Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Ann Ig ; 34(6): 558-571, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36040398

RESUMEN

Background: The Coronavirus disease 2019 (COVID-19) pandemic negatively impacted nursing students' opportunity to gain experience through clinical placement, potentially threatening their readiness for practice and their clinical competence. The aim of this study was to explore whether and to what extent the third-year undergraduate nursing students perceived that their readiness for practice was impacted by changes to clinical placement and classroom learning implemented in response to the COVID-19 pandemic. Study design: Cross-sectional study. Methods: The study was conducted in a university of North-western Italy that provides nursing education across five sites. All sites stopped in-person classroom learning at the beginning of March 2020, but each site was free to decide whether to continue in-person clinical placement based on the local epidemiological situation. All 228 third-year nursing students who completed their degree by June 2020 were invited to participate. Data were collected via online questionnaire, which included the question "What impact do you think that COVID-19 safety measures employed by your nursing programme had on your readiness for practice?" Answers were given on a 5-point Likert scale (none, minimal, moderate, major, and severe). Explanatory variables were collected at the individual, nursing programme, and university site levels. Results: A total of 126 (response rate 55.3%) nursing students completed the questionnaire. Overall, 84 (66.7%) perceived that COVID-19 safety measures had a moderate to severe impact on their readiness for practice. These students often had lower grade point averages (p=0.037) and received no clinical placement during the pandemic (72.6% vs 90.5% of students who reported no or minimal impact, p=0.022). Average duration of third-year clinical placement was also lower among these students, though it was not statistically significant. No differences emerged at the university site level. Conclusions: Despite important advances in technology-based educational activities, clinical placement remains the best educational strategy to allow nursing students to feel prepared to work effectively during a pandemic.


Asunto(s)
COVID-19 , Bachillerato en Enfermería , Estudiantes de Enfermería , COVID-19/epidemiología , Estudios Transversales , Humanos , Pandemias
2.
Clin Exp Immunol ; 179(3): 398-413, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25302633

RESUMEN

Several ß cell antigens recognized by T cells in the non-obese diabetic (NOD) mouse model of type 1 diabetes (T1D) are also T cell targets in the human disease. While numerous antigen-specific therapies prevent diabetes in NOD mice, successful translation of rodent findings to patients has been difficult. A human leucocyte antigen (HLA)-transgenic mouse model incorporating human ß cell-specific T cells might provide a better platform for evaluating antigen-specific therapies. The ability to study such T cells is limited by their low frequency in peripheral blood and the difficulty in obtaining islet-infiltrating T cells from patients. We have worked to overcome this limitation by using lentiviral transduction to 'reprogram' primary human CD8 T cells to express three T cell receptors (TCRs) specific for a peptide derived from the ß cell antigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP265-273 ) and recognized in the context of the human class I major histocompatibility complex (MHC) molecule HLA-A2. The TCRs bound peptide/MHC multimers with a range of avidities, but all bound with at least 10-fold lower avidity than the anti-viral TCR used for comparison. One exhibited antigenic recognition promiscuity. The ß cell-specific human CD8 T cells generated by lentiviral transduction with one of the TCRs released interferon (IFN)-γ in response to antigen and exhibited cytotoxic activity against peptide-pulsed target cells. The cells engrafted in HLA-A2-transgenic NOD-scid IL2rγ(null) mice and could be detected in the blood, spleen and pancreas up to 5 weeks post-transfer, suggesting the utility of this approach for the evaluation of T cell-modulatory therapies for T1D and other T cell-mediated autoimmune diseases.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Vectores Genéticos/genética , Inmunoterapia Adoptiva/métodos , Células Secretoras de Insulina/inmunología , Lentivirus/genética , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD8-positivos/trasplante , Supervivencia Celular , Glucosa-6-Fosfatasa/inmunología , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Humanos , Células Jurkat , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Transgénicos , Fragmentos de Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina-2/genética , Linfocitos T Citotóxicos/trasplante
3.
Nat Genet ; 25(2): 217-22, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10835641

