Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 554(7693): 544-548, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29443960

RESUMEN

Therapeutic antibodies that block the programmed death-1 (PD-1)-programmed death-ligand 1 (PD-L1) pathway can induce robust and durable responses in patients with various cancers, including metastatic urothelial cancer. However, these responses only occur in a subset of patients. Elucidating the determinants of response and resistance is key to improving outcomes and developing new treatment strategies. Here we examined tumours from a large cohort of patients with metastatic urothelial cancer who were treated with an anti-PD-L1 agent (atezolizumab) and identified major determinants of clinical outcome. Response to treatment was associated with CD8+ T-effector cell phenotype and, to an even greater extent, high neoantigen or tumour mutation burden. Lack of response was associated with a signature of transforming growth factor ß (TGFß) signalling in fibroblasts. This occurred particularly in patients with tumours, which showed exclusion of CD8+ T cells from the tumour parenchyma that were instead found in the fibroblast- and collagen-rich peritumoural stroma; a common phenotype among patients with metastatic urothelial cancer. Using a mouse model that recapitulates this immune-excluded phenotype, we found that therapeutic co-administration of TGFß-blocking and anti-PD-L1 antibodies reduced TGFß signalling in stromal cells, facilitated T-cell penetration into the centre of tumours, and provoked vigorous anti-tumour immunity and tumour regression. Integration of these three independent biological features provides the best basis for understanding patient outcome in this setting and suggests that TGFß shapes the tumour microenvironment to restrain anti-tumour immunity by restricting T-cell infiltration.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD8-positivos/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias Urológicas/tratamiento farmacológico , Neoplasias Urológicas/inmunología , Urotelio/patología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Anticuerpos Monoclonales Humanizados , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Estudios de Cohortes , Colágeno/metabolismo , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Inmunoterapia , Ratones , Mutación , Metástasis de la Neoplasia , Fenotipo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Resultado del Tratamiento , Microambiente Tumoral/inmunología , Neoplasias Urológicas/genética , Neoplasias Urológicas/patología , Urotelio/efectos de los fármacos , Urotelio/inmunología
2.
Nature ; 527(7577): 240-4, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26560302

RESUMEN

Animals have evolved homeostatic responses to changes in oxygen availability that act on different timescales. Although the hypoxia-inducible factor (HIF) transcriptional pathway that controls long-term responses to low oxygen (hypoxia) has been established, the pathway that mediates acute responses to hypoxia in mammals is not well understood. Here we show that the olfactory receptor gene Olfr78 is highly and selectively expressed in oxygen-sensitive glomus cells of the carotid body, a chemosensory organ at the carotid artery bifurcation that monitors blood oxygen and stimulates breathing within seconds when oxygen declines. Olfr78 mutants fail to increase ventilation in hypoxia but respond normally to hypercapnia. Glomus cells are present in normal numbers and appear structurally intact, but hypoxia-induced carotid body activity is diminished. Lactate, a metabolite that rapidly accumulates in hypoxia and induces hyperventilation, activates Olfr78 in heterologous expression experiments, induces calcium transients in glomus cells, and stimulates carotid sinus nerve activity through Olfr78. We propose that, in addition to its role in olfaction, Olfr78 acts as a hypoxia sensor in the breathing circuit by sensing lactate produced when oxygen levels decline.


Asunto(s)
Ácido Láctico/metabolismo , Neuronas Receptoras Olfatorias/metabolismo , Oxígeno/metabolismo , Receptores Odorantes/metabolismo , Respiración , Animales , Señalización del Calcio , Cuerpo Carotídeo/citología , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/metabolismo , Seno Carotídeo/inervación , Femenino , Células HEK293 , Humanos , Hipercapnia/genética , Hipercapnia/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Ácido Láctico/farmacología , Ratones , Oxígeno/sangre , Receptores Odorantes/deficiencia
3.
Stem Cells ; 36(2): 265-277, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29086457

RESUMEN

The ability to differentiate human pluripotent stem cells (hPSCs) into cardiomyocytes (CMs) makes them an attractive source for repairing injured myocardium, disease modeling, and drug testing. Although current differentiation protocols yield hPSC-CMs to >90% efficiency, hPSC-CMs exhibit immature characteristics. With the goal of overcoming this limitation, we tested the effects of varying passive stretch on engineered heart muscle (EHM) structural and functional maturation, guided by computational modeling. Human embryonic stem cells (hESCs, H7 line) or human induced pluripotent stem cells (IMR-90 line) were differentiated to hPSC-derived cardiomyocytes (hPSC-CMs) in vitro using a small molecule based protocol. hPSC-CMs were characterized by troponin+ flow cytometry as well as electrophysiological measurements. Afterwards, 1.2 × 106 hPSC-CMs were mixed with 0.4 × 106 human fibroblasts (IMR-90 line) (3:1 ratio) and type-I collagen. The blend was cast into custom-made 12-mm long polydimethylsiloxane reservoirs to vary nominal passive stretch of EHMs to 5, 7, or 9 mm. EHM characteristics were monitored for up to 50 days, with EHMs having a passive stretch of 7 mm giving the most consistent formation. Based on our initial macroscopic observations of EHM formation, we created a computational model that predicts the stress distribution throughout EHMs, which is a function of cellular composition, cellular ratio, and geometry. Based on this predictive modeling, we show cell alignment by immunohistochemistry and coordinated calcium waves by calcium imaging. Furthermore, coordinated calcium waves and mechanical contractions were apparent throughout entire EHMs. The stiffness and active forces of hPSC-derived EHMs are comparable with rat neonatal cardiomyocyte-derived EHMs. Three-dimensional EHMs display increased expression of mature cardiomyocyte genes including sarcomeric protein troponin-T, calcium and potassium ion channels, ß-adrenergic receptors, and t-tubule protein caveolin-3. Passive stretch affects the structural and functional maturation of EHMs. Based on our predictive computational modeling, we show how to optimize cell alignment and calcium dynamics within EHMs. These findings provide a basis for the rational design of EHMs, which enables future scale-up productions for clinical use in cardiovascular tissue engineering. Stem Cells 2018;36:265-277.


Asunto(s)
Biología Computacional/métodos , Miocardio/citología , Línea Celular , Citometría de Flujo , Humanos , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Ingeniería de Tejidos/métodos
5.
Circulation ; 135(19): 1832-1847, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28167635

RESUMEN

BACKGROUND: Advancing structural and functional maturation of stem cell-derived cardiomyocytes remains a key challenge for applications in disease modeling, drug screening, and heart repair. Here, we sought to advance cardiomyocyte maturation in engineered human myocardium (EHM) toward an adult phenotype under defined conditions. METHODS: We systematically investigated cell composition, matrix, and media conditions to generate EHM from embryonic and induced pluripotent stem cell-derived cardiomyocytes and fibroblasts with organotypic functionality under serum-free conditions. We used morphological, functional, and transcriptome analyses to benchmark maturation of EHM. RESULTS: EHM demonstrated important structural and functional properties of postnatal myocardium, including: (1) rod-shaped cardiomyocytes with M bands assembled as a functional syncytium; (2) systolic twitch forces at a similar level as observed in bona fide postnatal myocardium; (3) a positive force-frequency response; (4) inotropic responses to ß-adrenergic stimulation mediated via canonical ß1- and ß2-adrenoceptor signaling pathways; and (5) evidence for advanced molecular maturation by transcriptome profiling. EHM responded to chronic catecholamine toxicity with contractile dysfunction, cardiomyocyte hypertrophy, cardiomyocyte death, and N-terminal pro B-type natriuretic peptide release; all are classical hallmarks of heart failure. In addition, we demonstrate the scalability of EHM according to anticipated clinical demands for cardiac repair. CONCLUSIONS: We provide proof-of-concept for a universally applicable technology for the engineering of macroscale human myocardium for disease modeling and heart repair from embryonic and induced pluripotent stem cell-derived cardiomyocytes under defined, serum-free conditions.


Asunto(s)
Células Madre Embrionarias/trasplante , Insuficiencia Cardíaca/terapia , Células Madre Pluripotentes Inducidas/trasplante , Miocitos Cardíacos/trasplante , Ingeniería de Tejidos/métodos , Remodelación Ventricular/fisiología , Animales , Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Insuficiencia Cardíaca/patología , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/fisiología , Impresión Tridimensional , Ratas , Ratas Desnudas
6.
Nature ; 484(7394): 367-70, 2012 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-22495303

RESUMEN

Understanding the molecular and cellular mechanisms that mediate magnetosensation in vertebrates is a formidable scientific problem. One hypothesis is that magnetic information is transduced into neuronal impulses by using a magnetite-based magnetoreceptor. Previous studies claim to have identified a magnetic sense system in the pigeon, common to avian species, which consists of magnetite-containing trigeminal afferents located at six specific loci in the rostral subepidermis of the beak. These studies have been widely accepted in the field and heavily relied upon by both behavioural biologists and physicists. Here we show that clusters of iron-rich cells in the rostro-medial upper beak of the pigeon Columbia livia are macrophages, not magnetosensitive neurons. Our systematic characterization of the pigeon upper beak identified iron-rich cells in the stratum laxum of the subepidermis, the basal region of the respiratory epithelium and the apex of feather follicles. Using a three-dimensional blueprint of the pigeon beak created by magnetic resonance imaging and computed tomography, we mapped the location of iron-rich cells, revealing unexpected variation in their distribution and number--an observation that is inconsistent with a role in magnetic sensation. Ultrastructure analysis of these cells, which are not unique to the beak, showed that their subcellular architecture includes ferritin-like granules, siderosomes, haemosiderin and filopodia, characteristics of iron-rich macrophages. Our conclusion that these cells are macrophages and not magnetosensitive neurons is supported by immunohistological studies showing co-localization with the antigen-presenting molecule major histocompatibility complex class II. Our work necessitates a renewed search for the true magnetite-dependent magnetoreceptor in birds.


Asunto(s)
Pico/citología , Columbidae/anatomía & histología , Hierro/metabolismo , Macrófagos/metabolismo , Campos Magnéticos , Sensación , Migración Animal , Animales , Pico/anatomía & histología , Columbidae/fisiología , Plumas/citología , Plumas/ultraestructura , Ferrocianuros/análisis , Inmunohistoquímica , Hierro/análisis , Macrófagos/ultraestructura , Imagen por Resonancia Magnética , Neuronas/metabolismo , Orientación , Mucosa Respiratoria/citología , Mucosa Respiratoria/ultraestructura , Tomografía Computarizada de Emisión de Fotón Único
7.
J Mol Cell Cardiol ; 102: 94-107, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27914791

RESUMEN

Sarcomere assembly is a highly orchestrated and dynamic process which adapts, during perinatal development, to accommodate growth of the heart. Sarcomeric components, including titin, undergo an isoform transition to adjust ventricular filling. Many sarcomeric genes have been implicated in congenital cardiomyopathies, such that understanding developmental sarcomere transitions will inform the aetiology and treatment. We sought to determine whether Thymosin ß4 (Tß4), a peptide that regulates the availability of actin monomers for polymerization in non-muscle cells, plays a role in sarcomere assembly during cardiac morphogenesis and influences adult cardiac function. In Tß4 null mice, immunofluorescence-based sarcomere analyses revealed shortened thin filament, sarcomere and titin spring length in cardiomyocytes, associated with precocious up-regulation of the short titin isoforms during the postnatal splicing transition. By magnetic resonance imaging, this manifested as diminished stroke volume and limited contractile reserve in adult mice. Extrapolating to an in vitro cardiomyocyte model, the altered postnatal splicing was corrected with addition of synthetic Tß4, whereby normal sarcomere length was restored. Our data suggest that Tß4 is required for setting correct sarcomere length and for appropriate splicing of titin, not only in the heart but also in skeletal muscle. Distinguishing between thin filament extension and titin splicing as the primary defect is challenging, as these events are intimately linked. The regulation of titin splicing is a previously unrecognised role of Tß4 and gives preliminary insight into a mechanism by which titin isoforms may be manipulated to correct cardiac dysfunction.


Asunto(s)
Conectina/genética , Empalme del ARN , Sarcómeros/metabolismo , Timosina/deficiencia , Animales , Ecocardiografía , Corazón/diagnóstico por imagen , Corazón/fisiopatología , Hemodinámica , Masculino , Ratones , Ratones Noqueados , Contracción Miocárdica/genética , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Sarcómeros/ultraestructura
8.
Circ Res ; 117(8): 720-30, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26291556

RESUMEN

RATIONALE: Tissue engineering approaches may improve survival and functional benefits from human embryonic stem cell-derived cardiomyocyte transplantation, thereby potentially preventing dilative remodeling and progression to heart failure. OBJECTIVE: Assessment of transport stability, long-term survival, structural organization, functional benefits, and teratoma risk of engineered heart muscle (EHM) in a chronic myocardial infarction model. METHODS AND RESULTS: We constructed EHMs from human embryonic stem cell-derived cardiomyocytes and released them for transatlantic shipping following predefined quality control criteria. Two days of shipment did not lead to adverse effects on cell viability or contractile performance of EHMs (n=3, P=0.83, P=0.87). One month after ischemia/reperfusion injury, EHMs were implanted onto immunocompromised rat hearts to simulate chronic ischemia. Bioluminescence imaging showed stable engraftment with no significant cell loss between week 2 and 12 (n=6, P=0.67), preserving ≤25% of the transplanted cells. Despite high engraftment rates and attenuated disease progression (change in ejection fraction for EHMs, -6.7±1.4% versus control, -10.9±1.5%; n>12; P=0.05), we observed no difference between EHMs containing viable and nonviable human cardiomyocytes in this chronic xenotransplantation model (n>12; P=0.41). Grafted cardiomyocytes showed enhanced sarcomere alignment and increased connexin 43 expression at 220 days after transplantation. No teratomas or tumors were found in any of the animals (n=14) used for long-term monitoring. CONCLUSIONS: EHM transplantation led to high engraftment rates, long-term survival, and progressive maturation of human cardiomyocytes. However, cell engraftment was not correlated with functional improvements in this chronic myocardial infarction model. Most importantly, the safety of this approach was demonstrated by the lack of tumor or teratoma formation.


Asunto(s)
Células Madre Embrionarias/trasplante , Supervivencia de Injerto , Trasplante de Corazón/métodos , Infarto del Miocardio/cirugía , Miocitos Cardíacos/trasplante , Músculos Papilares/trasplante , Ingeniería de Tejidos/métodos , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Supervivencia Celular , Conexina 43/metabolismo , Modelos Animales de Enfermedad , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Trasplante de Corazón/efectos adversos , Xenoinjertos , Humanos , Inmunosupresores/farmacología , Masculino , Contracción Miocárdica , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Músculos Papilares/inmunología , Músculos Papilares/metabolismo , Músculos Papilares/patología , Músculos Papilares/fisiopatología , Ratas Desnudas , Ratas Sprague-Dawley , Volumen Sistólico , Factores de Tiempo , Transfección
9.
Nature ; 474(7353): 640-4, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21654746

RESUMEN

A significant bottleneck in cardiovascular regenerative medicine is the identification of a viable source of stem/progenitor cells that could contribute new muscle after ischaemic heart disease and acute myocardial infarction. A therapeutic ideal--relative to cell transplantation--would be to stimulate a resident source, thus avoiding the caveats of limited graft survival, restricted homing to the site of injury and host immune rejection. Here we demonstrate in mice that the adult heart contains a resident stem or progenitor cell population, which has the potential to contribute bona fide terminally differentiated cardiomyocytes after myocardial infarction. We reveal a novel genetic label of the activated adult progenitors via re-expression of a key embryonic epicardial gene, Wilm's tumour 1 (Wt1), through priming by thymosin ß4, a peptide previously shown to restore vascular potential to adult epicardium-derived progenitor cells with injury. Cumulative evidence indicates an epicardial origin of the progenitor population, and embryonic reprogramming results in the mobilization of this population and concomitant differentiation to give rise to de novo cardiomyocytes. Cell transplantation confirmed a progenitor source and chromosome painting of labelled donor cells revealed transdifferentiation to a myocyte fate in the absence of cell fusion. Derived cardiomyocytes are shown here to structurally and functionally integrate with resident muscle; as such, stimulation of this adult progenitor pool represents a significant step towards resident-cell-based therapy in human ischaemic heart disease.


Asunto(s)
Células Madre Adultas/citología , Diferenciación Celular , Lesiones Cardíacas , Miocitos Cardíacos/citología , Animales , Reprogramación Celular , Regulación de la Expresión Génica , Ratones , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Timosina/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo
10.
PLoS Genet ; 9(1): e1003094, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23382688

RESUMEN

The ribosome is an evolutionarily conserved organelle essential for cellular function. Ribosome construction requires assembly of approximately 80 different ribosomal proteins (RPs) and four different species of rRNA. As RPs co-assemble into one multi-subunit complex, mutation of the genes that encode RPs might be expected to give rise to phenocopies, in which the same phenotype is associated with loss-of-function of each individual gene. However, a more complex picture is emerging in which, in addition to a group of shared phenotypes, diverse RP gene-specific phenotypes are observed. Here we report the first two mouse mutations (Rps7(Mtu) and Rps7(Zma)) of ribosomal protein S7 (Rps7), a gene that has been implicated in Diamond-Blackfan anemia. Rps7 disruption results in decreased body size, abnormal skeletal morphology, mid-ventral white spotting, and eye malformations. These phenotypes are reported in other murine RP mutants and, as demonstrated for some other RP mutations, are ameliorated by Trp53 deficiency. Interestingly, Rps7 mutants have additional overt malformations of the developing central nervous system and deficits in working memory, phenotypes that are not reported in murine or human RP gene mutants. Conversely, Rps7 mouse mutants show no anemia or hyperpigmentation, phenotypes associated with mutation of human RPS7 and other murine RPs, respectively. We provide two novel RP mouse models and expand the repertoire of potential phenotypes that should be examined in RP mutants to further explore the concept of RP gene-specific phenotypes.


Asunto(s)
Anemia de Diamond-Blackfan , Sistema Nervioso Central , Morfogénesis/genética , Proteínas Ribosómicas/genética , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/patología , Animales , Tamaño Corporal/genética , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Humanos , Memoria a Corto Plazo/fisiología , Ratones , Mutación , Fenotipo , Proteínas Ribosómicas/fisiología , Ribosomas/genética
11.
Circulation ; 130(11 Suppl 1): S77-86, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25200059

RESUMEN

BACKGROUND: Cell therapies offer the potential to improve cardiac function after myocardial infarction. Although injection of single-cell suspensions has proven safe, cell retention and survival rates are low. Tissue-engineered grafts allow cell delivery with minimal initial cell loss and mechanical support to the heart. However, graft performance cannot be easily compared, and optimal construct thickness, vascularization, and survival kinetics are unknown. METHODS AND RESULTS: Cardiac tissue slices (CTS) were generated by sectioning mouse hearts (n=40) expressing firefly luciferase and green fluorescent protein into slices of defined size and thickness using a vibrating blade microtome. Bioluminescence imaging of CTS transplanted onto hearts of immunodeficient mice demonstrated survival of ≤30% of transplanted cells. Cardiac slice perfusion was re-established within 3 days, likely through anastomosis of pre-existing vessels with the host vasculature and invasion of vessels from the host. Immunofluorescence showed a peak in cell death 3 days after transplantation and a gradual decline thereafter. MRI revealed preservation of contractile function and an improved ejection fraction 1 month after transplantation of CTS (28±2% CTS versus 22±2% control; P=0.05). Importantly, this effect was specific to CTS because transplantation of skeletal muscle tissue slices led to faster dilative remodeling and higher animal mortality. CONCLUSIONS: In summary, this is the first study to use CTS as a benchmark to validate and model tissue-engineered graft studies. CTS transplantation improved cell survival, established reperfusion, and enhanced cardiac function after myocardial infarction. These findings also confirm that dilative remodeling can be attenuated by topical transplantation of CTS but not skeletal muscle tissue grafts.


Asunto(s)
Células Madre Embrionarias/trasplante , Ventrículos Cardíacos/trasplante , Infarto del Miocardio/cirugía , Ingeniería de Tejidos , Trasplante de Tejidos/métodos , Animales , Animales Recién Nacidos , Femenino , Genes Reporteros , Supervivencia de Injerto , Proteínas Fluorescentes Verdes/genética , Humanos , Luciferasas de Luciérnaga/genética , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Modelos Animales , Contracción Miocárdica , Tamaño de los Órganos , Músculo Cuádriceps , Distribución Aleatoria
12.
Stem Cells ; 31(11): 2354-63, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24038578

RESUMEN

RATIONALE: Human embryonic stem cell (hESC) derivatives are attractive candidates for therapeutic use. The engraftment and survival of hESC derivatives as xenografts or allografts require effective immunosuppression to prevent immune cell infiltration and graft destruction. OBJECTIVE: To test the hypothesis that a short-course, dual-agent regimen of two costimulation-adhesion blockade agents can induce better engraftment of hESC derivatives compared to current immunosuppressive agents. METHODS AND RESULTS: We transduced hESCs with a double fusion reporter gene construct expressing firefly luciferase (Fluc) and enhanced green fluorescent protein, and differentiated these cells to endothelial cells (hESC-ECs). Reporter gene expression enabled longitudinal assessment of cell engraftment by bioluminescence imaging. Costimulation-adhesion therapy resulted in superior hESC-EC and mouse EC engraftment compared to cyclosporine therapy in a hind limb model. Costimulation-adhesion therapy also promoted robust hESC-EC and hESC-derived cardiomyocyte survival in an ischemic myocardial injury model. Improved hESC-EC engraftment had a cardioprotective effect after myocardial injury, as assessed by magnetic resonance imaging. Mechanistically, costimulation-adhesion therapy is associated with systemic and intragraft upregulation of T-cell immunoglobulin and mucin domain 3 (TIM3) and a reduced proinflammatory cytokine profile. CONCLUSIONS: Costimulation-adhesion therapy is a superior alternative to current clinical immunosuppressive strategies for preventing the post-transplant rejection of hESC derivatives. By extending the window for cellular engraftment, costimulation-adhesion therapy enhances functional preservation following ischemic injury. This regimen may function through a TIM3-dependent mechanism.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Ciclosporina/farmacología , Células Madre Embrionarias/trasplante , Rechazo de Injerto/prevención & control , Inmunoconjugados/farmacología , Inmunosupresores/farmacología , Prednisona/farmacología , Abatacept , Animales , Cardiotónicos/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Madre Embrionarias/citología , Células Madre Embrionarias/inmunología , Células Endoteliales/inmunología , Células Endoteliales/trasplante , Rechazo de Injerto/inmunología , Humanos , Tolerancia Inmunológica , Terapia de Inmunosupresión/métodos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/cirugía , Distribución Aleatoria
14.
Neuroimage ; 60(2): 1149-55, 2012 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-22266177

RESUMEN

Early inflammation following status epilepticus has been implicated in the development of epilepsy and the evolution of brain injury, yet its precise role remains unclear. The development of non-invasive imaging markers of inflammation would enable researchers to test this hypothesis in vivo and study its temporal progression in relation to epileptogenic insults. In this study we have investigated the potential of a targeted magnetic resonance imaging contrast agent--vascular cell adhesion molecule 1 antibody labelled iron oxide--to image the inflammatory process following status epilepticus in the rat lithium-pilocarpine model. Intravascular administration of the targeted contrast agent was performed at approximately 1 day following status epilepticus. The control group received diazepam prior to pilocarpine to prevent status epilepticus. Magnetic resonance imaging of rats was performed before and after contrast administration. Comparison with quantitative T2 measurements was also performed. At the end of the study, brains were removed for ex vivo magnetic resonance imaging and histology. Marked focal hypointensities caused by contrast agent binding were observed on in vivo magnetic resonance images in the post status epilepticus group. In particular these occurred in the periventricular organs, the hippocampus and the cerebral cortex. Relatively little contrast agent binding was observed in the control group. T2 relaxation times were not significantly increased for the hippocampus or the cerebral cortex in post status epilepticus animals. These results demonstrate the feasibility of in vivo imaging of seizure-induced inflammation in an animal model of epilepsy. The antibody targeted MRI contrast agent identified regions of acute inflammation following status epilepticus and may provide an early marker of brain injury. This technique could be used to determine the role of inflammation in models of epileptogenesis and to study the potential for anti-inflammatory therapeutic interventions.


Asunto(s)
Encéfalo/patología , Medios de Contraste , Compuestos Férricos , Inflamación/etiología , Inflamación/patología , Imagen por Resonancia Magnética/métodos , Estado Epiléptico/complicaciones , Molécula 1 de Adhesión Celular Vascular , Animales , Masculino , Microesferas , Ratas , Ratas Sprague-Dawley
15.
Geroscience ; 44(2): 731-746, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35107705

RESUMEN

The prevalence of cardiovascular disease increases exponentially with age, highlighting the contribution of aging mechanisms to cardiac diseases. Although model organisms which share human disease pathologies can elucidate mechanisms driving disease, they do not provide us with innate examples how cardiac aging might be slowed or attenuated. The identification of animal models that preserve cardiac function throughout most of life offers an alternative approach to study mechanisms which might slow cardiac aging. One such species may be the naked mole-rat (NMR), a mouse-sized (40 g) rodent with extraordinary longevity (> 37 years), and constant mortality hazard over its four decades of life. We used a cross-sectional study design to measure a range of physiological parameters in NMRs between 2 and 34 years of age and compared these findings with those of mice aged between 3 months and 2.5 years. We observed a rapid decline in body fat content and bone mineral density in old mice, but no changes in NMRs. Similarly, rhythm disorders (premature atrial and ventricular complexes) occurred in aged mice but not in NMRs. Magnetic resonance and ultrasound imaging showed age-dependent increases in cardiac hypertrophy and diastolic dysfunction in mice which were absent in NMRs. Finally, cardiac stress tests showed an age-dependent decline in normalized cardiac output in mice, which was absent in NMRs. Unlike mice, that manifest several aspects of human cardiac aging, NMRs maintain cardiac function and reserve capacity throughout their long lives and may offer insights on how to delay or prevent cardiac aging.


Asunto(s)
Longevidad , Ratas Topo , Envejecimiento/fisiología , Animales , Composición Corporal , Estudios Transversales , Longevidad/fisiología , Ratones , Ratas Topo/fisiología
16.
Genetics ; 220(1)2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34791228

RESUMEN

Dietary interventions can dramatically affect physiological health and organismal lifespan. The degree to which organismal health is improved depends upon genotype and the severity of dietary intervention, but neither the effects of these factors, nor their interaction, have been quantified in an outbred population. Moreover, it is not well understood what physiological changes occur shortly after dietary change and how these may affect the health of an adult population. In this article, we investigated the effect of 6-month exposure of either caloric restriction (CR) or intermittent fasting (IF) on a broad range of physiological traits in 960 1-year old Diversity Outbred mice. We found CR and IF affected distinct aspects of physiology and neither the magnitude nor the direction (beneficial or detrimental) of effects were concordant with the severity of the intervention. In addition to the effects of diet, genetic variation significantly affected 31 of 36 traits (heritabilities ranged from 0.04 to 0.65). We observed significant covariation between many traits that was due to both diet and genetics and quantified these effects with phenotypic and genetic correlations. We genetically mapped 16 diet-independent and 2 diet-dependent significant quantitative trait loci, both of which were associated with cardiac physiology. Collectively, these results demonstrate the degree to which diet and genetics interact to shape the physiological health of adult mice following 6 months of dietary intervention.


Asunto(s)
Restricción Calórica
17.
Med Phys ; 38(7): 3932-43, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21858990

RESUMEN

PURPOSE: Our aim was to compare different magnet arrangements for magnetic cell delivery to human lower leg arteries and investigate the theoretical targeting efficiency under realistic flow conditions as a possible treatment after angioplasty. Additionally the potential of scaling down or translating the magnetic actuation device for preclinical studies was explored. METHODS: Using finite element methods, the magnetic field distribution was calculated in 3D for the optimization of magnet arrangements. Computational fluid dynamics simulations were performed for the human posterior tibial artery with the geometry and boundary condition data derived from magnetic resonance imaging (MRI) studies. These simulations were used to trace the trajectories of cells for an optimized magnet arrangement. Additionally the behavior of cells close to the vessel wall was investigated using a fluid-structure interaction model. RESULTS: The optimal magnet for the lower leg arteries was a Halbach cylinder k3 variety (12 elements with 900 rotation steps for the magnetization orientation). With this magnet, numerical simulations predict a targeting efficiency of 6.25% could be achieved in the posterior tibial artery for cells containing 150 pg iron. Similar simulations, which were scaled down to rabbit dimensions while keeping the forces acting on a cell constant, lead to similar predicted targeting efficiencies. Fluid dynamic and fluid-structure interaction simulations predict that magnetically labeled cells within a 0.5% radii distance to the vessel wall would be attracted and remain at the wall under physiological flow conditions. CONCLUSIONS: First pass capture of magnetically labeled cells under pulsatile flow conditions in human lower leg arteries leads to low targeting efficiencies. However, this can be increased to almost 100% by stopping the blood flow for 5 min. A magnetic actuation device can be designed for animal models that generate magnetic forces achievable for cells in human leg arteries.


Asunto(s)
Trasplante de Células/métodos , Células Endoteliales/efectos de la radiación , Células Endoteliales/trasplante , Magnetismo/métodos , Modelos Biológicos , Enfermedad Arterial Periférica/fisiopatología , Enfermedad Arterial Periférica/cirugía , Animales , Células Cultivadas , Simulación por Computador , Campos Electromagnéticos , Células Endoteliales/fisiología , Femenino , Análisis de Elementos Finitos , Humanos , Masculino , Persona de Mediana Edad , Conejos
18.
J Magn Reson Imaging ; 32(4): 869-77, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20882617

RESUMEN

PURPOSE: To establish the accuracy, intra- and inter-observer variabilities of four different segmentation methods for measuring cardiac functional parameters in healthy and infarcted rat hearts. MATERIALS AND METHODS: Six Wistar rats were imaged before and after myocardial infarction using an electrocardiogram and respiratory-gated spoiled gradient echo sequence. Blinded and randomized datasets were analyzed by various semi-automatic and manual segmentation methods to compare their measurement bias and variability. In addition, the accuracy of these methods was assessed by comparison with reference measurements acquired from high-resolution three-dimensional (3D) datasets of a heart phantom. RESULTS: Relative inter- and intra-observer variability were found to be similar for all four methods. Semi-automatic segmentation methods reduced analysis time by up to 70%, while yielding similar measurement bias and variability compared with manual segmentation. Semi-automatic methods were found to underestimate the ejection fraction for healthy hearts compared with manual segmentation while overestimating them in infarcted hearts. However, semi-automatic segmentation of short axis slices agreed better with 3D reference scans of a heart phantom compared with manual segmentation. CONCLUSION: Semi-automatic segmentation methods are faster than manual segmentation, while offering a similar intra- and inter-observer variability. However, a potential bias has been observed between healthy and infarcted hearts for different methods, which should also be considered when selecting the most appropriate analysis technique.


Asunto(s)
Corazón/fisiología , Imagen por Resonancia Cinemagnética/métodos , Imagen por Resonancia Magnética/métodos , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/patología , Miocardio/patología , Animales , Automatización , Electrocardiografía/métodos , Procesamiento de Imagen Asistido por Computador , Masculino , Variaciones Dependientes del Observador , Fantasmas de Imagen , Ratas , Ratas Wistar , Reproducibilidad de los Resultados
19.
Neoplasia ; 21(10): 1036-1050, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31521051

RESUMEN

Cancer immunotherapies have demonstrated durable responses in a range of different cancers. However, only a subset of patients responds to these therapies. We set out to test if non-invasive imaging of tumor perfusion and vascular inflammation may be able to explain differences in T-cell infiltration in pre-clinical tumor models, relevant for treatment outcomes. Tumor perfusion and vascular cell adhesion molecule (VCAM-1) density were quantified using magnetic resonance imaging (MRI) and correlated with infiltration of adoptively transferred and endogenous T-cells. MRI biomarkers were evaluated for their ability to detect tumor rejection 3 days after T-cell transfer. Baseline levels of these markers were used to assess their ability to predict PD-L1 treatment response. We found correlations between MRI-derived VCAM-1 density and infiltration of endogenous or adoptively transferred T-cells in some preclinical tumor models. Blocking T-cell binding to endothelial cell adhesion molecules (VCAM-1/ICAM) prevented T-cell mediated tumor rejection. Tumor rejection could be detected 3 days after adoptive T-cell transfer prior to tumor volume changes by monitoring the extracellular extravascular volume fraction. Imaging tumor perfusion and VCAM-1 density before treatment initiation was able to predict the response of MC38 tumors to PD-L1 blockade. These results indicate that MRI based assessment of tumor perfusion and VCAM-1 density can inform about the permissibility of the tumor vasculature for T-cell infiltration which may explain some of the observed variance in treatment response for cancer immunotherapies.


Asunto(s)
Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias/diagnóstico , Neoplasias/metabolismo , Imagen de Perfusión , Linfocitos T/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Antineoplásicos Inmunológicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Biomarcadores , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Femenino , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Imagen por Resonancia Magnética , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Tomografía de Emisión de Positrones , Linfocitos T/inmunología , Linfocitos T/patología
20.
Cell Death Differ ; 26(11): 2416-2429, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30824837

RESUMEN

Emerging research suggests that multiple tumor compartments can influence treatment responsiveness and relapse, yet the search for therapeutic resistance mechanisms remains largely focused on acquired genomic alterations in cancer cells. Here we show how treatment-induced changes occur in multiple tumor compartments during tumor relapse and can reduce benefit of follow-on therapies. By using serial biopsies, next-generation sequencing, and single-cell transcriptomics, we tracked the evolution of multiple cellular compartments within individual lesions during first-line treatment response, relapse, and second-line therapeutic interventions in an autochthonous model of melanoma. We discovered that although treatment-relapsed tumors remained genetically stable, they converged on a shared resistance phenotype characterized by dramatic changes in tumor cell differentiation state, immune infiltration, and extracellular matrix (ECM) composition. Similar alterations in tumor cell differentiation were also observed in more than half of our treatment-relapsed patient tumors. Tumor cell-state changes were coincident with ECM remodeling and increased tumor stiffness, which alone was sufficient to alter tumor cell fate and reduce treatment responses in melanoma cell lines in vitro. Despite the absence of acquired mutations in the targeted pathway, resistant tumors showed significantly decreased responsiveness to second-line therapy intervention within the same pathway. The ability to preclinically model relapse and refractory settings-while capturing dynamics within and crosstalk between all relevant tumor compartments-provides a unique opportunity to better design and sequence appropriate clinical interventions.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Matriz Extracelular/patología , Melanoma/tratamiento farmacológico , Melanoma/patología , Animales , Azetidinas/farmacología , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN/genética , Resistencia a Antineoplásicos/fisiología , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Melanoma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Piperidinas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Vemurafenib/farmacología , Secuenciación del Exoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA