their overexposure. The number of reported cases has decreased for all types of radiation use, but the medical one. 64% of retrieved overexposure cases occurred with the use of radiation therapy and fluoroscopy. Additionally, the types of reported accidents differed significantly across regions. CONCLUSIONS: This review provides an updated and broader view of reported radiation overexposures. It suggests an overall decline in reported radiation overexposures over 1980-2013. The greatest share of reported overexposures occurred in the medical fields using radiation therapy and fluoroscopy; this larger number of reported overexposures accidents indicates the potential need for enhanced quality assurance programs. Our data also highlights variations in characteristics of reported accidents by region. The main limitation of this study is the likely underreporting of radiation overexposures. Ensuring a comprehensive monitoring and reporting of radiation overexposures is paramount to inform and tailor prevention interventions to local needs.


Asunto(s)
Exposición a la Radiación/historia , Exposición a la Radiación/estadística & datos numéricos , Liberación de Radiactividad Peligrosa/historia , Liberación de Radiactividad Peligrosa/estadística & datos numéricos , Fluoroscopía/efectos adversos , Fluoroscopía/estadística & datos numéricos , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Radioterapia/efectos adversos , Radioterapia/estadística & datos numéricos
3.
Cell Transplant ; 24(2): 277-86, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24143883

RESUMEN

Fecal incontinence (FI) remains a socially isolating condition with profound impact on quality of life for which autologous myoblast cell therapy represents an attractive treatment option. We developed an animal model of FI and investigated the possibility of improving sphincter function by intrasphincteric injection of syngeneic myoblasts. Several types of anal cryoinjuries were evaluated on anesthetized Fischer rats receiving analgesics. The minimal lesion yielding sustainable anal sphincter deficiency was a 90° cryoinjury of the sphincter, repeated after a 24-h interval. Anal sphincter pressure was evaluated longitudinally by anorectal manometry under local electrostimulation. Myoblasts were prepared using a protocol mimicking a clinical-grade process and further transduced with a GFP-encoding lentiviral vector before intrasphincteric injection. Experimental groups were uninjured controls, cryoinjured + PBS, and cryoinjured + myoblasts (different doses or injection site). Myoblast injection was well tolerated. Transferred myoblasts expressing GFP integrated into the sphincter and differentiated in situ into dystrophin-positive mature myofibers. Posttreatment sphincter pressures increased over time. At day 60, pressures in the treated group were significantly higher than those of PBS-injected controls and not significantly different from those of normal rats. Longitudinal follow-up showed stability of the therapeutic effect on sphincter function over a period of 6 months. Intrasphincteric myoblast injections at the lesion borders were equally as effective as intralesion administration, but an injection opposite to the lesion was not. These results provide proof of principle for myoblast cell therapy to treat FI in a rat model. This strategy is currently being evaluated in humans in a randomized double-blind placebo-controlled clinical trial.


Asunto(s)
Canal Anal/fisiología , Incontinencia Fecal/terapia , Mioblastos/trasplante , Canal Anal/patología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Modelos Animales de Enfermedad , Estimulación Eléctrica , Incontinencia Fecal/patología , Femenino , Humanos , Contracción Muscular , Mioblastos/citología , Ratas , Recuperación de la Función
4.
Cell Transplant ; 23(12): 1475-87, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25565635

RESUMEN

Genetic alterations have recently been described as emerging during the culture of embryonic stem cells or induced pluripotent stem cells, raising concerns about their safety in future clinical use. Myoblasts are adult stem cells with important therapeutic potential that have been used in clinical trials for almost 20 years, but their genome integrity has not yet been established. Here we produced 10 human myoblast preparations and investigated their genomic stability. At the third passage, half of the preparations had a normal karyotype and half showed one to four alterations/30 metaphases. Chromosome 2 trisomy was found in 1-2/30 metaphases and/or 2/100 nuclei by FISH in 3/10 samples, and there was no other recurrent anomaly. When prolonging cultures, these erratic abnormalities were never associated with a growth advantage. Cellular senescence was manifested in all samples by growth arrest before passage 15. Expression of TERT was always negative. Molecular analysis of individual p53 transcripts did not reveal tumorigenic mutations. CGH array (10 samples) and exome sequencing (one sample) failed to detect copy number variations or accumulation of mutations, respectively. Myoblasts did not grow either in soft agar or in vivo after injection in immunodeficient mice. Hence, occasional genomic abnormalities may occur during myoblast culture but are not associated with risk of transformation.


Asunto(s)
Transformación Celular Neoplásica , Inestabilidad Cromosómica , Mioblastos/metabolismo , Mioblastos/patología , Animales , Carcinogénesis , Proliferación Celular , Células Cultivadas , Senescencia Celular , Hibridación Genómica Comparativa , Femenino , Dosificación de Gen , Humanos , Inmunofenotipificación , Cariotipificación , Masculino , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Fenotipo
5.
Stem Cell Res Ther ; 4(1): 1, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23290259

RESUMEN

INTRODUCTION: This study investigated the promising effect of a new Platelet Glue obtained from Cryoprecipitation of Apheresis Platelet products (PGCAP) used in combination with Mesenchymal Stromal Cells (MSC) loaded on ceramic biomaterials to provide novel strategies enhancing bone repair. METHODS: PGCAP growth factor content was analyzed by ELISA and compared to other platelet and plasma-derived products. MSC loaded on biomaterials (65% hydroxyapatite/35% beta-TCP or 100% beta-TCP) were embedded in PGCAP and grown in presence or not of osteogenic induction medium for 21 days. Biomaterials were then implanted subcutaneously in immunodeficient mice for 28 days. Effect of PGCAP on MSC was evaluated in vitro by proliferation and osteoblastic gene expression analysis and in vivo by histology and immunohistochemistry. RESULTS: We showed that PGCAP, compared to other platelet-derived products, allowed concentrating large amount of growth factors and cytokines which promoted MSC and osteoprogenitor proliferation. Next, we found that PGCAP improves the proliferation of MSC and osteogenic-induced MSC. Furthermore, we demonstrated that PGCAP up-regulates the mRNA expression of osteogenic markers (Collagen type I, Osteonectin, Osteopontin and Runx2). In vivo, type I collagen expressed in ectopic bone-like tissue was highly enhanced in biomaterials embedded in PGCAP in the absence of osteogenic pre-induction. Better results were obtained with 65% hydroxyapatite/35% beta-TCP biomaterials as compared to 100% beta-TCP. CONCLUSIONS: We have demonstrated that PGCAP is able to enhance in vitro MSC proliferation, osteoblastic differentiation and in vivo bone formation in the absence of osteogenic pre-induction. This clinically adaptable platelet glue could be of interest for improving bone repair.


Asunto(s)
Materiales Biocompatibles/farmacología , Plaquetas/efectos de los fármacos , Huesos/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Adhesivos/farmacología , Animales , Biomarcadores/metabolismo , Plaquetas/metabolismo , Huesos/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , ARN Mensajero/metabolismo , Ingeniería de Tejidos/métodos
6.
Int Urogynecol J ; 22(2): 183-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20821309

RESUMEN

INTRODUCTION AND HYPOTHESIS: Cell therapy for stress urinary incontinence (SUI) management has been experienced with encouraging results. METHODS: We conducted an open prospective study on 12 women presenting severe SUI with fixed urethra, after previous failed surgical management. Patients underwent intrasphincteric injections of autologous progenitor muscular cells isolated from a biopsy of deltoid muscle. Primary endpoint focused on safety (measurement of Q(max) variation after 3 months). Secondary endpoints assessed side effects and efficacy. RESULTS: No variation was diagnosed on Q(max) measurements. Efficacy data show that three of 12 patients are dry at 12 months, seven other patients are improved on pad test but not on voiding diary, and two patients were slightly worsened by the procedure. Quality of life was improved in half of patients. CONCLUSIONS: Cell therapy for severe multioperated cases of SUI is a mini-invasive, feasible, and safe procedure that can improve urinary condition in as a second line therapy.


Asunto(s)
Mioblastos/trasplante , Incontinencia Urinaria de Esfuerzo/terapia , Adulto , Anciano , Femenino , Humanos , Inyecciones , Persona de Mediana Edad , Estudios Prospectivos , Calidad de Vida , Trasplante Autólogo , Resultado del Tratamiento
7.
Leuk Lymphoma ; 48(10): 2032-41, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17917971

RESUMEN

Osteolytic bone lesions are common in patients with multiple myeloma (MM), a clonal plasma cell disorder, and result from increased osteoclastic bone resorption and decreased osteoblastic bone formation. Because mesenchymal stem cells (MSCs) are committed towards cells of the osteoblast lineage, we compared the in vitro characteristics of MSCs from the bone marrow of 18 MM patients (MM-MSCs) and eight normal donors (ND-MSCs). MM-MSCs displayed deficient growth that could be explained in part by the reduced expression of several growth factor receptors on the surface of MM-MSCs compared with ND-MSCs. Receptor downregulation was observed on RT-PCR analysis. A major finding was an approximately fivefold higher expression of osteoblast inhibitor DKK1 at transcript and protein levels in MM-MSCs than ND-MSCs. These data suggest that defective osteoblast function in patients with advanced MM may be related not only to factors released by tumor myeloma cells but also to MSC abnormalities.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Células Madre Mesenquimatosas/citología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Anciano , Huesos/metabolismo , Diferenciación Celular , Femenino , Humanos , Inmunofenotipificación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-6/metabolismo , Masculino , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteólisis , Sindecano-1/biosíntesis
8.
J Cell Physiol ; 205(2): 228-36, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15887229

RESUMEN

Mesenchymal stem cells (MSCs) are considered as emergent "universal" cells and various tissue repair programs using MSCs are in development. In vitro expansion of MSCs is conventionally achieved in medium containing fetal calf serum (FCS) and is increased by addition of growth factors. However, for widespread clinical applications, contact of MSCs with FCS must be minimized since it is a putative source of prion or virus transmission. Therefore, because platelets are a natural source of growth factors, we sought to investigate in vitro MSC expansion in response to platelet lysates (PL) obtained from platelet-rich plasma. Human MSCs were expanded in FCS (+/-bFGF)- or PL-supplemented medium through a process of subculture. We demonstrated that PL-containing medium is enriched by growth factors (platelet-derived growth factors (PDGFs), basic fibroblast growth factor (bFGF), transforming growth factor (TGF-beta), insulin-like growth factor-1 (IGF-1) ...) and showed that PL is able to promote MSC expansion, to decrease the time required to reach confluence, and to increase CFU-F size, as compared to the FCS medium. Furthermore, we demonstrated that MSCs cultured in the presence of PL maintain their osteogenic, chondrogenic, and adipogenic differentiation properties and retain their immunosuppressive activity. Therefore, we propose that PL may be a powerful and safe substitute for FCS in development of tissue- and cellular-engineered products in clinical settings using MSCs.


Asunto(s)
Plaquetas/fisiología , Sustitutos Sanguíneos/efectos adversos , Extractos Celulares/farmacología , Proliferación Celular/efectos de los fármacos , Tratamiento Basado en Trasplante de Células y Tejidos , Células Madre Mesenquimatosas/fisiología , Animales , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Sustancias de Crecimiento/análisis , Humanos , Inmunofenotipificación , Cinética , Factores de Tiempo
9.
J Gene Med ; 4(6): 581-91, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12439850

RESUMEN

Rheumatoid arthritis (RA) is a severe autoimmune systemic disease. Chronic synovial inflammation results in destruction of the joints. No conventional treatment is efficient in RA. Gene therapy of RA targets mainly the players of inflammation or articular destruction: TNF-alpha or IL-1 blocking agents (such as anti-TNF-alpha monoclonal antibodies, soluble TNF-alpha receptor, type II soluble receptor of IL-1, IL-1 receptor antagonist), antiinflammatory cytokines (such as IL-4, IL-10, IL-1), and growth factors. In this polyarticular disease, the vector expressing the therapeutic protein can be administered as a local (intra-articular injection) or a systemic treatment (extra-articular injection). All the main vectors have been used in experimental models, including the more recent lentivirus and adeno-associated virus. Ex vivo gene transfer was performed with synovial cells, fibroblasts, T cells, dendritic cells, and different cells from xenogeneic origin. In vivo gene therapy is simpler, although a less controlled method. Clinical trials in human RA have started with ex vivo retrovirus-expressing IL-1 receptor antagonists and have demonstrated the feasibility of the strategy of gene therapy. The best target remains to be determined and extensive research has to be conducted in preclinical studies.


Asunto(s)
Artritis Reumatoide/terapia , Terapia Genética , Ensayos Clínicos como Asunto , Vectores Genéticos , Humanos , Inflamación
10.
Eur J Immunol ; 32(9): 2437-49, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12207328

RESUMEN

The common gamma (gamma c) chain, shared by Th1 and Th2 cytokines, is fundamental for the activation of hematopoietic cells, but its role in non-hematopoietic tissues has not been explored. Here we show that in normal lung fibroblasts IL-4 and IL-13 induce the expression of the gamma c chain and its association with Janus kinase (JAK) 3, while lung myofibroblasts constitutively express a gamma c chain displaying a limited association with JAK3. In the latter cells, without exogenous cytokines, gamma c/JAK3 controls, through autocrine loops, tyrosine kinase (TYK) 2 phosphorylation and the balance between functional (IL-4Ralpha, IL-13Ralpha 1) and decoy (IL-13Ralpha 2) high-affinity receptors. Moreover, JAK3 is also associated with a pre-phosphorylated IL-4Ralpha and CD40. This novel "heterotrimer" (p-IL-4Ralpha, CD40/JAK3) is functional and controls STAT3 phosphorylation and CD40 expression, as shown by use of the specific JAK3 inhibitor WHI-P31. In basal culture conditions, CD40 signaling could be induced by the transient establishment of inter-fibroblastic CD40/CD40 ligand (CD40L) functional bridges. Indeed, powerful pro-inflammatory stimuli such as lipopolysaccharide and thrombin can rapidly mobilize CD40L at the surface of lung myofibroblasts. These interactions are modified by IL-13, which triggers the formation of a new type of functional receptor (p-IL-4Ralpha /IL-13Ralpha 1/gamma c) and also the recruitment and the phosphorylation of JAK3. Treatment with JAK3 inhibitors blocks IL-13-induced phosphorylation of JAK2, TYK2 and STAT3, but not of JAK1 and STAT6. These data underline (1) the pivotal role of the gamma c chain, CD40/CD40L, JAK3 and IL-13 in the inflammatory-like activation of lung myofibroblasts, (2) the cell-type restraint effects of IL-13 on these cells, and (3) the potential usefulness of JAK3 inhibitors in the treatment of asthma.


Asunto(s)
Ligando de CD40/fisiología , Fibroblastos/fisiología , Inflamación/metabolismo , Interleucina-13/farmacología , Interleucina-4/farmacología , Lipopolisacáridos/farmacología , Pulmón/citología , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas , Receptores de Interleucina-7/fisiología , Trombina/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Antiasmáticos/farmacología , Comunicación Autocrina , Antígenos CD40/fisiología , Ligando de CD40/biosíntesis , Ligando de CD40/genética , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Proteínas de Unión al ADN/metabolismo , Diseño de Fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Subunidad gamma Común de Receptores de Interleucina , Subunidad alfa1 del Receptor de Interleucina-13 , Janus Quinasa 2 , Janus Quinasa 3 , Sustancias Macromoleculares , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Tirosina Quinasas/metabolismo , Proteínas/metabolismo , Receptores de Interleucina/clasificación , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Receptores de Interleucina-13 , Receptores de Interleucina-4/clasificación , Receptores de Interleucina-4/genética , Receptores de Interleucina-4/metabolismo , Receptores de Interleucina-7/biosíntesis , Receptores de Interleucina-7/genética , Proteínas Recombinantes de Fusión/farmacología , Factor de Transcripción STAT3 , Transducción de Señal , TYK2 Quinasa , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...