RESUMEN

Gene-transfer vectors based on lentiviruses are distinguished by their ability to transduce non-dividing cells. The HIV-1 proteins Matrix, Vpr and Integrase have been implicated in the nuclear import of the viral genome in non-dividing cells. Here we show that a sequence within pol is also required in cis. It contains structural elements previously associated with the progress of reverse transcription in target cells. We restored these elements in cis within late-generation lentiviral vectors. The new vector transduced to a much higher efficiency several types of human primary cells, when both growing and growth-arrested, including haematopoietic stem cells assayed by long-term repopulation of NOD/SCID mice. On in vivo administration into SCID mice, the vector induced higher plasma levels of human clotting factor IX (F.IX) than non-modified vector. Our results indicate that nuclear translocation of the genome is a rate-limiting step in lentiviral infection of both dividing and non-dividing cells, and that it depends on protein and nucleic acid sequence determinants. Full rescue of this step in lentivirus-based vectors improves performance for gene-therapy applications.


Asunto(s)
Núcleo Celular/genética , Productos del Gen pol/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , VIH-1/genética , Transducción Genética/genética , Animales , Secuencia de Bases , División Celular , Núcleo Celular/metabolismo , Núcleo Celular/virología , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Factor IX/análisis , Factor IX/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Productos del Gen pol/fisiología , Genes Virales/genética , Genes Virales/fisiología , VIH-1/fisiología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Linfocitos/citología , Linfocitos/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones SCID , Datos de Secuencia Molecular , ARN Viral/genética , ARN Viral/metabolismo , Integración Viral
4.
Gene Ther ; 16(3): 340-8, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19112449

RESUMEN

The effectiveness of genetic engineering with lentivectors to protect transplanted cells from allogeneic rejection was examined using, as a model, type 1 diabetes treatment with beta-cell transplantation, whose widespread use has been limited by the requirement for sustained immunosuppressive treatment to prevent graft rejection. We examined whether lentivectors expressing select immunosuppressive proteins encoded by the adenoviral genome early region 3 (AdE3) would protect transplanted beta-cells from an alloimmune attack. The insulin-producing beta-cell line beta TC-tet (C3HeB/FeJ-derived) was transduced with lentiviruses encoding the AdE3 proteins gp19K and RID alpha/beta. The efficiency of lentiviral transduction of beta TC-tet cells exceeded 85%. Lentivector expression of gp19K decreased surface class I major histocompatibility complex expression by over 90%, whereas RID alpha/beta expression inhibited cytokine-induced Fas upregulation by over 75%. beta TC-tet cells transduced with gp19K and RID alpha/beta lentivectors, but not with a control lentivector, provided prolonged correction of hyperglycemia after transplantation into diabetic BALB/c severe combined immunodeficient mice reconstituted with allogeneic immune effector cells or into diabetic allogeneic BALB/c mice. Thus, genetic engineering of beta-cells using gp19K- and RID alpha/beta-expressing lentiviral vectors may provide an alternative that has the potential to eliminate or reduce treatment with the potent immunosuppressive agents necessary at present for prolonged engraftment with transplanted islets.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Ingeniería Genética/métodos , Rechazo de Injerto/prevención & control , Células Secretoras de Insulina/inmunología , Trasplante de Islotes Pancreáticos/métodos , Proteínas E3 de Adenovirus/genética , Proteínas E3 de Adenovirus/inmunología , Proteínas Precoces de Adenovirus/genética , Proteínas Precoces de Adenovirus/inmunología , Animales , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos , Rechazo de Injerto/inmunología , Tolerancia Inmunológica , Lentivirus/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Transducción Genética
5.
J Cell Biol ; 127(6 Pt 1): 1743-54, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7528222

RESUMEN

Hepatocyte growth factor (HGF) is a mesenchymal derived growth factor known to induce proliferation and "scattering" of epithelial and endothelial cells. Its receptor is the tyrosine kinase encoded by the c-MET protooncogene. Here we show that highly purified recombinant HGF stimulates hemopoietic progenitors to form colonies in vitro. In the presence of erythropoietin, picomolar concentrations of HGF induced the formation of erythroid burst-forming unit colonies from CD34-positive cells purified from human bone marrow, peripheral blood, or umbilical cord blood. The growth stimulatory activity was restricted to the erythroid lineage. HGF also stimulated the formation of multipotent CFU-GEMM colonies. This effect is synergized by stem cell factor, the ligand of the tyrosine kinase receptor encoded by the c-KIT protooncogene, which is active on early hemopoietic progenitors. By flow cytometry analysis, the receptor for HGF was found to be expressed on the cell surface in a fraction of CD34+ progenitors. Moreover, in situ hybridization experiments showed that HGF receptor mRNA is highly expressed in embryonic erythroid cells (megaloblasts). HGF mRNA was also found to be produced in the embryonal liver. These data show that HGF plays a direct role in the control of proliferation and differentiation of erythroid progenitors, and they suggest that it may be one of the long-sought mediators of paracrine interactions between stromal and hemopoietic cells within the hemopoietic microenvironment.


Asunto(s)
Células Precursoras Eritroides/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Animales , Antígenos CD , Antígenos CD34 , Baculoviridae/genética , Médula Ósea/embriología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Factores de Crecimiento de Célula Hematopoyética/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Factor de Crecimiento de Hepatocito/genética , Humanos , Hibridación in Situ , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-kit , Proteínas Proto-Oncogénicas c-met , ARN Mensajero/análisis , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores del Factor Estimulante de Colonias/metabolismo , Proteínas Recombinantes/farmacología , Spodoptera/citología , Factor de Células Madre
6.
Mol Biol Cell ; 12(5): 1341-52, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11359926

RESUMEN

Macrophage stimulating protein (MSP), also known as hepatocyte growth factor-like, is a soluble cytokine that belongs to the family of the plasminogen-related growth factors (PRGFs). PRGFs are alpha/beta heterodimers that bind to transmembrane tyrosine kinase receptors. MSP was originally isolated as a chemotactic factor for peritoneal macrophages. Through binding to its receptor, encoded by the RON gene, it stimulates dissociation of epithelia and works as an inflammatory mediator by repressing the production of nitric oxide (NO). Here, we identify a novel role for MSP in the central nervous system. As a paradigm to analyze this function we chose the hypoglossal system of adult mice. We demonstrate in vivo that either administration of exogenous MSP or transplantation of MSP-producing cells at the proximal stump of the resected nerve is sufficient to prevent motoneuron atrophy upon axotomy. We also show that the MSP gene is expressed in the tongue, the target of the hypoglossal nerve, and that MSP induces biosynthesis of Ron receptor in the motoneuron somata. Finally, we show that MSP suppresses NO production in the injured hypoglossal nuclei. Together, these data suggest that MSP is a novel neurotrophic factor for cranial motoneurons and, by regulating the production of NO, may have a role in brain plasticity and regeneration.


Asunto(s)
Encéfalo/metabolismo , Sustancias de Crecimiento/fisiología , Factor de Crecimiento de Hepatocito , Neuronas Motoras/metabolismo , Factores de Crecimiento Nervioso/farmacología , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Axotomía , Colina O-Acetiltransferasa/metabolismo , Sustancias de Crecimiento/genética , Sustancias de Crecimiento/metabolismo , Nervio Hipogloso/citología , Nervio Hipogloso/metabolismo , Inmunohistoquímica , Hibridación in Situ , Ratones , Modelos Neurológicos , Neuronas Motoras/efectos de los fármacos , Factores de Crecimiento Nervioso/genética , Neuronas/química , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Superficie Celular/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Lengua/inervación , Lengua/metabolismo
7.
Mol Biol Cell ; 7(4): 495-504, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8730094

RESUMEN

The mammalian RON and the avian sea genes encode tyrosine kinase receptors of poorly characterized biological functions. We recently identified macrophage-stimulating protein as the ligand for Ron; no ligand has yet been found for Sea. In this work we investigated the biological response to macrophage-stimulating protein in mouse liver progenitor cells expressing Ron. These cells were also transfected with a chimeric cDNA encoding the cytoplasmic domain of Sea, fused to the extracellular domain of Trk (nerve growth factor receptor). In the presence of nanomolar concentrations of the respective ligands, both receptors induced cell "scattering", extracellular matrix invasion, and DNA synthesis. When liver progenitor cells were grown in a tri-dimensional type-I collagen matrix, ligand-induced stimulation of either Ron or Sea induced sprouting of branched cell cords, evolving into ductular-like tubules. The motogenic, mitogenic, and morphogenic responses were also elicited by triggering the structurally related hepatocyte growth factor receptor (Met) but not epidermal growth factor or platelet-derived growth factor receptors. These data show that Ron, Sea, and Met belong to a receptor subfamily that elicits a distinctive biological response in epithelial cells.


Asunto(s)
Proteínas Aviares , Hígado/citología , Proteínas Tirosina Quinasas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Superficie Celular/fisiología , Células Madre/citología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , Diferenciación Celular/fisiología , Células Cultivadas , Cricetinae , Replicación del ADN , Técnicas In Vitro , Ratones , Microscopía Electrónica , Datos de Secuencia Molecular , Fenotipo , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptor trkA , Receptores de Superficie Celular/genética , Receptores de Factor de Crecimiento Nervioso/genética , Proteínas Recombinantes de Fusión/genética , Transfección
8.
J Thromb Haemost ; 15(11): 2188-2197, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28834196

RESUMEN

Essentials The Royal disease (RD) is a form of hemophilia B predicted to be caused by a splicing mutation. We generated an iPSC-based model of the disease allowing mechanistic studies at the RNA level. F9 mRNA analysis in iPSC-derived hepatocyte-like cells showed the predicted abnormal splicing. Mutated F9 mRNA level was very low but we also found traces of wild type transcripts. SUMMARY: Background The royal disease is a form of hemophilia B (HB) that affected many descendants of Queen Victoria in the 19th and 20th centuries. It was found to be caused by the mutation F9 c.278-3A>G. Objective To generate a physiological cell model of the disease and to study F9 expression at the RNA level. Methods Using fibroblasts from skin biopsies of a previously identified hemophilic patient bearing the F9 c.278-3A>G mutation and his mother, we generated induced pluripotent stem cells (iPSCs). Both the patient's and mother's iPSCs were differentiated into hepatocyte-like cells (HLCs) and their F9 mRNA was analyzed using next-generation sequencing (NGS). Results and Conclusion We demonstrated the previously predicted aberrant splicing of the F9 transcript as a result of an intronic nucleotide substitution leading to a frameshift and the generation of a premature termination codon (PTC). The F9 mRNA level in the patient's HLCs was significantly reduced compared with that of his mother, suggesting that mutated transcripts undergo nonsense-mediated decay (NMD), a cellular mechanism that degrades PTC-containing mRNAs. We also detected small proportions of correctly spliced transcripts in the patient's HLCs, which, combined with genetic variability in splicing and NMD machineries, could partially explain some clinical variability among affected members of the European royal families who had lifespans above the average. This work allowed the demonstration of the pathologic consequences of an intronic mutation in the F9 gene and represents the first bona fide cellular model of HB allowing the study of rare mutations at the RNA level.


Asunto(s)
Factor IX/genética , Hemofilia B/genética , Hepatocitos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , ARN Mensajero/genética , Adolescente , Empalme Alternativo , Diferenciación Celular , Línea Celular , Factor IX/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Hemofilia B/sangre , Hemofilia B/diagnóstico , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Fenotipo , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN
9.
Oncogene ; 11(12): 2627-37, 1995 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-8545120

RESUMEN

We previously showed that the proto-oncogene RON encodes the tyrosine kinase receptor for Macrophage Stimulating Protein (MSP), originally isolated as a chemotactic factor for peritoneal macrophages. To elucidate the biological role of MSP we studied the expression of the Ron receptor in vivo, and the response to the factor in vitro. RON specific transcripts were detectable in mouse liver from early embryonal life (day 12.5 p.c.) through adult life. Adrenal gland, spinal ganglia, skin, lung and--unexpectedly--ossification centers of developing mandible, clavicle and ribs were also positive at later stages (day 13.5-16.5 p.c.). From day 17.5 RON was expressed in the gut epithelium and in a specific area of the central nervous system, corresponding to the nucleus of the hypoglossus. In adult mouse tissues RON transcripts were observed in brain, adrenal glands, gastro-intestinal tract, testis and kidney. Epithelial, osteoclast-like and neuroendocrine cells express the Ron receptor and respond to MSP in vitro. In the neuroendocrine PC12 cell line, while NGF induced growth arrest and morphological differentiation, MSP behaved as a strong mitogen. These findings show that the Ron receptor and its ligand are involved in the development of epithelial tissues, bones, and neuroendocrine derivatives driving cells towards the proliferation program.


Asunto(s)
Desarrollo Óseo , Sistemas Neurosecretores/embriología , Proto-Oncogenes/fisiología , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Superficie Celular/genética , Animales , Secuencia de Bases , Clonación Molecular , Epitelio/embriología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met , ARN Mensajero/análisis , Transcripción Genética
10.
Oncogene ; 19(27): 3041-9, 2000 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-10871856

RESUMEN

Scatter Factors control a complex genetic program known as 'invasive growth'. HGF (Scatter factor 1) and MSP (Scatter Factor 2) bind to tyrosine kinase receptors encoded by the proto-oncogenes MET and RON. Using the appropriate 'kinase inactive' mutant receptors, we show that ligand-induced activation of Met results in transphosphorylation of Ron, and vice versa. Transphosphorylation is direct, as it occurs in Met or Ron receptors lacking the docking sites for signal transducers. Phosphate groups are transferred to the tyrosine phosphorylation sites responsible both for kinase up-regulation (Met: Y1234/Y1235 and Ron: Y1238/Y1239) and for generation of signal transducer docking sites (Met: Y1349/Y1356 and Ron Y1353/Y1360). The transphosphorylation specifically takes place for the receptor subfamily, as it is not observed between Met or Ron and ErbB1, ErbB2 or TrkA. Cross-linking experiments show that non-covalent Met-Ron complexes are present on the cell surface, before ligand-induced dimerization. Co-expression of a kinase inactive Ron receptor with naturally-occurring oncogenic Met mutants suppresses the transforming phenotype, suggesting a dominant negative role for the inefficient kinase partner. These data show that, while specific for their ligands, scatter factor receptors cross-talk and cooperate in intracellular signaling.


Asunto(s)
Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Superficie Celular/metabolismo , Células 3T3 , Animales , Western Blotting , Células COS , Dimerización , Regulación de la Expresión Génica , Ratones , Mutagénesis Sitio-Dirigida , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Superficie Celular/genética , Células Tumorales Cultivadas
11.
J Thromb Haemost ; 3(9): 2022-31, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16102109

RESUMEN

BACKGROUND: The deficiency of factor VIII, a co-factor in the intrinsic coagulation pathway results in hemophilia A. Although FVIII is synthesized largely in the liver, the specific liver cell type(s) responsible for FVIII production is controversial. OBJECTIVE: This study aimed to determine the cellular origin of FVIII synthesis and release in mouse models. METHODS: We transplanted cells into the peritoneal cavity of hemophilia A knockout mice. Plasma FVIII activity was measured using a Chromogenix assay 2-7 days after cell transplantation, and phenotypic correction was determined with tail-clip challenge 7 days following cell transplantation. Transplanted cells were identified by histologic and molecular assays. RESULTS: Untreated hemophilia A mice, as well as mice treated with the hepatocyte-enriched fraction, showed extensive mortality following tail-clip challenge. In contrast, recipients of unfractionated liver cells (mixture of hepatocytes, liver sinusoidal endothelial cells (LSEC), Kupffer cells, and hepatic stellate cells) or of the cell fraction enriched in LSECs survived tail-clip challenge (P < 0.001). FVIII was secreted in the blood stream in recipients of unfractionated liver cells, LSECs and pancreatic islet-derived MILE SVEN 1 (MS1) endothelial cells. Although transplanted hepatocytes maintained functional integrity in the peritoneal cavity, these cells did not produce detectable plasma FVIII activity. CONCLUSIONS: The assay of cell transplantation in the peritoneal cavity showed that endothelial cells but not hepatocytes produced phenotypic correction in hemophilia A mice. Therefore, endothelial cells should be suitable additional targets for cell and gene therapy in hemophilia A.


Asunto(s)
Trasplante de Células , Células Endoteliales/trasplante , Factor VIII/biosíntesis , Hemofilia A/terapia , Hígado/citología , Animales , Células Endoteliales/metabolismo , Factor VIII/administración & dosificación , Supervivencia de Injerto , Hepatocitos/trasplante , Hígado/metabolismo , Ratones , Ratones Noqueados
12.
Hum Gene Ther ; 13(2): 243-60, 2002 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-11812281

RESUMEN

Safe and efficient genetic modification of liver cells could enable new therapies for a variety of hepatic and systemic diseases. Lentiviral vectors are promising tools for in vivo gene delivery. Previous data suggested that recruitment into the cell cycle was required for transduction of hepatocytes in vivo. We developed an improved vector design that enhanced nuclear translocation in target cells and significantly improved gene transfer performance. Using the new vector and a panel of internal promoters, we showed that rat hepatocytes were transduced ex vivo to high frequency without requirement for proliferation. On intravenous administration of vector into adult severe combined immunodeficient (SCID) mice, we found high levels (up to 30%) of transduction of parenchymal and nonparenchymal cells of the liver, integration of the vector genome in liver DNA and stable expression of the marker green fluorescent protein (GFP)-encoding gene without signs of toxicity. Coadministration of vectors and 5'-bromo-2'-deoxyuridine in vivo proved that cell cycling was not required for efficient transduction of hepatocytes. In addition to the liver, the spleen and the bone marrow were transduced effectively by systemic delivery of vector. GFP expression was observed in all these organs when driven by the cytomegalovirus promoter and by the phosphoglycerate kinase gene promoter. Using the promoter of the albumin gene, we could restrict expression to hepatocytes. By a single vector injection into the bloodstream of SCID mice, we achieved therapeutic-range levels of the human clotting factor IX, stable in the plasma for up to 1 year (the longest time tested), indicating the potential efficacy of improved lentiviral vectors for the gene therapy of hemophilias and other diseases.


Asunto(s)
Vectores Genéticos , Hepatocitos , Lentivirus/genética , Regiones Promotoras Genéticas , Transducción Genética , Animales , Factor IX/genética , Femenino , Expresión Génica , Genes pol , Terapia Genética/métodos , Células HeLa , Factor de Crecimiento de Hepatocito , Humanos , Ratones , Ratones SCID , Ratas , Transgenes
14.
J Biol Chem ; 270(2): 603-11, 1995 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-7822285

RESUMEN

Hepatocyte growth factor (HGF) is a paracrine inducer of morphogenesis and invasive growth in epithelial and endothelial cells. HGF is secreted by mesenchymal cells as an inactive precursor (pro-HGF). The crucial step for HGF activation is the extracellular hydrolysis of the Arg494-Val495 bond, which converts pro-HGF into alpha beta-HGF, the high-affinity ligand for the Met receptor. We previously reported that the urokinase-type plasminogen activator (uPA) activates pro-HGF in vitro. We now show that this is a stoichiometric reaction, and provide evidence for its occurrence in tissue culture. Activation involves the formation of a stable complex between pro-HGF and uPA. This complex was isolated from the in vitro reaction of pure uPA with recombinant pro-HGF, as well as from the membrane of target cells, after sequential addition of uPA and pro-HGF. On the cell membrane, the uPA-HGF complex was bound to the Met receptor. Monocytic cell lines, and primary monocytes after adhesion, activated efficiently pro-HGF both on their surface and in the culture medium. This activation was inhibited by anti-catalytic anti-uPA antibodies, and occurred by a stoichiometric reaction. The stoichiometry of the activation reaction suggests that the biological effects of HGF can be titrated in vivo by the level of uPA activity. Adequate amounts of uPA can be locally provided by the macrophages, which would condition the tissue microenvironment by rendering HGF bioavailable to its target cells.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Precursores de Proteínas/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Células Cultivadas , Humanos , Cinética , Procesamiento Proteico-Postraduccional , Relación Estructura-Actividad
15.
EMBO J ; 13(15): 3524-32, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-8062829

RESUMEN

RON, a cDNA homologous to the hepatocyte growth factor (HGF) receptor gene (MET), encodes a putative tyrosine kinase. Here we show that the RON gene is expressed in several epithelial tissues as well as in granulocytes and monocytes. The major RON transcript is translated into a glycosylated single chain precursor, cleaved into a 185 kDa heterodimer (p185RON) of 35 (alpha) and 150 kDa (beta) disulfide-linked chains, before exposure at the cell surface. The Ron beta-chain displays intrinsic tyrosine kinase activity in vitro, after immunoprecipitation by specific antibodies. In vivo, tyrosine phosphorylation of p185RON is induced by stimulation with macrophage stimulating protein (MSP), a protease-like factor containing four 'kringle' domains, homologous to HGF. In epithelial cells, MSP-induced tyrosine phosphorylation of p185RON is followed by DNA synthesis. p185RON is not activated by HGF, nor is the HGF receptor activated by MSP in biochemical and biological assays. p185RON is also activated by a pure recombinant protein containing only the N-terminal two kringles of MSP. These data show that p185RON is a tyrosine kinase activated by MSP and that it is member of a family of growth factor receptors with distinct specificities for structurally related ligands.


Asunto(s)
Sustancias de Crecimiento/farmacología , Conformación Proteica , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Superficie Celular/metabolismo , Secuencia de Bases , Movimiento Celular/efectos de los fármacos , Clonación Molecular , Replicación del ADN/efectos de los fármacos , Células Epiteliales , Sustancias de Crecimiento/genética , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Kringles , Datos de Secuencia Molecular , Especificidad de Órganos , Fosforilación/efectos de los fármacos , Precursores de Proteínas/biosíntesis , ARN Mensajero/análisis , Proteínas Tirosina Quinasas Receptoras/química , Receptores de Superficie Celular/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Transcripción Genética , Células Tumorales Cultivadas , Tirosina/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 15(11): 1857-65, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7583565

RESUMEN

Hepatocyte growth factor (HGF) is a powerful motogen and mitogen for epithelial cells. The factor is a 90-kD heterodimer composed of an alpha chain containing four kringle motifs and a beta chain showing structural homologies with serine proteases. It is, however, devoid of enzymatic activity. Recently, it has been reported that HGF activates migration and proliferation of endothelial cells and is angiogenic. In this article we discuss (1) the molecular domains of HGF required to activate in vitro and in vivo endothelial cells, studied by use of molecular mutants, and (2) the characteristics of the angiogenic response to HGF in an experimental model system of implanted reconstituted basement membrane (Matrigel). Two groups of mutants were made and used in vitro and in vivo: one with deletions of kringle domains and one with substitution at the cleavage site of the HGF precursor. In vitro, HGF variants containing only the first two (HGF-NK2) or the first three kringles (HGF-NK3) of the alpha chain did not induce proliferation of endothelial cells even if used at concentration 160-fold higher than that optimal for HGF (0.05 nmol/L). High concentrations of these mutants (4 to 8 nmol/L) activated a little endothelial cell motogenic response that was 60% lower than that elicited by HGF. Substitution of Arg 489 with Gln 489 in the HGF precursor generated an uncleavable single-chain factor, unable to induce either endothelial cell migration or proliferation. In vivo, HGF induced a dose-dependent angiogenic response, which was enhanced by heparin.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Animales , Secuencia de Bases , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Sinergismo Farmacológico , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Activación Enzimática , Femenino , Heparina/farmacología , Factor de Crecimiento de Hepatocito/genética , Humanos , Ratones , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Mutación
17.
Gene Ther ; 10(8): 630-6, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12692591

RESUMEN

Efficient gene transduction in cardiomyocytes is a task that can be accomplished only by viral vectors. Up to now, the most commonly used vectors for this purpose have been adenoviral-derived ones. Recently, it has been demonstrated that lentiviral vectors can transduce growth-arrested cells, such as hematopoietic stem cells. Moreover, a modified form of lentiviral vector (the 'advanced' generation), containing an mRNA-stabilizer sequence and a nuclear import sequence, has been shown to significantly improve gene transduction in growth-arrested cells as compared to the third-generation vector. Therefore, we tested whether the 'advanced' generation lentivirus is capable of infecting and transducing cardiomyocytes both in vitro and in vivo, comparing efficacy in vitro against the third-generation of the same vector. Here we report that 'advanced' generation lentiviral vectors infected most (>80%) cardiomyocytes in culture, as demonstrated by immunofluorescence and FACS analyses: in contrast the percentage of cardiomyocytes infected by third-generation lentivirus was three- to four-fold lower. Moreover, 'advanced' generation lentivirus was also capable of infecting and inducing stable gene expression in adult myocardium in vivo. Thus, 'advanced' generation lentiviral vectors can be used for both in vitro and in vivo gene expression studies in the cardiomyocyte.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Terapia Genética/métodos , Vectores Genéticos/farmacología , Lentivirus/genética , Miocitos Cardíacos/metabolismo , Transducción Genética/métodos , Animales , Línea Celular , Citometría de Flujo , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Microscopía Fluorescente , Ratas
18.
Proc Natl Acad Sci U S A ; 98(19): 10733-8, 2001 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-11535818

RESUMEN

The concept of tissue-restricted differentiation of postnatal stem cells has been challenged by recent evidence showing pluripotency for hematopoietic, mesenchymal, and neural stem cells. Furthermore, rare but well documented examples exist of already differentiated cells in developing mammals that change fate and trans-differentiate into another cell type. Here, we report that endothelial cells, either freshly isolated from embryonic vessels or established as homogeneous cells in culture, differentiate into beating cardiomyocytes and express cardiac markers when cocultured with neonatal rat cardiomyocytes or when injected into postischemic adult mouse heart. Human umbilical vein endothelial cells also differentiate into cardiomyocytes under similar experimental conditions and transiently coexpress von Willebrand factor and sarcomeric myosin. In contrast, neural stem cells, which efficiently differentiate into skeletal muscle, differentiate into cardiomyocytes at a low rate. Fibroblast growth factor 2 and bone morphogenetic protein 4, which activate cardiac differentiation in embryonic cells, do not activate cardiogenesis in endothelial cells or stimulate trans-differentiation in coculture, suggesting that different signaling molecules are responsible for cardiac induction during embryogenesis and in successive periods of development. The fact that endothelial cells can generate cardiomyocytes sheds additional light on the plasticity of endothelial cells during development and opens perspectives for cell autologous replacement therapies.


Asunto(s)
Endotelio Vascular/citología , Corazón/fisiología , Miocardio/citología , Regeneración/fisiología , Animales , Aorta/citología , Diferenciación Celular , Células Cultivadas , Humanos , Ratones , Isquemia Miocárdica , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA