Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Med Genet A ; 176(12): 2924-2929, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30302932

RESUMEN

This report summarizes and highlights the fifth International RASopathies Symposium: When Development and Cancer Intersect, held in Orlando, Florida in July 2017. The RASopathies comprise a recognizable pattern of malformation syndromes that are caused by germ line mutations in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) pathway. Because of their common underlying pathogenetic etiology, there is significant overlap in their phenotypic features, which includes craniofacial dysmorphology, cardiac, cutaneous, musculoskeletal, gastrointestinal and ocular abnormalities, neurological and neurocognitive issues, and a predisposition to cancer. The RAS pathway is a well-known oncogenic pathway that is commonly found to be activated in somatic malignancies. As in somatic cancers, the RASopathies can be caused by various pathogenetic mechanisms that ultimately impact or alter the normal function and regulation of the MAPK pathway. As such, the RASopathies represent an excellent model of study to explore the intersection of the effects of dysregulation and its consequence in both development and oncogenesis.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Proteínas ras/genética , Animales , Regulación de la Expresión Génica , Estudios de Asociación Genética/métodos , Desarrollo Humano , Humanos , Modelos Biológicos , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Organogénesis/genética , Transducción de Señal , Síndrome , Proteínas ras/metabolismo
2.
Nature ; 492(7428): 252-5, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23143332

RESUMEN

The innate immune response is essential for combating infectious disease. Macrophages and other cells respond to infection by releasing cytokines, such as interleukin-1ß (IL-1ß), which in turn activate a well-described, myeloid-differentiation factor 88 (MYD88)-mediated, nuclear factor-κB (NF-κB)-dependent transcriptional pathway that results in inflammatory-cell activation and recruitment. Endothelial cells, which usually serve as a barrier to the movement of inflammatory cells out of the blood and into tissue, are also critical mediators of the inflammatory response. Paradoxically, the cytokines vital to a successful immune defence also have disruptive effects on endothelial cell-cell interactions and can trigger degradation of barrier function and dissociation of tissue architecture. The mechanism of this barrier dissolution and its relationship to the canonical NF-κB pathway remain poorly defined. Here we show that the direct, immediate and disruptive effects of IL-1ß on endothelial stability in a human in vitro cell model are NF-κB independent and are instead the result of signalling through the small GTPase ADP-ribosylation factor 6 (ARF6) and its activator ARF nucleotide binding site opener (ARNO; also known as CYTH2). Moreover, we show that ARNO binds directly to the adaptor protein MYD88, and thus propose MYD88-ARNO-ARF6 as a proximal IL-1ß signalling pathway distinct from that mediated by NF-κB. Finally, we show that SecinH3, an inhibitor of ARF guanine nucleotide-exchange factors such as ARNO, enhances vascular stability and significantly improves outcomes in animal models of inflammatory arthritis and acute inflammation.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de Interleucina/metabolismo , Factor 6 de Ribosilación del ADP , Adyuvantes Inmunológicos/farmacología , Animales , Artritis/patología , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Línea Celular , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Interleucina-1beta/farmacología , FN-kappa B/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Purinas/farmacología , Transducción de Señal , Tiofenos/farmacología
3.
Circulation ; 131(3): 289-99, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25486933

RESUMEN

BACKGROUND: Cerebral cavernous malformation (CCM) is a hemorrhagic stroke disease affecting up to 0.5% of North Americans that has no approved nonsurgical treatment. A subset of patients have a hereditary form of the disease due primarily to loss-of-function mutations in KRIT1, CCM2, or PDCD10. We sought to identify known drugs that could be repurposed to treat CCM. METHODS AND RESULTS: We developed an unbiased screening platform based on both cellular and animal models of loss of function of CCM2. Our discovery strategy consisted of 4 steps: an automated immunofluorescence and machine-learning-based primary screen of structural phenotypes in human endothelial cells deficient in CCM2, a secondary screen of functional changes in endothelial stability in these same cells, a rapid in vivo tertiary screen of dermal microvascular leak in mice lacking endothelial Ccm2, and finally a quaternary screen of CCM lesion burden in these same mice. We screened 2100 known drugs and bioactive compounds and identified 2 candidates, cholecalciferol (vitamin D3) and tempol (a scavenger of superoxide), for further study. Each drug decreased lesion burden in a mouse model of CCM vascular disease by ≈50%. CONCLUSIONS: By identifying known drugs as potential therapeutics for CCM, we have decreased the time, cost, and risk of bringing treatments to patients. Each drug also prompts additional exploration of biomarkers of CCM disease. We further suggest that the structure-function screening platform presented here may be adapted and scaled to facilitate drug discovery for diverse loss-of-function genetic vascular disease.


Asunto(s)
Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos/métodos , Hemangioma Cavernoso del Sistema Nervioso Central/tratamiento farmacológico , Animales , Células Cultivadas , Neoplasias del Sistema Nervioso Central/patología , Colecalciferol/farmacología , Colecalciferol/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales/métodos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Resultado del Tratamiento
4.
Hum Mol Genet ; 23(23): 6223-34, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24990152

RESUMEN

Cerebral cavernous malformation (CCM) is a disease of vascular malformations known to be caused by mutations in one of three genes: CCM1, CCM2 or CCM3. Despite several studies, the mechanism of CCM lesion onset remains unclear. Using a Ccm1 knockout mouse model, we studied the morphogenesis of early lesion formation in the retina in order to provide insight into potential mechanisms. We demonstrate that lesions develop in a stereotypic location and pattern, preceded by endothelial hypersprouting as confirmed in a zebrafish model of disease. The vascular defects seen with loss of Ccm1 suggest a defect in endothelial flow response. Taken together, these results suggest new mechanisms of early CCM disease pathogenesis and provide a framework for further study.


Asunto(s)
Hemangioma Cavernoso del Sistema Nervioso Central/patología , Proteínas Asociadas a Microtúbulos/genética , Proteínas Proto-Oncogénicas/genética , Retina/patología , Animales , Animales Modificados Genéticamente , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Hemangioma Cavernoso del Sistema Nervioso Central/metabolismo , Humanos , Proteína KRIT1 , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Pez Cebra
5.
J Immunol ; 192(12): 6045-52, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24835390

RESUMEN

The vascular endothelium responds to infection by destabilizing endothelial cell-cell junctions to allow fluid and cells to pass into peripheral tissues, facilitating clearance of infection and tissue repair. During sepsis, endotoxin and other proinflammatory molecules induce excessive vascular leak, which can cause organ dysfunction, shock, and death. Current therapies for sepsis are limited to antibiotics and supportive care, which are often insufficient to reduce morbidity and prevent mortality. Previous attempts at blocking inflammatory cytokine responses in humans proved ineffective at reducing the pathologies associated with sepsis, highlighting the need for a new therapeutic strategy. The small GTPase ARF6 is activated by a MyD88-ARNO interaction to induce vascular leak through disruption of endothelial adherens junctions. In this study, we show that the MyD88-ARNO-ARF6-signaling axis is responsible for LPS-induced endothelial permeability and is a destabilizing convergence point used by multiple inflammatory cues. We also show that blocking ARF6 with a peptide construct of its N terminus is sufficient to reduce vascular leak and enhance survival during endotoxic shock, without inhibiting the host cytokine response. Our data highlight the therapeutic potential of blocking ARF6 and reducing vascular leak for the treatment of inflammatory conditions, such as endotoxemia.


Asunto(s)
Factores de Ribosilacion-ADP/inmunología , Uniones Adherentes/inmunología , Permeabilidad Capilar/inmunología , Células Endoteliales/inmunología , Choque Séptico/inmunología , Transducción de Señal/inmunología , Factor 6 de Ribosilación del ADP , Uniones Adherentes/patología , Animales , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Células Endoteliales/patología , Femenino , Proteínas Activadoras de GTPasa/inmunología , Humanos , Lipopolisacáridos/toxicidad , Masculino , Ratones , Factor 88 de Diferenciación Mieloide/inmunología , Choque Séptico/inducido químicamente , Choque Séptico/patología , Transducción de Señal/efectos de los fármacos
6.
Nat Genet ; 56(7): 1482-1493, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38811841

RESUMEN

Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) is a powerful tool for introducing targeted mutations in DNA, but recent studies have shown that it can have unintended effects such as structural changes. However, these studies have not yet looked genome wide or across data types. Here we performed a phenotypic CRISPR-Cas9 scan targeting 17,065 genes in primary human cells, revealing a 'proximity bias' in which CRISPR knockouts show unexpected similarities to unrelated genes on the same chromosome arm. This bias was found to be consistent across cell types, laboratories, Cas9 delivery methods and assay modalities, and the data suggest that it is caused by telomeric truncations of chromosome arms, with cell cycle and apoptotic pathways playing a mediating role. Additionally, a simple correction is demonstrated to mitigate this pervasive bias while preserving biological relationships. This previously uncharacterized effect has implications for functional genomic studies using CRISPR-Cas9, with applications in discovery biology, drug-target identification, cell therapies and genetic therapeutics.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Humanos , Edición Génica/métodos , Mapeo Cromosómico/métodos , Genoma Humano
7.
Mol Pharm ; 9(6): 1599-611, 2012 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-22497592

RESUMEN

Poly(amidoamine) (PAMAM) dendrimers have been proposed for a variety of biomedical applications and are increasingly studied as model nanomaterials for such use. The dendritic structure features both modular synthetic control of molecular size and shape and presentation of multiple equivalent terminal groups. These properties make PAMAM dendrimers highly functionalizable, versatile single-molecule nanoparticles with a high degree of consistency and low polydispersity. Recent nanotoxicological studies showed that intravenous administration of amine-terminated PAMAM dendrimers to mice was lethal, causing a disseminated intravascular coagulation-like condition. To elucidate the mechanisms underlying this coagulopathy, in vitro assessments of platelet functions in contact with PAMAM dendrimers were undertaken. This study demonstrates that cationic G7 PAMAM dendrimers activate platelets and dramatically alter their morphology. These changes to platelet morphology and activation state substantially altered platelet function, including increased aggregation and adherence to surfaces. Surprisingly, dendrimer exposure also attenuated platelet-dependent thrombin generation, indicating that not all platelet functions remained intact. These findings provide additional insight into PAMAM dendrimer effects on blood components and underscore the necessity for further research on the effects and mechanisms of PAMAM-specific and general nanoparticle toxicity in blood.


Asunto(s)
Plaquetas/efectos de los fármacos , Dendrímeros/efectos adversos , Plaquetas/metabolismo , Células Cultivadas , Citometría de Flujo , Humanos , Microscopía Confocal , Nanopartículas/efectos adversos , Nanotecnología , Activación Plaquetaria/efectos de los fármacos , Pruebas de Función Plaquetaria , Trombina/metabolismo
8.
Nat Protoc ; 11(9): 1757-74, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27560178

RESUMEN

In morphological profiling, quantitative data are extracted from microscopy images of cells to identify biologically relevant similarities and differences among samples based on these profiles. This protocol describes the design and execution of experiments using Cell Painting, which is a morphological profiling assay that multiplexes six fluorescent dyes, imaged in five channels, to reveal eight broadly relevant cellular components or organelles. Cells are plated in multiwell plates, perturbed with the treatments to be tested, stained, fixed, and imaged on a high-throughput microscope. Next, an automated image analysis software identifies individual cells and measures ∼1,500 morphological features (various measures of size, shape, texture, intensity, and so on) to produce a rich profile that is suitable for the detection of subtle phenotypes. Profiles of cell populations treated with different experimental perturbations can be compared to suit many goals, such as identifying the phenotypic impact of chemical or genetic perturbations, grouping compounds and/or genes into functional pathways, and identifying signatures of disease. Cell culture and image acquisition takes 2 weeks; feature extraction and data analysis take an additional 1-2 weeks.


Asunto(s)
Colorantes Fluorescentes/metabolismo , Imagen Molecular/métodos , Coloración y Etiquetado/métodos , Línea Celular Tumoral , Forma de la Célula , Tamaño de la Célula , Humanos , Procesamiento de Imagen Asistido por Computador
9.
Biomark Med ; 10(3): 255-64, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26861901

RESUMEN

AIM: To correlate cerebral cavernous malformations (CCMs) disease aggressiveness with peripheral blood biomarkers hypothesized mechanistically. PATIENTS & METHODS: A prospective case-control study enrolled 43 CCM patients, where 25-(OH) vitamin D, HDL and non-HDL cholesterol, CRP plasma levels and leukocyte ROCK activity were correlated with parameters of disease aggressiveness reflecting chronic and acute domains. RESULTS: Patients with one or more features of chronically aggressive disease (early age at symptom onset, two or more symptomatic bleeds, high lesion burden) had significantly lower 25-(OH) vitamin D and non-HDL cholesterol levels in comparison to patients without these features. CONCLUSION: Validation of these biomarkers and their potential treatment modulation may influence the clinical care of patients with CCM disease.


Asunto(s)
HDL-Colesterol/sangre , Hemangioma Cavernoso del Sistema Nervioso Central/sangre , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Vitamina D/sangre , Enfermedad Aguda , Adulto , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , Enfermedad Crónica , Demografía , Femenino , Humanos , Leucocitos/enzimología , Masculino , Persona de Mediana Edad , Quinasas Asociadas a rho/metabolismo
10.
Biotechnol Prog ; 31(1): 248-57, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25376776

RESUMEN

Angiogenesis is regulated by chemical and mechanical factors in vivo. The regulatory role of mechanical factors and how chemical and mechanical angiogenic regulators work in concert remains to be explored. We investigated the effect of cyclic uniaxial stretch (20%, 1 Hz), with and without the stimulation of vascular endothelial growth factor (VEGF), on sprouting angiogenesis by employing a stretchable three-dimensional cell culture model. When compared to static controls, stretch alone significantly increased the density of endothelial sprouts, and these sprouts aligned perpendicular to the direction of stretch. The Rho-associated kinase (ROCK) inhibitor Y27632 suppressed stretch-induced sprouting angiogenesis and associated sprout alignment. While VEGF is a potent angiogenic stimulus through ROCK-dependent pathways, the combination of VEGF and stretch did not have an additive effect on angiogenesis. In the presence of VEGF stimulation, the ROCK inhibitor suppressed stretch-induced sprout alignment but did not affect stretch-induced sprout density; in contrast, the receptor tyrosine kinase (RTK) inhibitor sunitinib had no effect on stretch-induced alignment but trended toward suppressed stretch-induced sprout density. Our results suggest that the formation of sprouts and their directionality do not have completely identical regulatory pathways, and thus it is possible to separately manipulate the number and pattern of new sprouts.


Asunto(s)
Fenómenos Biomecánicos/fisiología , Proliferación Celular/fisiología , Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Amidas/farmacología , Animales , Aorta/citología , Bovinos , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Piridinas/farmacología , Estrés Mecánico , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
11.
PLoS One ; 10(10): e0140370, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26469335

RESUMEN

Vitamin D is a known modulator of inflammation. Native dietary vitamin D3 is thought to be bio-inactive, and beneficial vitamin D3 effects are thought to be largely mediated by the metabolite 1,25(OH)2D3. Reduced serum levels of the most commonly measured precursor metabolite, 25(OH)D3, is linked to an increased risk of multiple inflammatory diseases, including: cardiovascular disease, arthritis, multiple sclerosis, and sepsis. Common to all of these diseases is the disruption of endothelial stability and an enhancement of vascular leak. We previously performed an unbiased chemical suppressor screen on a genetic model of vascular instability, and identified cholecalciferol (D3, dietary Vitamin D3) as a factor that had profound and immediate stabilizing and therapeutic effects in that model. In this manuscript we show that the presumed inactive sterol, D3, is actually a potent and general mediator of endothelial stability at physiologically relevant concentrations. We further demonstrate that this phenomenon is apparent in vitamin D3 metabolites 25(OH)D3 and 1,25(OH)2D3, and that the effects are independent of the canonical transcription-mediated vitamin D pathway. Our data suggests the presence of an alternative signaling modality by which D3 acts directly on endothelial cells to prevent vascular leak. The finding that D3 and its metabolites modulate endothelial stability may help explain the clinical correlations between low serum vitamin D levels and the many human diseases with well-described vascular dysfunction phenotypes.


Asunto(s)
Colecalciferol/farmacología , Endotelio Vascular/efectos de los fármacos , Vitaminas/farmacología , Animales , Permeabilidad Capilar , Células Cultivadas , Colecalciferol/análogos & derivados , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Ratones
12.
Diabetes ; 64(12): 4247-59, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26340930

RESUMEN

Diabetic retinopathy (DR) is the leading cause of blindness in the working-age population in the U.S. The vision-threatening processes of neuroglial and vascular dysfunction in DR occur in concert, driven by hyperglycemia and propelled by a pathway of inflammation, ischemia, vasodegeneration, and breakdown of the blood retinal barrier. Currently, no therapies exist for normalizing the vasculature in DR. Here, we show that a single intravitreal dose of adeno-associated virus serotype 2 encoding a more stable, soluble, and potent form of angiopoietin 1 (AAV2.COMP-Ang1) can ameliorate the structural and functional hallmarks of DR in Ins2Akita mice, with sustained effects observed through six months. In early DR, AAV2.COMP-Ang1 restored leukocyte-endothelial interaction, retinal oxygenation, vascular density, vascular marker expression, vessel permeability, retinal thickness, inner retinal cellularity, and retinal neurophysiological response to levels comparable with nondiabetic controls. In late DR, AAV2.COMP-Ang1 enhanced the therapeutic benefit of intravitreally delivered endothelial colony-forming cells by promoting their integration into the vasculature and thereby stemming further visual decline. AAV2.COMP-Ang1 single-dose gene therapy can prevent neurovascular pathology, support vascular regeneration, and stabilize vision in DR.


Asunto(s)
Angiopoyetina 1/uso terapéutico , Proteína de la Matriz Oligomérica del Cartílago/uso terapéutico , Diabetes Mellitus Tipo 1/complicaciones , Retinopatía Diabética/terapia , Modelos Animales de Enfermedad , Terapia Genética , Retina/patología , Angiopoyetina 1/química , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Animales , Proteína de la Matriz Oligomérica del Cartílago/química , Proteína de la Matriz Oligomérica del Cartílago/genética , Proteína de la Matriz Oligomérica del Cartílago/metabolismo , Células Cultivadas , Terapia Combinada/efectos adversos , Cruzamientos Genéticos , Retinopatía Diabética/inmunología , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/trasplante , Terapia Genética/efectos adversos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inyecciones Intravítreas , Leucocitos/citología , Leucocitos/inmunología , Leucocitos/metabolismo , Leucocitos/patología , Ratones Endogámicos C57BL , Ratones Mutantes , Estabilidad Proteica , Distribución Aleatoria , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Retina/inmunología , Retina/metabolismo , Solubilidad
13.
Biotechnol Prog ; 30(4): 879-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24574264

RESUMEN

How mechanical factors affect angiogenesis and how they and chemical angiogenic factors work in concert remain not yet well-understood. This study investigated the interactive effects of cyclic uniaxial stretch and two potent proangiogenic molecules [basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF)] on angiogenesis using a stretchable three-dimensional (3-D) cell culture model. Endothelial cells seeded atop a 3-D collagen gel underwent sprouting angiogenesis while being subjected to either 10 or 20% cyclic uniaxial stretch at a frequency of either 1/12 or 1 Hz, in conjunction with an elevated concentration of bFGF or VEGF. Without the presence of additional growth factors, 10 and 20% stretch at 1 Hz induced angiogenesis and the perpendicular alignment of new sprouts, and both inductive effects were abolished by cytochalasin D (an actin polymerization inhibitor). While "10% stretch at 1 Hz," "20% stretch at 1 Hz," bFGF, and VEGF were strong angiogenesis stimulants individually, only the combination of "20% stretch at 1 Hz" and bFGF had an additive effect on inducing new sprouts. Interestingly, the combination of "20% stretch at a lower frequency (1/12 Hz)" and bFGF decreased sprouting angiogenesis, even though the level of perpendicular alignment of new sprouts was the same for both stretch frequencies. Taken together, these results demonstrate that both stretch frequency and magnitude, along with interactions with various growth factors, are essential in mediating formation of endothelial sprouts and vascular patterning. Furthermore, work in this area is warranted to elucidate synergistic or competitive signaling mechanisms.


Asunto(s)
Citoesqueleto de Actina/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/administración & dosificación , Neovascularización Fisiológica/genética , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Citoesqueleto de Actina/metabolismo , Animales , Bovinos , Línea Celular , Células Endoteliales/efectos de los fármacos , Humanos , Estrés Mecánico , Ingeniería de Tejidos
14.
Ann Biomed Eng ; 39(1): 337-46, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20737290

RESUMEN

Elastin is an essential component of arteries which provides structural integrity and instructs smooth muscle cells to adopt a quiescent state. Despite interaction of endothelial cells with elastin in the internal elastic lamina, the potential for exploiting this interaction therapeutically has not been explored in detail. In this study, we show that tropoelastin (a precursor of elastin) stimulates endothelial cell migration and adhesion more than smooth muscle cells. The biological activity of tropoelastin on endothelial cells is contained in the VGVAPG domain and in the carboxy-terminal 17-amino acids. We show that the effects of the carboxy-terminal 17 amino acids, but not those of VGVAPG, are mediated by integrin α(V)ß(3). We demonstrate that tropoelastin covalently linked to stainless steel disks promotes adhesion of endothelial progenitor cells and endothelial cells to the metal surfaces. The adherent cells on the tropoelastin-coated metal surfaces form monolayers that can withstand and respond to arterial shear stress. Because of the unique effects of tropoelastin on endothelial and smooth muscle cells, coating intravascular devices with tropoelastin may stimulate their endothelialization, inhibit smooth muscle hyperplasia, and improve device performance.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Tropoelastina/administración & dosificación , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos
15.
J Clin Invest ; 121(5): 1871-81, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21490399

RESUMEN

Cerebral cavernous malformations (CCMs) are a common type of vascular malformation in the brain that are a major cause of hemorrhagic stroke. This condition has been independently linked to 3 separate genes: Krev1 interaction trapped (KRIT1), Cerebral cavernous malformation 2 (CCM2), and Programmed cell death 10 (PDCD10). Despite the commonality in disease pathology caused by mutations in these 3 genes, we found that the loss of Pdcd10 results in significantly different developmental, cell biological, and signaling phenotypes from those seen in the absence of Ccm2 and Krit1. PDCD10 bound to germinal center kinase III (GCKIII) family members, a subset of serine-threonine kinases, and facilitated lumen formation by endothelial cells both in vivo and in vitro. These findings suggest that CCM may be a common tissue manifestation of distinct mechanistic pathways. Nevertheless, loss of heterozygosity (LOH) for either Pdcd10 or Ccm2 resulted in CCMs in mice. The murine phenotype induced by loss of either protein reproduced all of the key clinical features observed in human patients with CCM, as determined by direct comparison with genotype-specific human surgical specimens. These results suggest that CCM may be more effectively treated by directing therapies based on the underlying genetic mutation rather than treating the condition as a single clinical entity.


Asunto(s)
Hemangioma Cavernoso del Sistema Nervioso Central/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Modelos Genéticos , Mutación , Animales , Proteínas Reguladoras de la Apoptosis , Encéfalo/embriología , Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genotipo , Humanos , Proteína KRIT1 , Pérdida de Heterocigocidad , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas Asociadas a Microtúbulos/genética , Fenotipo , Proteínas Proto-Oncogénicas/genética , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA