Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Semin Cell Dev Biol ; 29: 125-47, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24662892

RESUMEN

Lamin A-related progeroid syndromes are genetically determined, extremely rare and severe. In the past ten years, our knowledge and perspectives for these diseases has widely progressed, through the progressive dissection of their pathophysiological mechanisms leading to precocious and accelerated aging, from the genes mutations discovery until therapeutic trials in affected children. A-type lamins are major actors in several structural and functional activities at the nuclear periphery, as they are major components of the nuclear lamina. However, while this is usually poorly considered, they also play a key role within the rest of the nucleoplasm, whose defects are related to cell senescence. Although nuclear shape and nuclear envelope deformities are obvious and visible events, nuclear matrix disorganization and abnormal composition certainly represent the most important causes of cell defects with dramatic pathological consequences. Therefore, lamin-associated diseases should be better referred as laminopathies instead of envelopathies, this later being too restrictive, considering neither the key structural and functional roles of soluble lamins in the entire nucleoplasm, nor the nuclear matrix contribution to the pathophysiology of lamin-associated disorders and in particular in defective lamin A processing-associated aging diseases. Based on both our understanding of pathophysiological mechanisms and the biological and clinical consequences of progeria and related diseases, therapeutic trials have been conducted in patients and were terminated less than 10 years after the gene discovery, a quite fast issue for a genetic disease. Pharmacological drugs have been repurposed and used to decrease the toxicity of the accumulated, unprocessed and truncated prelaminA in progeria. To date, none of them may be considered as a cure for progeria and these clinical strategies were essentially designed toward reducing a subset of the most dramatic and morbid features associated to progeria. New therapeutic strategies under study, in particular targeting the protein expression pathway at the mRNA level, have shown a remarkable efficacy both in vitro in cells and in vivo in mice models. Strategies intending to clear the toxic accumulated proteins from the nucleus are also under evaluation. However, although exceedingly rare, improving our knowledge of genetic progeroid syndromes and searching for innovative and efficient therapies in these syndromes is of paramount importance as, even before they can be used to save lives, they may significantly (i) expand the affected childrens' lifespan and preserve their quality of life; (ii) improve our understanding of aging-related disorders and other more common diseases; and (iii) expand our fundamental knowledge of physiological aging and its links with major physiological processes such as those involved in oncogenesis.


Asunto(s)
Envejecimiento Prematuro/patología , Lamina Tipo A/genética , Lámina Nuclear/genética , Progeria/patología , Envejecimiento Prematuro/genética , Animales , Ataxia Telangiectasia/genética , Senescencia Celular/genética , Reparación del ADN/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Progeria/genética , Procesamiento Proteico-Postraduccional , Investigación Biomédica Traslacional
2.
Semin Cell Dev Biol ; 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24685615

RESUMEN

The Publisher regrets that this article is an accidental duplication of an article that has already been published, http://dx.doi.org/10.1016/j.semcdb.2014.03.022. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

3.
J Biol Chem ; 288(13): 8991-9000, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23389031

RESUMEN

The melanoma cell adhesion molecule (CD146) contains a circulating proteolytic variant (sCD146), which is involved in inflammation and angiogenesis. Its circulating level is modulated in different pathologies, but its intracellular transduction pathways are still largely unknown. Using peptide pulldown and mass spectrometry, we identified angiomotin as a sCD146-associated protein in endothelial progenitor cells (EPC). Interaction between angiomotin and sCD146 was confirmed by enzyme-linked immunosorbent assay (ELISA), homogeneous time-resolved fluorescence, and binding of sCD146 on both immobilized recombinant angiomotin and angiomotin-transfected cells. Silencing angiomotin in EPC inhibited sCD146 angiogenic effects, i.e. EPC migration, proliferation, and capacity to form capillary-like structures in Matrigel. In addition, sCD146 effects were inhibited by the angiomotin inhibitor angiostatin and competition with recombinant angiomotin. Finally, binding of sCD146 on angiomotin triggered the activation of several transduction pathways that were identified by antibody array. These results delineate a novel signaling pathway where sCD146 binds to angiomotin to stimulate a proangiogenic response. This result is important to find novel target cells of sCD146 and for the development of therapeutic strategies based on EPC in the treatment of ischemic diseases.


Asunto(s)
Antígeno CD146/sangre , Endotelio Vascular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Patológica , Células Madre/citología , Angiomotinas , Angiostatinas/metabolismo , Capilares/metabolismo , Colágeno/química , Combinación de Medicamentos , Células Endoteliales/citología , Ensayo de Inmunoadsorción Enzimática/métodos , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Laminina/química , Espectrometría de Masas/métodos , Proteínas de Microfilamentos , Microscopía Fluorescente/métodos , Proteoglicanos/química , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Espectrometría de Fluorescencia/métodos , Cicatrización de Heridas
4.
Arterioscler Thromb Vasc Biol ; 32(4): 1054-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22328775

RESUMEN

OBJECTIVE: Cellular microparticles (MP) are promising biomarkers in many pathological situations. Although flow cytometry (FCM) is widely used for their measurement, it has raised controversies because the smallest MP size falls below the detection limit of standard FCM (sd-FCM). Following recent technological improvements leading to high sensitivity FCM (hs-FCM), our objectives were (1) to evaluate the potential of hs-FCM for extended MP detection, (2) to set up a standardized protocol for MP enumeration, and (3) to compare MP counts obtained with both sensitivity levels. METHODS AND RESULTS: Compared with sd-FCM, hs-FCM displayed improved forward scatter resolution and lower background noise, allowing us to discriminate previously undetectable small MP in plasma samples. Using fluorescent beads with appropriate sizes (0.1/0.3/0.5/0.9 µm) and relative amounts, a new standardized hs-FCM MP protocol was set up and provided reproducible MP counts. Applied to coronary patient samples, it resulted into 8- to 20-fold increases in MP counts as compared with sd-FCM. Interestingly, the ratio between small and large MP varied according to clinical status but also depending on MP subset, suggesting access to new biological information. CONCLUSIONS: Recent improvements in FCM provide access to previously undetectable MP and represent a new opportunity to enhance their impact as biomarkers in clinical practice.


Asunto(s)
Micropartículas Derivadas de Células/patología , Enfermedad Coronaria/patología , Citometría de Flujo , Biomarcadores/sangre , Calibración , Micropartículas Derivadas de Células/química , Enfermedad Coronaria/sangre , Citometría de Flujo/métodos , Citometría de Flujo/normas , Humanos , Tamaño de la Partícula , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
5.
Blood ; 115(18): 3843-51, 2010 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-20185588

RESUMEN

CD146, an endothelial molecule involved in permeability and monocyte transmigration, has recently been reported to promote vessel growth. As CD146 is also detectable as a soluble form (sCD146), we hypothesized that sCD146 could stimulate angiogenesis. Experiments of Matrigel plugs in vivo showed that sCD146 displayed chemotactic activity on endogenous endothelial cells, and exogenously injected late endothelial progenitor cells (EPCs). Recruited endothelial cells participated in formation of vascular-like structures. In vitro, sCD146 enhanced angiogenic properties of EPCs, with an increased cell migration, proliferation, and capacity to establish capillary-like structures. Effects were additive with those of vascular endothelial growth factor (VEGF), and sCD146 enhanced VEGFR2 expression and VEGF secretion. Consistent with a proangiogenic role, gene expression profiling of sCD146-stimulated EPCs revealed an up-regulation of endothelial nitric oxide synthase, urokinase plasminogen activator, matrix metalloproteinase 2, and VEGFR2. Silencing membrane-bound CD146 inhibited responses. The potential therapeutic interest of sCD146 was tested in a model of hind limb ischemia. Local injections of sCD146 significantly reduced auto-amputation, tissue necrosis, fibrosis, inflammation, and increased blood flow. Together, these findings establish that sCD146 displays chemotactic and angiogenic properties and promotes efficient neovascularization in vivo. Recombinant human sCD146 might thus support novel strategies for therapeutic angiogenesis in ischemic diseases.


Asunto(s)
Biomarcadores/metabolismo , Antígeno CD146/metabolismo , Células Endoteliales/metabolismo , Miembro Posterior/irrigación sanguínea , Isquemia/metabolismo , Neovascularización Fisiológica , Animales , Western Blotting , Antígeno CD146/genética , Citometría de Flujo , Perfilación de la Expresión Génica , Miembro Posterior/metabolismo , Humanos , Isquemia/etiología , Isquemia/terapia , Masculino , Ratones , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cicatrización de Heridas
6.
Circ Res ; 107(1): 66-75, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20448216

RESUMEN

RATIONALE: CD146, a transmembrane immunoglobulin mainly expressed at the intercellular junction of endothelial cells, is involved in cell-cell cohesion, paracellular permeability, monocyte transmigration and angiogenesis. CD146 exists as 2 isoforms, short (sh) and long (lg), but which isoform is involved remains undefined. OBJECTIVE: The recently described role of CD146 in angiogenesis prompted us to investigate which isoform was involved in this process in human late endothelial progenitors (EPCs), with the objective of increasing their proangiogenic potential. METHODS AND RESULTS: Immunofluorescence experiments showed that, in subconfluent EPCs, shCD146 was localized in the nucleus and at the migrating edges of the membrane, whereas lgCD146 was intracellular. In confluent cells, shCD146 was redistributed at the apical membrane and lgCD146 was directed toward the junction. In contrast to lgCD146, shCD146 was overexpressed in EPCs as compared to mature endothelial cells and upregulated by vascular endothelial growth factor and SDF-1 (stromal cell-derived factor 1). Study of the properties of both isoforms in vitro provided evidence that shCD146 was involved in EPC adhesion to activated endothelium, migration, and proliferation, with a paracrine secretion of interleukin-8 or angiopoietin 2, whereas lgCD146 was implicated in stabilization of capillary-like structures in Matrigel and transendothelial permeability. In an animal model of hindlimb ischemia, transplantation of shCD146-modified EPCs selectively promoted both EPC engraftment and blood flow. CONCLUSIONS: Altogether, these findings establish that CD146 isoforms display distinct functions in vessels regeneration. Selective improvement of therapeutic angiogenesis by shCD146 overexpression suggests a potential interest of shCD146-transduced EPCs for the treatment of peripheral ischemic disease.


Asunto(s)
Antígeno CD146/fisiología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Neovascularización Fisiológica/fisiología , Células Madre/fisiología , Animales , Antígeno CD146/biosíntesis , Endotelio Vascular/trasplante , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/metabolismo , Isquemia/patología , Isquemia/cirugía , Ratones , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/fisiología , Trasplante de Células Madre/métodos
7.
Cells ; 11(4)2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35203262

RESUMEN

Progeroid syndromes (PS), including Hutchinson-Gilford Progeria Syndrome (HGPS), are premature and accelerated aging diseases, characterized by clinical features mimicking physiological aging. Most classical HGPS patients carry a de novo point mutation within exon 11 of the LMNA gene encoding A-type lamins. This mutation activates a cryptic splice site, leading to the production of a truncated prelamin A, called prelamin A ∆50 or progerin, that accumulates in HGPS cell nuclei and is a hallmark of the disease. Some patients with PS carry other LMNA mutations and are named "HGPS-like" patients. They produce progerin and/or other truncated prelamin A isoforms (∆35 and ∆90). We previously found that MG132, a proteasome inhibitor, induced progerin clearance in classical HGPS through autophagy activation and splicing regulation. Here, we show that MG132 induces aberrant prelamin A clearance and improves cellular phenotypes in HGPS-like patients' cells other than those previously described in classical HGPS. These results provide preclinical proof of principle for the use of a promising class of molecules toward a potential therapy for children with HGPS-like or classical HGPS.


Asunto(s)
Progeria , Núcleo Celular , Humanos , Leupeptinas/farmacología , Leupeptinas/uso terapéutico , Fenotipo , Progeria/tratamiento farmacológico , Progeria/genética
8.
Arterioscler Thromb Vasc Biol ; 29(5): 746-53, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19229070

RESUMEN

OBJECTIVES: During inflammation, cell adhesion molecules are modulated or redistributed for leukocyte transmigration. Among molecules at the interendothelial junction, CD146 is involved in cell-cell cohesion and permeability, but its role in monocyte transmigration is unknown. METHODS AND RESULTS: TNF enhanced CD146 expression at the junction and apical membrane of human umbilical veins endothelial cells (HUVECs) through CD146 synthesis and intracellular store redistribution. In addition, TNF increased the release of a soluble form (sCD146) through a metalloproteinase-dependent mechanism. The redistribution of CD146 to the junction led us to investigate its role in monocyte transmigration using THP1 and freshly isolated monocytes. Evidence that CD146 contributes to monocyte transmigration was provided by inhibition experiments using anti-CD146 antibodies and CD146 siRNA in HUVECs. In addition, sCD146 specifically bound both monocytes and HUVECs and dose-dependently increased monocyte transmigration. Assessment of sCD146 binding on immobilized CD146 failed to evidence any homophilic interaction. Together, our data suggest endothelial CD146 binds heterophilically with a yet unknown ligand on monocytes. CONCLUSIONS: Our results demonstrate that CD146 is regulated by the inflammatory cytokine TNF and that CD146 and sCD146 are both involved in monocyte transendothelial migration during inflammation.


Asunto(s)
Quimiotaxis/inmunología , Inflamación/inmunología , Monocitos/inmunología , Antígeno CD146/inmunología , Línea Celular Tumoral , Células Endoteliales/fisiología , Humanos , Factor de Necrosis Tumoral alfa/inmunología , Venas Umbilicales/citología
9.
Nat Commun ; 11(1): 4110, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32807790

RESUMEN

Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disease in children that leads to early death. Smooth muscle cells (SMCs) are the most affected cells in HGPS individuals, although the reason for such vulnerability remains poorly understood. In this work, we develop a microfluidic chip formed by HGPS-SMCs generated from induced pluripotent stem cells (iPSCs), to study their vulnerability to flow shear stress. HGPS-iPSC SMCs cultured under arterial flow conditions detach from the chip after a few days of culture; this process is mediated by the upregulation of metalloprotease 13 (MMP13). Importantly, double-mutant LmnaG609G/G609GMmp13-/- mice or LmnaG609G/G609GMmp13+/+ mice treated with a MMP inhibitor show lower SMC loss in the aortic arch than controls. MMP13 upregulation appears to be mediated, at least in part, by the upregulation of glycocalyx. Our HGPS-SMCs chip represents a platform for developing treatments for HGPS individuals that may complement previous pre-clinical and clinical treatments.


Asunto(s)
Metaloproteinasa 13 de la Matriz/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Biotecnología/métodos , Enfermedades Cardiovasculares/metabolismo , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Masculino , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Ratones Mutantes , Miocitos del Músculo Liso/efectos de los fármacos , Progeria/metabolismo , Progeria/patología , Proteómica/métodos
10.
Nat Commun ; 11(1): 4589, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917887

RESUMEN

Mandibuloacral dysplasia syndromes are mainly due to recessive LMNA or ZMPSTE24 mutations, with cardinal nuclear morphological abnormalities and dysfunction. We report five homozygous null mutations in MTX2, encoding Metaxin-2 (MTX2), an outer mitochondrial membrane protein, in patients presenting with a severe laminopathy-like mandibuloacral dysplasia characterized by growth retardation, bone resorption, arterial calcification, renal glomerulosclerosis and severe hypertension. Loss of MTX2 in patients' primary fibroblasts leads to loss of Metaxin-1 (MTX1) and mitochondrial dysfunction, including network fragmentation and oxidative phosphorylation impairment. Furthermore, patients' fibroblasts are resistant to induced apoptosis, leading to increased cell senescence and mitophagy and reduced proliferation. Interestingly, secondary nuclear morphological defects are observed in both MTX2-mutant fibroblasts and mtx-2-depleted C. elegans. We thus report the identification of a severe premature aging syndrome revealing an unsuspected link between mitochondrial composition and function and nuclear morphology, establishing a pathophysiological link with premature aging laminopathies and likely explaining common clinical features.


Asunto(s)
Acroosteólisis/metabolismo , Predisposición Genética a la Enfermedad/genética , Lipodistrofia/metabolismo , Mandíbula/anomalías , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Acroosteólisis/diagnóstico por imagen , Acroosteólisis/genética , Acroosteólisis/patología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Animales , Apoptosis , Caenorhabditis elegans , Proliferación Celular , Niño , Regulación hacia Abajo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Genotipo , Homocigoto , Humanos , Lipodistrofia/diagnóstico por imagen , Lipodistrofia/genética , Lipodistrofia/patología , Masculino , Mandíbula/diagnóstico por imagen , Proteínas de la Membrana/genética , Metaloendopeptidasas , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas Mitocondriales/genética , Mutación , Fenotipo , Piel , Secuenciación Completa del Genoma
12.
Aging Cell ; 17(4): e12766, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29696758

RESUMEN

Hereditary and sporadic laminopathies are caused by mutations in genes encoding lamins, their partners, or the metalloprotease ZMPSTE24/FACE1. Depending on the clinical phenotype, they are classified as tissue-specific or systemic diseases. The latter mostly manifest with several accelerated aging features, as in Hutchinson-Gilford progeria syndrome (HGPS) and other progeroid syndromes. MicroRNAs are small noncoding RNAs described as powerful regulators of gene expression, mainly by degrading target mRNAs or by inhibiting their translation. In recent years, the role of these small RNAs has become an object of study in laminopathies using in vitro or in vivo murine models as well as cells/tissues of patients. To date, few miRNAs have been reported to exert protective effects in laminopathies, including miR-9, which prevents progerin accumulation in HGPS neurons. The recent literature has described the potential implication of several other miRNAs in the pathophysiology of laminopathies, mostly by exerting deleterious effects. This review provides an overview of the current knowledge of the functional relevance and molecular insights of miRNAs in laminopathies. Furthermore, we discuss how these discoveries could help to better understand these diseases at the molecular level and could pave the way toward identifying new potential therapeutic targets and strategies based on miRNA modulation.


Asunto(s)
Envejecimiento Prematuro/genética , Laminas/metabolismo , MicroARNs/genética , Progeria/genética , Envejecimiento Prematuro/patología , Animales , Humanos , Progeria/patología
13.
Nucleus ; 9(1): 246-257, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29619863

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a sporadic, autosomal dominant disorder characterized by premature and accelerated aging symptoms leading to death at the mean age of 14.6 years usually due to cardiovascular complications. HGPS is caused by a de novo point mutation in the LMNA gene encoding the intermediate filament proteins lamins A and C which are structural components of the nuclear lamina. This mutation leads to the production of a truncated toxic form of lamin A, issued from aberrant splicing and called progerin. Progerin accumulates in HGPS cells' nuclei and is a hallmark of the disease. Small amounts of progerin are also produced during normal aging. HGPS cells and animal preclinical models have provided insights into the molecular and cellular pathways that underlie the disease and have also highlighted possible mechanisms involved in normal aging. This review reports recent medical advances and treatment approaches for patients affected with HGPS.


Asunto(s)
Progeria/tratamiento farmacológico , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/patología , Humanos , Mutación , Progeria/genética , Progeria/patología
14.
EMBO Mol Med ; 9(9): 1294-1313, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28674081

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a lethal premature and accelerated aging disease caused by a de novo point mutation in LMNA encoding A-type lamins. Progerin, a truncated and toxic prelamin A issued from aberrant splicing, accumulates in HGPS cells' nuclei and is a hallmark of the disease. Small amounts of progerin are also produced during normal aging. We show that progerin is sequestered into abnormally shaped promyelocytic nuclear bodies, identified as novel biomarkers in late passage HGPS cell lines. We found that the proteasome inhibitor MG132 induces progerin degradation through macroautophagy and strongly reduces progerin production through downregulation of SRSF-1 and SRSF-5 accumulation, controlling prelamin A mRNA aberrant splicing. MG132 treatment improves cellular HGPS phenotypes. MG132 injection in skeletal muscle of LmnaG609G/G609G mice locally reduces SRSF-1 expression and progerin levels. Altogether, we demonstrate progerin reduction based on MG132 dual action and shed light on a promising class of molecules toward a potential therapy for children with HGPS.


Asunto(s)
Autofagia/efectos de los fármacos , Leupeptinas/administración & dosificación , Progeria/tratamiento farmacológico , Empalme del ARN/efectos de los fármacos , Animales , Femenino , Humanos , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Masculino , Ratones , Ratones Noqueados , Progeria/genética , Progeria/metabolismo , Progeria/fisiopatología , Proteolisis/efectos de los fármacos , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo
15.
PLoS One ; 12(8): e0183136, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28806747

RESUMEN

The type V intermediate filament lamins are the principal components of the nuclear matrix, including the nuclear lamina. Lamins are divided into A-type and B-type, which are encoded by three genes, LMNA, LMNB1, and LMNB2. The alternative splicing of LMNA produces two major A-type lamins, lamin A and lamin C. Previous studies have suggested that lamins are involved in cancer development and progression. A-type lamins have been proposed as biomarkers for cancer diagnosis, prognosis, and/or follow-up. The aim of the present study was to investigate lamins in cancer cells from metastatic pleural effusions using immunofluorescence, western blotting, and flow cytometry. In a sub-group of lung adenocarcinomas, we found reduced expression of lamin A but not of lamin C. The reduction in lamin A expression was correlated with the loss of epithelial membrane antigen (EMA)/MUC-1, an epithelial marker that is involved in the epithelial to mesenchymal transition (EMT). Finally, the lamin A expression was inversely correlated with the number of metastatic sites and the WHO Performance status, and association of pleural, bone and lung metastatic localizations was more frequent when lamin A expression was reduced. In conclusion, low lamin A but not lamin C expression in pleural metastatic cells could represent a major actor in the development of metastasis, associated with EMT and could account for a pejorative factor correlated with a poor Performance status.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Lamina Tipo A/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Derrame Pleural/metabolismo , Derrame Pleural/patología , Adenocarcinoma del Pulmón , Anciano , Anciano de 80 o más Años , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Persona de Mediana Edad , Mucina-1/metabolismo , Metástasis de la Neoplasia , Organización Mundial de la Salud
16.
Cells ; 5(3)2016 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-27409638

RESUMEN

Progeroid laminopathies, including Hutchinson-Gilford Progeria Syndrome (HGPS, OMIM #176670), are premature and accelerated aging diseases caused by defects in nuclear A-type Lamins. Most HGPS patients carry a de novo point mutation within exon 11 of the LMNA gene encoding A-type Lamins. This mutation activates a cryptic splice site leading to the deletion of 50 amino acids at its carboxy-terminal domain, resulting in a truncated and permanently farnesylated Prelamin A called Prelamin A Δ50 or Progerin. Some patients carry other LMNA mutations affecting exon 11 splicing and are named "HGPS-like" patients. They also produce Progerin and/or other truncated Prelamin A isoforms (Δ35 and Δ90) at the transcriptional and/or protein level. The results we present show that morpholino antisense oligonucleotides (AON) prevent pathogenic LMNA splicing, markedly reducing the accumulation of Progerin and/or other truncated Prelamin A isoforms (Prelamin A Δ35, Prelamin A Δ90) in HGPS-like patients' cells. Finally, a patient affected with Mandibuloacral Dysplasia type B (MAD-B, carrying a homozygous mutation in ZMPSTE24, encoding an enzyme involved in Prelamin A maturation, leading to accumulation of wild type farnesylated Prelamin A), was also included in this study. These results provide preclinical proof of principle for the use of a personalized antisense approach in HGPS-like and MAD-B patients, who may therefore be eligible for inclusion in a therapeutic trial based on this approach, together with classical HGPS patients.

17.
Cardiovasc Res ; 111(3): 240-51, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27170199

RESUMEN

AIMS: Endothelial colony-forming cells (ECFC) constitute an endothelial progenitor fraction with a promising interest for the treatment of ischaemic cardiovascular diseases. As soluble CD146 (sCD146) is a new factor promoting angiogenesis, we examined whether sCD146 priming could improve the therapeutic potential of ECFC and defined the involved mechanism. METHODS AND RESULTS: We investigated the effects of sCD146 priming on regenerative properties of ECFC in vivo. In a mouse model of hindlimb ischaemia, the homing of radiolabelled cells to ischaemic tissue was assessed by SPECT-CT imaging. Soluble CD146 priming did not modify the number of engrafted ECFC but improved their survival capacity, leading to an enhanced revascularization. The mechanism of action of sCD146 on ECFC was studied in vitro. We showed that sCD146 acts in ECFC through a signalosome, located in lipid rafts, containing angiomotin, the short isoform of CD146 (shCD146), VEGFR1, VEGFR2, and presenilin-1. Soluble CD146 induced a sequential proteolytic cleavage of shCD146, with an extracellular shedding followed by an intramembrane cleavage mediated by matrix metalloprotease (MMP)/ADAM and presenilin-1, respectively. The generated intracellular part of shCD146 was directed towards the nucleus where it associated with the transcription factor CSL and modulated the transcription of genes involved in cell survival (FADD, Bcl-xl) and angiogenesis (eNOS). This effect was dependent on both VEGFR1 and VEGFR2, which were rapidly phosphorylated by sCD146. CONCLUSIONS: These findings establish that activation of the proteolytic processing of shCD146, in particular by sCD146, constitutes a promising pathway to improve endothelial progenitors' regenerative properties for the treatment of cardiovascular diseases.


Asunto(s)
Proteínas ADAM/metabolismo , Células Progenitoras Endoteliales/trasplante , Isquemia/cirugía , Metaloproteinasas de la Matriz Asociadas a la Membrana/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Regeneración , Animales , Antígeno CD146/metabolismo , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/enzimología , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Miembro Posterior , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatología , Microdominios de Membrana/metabolismo , Ratones , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Presenilina-1/metabolismo , Isoformas de Proteínas , Proteolisis , Transducción de Señal , Factores de Tiempo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
19.
J Am Coll Cardiol ; 56(13): 1024-31, 2010 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-20846600

RESUMEN

OBJECTIVES: We aimed to investigate whether clopidogrel-induced inhibition of platelet reactivity could reduce the level of circulating endothelial cells (CEC), reflecting the endothelial injury induced by percutaneous coronary intervention (PCI). BACKGROUND: Clopidogrel loading dose before percutaneous coronary angioplasty (PCI) reduces platelet activation through a selective and irreversible blockade of the adenosine diphosphate (ADP) receptor P2Y(12). The impact of clopidogrel on endothelial cells has been scarcely studied. METHODS: A total of 149 patients undergoing PCI for stable angina were enrolled. Levels of CEC were measured at baseline (H0) and 6 (H6) and 24 (H24) h after the procedure using a CD146-based immunomagnetic separation assay. The CEC delta-change (CEC at H6 - CEC at H0) was analyzed according to ADP receptor P2Y(12) blockade, assessed by a vasodilator-stimulated phosphoprotein (VASP) assay after a 600-mg loading dose of clopidogrel. RESULTS: The PCI induced a significant rise in CEC levels 6 h after the procedure. The CEC peak value was significantly higher in patients with high on-treatment platelet reactivity (VASP index ≥50%: 59.6 ± 27.5 cells/ml) as compared with good responders (VASP index <50%: 27 ± 22 cells/ml; p = 0.04). The endothelial injury, assessed by CEC delta-change between H6 and H0, was significantly higher in the high on-treatment platelet reactivity group compared with the good responders group (52.6 ± 25.6 vs. 18.6 ± 23.5, respectively; p < 0.001) and correlated with the VASP index (r = 0.59; p < 0.001). In multivariate analysis, VASP group, the number of diseased vessels, and the number of implanted stents independently predicted the endothelial injury (p < 0.001). CONCLUSIONS: Optimal ADP receptor P2Y(12) blockade reduces the endothelial injury during PCI. This protective effect of clopidogrel on endothelial cells could add to the clinical benefit associated with this drug.


Asunto(s)
Angina de Pecho/terapia , Angioplastia Coronaria con Balón/efectos adversos , Endotelio Vascular/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Ticlopidina/análogos & derivados , Enfermedades Vasculares/diagnóstico , Plaquetas/efectos de los fármacos , Clopidogrel , Células Endoteliales , Endotelio Vascular/lesiones , Femenino , Humanos , Masculino , Valor Predictivo de las Pruebas , Antagonistas del Receptor Purinérgico P2 , Receptores Purinérgicos P2Y12 , Ticlopidina/farmacología , Enfermedades Vasculares/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA