Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 12(4)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38609317

RESUMEN

BACKGROUND: Chimeric antigen receptor (CAR) T-cell therapy target receptor tyrosine kinase-like orphan receptor 1 (ROR1) is broadly expressed in hematologic and solid tumors, however clinically-characterized ROR1-CAR T cells with single chain variable fragment (scFv)-R12 targeting domain failed to induce durable remissions, in part due to the immunosuppressive tumor microenvironment (TME). Herein, we describe the development of an improved ROR1-CAR with a novel, fully human scFv9 targeting domain, and augmented with TGFßRIIDN armor protective against a major TME factor, transforming growth factor beta (TGFß). METHODS: CAR T cells were generated by lentiviral transduction of enriched CD4+ and CD8+ T cells, and the novel scFv9-based ROR1-CAR-1 was compared with the clinically-characterized ROR1-R12-scFv-based CAR-2 in vitro and in vivo. RESULTS: CAR-1 T cells exhibited greater CAR surface density than CAR-2 when normalized for %CAR+, and produced more interferon (IFN)-γ tumor necrosis factor (TNF)-α and interleukin (IL)-2 in response to hematologic (Jeko-1, RPMI-8226) and solid (OVCAR-3, Capan-2, NCI-H226) tumor cell lines in vitro. In vivo, CAR-1 and CAR-2 both cleared hematologic Jeko-1 lymphoma xenografts, however only CAR-1 fully rejected ovarian solid OVCAR-3 tumors, concordantly with greater expansion of CD8+ and CD4+CAR T cells, and enrichment for central and effector memory phenotype. When equipped with TGFß-protective armor TGFßRIIDN, CAR-1 T cells resisted TGFß-mediated pSmad2/3 phosphorylation, as compared with CAR-1 alone. When co-cultured with ROR-1+ AsPC-1 pancreatic cancer line in the presence of TGFß1, armored CAR-1 demonstrated improved recovery of killing function, IFN-γ, TNF-α and IL-2 secretion. In mouse AsPC-1 pancreatic tumor xenografts overexpressing TGFß1, armored CAR-1, in contrast to CAR-1 alone, achieved complete tumor remissions, and yielded accelerated expansion of CAR+ T cells, diminished circulating active TGFß1, and no apparent toxicity or weight loss. Unexpectedly, in AsPC-1 xenografts without TGFß overexpression, TGFß1 production was specifically induced by ROR-1-CAR T cells interaction with ROR-1 positive tumor cells, and the TGFßRIIDN armor conferred accelerated tumor clearance. CONCLUSIONS: The novel fully human TGFßRIIDN-armored ROR1-CAR-1 T cells are highly potent against ROR1-positive tumors, and withstand the inhibitory effects of TGFß in solid TME. Moreover, TGFß1 induction represents a novel, CAR-induced checkpoint in the solid TME, which can be circumvented by co-expressing the TGßRIIDN armor on T cells.


Asunto(s)
Neoplasias Ováricas , Neoplasias Pancreáticas , Humanos , Animales , Femenino , Ratones , Apoptosis , Linfocitos T CD8-positivos , Línea Celular Tumoral , Factor de Crecimiento Transformador beta , Microambiente Tumoral , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética
2.
Nat Commun ; 10(1): 2061, 2019 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-31053734

RESUMEN

Nephrotoxicity is a critical adverse event that leads to discontinuation of kinase inhibitor (KI) treatment. Here we show, through meta-analyses of FDA Adverse Event Reporting System, that dasatinib is associated with high risk for glomerular toxicity that is uncoupled from hypertension, suggesting a direct link between dasatinib and podocytes. We further investigate the cellular effects of dasatinib and other comparable KIs with varying risks of nephrotoxicity. Dasatinib treated podocytes show significant changes in focal adhesions, actin cytoskeleton, and morphology that are not observed with other KIs. We use phosphoproteomics and kinome profiling to identify the molecular mechanisms of dasatinib-induced injury to the actin cytoskeleton, and atomic force microscopy to quantify impairment to cellular biomechanics. Furthermore, chronic administration of dasatinib in mice causes reversible glomerular dysfunction, loss of stress fibers, and foot process effacement. We conclude that dasatinib induces nephrotoxicity through altered podocyte actin cytoskeleton, leading to injurious cellular biomechanics.


Asunto(s)
Citoesqueleto de Actina/efectos de los fármacos , Antineoplásicos/efectos adversos , Dasatinib/efectos adversos , Podocitos/patología , Inhibidores de Proteínas Quinasas/efectos adversos , Insuficiencia Renal Crónica/patología , Sistemas de Registro de Reacción Adversa a Medicamentos/estadística & datos numéricos , Animales , Línea Celular , Modelos Animales de Enfermedad , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Ratones , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Insuficiencia Renal Crónica/inducido químicamente , Estados Unidos , United States Food and Drug Administration
3.
Biomech Model Mechanobiol ; 18(5): 1297-1309, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30968216

RESUMEN

Multiscale models that couple agent-based modeling (ABM) and finite-element modeling (FEM) allow the dynamic simulation of tissue remodeling and wound healing, with mechanical environment influencing cellular behaviors even as tissue remodeling modifies mechanics. One of the challenges in coupling ABM to FEM is that these two domains typically employ grid or element sizes that differ by several orders of magnitude. Here, we develop and demonstrate an interpolation-based method for mapping between ABM and FEM domains of different resolutions that is suitable for linear and nonlinear FEM meshes and balances accuracy with computational demands. We then explore the effects of refining the FEM mesh and the ABM grid in the setting of a fully coupled model. ABM grid refinement studies showed unexpected effects of grid size whenever cells were present at a high enough density for crowding to affect proliferation and migration. In contrast to an FE-only model, refining the FE mesh in the coupled model increased strain differences between adjacent finite elements. Allowing strain-dependent feedback on collagen turnover magnified the effects of regional heterogeneity, producing highly nonlinear spatial and temporal responses. Our results suggest that the choice of both ABM grid and FEM mesh density in coupled models must be guided by experimental data and accompanied by careful grid and mesh refinement studies in the individual domains as well as the fully coupled model.


Asunto(s)
Análisis de Elementos Finitos , Modelos Biológicos , Análisis de Sistemas , Cicatrización de Heridas , Animales , Simulación por Computador , Ratas , Factores de Tiempo
4.
Front Physiol ; 10: 1481, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31920691

RESUMEN

Wound healing and fibrosis following myocardial infarction (MI) is a dynamic process involving many cell types, extracellular matrix (ECM), and inflammatory cues. As both incidence and survival rates for MI increase, management of post-MI recovery and associated complications are an increasingly important focus. Complexity of the wound healing process and the need for improved therapeutics necessitate a better understanding of the biochemical cues that drive fibrosis. To study the progression of cardiac fibrosis across spatial and temporal scales, we developed a novel hybrid multiscale model that couples a logic-based differential equation (LDE) model of the fibroblast intracellular signaling network with an agent-based model (ABM) of multi-cellular tissue remodeling. The ABM computes information about cytokine and growth factor levels in the environment including TGFß, TNFα, IL-1ß, and IL-6, which are passed as inputs to the LDE model. The LDE model then computes the network signaling state of individual cardiac fibroblasts within the ABM. Based on the current network state, fibroblasts make decisions regarding cytokine secretion and deposition and degradation of collagen. Simulated fibroblasts respond dynamically to rapidly changing extracellular environments and contribute to spatial heterogeneity in model predicted fibrosis, which is governed by many parameters including cell density, cell migration speeds, and cytokine levels. Verification tests confirmed that predictions of the coupled model and network model alone were consistent in response to constant cytokine inputs and furthermore, a subset of coupled model predictions were validated with in vitro experiments with human cardiac fibroblasts. This multiscale framework for cardiac fibrosis will allow for systematic screening of the effects of molecular perturbations in fibroblast signaling on tissue-scale extracellular matrix composition and organization.

5.
Am J Physiol Heart Circ Physiol ; 315(4): H1041-H1050, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30028201

RESUMEN

Previous studies have suggested that the composition and global mechanical properties of the scar tissue that forms after a myocardial infarction (MI) are key determinants of long-term survival, and emerging therapies such as biomaterial injection are designed in part to alter those mechanical properties. However, recent evidence suggests that local mechanics regulate scar formation post-MI, so that perturbing infarct mechanics could have unexpected consequences. We therefore tested the effect of changes in local mechanical environment on scar collagen turnover, accumulation, and alignment in 77 Sprague-Dawley rats at 1, 2, 3 and 6 wk post-MI by sewing a Dacron patch to the epicardium to eliminate circumferential strain while permitting continued longitudinal stretching with each heart beat. We found that collagen in healing infarcts aligned parallel to regional strain and perpendicular to the preinfarction muscle and collagen fiber direction, strongly supporting our hypothesis that mechanical environment is the primary determinant of scar collagen alignment. Mechanical reinforcement reduced levels of carboxy-terminal propeptide of type I procollagen (PICP; a biomarker for collagen synthesis) in samples collected by microdialysis significantly, particularly in the first 2 wk. Reinforcement also reduced carboxy-terminal telopeptide of type I collagen (ICTP; a biomarker for collagen degradation), particularly at later time points. These alterations in collagen turnover produced no change in collagen area fraction as measured by histology but significantly reduced wall thickness in the reinforced scars compared with untreated controls. Our findings confirm the importance of regional mechanics in regulating scar formation after infarction and highlight the potential for therapies that reduce stretch to also reduce wall thickness in healing infarcts. NEW & NOTEWORTHY This study shows that therapies such as surgical reinforcement, which reduce stretch in healing infarcts, can also reduce collagen synthesis and wall thickness and modify collagen alignment in postinfarction scars.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos/instrumentación , Colágeno/metabolismo , Infarto del Miocardio/cirugía , Miocardio/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Cicatrización de Heridas , Animales , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Fragmentos de Péptidos/metabolismo , Péptidos/metabolismo , Tereftalatos Polietilenos , Procolágeno/metabolismo , Ratas Sprague-Dawley , Factores de Tiempo
6.
Biophys J ; 114(11): 2717-2731, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29874620

RESUMEN

Atomic force microscopy (AFM) is used to study mechanical properties of biological materials at submicron length scales. However, such samples are often structurally heterogeneous even at the local level, with different regions having distinct mechanical properties. Physical or chemical disruption can isolate individual structural elements but may alter the properties being measured. Therefore, to determine the micromechanical properties of intact heterogeneous multilayered samples indented by AFM, we propose the Hybrid Eshelby Decomposition (HED) analysis, which combines a modified homogenization theory and finite element modeling to extract layer-specific elastic moduli of composite structures from single indentations, utilizing knowledge of the component distribution to achieve solution uniqueness. Using finite element model-simulated indentation of layered samples with micron-scale thickness dimensions, biologically relevant elastic properties for incompressible soft tissues, and layer-specific heterogeneity of an order of magnitude or less, HED analysis recovered the prescribed modulus values typically within 10% error. Experimental validation using bilayer spin-coated polydimethylsiloxane samples also yielded self-consistent layer-specific modulus values whether arranged as stiff layer on soft substrate or soft layer on stiff substrate. We further examined a biophysical application by characterizing layer-specific microelastic properties of full-thickness mouse aortic wall tissue, demonstrating that the HED-extracted modulus of the tunica media was more than fivefold stiffer than the intima and not significantly different from direct indentation of exposed media tissue. Our results show that the elastic properties of surface and subsurface layers of microscale synthetic and biological samples can be simultaneously extracted from the composite material response to AFM indentation. HED analysis offers a robust approach to studying regional micromechanics of heterogeneous multilayered samples without destructively separating individual components before testing.


Asunto(s)
Elasticidad , Microscopía de Fuerza Atómica , Animales , Aorta/citología , Aorta/diagnóstico por imagen , Dimetilpolisiloxanos , Análisis de Elementos Finitos , Ratones , Nylons
7.
Cell Physiol Biochem ; 44(2): 701-715, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29169155

RESUMEN

BACKGROUND/AIMS: Our previous studies demonstrated that intrinsic aortic smooth muscle cell (VSMC) stiffening plays a pivotal role in aortic stiffening in aging and hypertension. However, the underlying molecular mechanisms remain largely unknown. We here hypothesized that Rho kinase (ROCK) acts as a novel mediator that regulates intrinsic VSMC mechanical properties through the serum response factor (SRF) /myocardin pathway and consequently regulates aortic stiffness and blood pressure in hypertension. METHODS: Four-month old male spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats were studied. Aortic stiffness was measured by echography. Intrinsic mechanical properties of VSMCs were measured by atomic force microscopy (AFM) in vitro. RESULTS: Compared to WKY rats, SHR showed a significant increase in aortic stiffness and blood pressure, which is accompanied by a remarkable cell stiffening and ROCK activation in thoracic aortic (TA) VSMCs. Theses alterations in SHR were abolished by Y-27632, a specific inhibitor of ROCK. Additionally, boosted filamentous/globular actin ratio was detected in TA VSMCs from SHR versus WKY rats, resulting in an up-regulation of SRF and myocardin expression and its downstream stiffness-associated genes including α-smooth muscle actin, SM22, smoothelin and myosin heavy chain 11. Reciprocally, these alterations in SHR TA VSMCs were also suppressed by Y-27632. Furthermore, a specific inhibitor of SRF/myocardin, CCG-100602, showed a similar effect to Y-27632 in SHR in both TA VSMCs stiffness in vitro and aorta wall stiffness in vivo. CONCLUSION: ROCK is a novel mediator modulating aortic VSMC stiffness through SRF/myocardin signaling which offers a therapeutic target to reduce aortic stiffening in hypertension.


Asunto(s)
Actinas/metabolismo , Hipertensión/fisiopatología , Músculo Liso Vascular/fisiología , Proteínas Nucleares/metabolismo , Factor de Respuesta Sérica/metabolismo , Transactivadores/metabolismo , Quinasas Asociadas a rho/metabolismo , Amidas/farmacología , Animales , Aorta Torácica/citología , Aorta Torácica/fisiología , Presión Sanguínea , Proteínas del Citoesqueleto/metabolismo , Ecocardiografía , Hipertensión/veterinaria , Masculino , Microscopía de Fuerza Atómica , Proteínas Musculares/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Cadenas Pesadas de Miosina/metabolismo , Ácidos Nipecóticos/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Piridinas/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Factor de Respuesta Sérica/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Ultrasonografía , Regulación hacia Arriba , Rigidez Vascular/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores
8.
Cardiovasc Res ; 113(2): 171-182, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28003268

RESUMEN

AIMS: Increased aortic stiffness is a fundamental manifestation of hypertension. However, the molecular mechanisms involved remain largely unknown. We tested the hypothesis that abnormal intrinsic vascular smooth muscle cell (VSMC) mechanical properties in large arteries, but not in distal arteries, contribute to the pathogenesis of aortic stiffening in hypertension, mediated by the serum response factor (SRF)/myocardin signalling pathway. METHODS AND RESULTS: Four month old male spontaneously hypertensive rats (SHR) and normotensive Wistar-Kyoto (WKY) rats were studied. Using atomic force microscopy, significant VSMC stiffening was observed in the large conducting aorta compared with the distal arteries in SHR (P < 0.001), however, this regional variation was not observed in WKY rats (P > 0.4). The increase of VSMC stiffness was accompanied by a parallel increase in the expression of SRF by 9.8-fold and of myocardin by 10.5-fold in thoracic aortic VSMCs from SHR vs. WKY rats, resulting in a significant increase of downstream stiffness-associated genes (all, P < 0.01 vs. WKY). Inhibition of SRF/myocardin expression selectively attenuated aortic VSMC stiffening, and normalized downstream targets in VSMCs isolated from SHR but not from WKY rats. In vivo, 2 weeks of treatment with SRF/myocardin inhibitor delivered by subcutaneous osmotic minipump significantly reduced aortic stiffness and then blood pressure in SHR but not in WKY rats, although concomitant changes in aortic wall remodelling were not detected during this time frame. CONCLUSIONS: SRF/myocardin pathway acts as a pivotal mediator of aortic VSMC mechanical properties and plays a central role in the pathological aortic stiffening in hypertension. Attenuation of aortic VSMC stiffening by pharmacological inhibition of SRF/myocardin signalling presents a novel therapeutic strategy for the treatment of hypertension by targeting the cellular contributors to aortic stiffness.


Asunto(s)
Antihipertensivos/administración & dosificación , Hipertensión/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Ácidos Nipecóticos/administración & dosificación , Proteínas Nucleares/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Rigidez Vascular/efectos de los fármacos , Animales , Aorta Torácica/metabolismo , Aorta Torácica/fisiopatología , Presión Arterial/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Hipertensión/genética , Hipertensión/fisiopatología , Infusiones Subcutáneas , Masculino , Microscopía de Fuerza Atómica , Músculo Liso Vascular/fisiopatología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Transducción de Señal/efectos de los fármacos , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Ann Biomed Eng ; 44(10): 2994-3006, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27090893

RESUMEN

Marfan syndrome (MFS) is an autosomal dominant disease of the connective tissue due to mutations in the fibrillin-1 gene (FBN1). This study aimed at characterizing microelastic properties of the ascending aortic wall and lung parenchyma tissues from wild type (WT) and age-matched Fbn1 hypomorphic mice (Fbn1(mgR/mgR) mice) to identify tissue-specific biomechanical effects of aging and disease in MFS. Atomic force microscopy was used to indent lung parenchyma and aortic wall tissues, using Hybrid Eshelby Decomposition analysis to extract layer-specific properties of the intima and media. The intima stiffened with age and was not different between WT and Fbn1(mgR/mgR) tissues, whereas the media layer of MFS aortas showed progressive structural and mechanical degradation with a modulus that was 50% softer than WT by 3.5 months of age. Similarly, MFS mice displayed progressive structural and mechanical deterioration of lung tissue, which was over 85% softer than WT by 3.5 months of age. Chronic treatment with the angiotensin type I receptor antagonist, losartan, attenuated the aorta and lung tissue degradation, resulting in structural and mechanical properties not significantly different from age-matched WT controls. By revealing micromechanical softening of elastin-rich aorta and lung tissues with disease progression in fibrillin-1 deficient mice, our findings support the use of losartan as a prophylactic treatment that may abrogate the life-threatening symptoms of MFS.


Asunto(s)
Aorta , Losartán/farmacología , Pulmón , Síndrome de Marfan , Animales , Aorta/metabolismo , Aorta/patología , Aorta/fisiopatología , Modelos Animales de Enfermedad , Fibrilina-1/genética , Fibrilina-1/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Síndrome de Marfan/tratamiento farmacológico , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Síndrome de Marfan/fisiopatología , Ratones , Ratones Transgénicos
10.
FASEB J ; 28(2): 644-54, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24174427

RESUMEN

Cardiac experimental biology and translational research would benefit from an in vitro surrogate for human heart muscle. This study investigated structural and functional properties and interventional responses of human engineered cardiac tissues (hECTs) compared to human myocardium. Human embryonic stem cell-derived cardiomyocytes (hESC-CMs, >90% troponin-positive) were mixed with collagen and cultured on force-sensing elastomer devices. hECTs resembled trabecular muscle and beat spontaneously (1.18 ± 0.48 Hz). Microstructural features and mRNA expression of cardiac-specific genes (α-MHC, SERCA2a, and ACTC1) were comparable to human myocardium. Optical mapping revealed cardiac refractoriness with loss of 1:1 capture above 3 Hz, and cycle length dependence of the action potential duration, recapitulating key features of cardiac electrophysiology. hECTs reconstituted the Frank-Starling mechanism, generating an average maximum twitch stress of 660 µN/mm(2) at Lmax, approaching values in newborn human myocardium. Dose-response curves followed exponential pharmacodynamics models for calcium chloride (EC50 1.8 mM) and verapamil (IC50 0.61 µM); isoproterenol elicited a positive chronotropic but negligible inotropic response, suggesting sarcoplasmic reticulum immaturity. hECTs were amenable to gene transfer, demonstrated by successful transduction with Ad.GFP. Such 3-D hECTs recapitulate an early developmental stage of human myocardium and promise to offer an alternative preclinical model for cardiology research.


Asunto(s)
Miocardio/citología , Ingeniería de Tejidos/métodos , Línea Celular , Electrofisiología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...