Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 144(3): 210-228, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-33951921

RESUMEN

BACKGROUND: Human induced pluripotent stem cells with normal (wild-type) or upregulated (overexpressed) levels of CCND2 (cyclin D2) expression were differentiated into cardiomyocytes (CCND2WTCMs or CCND2OECMs, respectively) and injected into infarcted pig hearts. METHODS: Acute myocardial infarction was induced by a 60-minute occlusion of the left anterior descending coronary artery. Immediately after reperfusion, CCND2WTCMs or CCND2OECMs (3×107 cells each) or an equivalent volume of the delivery vehicle was injected around the infarct border zone area. RESULTS: The number of the engrafted CCND2OECMs exceeded that of the engrafted CCND2WTCMs from 6- to 8-fold, rising from 1 week to 4 weeks after implantation. In contrast to the treatment with the CCND2WTCMs or the delivery vehicle, the administration of CCND2OECM was associated with significantly improved left ventricular function, as revealed by magnetic resonance imaging. This correlated with reduction of infarct size, fibrosis, ventricular hypertrophy, and cardiomyocyte apoptosis, and increase of vascular density and arterial density, as per histologic analysis of the treated hearts. Expression of cell proliferation markers (eg, Ki67, phosphorylated histone 3, and Aurora B kinase) was also significantly upregulated in the recipient cardiomyocytes from the CCND2OECM-treated than from the CCND2WTCM-treated pigs. The cell proliferation rate and the hypoxia tolerance measured in cultured human induced pluripotent stem cell cardiomyocytes were significantly greater after treatment with exosomes isolated from the CCND2OECMs (CCND2OEExos) than from the CCND2WTCMs (CCND2WTExos). As demonstrated by our study, CCND2OEExos can also promote the proliferation activity of postnatal rat and adult mouse cardiomyocytes. A bulk miRNA sequencing analysis of CCND2OEExos versus CCND2WTExos identified 206 and 91 miRNAs that were significantly upregulated and downregulated, respectively. Gene ontology enrichment analysis identified significant differences in the expression profiles of miRNAs from various functional categories and pathways, including miRNAs implicated in cell-cycle checkpoints (G2/M and G1/S transitions), or the mechanism of cytokinesis. CONCLUSIONS: We demonstrated that enhanced potency of CCND2OECMs promoted myocyte proliferation in both grafts and recipient tissue in a large mammal acute myocardial infarction model. These results suggest that CCND2OECMs transplantation may be a potential therapeutic strategy for the repair of infarcted hearts.


Asunto(s)
Diferenciación Celular/genética , Ciclina D2/genética , Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Trasplante de Células Madre , Animales , Biomarcadores , Técnicas de Cultivo de Célula , Proliferación Celular , Separación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/etiología , Miocitos Cardíacos/citología , Neovascularización Fisiológica/genética , Recuperación de la Función , Porcinos , Resultado del Tratamiento
2.
Heart Surg Forum ; 24(5): E868-E869, 2021 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-34623255

RESUMEN

Coronary artery aneurysm (CAA) is an aortic catastrophe with low prevalence. Giant CAA is even more uncommon, requiring surgical intervention. Giant CAA usually originates from the proximal segments of the right coronary and the anterior descending arteries. Here we report a rare case of giant left CAA with fistula formation treated with successful surgery.


Asunto(s)
Aneurisma Coronario/cirugía , Vasos Coronarios/cirugía , Fístula Vascular/cirugía , Procedimientos Quirúrgicos Vasculares/métodos , Angiografía por Tomografía Computarizada , Aneurisma Coronario/complicaciones , Aneurisma Coronario/diagnóstico , Angiografía Coronaria , Vasos Coronarios/diagnóstico por imagen , Ecocardiografía , Femenino , Humanos , Imagen por Resonancia Cinemagnética , Persona de Mediana Edad , Resultado del Tratamiento , Fístula Vascular/diagnóstico , Fístula Vascular/etiología
3.
Cancer Metastasis Rev ; 38(3): 493-506, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31705228

RESUMEN

Tumor suppressors are cellular proteins typically expressed in normal (non-cancer) cells that not only regulate such cellular functions as proliferation, migration and adhesion, but can also be secreted into extracellular space and serve as biomarkers for pathological conditions or tumor progression. KISS1, a precursor for several shorter peptides, known as metastin (Kisspeptin-54), Kisspeptin-14, Kisspeptin-13 and Kisspeptin-10, is one of those metastasis suppressor proteins, whose expression is commonly downregulated in the metastatic tumors of various origins. The commonly accepted role of KISS1 in metastatic tumor progression mechanism is the ability of this protein to suppress colonization of disseminated cancer cells in distant organs critical for the formation of the secondary tumor foci. Besides, recent evidence suggests involvement of KISS1 in the mechanisms of tumor angiogenesis, autophagy and apoptosis regulation, suggesting a possible role in both restricting and promoting cancer cell invasion. Here, we discuss the role of KISS1 in regulating metastases, the link between KISS1 expression and the autophagy-related biology of cancer cells and the perspectives of using KISS1 as a potential diagnostic marker for cancer progression as well as a new anti-cancer therapeutics.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Kisspeptinas/metabolismo , Animales , Autofagia/fisiología , Biomarcadores de Tumor/metabolismo , Femenino , Humanos
4.
Circulation ; 138(24): 2809-2816, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30030418

RESUMEN

BACKGROUND: Rodent hearts can regenerate myocardium lost to apical resection or myocardial infarction for up to 7 days after birth, but whether a similar window for myocardial regeneration also exists in large mammals is unknown. METHODS: Acute myocardial infarction (AMI) was surgically induced in neonatal pigs on postnatal days 1, 2, 3, 7, and 14 (ie, the P1, P2, P3, P7, and P14 groups, respectively). Cardiac systolic function was evaluated before AMI and at 30 days post-AMI via transthoracic echocardiography. Cardiomyocyte cell cycle activity was assessed via immunostaining for proliferation and mitosis markers, infarct size was evaluated histologically, and telomerase activity was measured by quantitative polymerase chain reaction. RESULTS: Systolic function at day 30 post-AMI was largely restored in P1 animals and partially restored in P2 animals, but significantly impaired when AMI was induced on postnatal day 3 or later. Hearts of P1 animals showed little evidence of scar formation or wall thinning on day 30 after AMI, with increased measures of cell-cycle activity seen 6 days after AMI (ie, postnatal day 7) compared with postnatal day 7 in noninfarcted hearts. CONCLUSIONS: The neonatal porcine heart is capable of regeneration after AMI during the first 2 days of life. This phenomenon is associated with induction of cardiomyocyte proliferation and is lost when cardiomyocytes exit the cell cycle shortly after birth.


Asunto(s)
Corazón/fisiología , Infarto del Miocardio/patología , Animales , Animales Recién Nacidos , Aurora Quinasa B/metabolismo , Ecocardiografía , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/fisiopatología , Mitosis , Miocardio/patología , Miocitos Cardíacos/metabolismo , Regeneración , Porcinos , Telomerasa/metabolismo
5.
Circulation ; 137(16): 1712-1730, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29233823

RESUMEN

BACKGROUND: Here, we generated human cardiac muscle patches (hCMPs) of clinically relevant dimensions (4 cm × 2 cm × 1.25 mm) by suspending cardiomyocytes, smooth muscle cells, and endothelial cells that had been differentiated from human induced-pluripotent stem cells in a fibrin scaffold and then culturing the construct on a dynamic (rocking) platform. METHODS: In vitro assessments of hCMPs suggest maturation in response to dynamic culture stimulation. In vivo assessments were conducted in a porcine model of myocardial infarction (MI). Animal groups included: MI hearts treated with 2 hCMPs (MI+hCMP, n=13), MI hearts treated with 2 cell-free open fibrin patches (n=14), or MI hearts with neither experimental patch (n=15); a fourth group of animals underwent sham surgery (Sham, n=8). Cardiac function and infarct size were evaluated by MRI, arrhythmia incidence by implanted loop recorders, and the engraftment rate by calculation of quantitative polymerase chain reaction measurements of expression of the human Y chromosome. Additional studies examined the myocardial protein expression profile changes and potential mechanisms of action that related to exosomes from the cell patch. RESULTS: The hCMPs began to beat synchronously within 1 day of fabrication, and after 7 days of dynamic culture stimulation, in vitro assessments indicated the mechanisms related to the improvements in electronic mechanical coupling, calcium-handling, and force generation, suggesting a maturation process during the dynamic culture. The engraftment rate was 10.9±1.8% at 4 weeks after the transplantation. The hCMP transplantation was associated with significant improvements in left ventricular function, infarct size, myocardial wall stress, myocardial hypertrophy, and reduced apoptosis in the periscar boarder zone myocardium. hCMP transplantation also reversed some MI-associated changes in sarcomeric regulatory protein phosphorylation. The exosomes released from the hCMP appeared to have cytoprotective properties that improved cardiomyocyte survival. CONCLUSIONS: We have fabricated a clinically relevant size of hCMP with trilineage cardiac cells derived from human induced-pluripotent stem cells. The hCMP matures in vitro during 7 days of dynamic culture. Transplantation of this type of hCMP results in significantly reduced infarct size and improvements in cardiac function that are associated with reduction in left ventricular wall stress. The hCMP treatment is not associated with significant changes in arrhythmogenicity.


Asunto(s)
Células Endoteliales/trasplante , Células Madre Pluripotentes Inducidas/trasplante , Infarto del Miocardio/cirugía , Miocardio/patología , Miocitos Cardíacos/trasplante , Miocitos del Músculo Liso/trasplante , Regeneración , Trasplante de Células Madre/métodos , Ingeniería de Tejidos/métodos , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Miocitos del Músculo Liso/patología , Recuperación de la Función , Regeneración/genética , Sus scrofa , Factores de Tiempo , Andamios del Tejido , Trasplante Heterólogo , Función Ventricular Izquierda , Remodelación Ventricular
6.
Circ Res ; 120(1): 150-165, 2017 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-28057791

RESUMEN

Current strategies for engineering cardiovascular cells and tissues have yielded a variety of sophisticated tools for studying disease mechanisms, for development of drug therapies, and for fabrication of tissue equivalents that may have application in future clinical use. These efforts are motivated by the need to extend traditional 2-dimensional (2D) cell culture systems into 3D to more accurately replicate in vivo cell and tissue function of cardiovascular structures. Developments in microscale devices and bioprinted 3D tissues are beginning to supplant traditional 2D cell cultures and preclinical animal studies that have historically been the standard for drug and tissue development. These new approaches lend themselves to patient-specific diagnostics, therapeutics, and tissue regeneration. The emergence of these technologies also carries technical challenges to be met before traditional cell culture and animal testing become obsolete. Successful development and validation of 3D human tissue constructs will provide powerful new paradigms for more cost effective and timely translation of cardiovascular tissue equivalents.


Asunto(s)
Miocitos Cardíacos/fisiología , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Sistema Cardiovascular/citología , Técnicas de Cultivo de Célula/métodos , Humanos
7.
Pediatr Cardiol ; 40(8): 1728-1734, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31549187

RESUMEN

We evaluated the efficacy of bioresorbable sternal reinforcement device (poly-L-lactide sternal pins) on sternal healing after median sternotomy in young children (with body weight less than 10 kg) with congenital heart disease (CHD). Data from 85 patients, who underwent CHD surgery through median sternotomy from October 2016 to May 2018, were collected and analyzed. Sternal pins were utilized in 85 patients (10 mm × 1 mm × 1 mm for patients with body weights less than 5 kg and 15 mm × 2 mm × 2 mm for those weighing between 5 and 10 kg) in addition to sternum closure with Ethicon PDSTMII running sutures (Group A), while 84 patients received the Ethicon sternal closure (Group B) with no pins. The occurrence of sternal dehiscence, anterior-posterior displacement, and high-low displacement was evaluated by physical examination and three-dimensional computed tomography at one month postoperatively. No anterior-posterior sternal displacement (0%) was observed in Group A, while 10 anterior-posterior displacements (11.9%) were observed in Group B (P < 0.01). The number of sternal caudal-cranial displacements in Groups A and B was 4 (4.71%) and 5 (5.35%), respectively (P = 0.870). While no sternal dehiscence (0%) was observed in Group A, 7 out of 84 patients (8.33%) in Group B exhibited obvious sternal dehiscence (P < 0.01). The bioresorbable poly-L-lactide sternal pins reduced an anterior-posterior sternal displacement and sternal dehiscence, which was accompanied by a significant improvement of an early sternal fixation.


Asunto(s)
Clavos Ortopédicos , Esternotomía/métodos , Esternón/cirugía , Estudios de Casos y Controles , Femenino , Cardiopatías Congénitas/cirugía , Humanos , Imagenología Tridimensional , Lactante , Masculino , Poliésteres/uso terapéutico , Esternotomía/estadística & datos numéricos , Dehiscencia de la Herida Operatoria/prevención & control , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
8.
Am J Physiol Heart Circ Physiol ; 314(2): H278-H284, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29101176

RESUMEN

Vascular endothelial growth factor (VEGF) is a well-characterized proangiogenic cytokine that has been shown to promote neovascularization in hearts of patients with ischemic heart disease but can also lead to adverse effects depending on the dose and mode of delivery. We investigated whether prolonged exposure to a low dose of VEGF could be achieved by encapsulating VEGF in polylactic coglycolic acid nanoparticles and whether treatment with VEGF-containing nanoparticles improved cardiac function and protected against left ventricular remodeling in the hearts of mice with experimentally induced myocardial infarction. Polylactic coglycolic acid nanoparticles with a mean diameter of ~113 nm were generated via double emulsion and loaded with VEGF; the encapsulation efficiency was 53.5 ± 1.7% (107.1 ± 3.3 ng VEGF/mg nanoparticles). In culture, VEGF nanoparticles released VEGF continuously for at least 31 days, and in a murine myocardial infarction model, VEGF nanoparticle administration was associated with significantly greater vascular density in the peri-infarct region, reductions in infarct size, and improvements in left ventricular contractile function 4 wk after treatment. Thus, our study provides proof of principle that nanoparticle-mediated delivery increases the angiogenic and therapeutic potency of VEGF for the treatment of ischemic heart disease. NEW & NOTEWORTHY Vascular endothelial growth factor (VEGF) is a well-characterized proangiogenic cytokine but has a short half-life and a rapid clearance rate. When encapsulated in nanoparticles, VEGF was released for 31 days and improved left ventricular function in infarcted mouse hearts. These observations indicate that our new platform increases the therapeutic potency of VEGF.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Infarto del Miocardio/tratamiento farmacológico , Nanopartículas , Neovascularización Fisiológica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Inductores de la Angiogénesis/química , Animales , Células Cultivadas , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Portadores de Fármacos , Composición de Medicamentos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Ratones Endogámicos NOD , Ratones SCID , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Recuperación de la Función , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/química
9.
Am J Physiol Heart Circ Physiol ; 315(2): H327-H339, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29631371

RESUMEN

The microenvironment of native heart tissue may be better replicated when cardiomyocytes are cultured in three-dimensional clusters (i.e., spheroids) than in monolayers or as individual cells. Thus, we differentiated human cardiac lineage-induced pluripotent stem cells in cardiomyocytes (hiPSC-CMs) and allowed them to form spheroids and spheroid fusions that were characterized in vitro and evaluated in mice after experimentally induced myocardial infarction (MI). Synchronized contractions were observed within 24 h of spheroid formation, and optical mapping experiments confirmed the presence of both Ca2+ transients and propagating action potentials. In spheroid fusions, the intraspheroid conduction velocity was 7.0 ± 3.8 cm/s on days 1- 2 after formation, whereas the conduction velocity between spheroids increased significantly ( P = 0.003) from 0.8 ± 1.1 cm/s on days 1- 2 to 3.3 ± 1.4 cm/s on day 7. For the murine MI model, five-spheroid fusions (200,000 hiPSC-CMs/spheroid) were embedded in a fibrin patch and the patch was transplanted over the site of infarction. Later (4 wk), echocardiographic measurements of left ventricular ejection fraction and fractional shortening were significantly greater in patch-treated animals than in animals that recovered without the patch, and the engraftment rate was 25.6% or 30% when evaluated histologically or via bioluminescence imaging, respectively. The exosomes released from the spheroid patch seemed to increase cardiac function. In conclusion, our results established the feasibility of using hiPSC-CM spheroids and spheroid fusions for cardiac tissue engineering, and, when fibrin patches containing hiPSC-CM spheroid fusions were evaluated in a murine MI model, the engraftment rate was much higher than the rates we have achieved via the direct intramyocardial injection. NEW & NOTEWORTHY Spheroids fuse in culture to produce structures with uniformly distributed cells. Furthermore, human cardiac lineage-induced pluripotent stem cells in cardiomyocytes in adjacent fused spheroids became electromechanically coupled as the fusions matured in vitro, and when the spheroids were combined with a biological matrix and administered as a patch over the infarcted region of mouse hearts, the engraftment rate exceeded 25%, and the treatment was associated with significant improvements in cardiac function via a paracrine mechanism, where exosomes released from the spheroid patch.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Infarto del Miocardio/terapia , Miocitos Cardíacos/trasplante , Esferoides Celulares/trasplante , Animales , Señalización del Calcio , Células Cultivadas , Células HEK293 , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Contracción Miocárdica , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Esferoides Celulares/metabolismo , Trasplante de Células Madre/métodos
10.
Cancer Rep (Hoboken) ; 6(2): e1708, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36253876

RESUMEN

BACKGROUND: Purine nucleoside phosphorylase (PNP) gene transfer represents a promising approach to treatment of head and neck malignancies. We tested recombinant adenovirus already in phase I/II clinical testing and leading-edge patient-derived xenografts (PDX) as a means to optimize this therapeutic strategy. METHODS: Our experiments investigated purine base cytotoxicity, PNP enzyme activity following treatment of malignant tissue, tumor mass regression, viral receptor studies, and transduction by tropism-modified adenovirus. RESULTS: Replication deficient vector efficiently transduced PDX cells and mediated significant anticancer effect following treatment with fludarabine phosphate in vivo. Either 6-methylpurine or 2-fluoroadenine (toxic molecules generated by the PNP approach) ablated head and neck cancer cell proliferation. High levels of adenovirus-3 specific receptors were detected in human tumor models, and vector was evaluated that utilizes this pathway. CONCLUSIONS: Our studies provide the scientific foundation necessary to improve PNP prodrug cleavage and advance a new treatment for head and neck cancer.


Asunto(s)
Neoplasias de Cabeza y Cuello , Purina-Nucleósido Fosforilasa , Humanos , Purina-Nucleósido Fosforilasa/genética , Purina-Nucleósido Fosforilasa/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Xenoinjertos , Vectores Genéticos , Terapia Genética , Adenoviridae/genética
11.
Curr Opin Cell Biol ; 15(3): 318-25, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12787774

RESUMEN

The nucleolus is the site of ribosomal RNA synthesis, processing and ribosome maturation. Various small ribonucleoproteins also undergo maturation in the nucleolus, involving RNA modification and RNA-protein assembly. Such steps and other activities of small ribonucleoproteins also take place in Cajal (coiled) bodies. Events of ribosome biogenesis are found solely in the nucleolus, which is the final destination of small nucleolar RNAs after their traffic through Cajal bodies. However, nucleoli are just a stopping point in the intricate cellular traffic for small nuclear RNAs and other ribonucleoproteins.


Asunto(s)
Nucléolo Celular/fisiología , Ribonucleoproteínas/fisiología , Ribosomas/fisiología , Animales , Humanos
12.
Front Pediatr ; 9: 611007, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33681097

RESUMEN

Introduction: The purpose of this study was to report our experience in the surgical reconstruction of the right ventricular outflow tract in double outlet right ventricle with a major coronary artery crossing the right ventricular outflow tract in the presence of mirror image-dextrocardia. Methods: From January 2005 to December 2019, 19 double outlet right ventricle patients (median age 4 years) with mirror image-dextrocardia and a major coronary artery crossing the right ventricular outflow tract received surgical repair. An autologous pericardial patch was used to enlarge the right ventricular outflow tract in four patients without pulmonary stenosis and three patients with mild pulmonary stenosis. A valved bovine jugular venous conduit was added to a hypoplastic native pathway in nine patients, among which six patients with moderate pulmonary stenosis received small-sized bovine jugular venous conduit implantation (diameter ≤ 16 mm). In comparison, a large-sized bovine jugular venous conduit (diameter >16 mm) was adopted in a total of three patients with severe pulmonary stenosis. Finally, three patients with preoperative pulmonary hypertension (mean pulmonary artery pressure ≥40 mmHg) did not undergo further intervention of right ventricular outflow tract due to the adequate outflow tract blood flow. Results: There was no hospital mortality. One patient with sub-pulmonary ventricular septal defect and concomitant severe pulmonary hypertension died from respiratory failure 11 months after the operation. Kaplan-Meier survival was 94% at 5, 10 years. Within a mean echocardiographic follow-up of 6.9 ± 3.6 years, a total of two patients received reintervention due to valvular stenosis of the bovine jugular venous conduit (pressure gradient > 50 mmHg at 4 and 9 years) after surgical operation. Actuarial freedom from reoperation was 90 and 72% at 5 and 10 years, respectively. During the last echocardiographic follow-up phase, all the survivors were in NYHA class I. Conclusions: Double outlet right ventricle with mirror image-dextrocardia is a rare and complicated congenital cardiac malformation. Surgical reconstruction of the right ventricular outflow tract should be individualized based on the degree of pulmonary stenosis and the specific anatomical features of each patient. Reconstructing the pulmonary artery using the various sizes of valved bovine jugular venous conduit is a safe and effective surgical method.

13.
Mol Imaging ; 9(2): 59-75, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20236604

RESUMEN

Genetic capsid labeling of conditionally replicative adenoviruses (CRAds) with fluorescent tags offers a potentially more accurate monitoring of those virotherapy agents in vivo. The capsid of an infectivity-enhanced CRAd, Ad5/3, delta 24, was genetically labeled with monomeric red fluorescent protein 1 (mRFP1) or its advanced derivative, "mCherry," to evaluate the utility of each red fluorescent reporter and the benefit of CRAd capsid labeling for noninvasive virus tracking in animal tumor models by a new spectral imaging approach. Either reporter was incorporated into the CRAd particles by genetic fusion to the viral capsid protein IX. Following intratumoral injection, localization and replication of each virus in orthotopic breast cancer xenografts were analyzed by spectral imaging and verified by quantitative polymerase chain reaction. Fluorescence in tumors increased up to 2,000-fold by day 4 and persisted for 5 to 7 weeks, showing oscillatory dynamics reflective of CRAd replication cycles. Capsid labeling in conjunction with spectral imaging thus enables direct visualization and quantification of CRAd particles in tumors prior to the reporter transgene expression. This allows for noninvasive control of CRAd delivery and distribution in tumors and facilitates quantitative assessment of viral replication. Although mCherry appeared to be superior to mRFP1 as an imaging tag, both reporters showed utility for CRAd imaging applications.


Asunto(s)
Adenoviridae/fisiología , Neoplasias de la Mama/virología , Cápside/fisiología , Proteínas Luminiscentes/biosíntesis , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Antineoplásicos/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Cápside/química , Cápside/metabolismo , Línea Celular Tumoral , Clonación Molecular , Femenino , Colorantes Fluorescentes/análisis , Humanos , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Ratones , Ratones Desnudos , Microscopía Fluorescente/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Reproducibilidad de los Resultados , Virión/química , Virión/genética , Virión/metabolismo , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Fluorescente Roja
14.
Matrix Biol Plus ; 8: 100034, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33543033

RESUMEN

Previously, our lab showed that the endoplasmic reticulum (ER) and calcium regulatory protein, calreticulin (CRT), is important for collagen transcription, secretion, and assembly into the extracellular matrix (ECM) and that ER CRT is critical for TGF-ß stimulation of type I collagen transcription through stimulation of ER calcium release and NFAT activation. Diabetes is the leading cause of end stage renal disease. TGF-ß is a key factor in the pathogenesis of diabetic nephropathy. However, the role of calreticulin (Calr) in fibrosis of diabetic nephropathy has not been investigated. In current work, we used both in vitro and in vivo approaches to assess the role of ER CRT in TGF-ß and glucose stimulated ECM production by renal tubule cells and in diabetic mice. Knockdown of CALR by siRNA in a human proximal tubular cell line (HK-2) showed reduced induction of soluble collagen when stimulated by TGF-ß or high glucose as compared to control cells, as well as a reduction in fibronectin and collagen IV transcript levels. CRT protein is increased in kidneys of mice made diabetic with streptozotocin and subjected to uninephrectomy to accelerate renal tubular injury as compared to controls. We used renal-targeted ultrasound delivery of Cre-recombinase plasmid to knockdown specifically CRT expression in the remaining kidney of uninephrectomized Calr fl/fl mice with streptozotocin-induced diabetes. This approach reduced CRT expression in the kidney, primarily in the tubular epithelium, by 30-55%, which persisted over the course of the studies. Renal function as measured by the urinary albumin/creatinine ratio was improved in the mice with knockdown of CRT as compared to diabetic mice injected with saline or subjected to ultrasound and injected with control GFP plasmid. PAS staining of kidneys and immunohistochemical analyses of collagen types I and IV show reduced glomerular and tubulointerstitial fibrosis. Renal sections from diabetic mice with CRT knockdown showed reduced nuclear NFAT in renal tubules and treatment of diabetic mice with 11R-VIVIT, an NFAT inhibitor, reduced proteinuria and renal fibrosis. These studies identify ER CRT as an important regulator of TGF-ß stimulated ECM production in the diabetic kidney, potentially through regulation of NFAT-dependent ECM transcription.

15.
Mol Imaging ; 8(5): 264-77, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19796604

RESUMEN

We sought to develop a cancer-targeted, infectivity-enhanced oncolytic adenovirus that embodies a capsid-labeling fusion for noninvasive dual-modality imaging of ovarian cancer virotherapy. A functional fusion protein composed of fluorescent and nuclear imaging tags was genetically incorporated into the capsid of an infectivity-enhanced conditionally replicative adenovirus. Incorporation of herpes simplex virus thymidine kinase (HSV-tk) and monomeric red fluorescent protein 1 (mRFP1) into the viral capsid and its genomic stability were verified by molecular analyses. Replication and oncolysis were evaluated in ovarian cancer cells. Fusion functionality was confirmed by in vitro gamma camera and fluorescent microscopy imaging. Comparison of tk-mRFP virus to single-modality controls revealed similar replication efficiency and oncolytic potency. Molecular fusion did not abolish enzymatic activity of HSV-tk as the virus effectively phosphorylated thymidine both ex vivo and in vitro. In vitro fluorescence imaging demonstrated a strong correlation between the intensity of fluorescent signal and cytopathic effect in infected ovarian cancer cells, suggesting that fluorescence can be used to monitor viral replication. We have in vitro validated a new infectivity-enhanced oncolytic adenovirus with a dual-imaging modality-labeled capsid, optimized for ovarian cancer virotherapy. The new agent could provide incremental gains toward climbing the barriers for achieving conditionally replicated adenovirus efficacy in human trials.


Asunto(s)
Adenoviridae/metabolismo , Adenoviridae/fisiología , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/terapia , Adenoviridae/genética , Cápside/metabolismo , Línea Celular , Línea Celular Tumoral , Femenino , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/fisiología , Microscopía Fluorescente , Simplexvirus/enzimología , Timidina Quinasa/genética , Timidina Quinasa/fisiología , Proteína Fluorescente Roja
16.
J Cell Biol ; 162(5): 821-32, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12939253

RESUMEN

All small nuclear RNAs (snRNAs) of the [U4/U6.U5] tri-snRNP localize transiently to nucleoli, as visualized by microscopy after injection of fluorescein-labeled transcripts into Xenopus laevis oocyte nuclei. Here, we demonstrate that these RNAs traffic to nucleoli independently of one another, because U4 snRNA deleted in the U6 base-pairing region still localizes to nucleoli. Furthermore, depletion of endogenous U6 snRNA does not affect nucleolar localization of injected U4 or U5. The wild-type U4 transcripts used here are functional: they exhibit normal nucleocytoplasmic traffic, associate with Sm proteins, form the [U4/U6] di-snRNP, and localize to nucleoli and Cajal bodies. The nucleolar localization element (NoLE) of U4 snRNA was mapped by mutagenesis. Neither the 5'-cap nor the 3'-region of U4, which includes the Sm protein binding site, are essential for nucleolar localization. The only region in U4 snRNA required for nucleolar localization is the 5'-proximal stem loop, which contains the binding site for the NHPX/15.5-kD protein. Even mutation of just five nucleotides, essential for binding this protein, impaired U4 nucleolar localization. Intriguingly, the NHPX/15.5-kD protein also binds the nucleolar localization element of box C/D small nucleolar RNAs, suggesting that this protein might mediate nucleolar localization of several small RNAs.


Asunto(s)
Nucléolo Celular/metabolismo , ARN Nuclear Pequeño/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Animales , Autoantígenos , Sitios de Unión , Cuerpos Enrollados/química , Humanos , Microinyecciones , Conformación de Ácido Nucleico , Oocitos/fisiología , Caperuzas de ARN/metabolismo , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/genética , Xenopus laevis , Proteínas Nucleares snRNP
17.
PLoS One ; 14(4): e0215226, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31026285

RESUMEN

BACKGROUND: A major obstacle to using recombinant adenoviral vectors in gene therapy is the natural ability of human adenovirus to activate the classical and alternate complement pathways. These innate immune responses contribute to hepatic adenoviral uptake following systemic delivery and enhance the humoral immune responses associated with adenoviral infection. METHODS: A recombinant Ad5 vector was genetically modified to display a peptide sequence ("rH17d'"), a known inhibitor of the classical complement pathway. The replication-defective vectors Ad5.HVR2-rH17d' and Ad5.HVR5-rH17d' were constructed by engineering the rH17d' peptide into the hypervariable region (HVR)-2 or HVR5 of their major capsid protein hexon. Control Ad5 vectors were created by incorporation of a 6-histidine (His6)-insert in either HVR2 or HVR5 (Ad5.HVR2-His6 and Ad5.HVR5-His6, respectively). All vectors encoded CMV promoter-controlled firefly luciferase (Luc). The four vectors were evaluated in TIB76 mouse liver cells and immunocompetent mice to compare infectivity and liver sequestration, respectively. RESULTS: In vitro studies demonstrated that preincubation of all the Ad5 vectors with fresh serum significantly increased their gene transfer relative to preincubation with PBS except Ad5.HVR5-rH17d', whose infectivity of liver cells showed no serum-mediated enhancement. In line with that, mice injected with Ad5.HVR2-rH17d' or Ad5.HVR5-rH17d' showed significantly lower luciferase expression levels in the liver as compared to the respective control vectors, whereas efficiency of tumor transduction by rH17d' and His6 vectors following their intratumoral injection was similar. CONCLUSIONS: Displaying a complement-inhibiting peptide on the Ad5 capsid surface by genetic modification of the hexon protein could be a suitable strategy for reducing Ad5 liver tropism (Ad5 sequestration by liver), which may be applicable to other gene therapy vectors with natural liver tropism.


Asunto(s)
Adenovirus Humanos/genética , Activación de Complemento/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Neoplasias/terapia , Adenovirus Humanos/inmunología , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Terapia Genética/efectos adversos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Humanos , Inmunidad Humoral/inmunología , Inyecciones Intralesiones , Hígado/citología , Hígado/inmunología , Masculino , Ratones , Neoplasias/inmunología , Péptidos/genética , Transducción Genética
18.
Cardiovasc Res ; 115(2): 343-356, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30107391

RESUMEN

Aims: The effectiveness of cell-based treatments for regenerative myocardial therapy is limited by low rates of cell engraftment. Y-27632 inhibits Rho-associated protein kinase (ROCK), which regulates the cytoskeletal changes associated with cell adhesion, and has been used to protect cultured cells during their passaging. Here, we investigated whether preconditioning of cardiomyocytes, derived from human-induced pluripotent stem cells (hiPSC-CM), with Y-27632 improves their survival and engraftment in a murine model of acute myocardial infarction (MI). Methods and results: After MI induction, mice were subjected to intramyocardial injections of phosphate-buffered saline, hiPSC-CM cultured under standard conditions (hiPSC-CM-RI), or Y-27632-preconditioned hiPSC-CM (hiPSC-CM+RI). The resulting engraftment rate calculated 4 weeks after implantation was significantly higher and the abundance of apoptotic transplanted cells was significantly lower in hiPSC-CM+RI recipients than in hiPSC-CM-RI animals. In cultured hiPSC-CM, Y-27632-preconditioning reversibly reduced contractile activity and the expression of troponin genes, while increasing their attachment to an underlying mouse cardiomyocyte (HL1) monolayer. Y-27632 preconditioning also increased the expression of N-cadherin and integrin ß1, the two cell junction proteins. hiPSC-CM+RI were also larger in cell area with greater cytoskeletal alignment and a more rod-like shape than hiPSC-CM-RI, both after transplantation (in vivo) and in culture. The effects of Y-27632 preconditioning on contractile activity and morphology of hiPSC-CMs in culture, as well as on their engraftment rate and apoptotic death in MI mouse grafts, could be recapitulated by hiPSC-CM treatment with the L-type calcium-channel blocker verapamil. Conclusion: Preconditioning with the ROCK inhibitor Y-27632 increased the engraftment of transplanted hiPSC-CM in a murine MI model, while reversibly impairing hiPSC-CM contractility and promoting adhesion.


Asunto(s)
Amidas/farmacología , Diferenciación Celular , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/trasplante , Infarto del Miocardio/cirugía , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/trasplante , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Supervivencia de Injerto/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Ratones Endogámicos NOD , Ratones SCID , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/enzimología , Fenotipo , Recuperación de la Función , Factores de Tiempo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
19.
J Mol Biol ; 366(4): 1142-60, 2007 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-17208253

RESUMEN

Mastadenoviruses represent one of the four major genera of the Adenoviridae family comprising a variety of mammalian pathogens including human adenovirus (Ad), whose genomes encode a gene for minor core protein V (pV), not found in other genera of Adenoviridae. Deletion of other genus-specific genes (gene IX and E3 genes) from the Ad type 5 (Ad5) genome has been studied experimentally in vitro and the results on biological characterization of the mutants support the phylogenetic evidence of those genes being non-essential for Ad viability. On this basis it seemed logical to suggest that a deletion of gene V from the Ad5 genome could also be tolerated. To test this hypothesis we constructed and rescued the first pV-deletion mutant of human Ad5. As compared to Ad5, this mutant formed small plaques, had dramatically reduced thermostability and lower infectivity. A subsequent thermoselection screen of the pV-deleted Ad5 allowed isolation of a suppressor mutant Ad5-dV/TSB with restored biological characteristics. Since replication and viral assembly of Ad5-dV/TSB could still occur in the absence of pV, we conclude that pV is a non-essential component of the virion. The observed rescue of the biological defects appears to be associated with a cluster of point mutations in the gene encoding the precursor for the other core protein, X/Mu. This finding, thus, suggests possible roles of pV and protein X/Mu precursor in viral assembly. It also provides an interesting insight into genetic events that mediate molecular adaptation of viruses to possible changes in the genetic background in the course of their evolutionary divergence. The possible mechanism of the observed genetic suppression is discussed.


Asunto(s)
Adenovirus Humanos/genética , Eliminación de Gen , Precursores de Proteínas/genética , Temperatura , Proteínas del Núcleo Viral/genética , Adenovirus Humanos/ultraestructura , Secuencia de Aminoácidos , Secuencia de Bases , Mapeo Cromosómico , Genes Virales , Genoma , Humanos , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Mutación , Mutación Missense , Mutación Puntual , Precursores de Proteínas/metabolismo , Homología de Secuencia de Aminoácido , Proteínas del Núcleo Viral/metabolismo , Replicación Viral
20.
Cancer Lett ; 417: 75-88, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29269086

RESUMEN

KISS1 tumor suppressor protein regulates cancer cell invasion via MMP9 metalloproteinase. Downregulation of KISS1 gene expression promotes progression of breast cancer and melanoma, resulting in the development of distant metastases. In the current study, we investigated whether restoration of KISS1 expression in KISS1-deficient human metastatic breast cancer cells holds potential as an advanced anticancer strategy. To this end we engineered an infectivity-enhanced conditionally-replicative human adenovirus type 5 encoding KISS1 as an "arming" transgene in the Ad5 E3 region for an ectopic KISS1 expression in transduced cancer cells. The oncolytic potential of the vector was examined using brain-invading metastatic clones of CN34 and MDA-MB-231 breast cancer cells, which supported high levels of AdKISS1 replication, correlating with a robust CRAd-mediated cytotoxicity. Secretion of cellular factors responsible for tumor angiogenesis, cell-to-cell communication and anti-tumoral immune responses upon KISS1 expression in breast cancer cells was analyzed by a RayBiotech Kiloplex Quantibody array. Overall, our results indicate that KISS1 transgene expression provides an important benefit for CRAd-mediated cytotoxicity in breast cancer cells and holds potential as an anticancer treatment in conjunction with oncolytic virotherapy of breast and other metastatic cancers.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Kisspeptinas/genética , Neovascularización Patológica/genética , Células A549 , Adenoviridae/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/genética , Células Cultivadas , Genes Supresores de Tumor , Vectores Genéticos/genética , Humanos , Kisspeptinas/metabolismo , Neovascularización Patológica/metabolismo , Viroterapia Oncolítica/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA