Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(9): e2310993121, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38386707

RESUMEN

How do vessels find optimal radii? Capillaries are known to adapt their radii to maintain the shear stress of blood flow at the vessel wall at a set point, yet models of adaptation purely based on average shear stress have not been able to produce complex loopy networks that resemble real microvascular systems. For narrow vessels where red blood cells travel in a single file, the shear stress on vessel endothelium peaks sharply when a red blood cell passes through. We show that stable shear-stress-based adaptation is possible if vessel shear stress set points are cued to the stress peaks. Model networks that respond to peak stresses alone can quantitatively reproduce the observed zebrafish trunk microcirculation, including its adaptive trajectory when hematocrit changes or parts of the network are amputated. Our work reveals the potential for mechanotransduction alone to generate stable hydraulically tuned microvascular networks.


Asunto(s)
Mecanotransducción Celular , Pez Cebra , Animales , Microvasos , Endotelio Vascular , Venas
2.
Circ Res ; 132(10): 1405-1424, 2023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-37167356

RESUMEN

SARS-CoV-2, the virus underlying COVID-19, has now been recognized to cause multiorgan disease with a systemic effect on the host. To effectively combat SARS-CoV-2 and the subsequent development of COVID-19, it is critical to detect, monitor, and model viral pathogenesis. In this review, we discuss recent advancements in microfluidics, organ-on-a-chip, and human stem cell-derived models to study SARS-CoV-2 infection in the physiological organ microenvironment, together with their limitations. Microfluidic-based detection methods have greatly enhanced the rapidity, accessibility, and sensitivity of viral detection from patient samples. Engineered organ-on-a-chip models that recapitulate in vivo physiology have been developed for many organ systems to study viral pathology. Human stem cell-derived models have been utilized not only to model viral tropism and pathogenesis in a physiologically relevant context but also to screen for effective therapeutic compounds. The combination of all these platforms, along with future advancements, may aid to identify potential targets and develop novel strategies to counteract COVID-19 pathogenesis.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Microfluídica , Sistemas Microfisiológicos
3.
Mol Ther ; 32(6): 1849-1874, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38584391

RESUMEN

The clinical potential of current FDA-approved chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapy is encumbered by its autologous nature, which presents notable challenges related to manufacturing complexities, heightened costs, and limitations in patient selection. Therefore, there is a growing demand for off-the-shelf universal cell therapies. In this study, we have generated universal CAR-engineered NKT (UCAR-NKT) cells by integrating iNKT TCR engineering and HLA gene editing on hematopoietic stem cells (HSCs), along with an ex vivo, feeder-free HSC differentiation culture. The UCAR-NKT cells are produced with high yield, purity, and robustness, and they display a stable HLA-ablated phenotype that enables resistance to host cell-mediated allorejection. These UCAR-NKT cells exhibit potent antitumor efficacy to blood cancers and solid tumors, both in vitro and in vivo, employing a multifaceted array of tumor-targeting mechanisms. These cells are further capable of altering the tumor microenvironment by selectively depleting immunosuppressive tumor-associated macrophages and myeloid-derived suppressor cells. In addition, UCAR-NKT cells demonstrate a favorable safety profile with low risks of graft-versus-host disease and cytokine release syndrome. Collectively, these preclinical studies underscore the feasibility and significant therapeutic potential of UCAR-NKT cell products and lay a foundation for their translational and clinical development.


Asunto(s)
Células Madre Hematopoyéticas , Inmunoterapia Adoptiva , Células T Asesinas Naturales , Receptores Quiméricos de Antígenos , Humanos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Animales , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Inmunoterapia Adoptiva/métodos , Ratones , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Edición Génica , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias/terapia , Neoplasias/inmunología , Línea Celular Tumoral , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
4.
Nat Methods ; 18(5): 551-556, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33574612

RESUMEN

Light-field microscopy has emerged as a technique of choice for high-speed volumetric imaging of fast biological processes. However, artifacts, nonuniform resolution and a slow reconstruction speed have limited its full capabilities for in toto extraction of dynamic spatiotemporal patterns in samples. Here, we combined a view-channel-depth (VCD) neural network with light-field microscopy to mitigate these limitations, yielding artifact-free three-dimensional image sequences with uniform spatial resolution and high-video-rate reconstruction throughput. We imaged neuronal activities across moving Caenorhabditis elegans and blood flow in a beating zebrafish heart at single-cell resolution with volumetric imaging rates up to 200 Hz.


Asunto(s)
Caenorhabditis elegans/fisiología , Aprendizaje Profundo , Corazón/fisiología , Procesamiento de Imagen Asistido por Computador/métodos , Microscopía/métodos , Animales , Conducta Animal , Fenómenos Biomecánicos , Actividad Motora/fisiología , Neuronas/fisiología , Pez Cebra
5.
Environ Res ; 248: 118242, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38242419

RESUMEN

Exposure to ultrafine particles (UFPs) has been associated with multiple adverse health effects. Inhaled UFPs could reach the gastrointestinal tract and influence the composition of the gut microbiome. We have previously shown that oral ingestion of UFPs alters the gut microbiome and promotes intestinal inflammation in hyperlipidemic Ldlr-/- mice. Particulate matter (PM)2.5 inhalation studies have also demonstrated microbiome shifts in normolipidemic C57BL/6 mice. However, it is not known whether changes in microbiome precede or follow inflammatory effects in the intestinal mucosa. We hypothesized that inhaled UFPs modulate the gut microbiome prior to the development of intestinal inflammation. We studied the effects of UFP inhalation on the gut microbiome and intestinal mucosa in two hyperlipidemic mouse models (ApoE-/- mice and Ldlr-/- mice) and normolipidemic C57BL/6 mice. Mice were exposed to PM in the ultrafine-size range by inhalation for 6 h a day, 3 times a week for 10 weeks at a concentration of 300-350 µg/m3.16S rRNA gene sequencing was performed to characterize sequential changes in the fecal microbiome during exposures, and changes in the intestinal microbiome at the end. PM exposure led to progressive differentiation of the microbiota over time, associated with increased fecal microbial richness and evenness, altered microbial composition, and differentially abundant microbes by week 10 depending on the mouse model. Cross-sectional analysis of the small intestinal microbiome at week 10 showed significant changes in α-diversity, ß-diversity, and abundances of individual microbial taxa in the two hyperlipidemic models. These alterations of the intestinal microbiome were not accompanied, and therefore could not be caused, by increased intestinal inflammation as determined by histological analysis of small and large intestine, cytokine gene expression, and levels of fecal lipocalin. In conclusion, 10-week inhalation exposures to UFPs induced taxonomic changes in the microbiome of various animal models in the absence of intestinal inflammation.


Asunto(s)
Contaminantes Atmosféricos , Microbioma Gastrointestinal , Ratones , Animales , Material Particulado/análisis , Contaminantes Atmosféricos/toxicidad , Exposición por Inhalación/análisis , ARN Ribosómico 16S , Estudios Transversales , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Inflamación/inducido químicamente
6.
Artículo en Inglés | MEDLINE | ID: mdl-38523716

RESUMEN

In numerical simulations of cardiac mechanics, coupling the heart to a model of the circulatory system is essential for capturing physiological cardiac behavior. A popular and efficient technique is to use an electrical circuit analogy, known as a lumped parameter network or zero-dimensional (0D) fluid model, to represent blood flow throughout the cardiovascular system. Due to the strong physical interaction between the heart and the blood circulation, developing accurate and efficient numerical coupling methods remains an active area of research. In this work, we present a modular framework for implicitly coupling three-dimensional (3D) finite element simulations of cardiac mechanics to 0D models of blood circulation. The framework is modular in that the circulation model can be modified independently of the 3D finite element solver, and vice versa. The numerical scheme builds upon a previous work that combines 3D blood flow models with 0D circulation models (3D fluid - 0D fluid). Here, we extend it to couple 3D cardiac tissue mechanics models with 0D circulation models (3D structure - 0D fluid), showing that both mathematical problems can be solved within a unified coupling scheme. The effectiveness, temporal convergence, and computational cost of the algorithm are assessed through multiple examples relevant to the cardiovascular modeling community. Importantly, in an idealized left ventricle example, we show that the coupled model yields physiological pressure-volume loops and naturally recapitulates the isovolumic contraction and relaxation phases of the cardiac cycle without any additional numerical techniques. Furthermore, we provide a new derivation of the scheme inspired by the Approximate Newton Method of Chan (1985), explaining how the proposed numerical scheme combines the stability of monolithic approaches with the modularity and flexibility of partitioned approaches.

7.
Int J Mol Sci ; 25(13)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-39000289

RESUMEN

Inflammatory bowel disease (IBD) is an immunologically complex disorder involving genetic, microbial, and environmental risk factors. Its global burden has continued to rise since industrialization, with epidemiological studies suggesting that ambient particulate matter (PM) in air pollution could be a contributing factor. Prior animal studies have shown that oral PM10 exposure promotes intestinal inflammation in a genetic IBD model and that PM2.5 inhalation exposure can increase intestinal levels of pro-inflammatory cytokines. PM10 and PM2.5 include ultrafine particles (UFP), which have an aerodynamic diameter of <0.10 µm and biophysical and biochemical properties that promote toxicity. UFP inhalation, however, has not been previously studied in the context of murine models of IBD. Here, we demonstrated that ambient PM is toxic to cultured Caco-2 intestinal epithelial cells and examined whether UFP inhalation affected acute colitis induced by dextran sodium sulfate and 2,4,6-trinitrobenzenesulfonic acid. C57BL/6J mice were exposed to filtered air (FA) or various types of ambient PM reaerosolized in the ultrafine size range at ~300 µg/m3, 6 h/day, 3-5 days/week, starting 7-10 days before disease induction. No differences in weight change, clinical disease activity, or histology were observed between the PM and FA-exposed groups. In conclusion, UFP inhalation exposure did not exacerbate intestinal inflammation in acute, chemically-induced colitis models.


Asunto(s)
Colitis , Sulfato de Dextran , Ratones Endogámicos C57BL , Material Particulado , Ácido Trinitrobencenosulfónico , Material Particulado/toxicidad , Animales , Colitis/inducido químicamente , Colitis/patología , Ratones , Humanos , Sulfato de Dextran/toxicidad , Células CACO-2 , Ácido Trinitrobencenosulfónico/toxicidad , Ácido Trinitrobencenosulfónico/efectos adversos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Células Epiteliales/metabolismo , Modelos Animales de Enfermedad , Masculino , Tamaño de la Partícula
8.
PLoS Comput Biol ; 17(7): e1009175, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34228702

RESUMEN

Biomechanical forces intimately contribute to cardiac morphogenesis. However, volumetric imaging to investigate the cardiac mechanics with high temporal and spatial resolution remains an imaging challenge. We hereby integrated light-field microscopy (LFM) with light-sheet fluorescence microscopy (LSFM), coupled with a retrospective gating method, to simultaneously access myocardial contraction and intracardiac blood flow at 200 volumes per second. While LSFM allows for the reconstruction of the myocardial function, LFM enables instantaneous acquisition of the intracardiac blood cells traversing across the valves. We further adopted deformable image registration to quantify the ventricular wall displacement and particle tracking velocimetry to monitor intracardiac blood flow. The integration of LFM and LSFM enabled the time-dependent tracking of the individual blood cells and the differential rates of segmental wall displacement during a cardiac cycle. Taken together, we demonstrated a hybrid system, coupled with our image analysis pipeline, to simultaneously capture the myocardial wall motion with intracardiac blood flow during cardiac development.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Corazón , Animales , Biología Computacional , Embrión no Mamífero/diagnóstico por imagen , Embrión no Mamífero/fisiología , Corazón/diagnóstico por imagen , Corazón/crecimiento & desarrollo , Corazón/fisiología , Procesamiento de Imagen Asistido por Computador , Microscopía Fluorescente , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/fisiología
9.
Cardiovasc Drugs Ther ; 36(2): 201-215, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33459922

RESUMEN

PURPOSE: HIV infection is consistently associated with an increased risk of atherosclerotic cardiovascular disease, but the underlying mechanisms remain elusive. HIV protein Tat, a transcriptional activator of HIV, has been shown to activate NF-κB signaling and promote inflammation in vitro. However, the atherogenic effects of HIV Tat have not been investigated in vivo. Macrophages are one of the major cell types involved in the initiation and progression of atherosclerosis. We and others have previously revealed the important role of IκB kinase ß (IKKß), a central inflammatory coordinator through activating NF-κB, in the regulation of macrophage functions and atherogenesis. This study investigated the impact of HIV Tat exposure on macrophage functions and atherogenesis. METHODS: To investigate the effects of Tat on macrophage IKKß activation and atherosclerosis development in vivo, myeloid-specific IKKß-deficient LDLR-deficient (IKKßΔMyeLDLR-/-) mice and their control littermates (IKKßF/FLDLR-/-) were exposed to recombinant HIV protein Tat. RESULTS: Exposure to Tat significantly increased atherosclerotic lesion size and plaque vulnerability in IKKßF/FLDLR-/- but not IKKßΔMyeLDLR-/- mice. Deficiency of myeloid IKKß attenuated Tat-elicited macrophage inflammatory responses and atherosclerotic lesional inflammation in IKKßΔMyeLDLR-/- mice. Further, RNAseq analysis demonstrated that HIV protein Tat affects the expression of many atherosclerosis-related genes in vitro in an IKKß-dependent manner. CONCLUSIONS: Our findings reveal atherogenic effects of HIV protein Tat in vivo and demonstrate a pivotal role of myeloid IKKß in Tat-driven atherogenesis.


Asunto(s)
Aterosclerosis , Infecciones por VIH , Animales , Aterosclerosis/metabolismo , Infecciones por VIH/complicaciones , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Inflamación/metabolismo , Lipoproteínas LDL , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas , Receptores de LDL/metabolismo
10.
Stroke ; 52(1): 260-270, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33161843

RESUMEN

BACKGROUND AND PURPOSE: Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is associated with an increased rate of cerebrovascular events including ischemic stroke and intracerebral hemorrhage. The mechanisms underlying cerebral endothelial susceptibility and response to SARS-CoV-2 are unknown yet critical to understanding the association of SARS-CoV-2 infection with cerebrovascular events. METHODS: Endothelial cells were isolated from human brain and analyzed by RNA sequencing. Human umbilical vein and human brain microvascular cells were used in both monolayer culture and endothelialized within a 3-dimensional printed vascular model of the middle cerebral artery. Gene expression levels were measured by quantitative polymerase chain reaction and direct RNA hybridization. Recombinant SARS-CoV-2 S protein and S protein-containing liposomes were used to measure endothelial binding by immunocytochemistry. RESULTS: ACE2 (angiotensin-converting enzyme-2) mRNA levels were low in human brain and monolayer endothelial cell culture. Within the 3-dimensional printed vascular model, ACE2 gene expression and protein levels were progressively increased by vessel size and flow rates. SARS-CoV-2 S protein-containing liposomes were detected in human umbilical vein endothelial cells and human brain microvascular endothelial cells in 3-dimensional middle cerebral artery models but not in monolayer culture consistent with flow dependency of ACE2 expression. Binding of SARS-CoV-2 S protein triggered 83 unique genes in human brain endothelial cells including upregulation of complement component C3. CONCLUSIONS: Brain endothelial cells are susceptible to direct SARS-CoV-2 infection through flow-dependent expression of ACE2. Viral S protein binding triggers a unique gene expression profile in brain endothelia that may explain the association of SARS-CoV-2 infection with cerebrovascular events.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/virología , Células Endoteliales/virología , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Transcriptoma , Encéfalo/metabolismo , Encéfalo/virología , COVID-19/metabolismo , Células Cultivadas , Circulación Cerebrovascular/fisiología , Células Endoteliales/metabolismo , Humanos , Modelos Anatómicos , Estrés Mecánico
11.
Curr Top Membr ; 87: 131-151, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34696883

RESUMEN

Living cells are exposed to multiple mechanical stimuli from the extracellular matrix or from surrounding cells. Mechanoreceptors are molecules that display status changes in response to mechanical stimulation, transforming physical cues into biological responses to help the cells adapt to dynamic changes of the microenvironment. Mechanical stimuli are responsible for shaping the tridimensional development and patterning of the organs in early embryonic stages. The development of the heart is one of the first morphogenetic events that occur in embryos. As the circulation is established, the vascular system is exposed to constant shear stress, which is the force created by the movement of blood. Both spatial and temporal variations in shear stress differentially modulate critical steps in heart development, such as trabeculation and compaction of the ventricular wall and the formation of the heart valves. Zebrafish embryos are small, transparent, have a short developmental period and allow for real-time visualization of a variety of fluorescently labeled proteins to recapitulate developmental dynamics. In this review, we will highlight the application of zebrafish models as a genetically tractable model for investigating cardiovascular development and regeneration. We will introduce our approaches to manipulate mechanical forces during critical stages of zebrafish heart development and in a model of vascular regeneration, as well as advances in imaging technologies to capture these processes at high resolution. Finally, we will discuss the role of molecules of the Plexin family and Piezo cation channels as major mechanosensors recently implicated in cardiac morphogenesis.


Asunto(s)
Mecanotransducción Celular , Pez Cebra , Animales , Modelos Animales , Morfogénesis , Estrés Mecánico
12.
J Vasc Res ; 56(6): 273-283, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31466069

RESUMEN

Cardiovascular diseases such as coronary heart disease, myocardial infarction, and cardiac arrhythmia are the leading causes of morbidity and mortality in developed countries and are steadily increasing in developing countries. Fundamental mechanistic studies at the molecular, cellular, and animal model levels are critical for the diagnosis and treatment of these diseases. Despite being phylogenetically distant from humans, zebrafish share remarkable similarity in the genetics and electrophysiology of the cardiovascular system. In the last 2 decades, the development and deployment of innovative genetic manipulation techniques greatly facilitated the application of zebrafish as an animal model for studying basic biology and diseases. Hemodynamic shear stress is intimately involved in vascular development and homeostasis. The critical mechanosensitive signaling pathways in cardiovascular development and pathophysiology previously studied in mammals have been recapitulated in zebrafish. In this short article, we reviewed recent knowledge about the role of mechanosensitive pathways such as Notch, PKCε/PFKFB3, and Wnt/Ang2 in cardiovas-cular development and homeostasis from studies in the -zebrafish model.


Asunto(s)
Sistema Cardiovascular/metabolismo , Hemodinámica , Mecanotransducción Celular , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Sistema Cardiovascular/embriología , Regulación del Desarrollo de la Expresión Génica , Homeostasis , Organogénesis , Estrés Mecánico , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
13.
14.
Am J Physiol Heart Circ Physiol ; 314(6): H1203-H1213, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29451816

RESUMEN

Calcific aortic vasculopathy correlates with bone loss in osteoporosis in an age-independent manner. Prior work suggests that teriparatide, the bone anabolic treatment for postmenopausal osteoporosis, may inhibit the onset of aortic calcification. Whether teriparatide affects the progression of preexisting aortic calcification, widespread among this patient population, is unknown. Female apolipoprotein E-deficient mice were aged for over 1 yr to induce aortic calcification, treated for 4.5 wk with daily injections of control vehicle (PBS), 40 µg/kg teriparatide (PTH40), or 400 µg/kg teriparatide (PTH400), and assayed for aortic calcification by microcomputed tomography (microCT) before and after treatment. In a followup cohort, aged female apolipoprotein E-deficient mice were treated with PBS or PTH400 and assayed for aortic calcification by serial microCT and micropositron emission tomography. In both cohorts, aortic calcification detected by microCT progressed similarly in all groups. Mean aortic 18F-NaF incorporation, detected by serial micropositron emission tomography, increased in the PBS-treated group (+14 ± 5%). In contrast, 18F-NaF incorporation decreased in the PTH400-treated group (-33 ± 20%, P = 0.03). Quantitative histochemical analysis by Alizarin red staining revealed a lower mineral surface area index in the PTH400-treated group compared with the PBS-treated group ( P = 0.04). Furthermore, Masson trichrome staining showed a significant increase in collagen deposition in the left ventricular myocardium of mice that received PTH400 [2.1 ± 0.6% vs. control mice (0.5 ± 0.1%), P = 0.02]. In summary, although teriparatide may not affect the calcium mineral content of aortic calcification, it reduces 18F-NaF uptake in calcified lesions, suggesting the possibility that it may reduce mineral surface area with potential impact on plaque stability. NEW & NOTEWORTHY Parathyroid hormone regulates bone mineralization and may also affect vascular calcification, which is an important issue, given that its active fragment, teriparatide, is widely used for the treatment of osteoporosis. To determine whether teriparatide alters vascular calcification, we imaged aortic calcification in mice treated with teriparatide and control mice. Although teriparatide did not affect the calcium content of cardiovascular deposits, it reduced their fluoride tracer uptake.


Asunto(s)
Aorta/efectos de los fármacos , Enfermedades de la Aorta/tratamiento farmacológico , Aterosclerosis/tratamiento farmacológico , Conservadores de la Densidad Ósea/farmacología , Hiperlipidemias/complicaciones , Teriparatido/farmacología , Calcificación Vascular/tratamiento farmacológico , Factores de Edad , Envejecimiento , Animales , Aorta/diagnóstico por imagen , Aorta/patología , Enfermedades de la Aorta/diagnóstico por imagen , Enfermedades de la Aorta/patología , Aortografía/métodos , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/etiología , Aterosclerosis/patología , Conservadores de la Densidad Ósea/toxicidad , Angiografía por Tomografía Computarizada , Modelos Animales de Enfermedad , Femenino , Fibrosis , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ratones Noqueados para ApoE , Placa Aterosclerótica , Tomografía de Emisión de Positrones , Teriparatido/toxicidad , Calcificación Vascular/diagnóstico por imagen , Calcificación Vascular/etiología , Calcificación Vascular/patología , Microtomografía por Rayos X
15.
PLoS Comput Biol ; 13(10): e1005828, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29084212

RESUMEN

Blood flow and mechanical forces in the ventricle are implicated in cardiac development and trabeculation. However, the mechanisms of mechanotransduction remain elusive. This is due in part to the challenges associated with accurately quantifying mechanical forces in the developing heart. We present a novel computational framework to simulate cardiac hemodynamics in developing zebrafish embryos by coupling 4-D light sheet imaging with a stabilized finite element flow solver, and extract time-dependent mechanical stimuli data. We employ deformable image registration methods to segment the motion of the ventricle from high resolution 4-D light sheet image data. This results in a robust and efficient workflow, as segmentation need only be performed at one cardiac phase, while wall position in the other cardiac phases is found by image registration. Ventricular hemodynamics are then quantified by numerically solving the Navier-Stokes equations in the moving wall domain with our validated flow solver. We demonstrate the applicability of the workflow in wild type zebrafish and three treated fish types that disrupt trabeculation: (a) chemical treatment using AG1478, an ErbB2 signaling inhibitor that inhibits proliferation and differentiation of cardiac trabeculation; (b) injection of gata1a morpholino oligomer (gata1aMO) suppressing hematopoiesis and resulting in attenuated trabeculation; (c) weak-atriumm58 mutant (wea) with inhibited atrial contraction leading to a highly undeveloped ventricle and poor cardiac function. Our simulations reveal elevated wall shear stress (WSS) in wild type and AG1478 compared to gata1aMO and wea. High oscillatory shear index (OSI) in the grooves between trabeculae, compared to lower values on the ridges, in the wild type suggest oscillatory forces as a possible regulatory mechanism of cardiac trabeculation development. The framework has broad applicability for future cardiac developmental studies focused on quantitatively investigating the role of hemodynamic forces and mechanotransduction during morphogenesis.


Asunto(s)
Técnicas de Imagen Cardíaca/métodos , Ventrículos Cardíacos/embriología , Mecanotransducción Celular/fisiología , Modelos Cardiovasculares , Morfogénesis/fisiología , Función Ventricular/fisiología , Animales , Velocidad del Flujo Sanguíneo/fisiología , Simulación por Computador , Ventrículos Cardíacos/anatomía & histología , Interpretación de Imagen Asistida por Computador , Imagenología Tridimensional , Estrés Mecánico , Pez Cebra
16.
PLoS Comput Biol ; 13(12): e1005892, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29244812

RESUMEN

In animals, gas exchange between blood and tissues occurs in narrow vessels, whose diameter is comparable to that of a red blood cell. Red blood cells must deform to squeeze through these narrow vessels, transiently blocking or occluding the vessels they pass through. Although the dynamics of vessel occlusion have been studied extensively, it remains an open question why microvessels need to be so narrow. We study occlusive dynamics within a model microvascular network: the embryonic zebrafish trunk. We show that pressure feedbacks created when red blood cells enter the finest vessels of the trunk act together to uniformly partition red blood cells through the microvasculature. Using mathematical models as well as direct observation, we show that these occlusive feedbacks are tuned throughout the trunk network to prevent the vessels closest to the heart from short-circuiting the network. Thus occlusion is linked with another open question of microvascular function: how are red blood cells delivered at the same rate to each micro-vessel? Our analysis shows that tuning of occlusive feedbacks increase the total dissipation within the network by a factor of 11, showing that uniformity of flows rather than minimization of transport costs may be prioritized by the microvascular network.


Asunto(s)
Microcirculación/fisiología , Microvasos/fisiología , Modelos Cardiovasculares , Animales , Animales Modificados Genéticamente , Velocidad del Flujo Sanguíneo/fisiología , Biología Computacional , Eritrocitos/fisiología , Retroalimentación Fisiológica , Hemorreología , Microvasos/anatomía & histología , Pez Cebra
17.
Curr Cardiol Rep ; 20(5): 35, 2018 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-29574550

RESUMEN

PURPOSE OF REVIEW: Real-time 3-dimensional (3-D) imaging of cardiovascular injury and regeneration remains challenging. We introduced a multi-scale imaging strategy that uses light-sheet illumination to enable applications of cardiovascular injury and repair in models ranging from zebrafish to rodent hearts. RECENT FINDINGS: Light-sheet imaging enables rapid data acquisition with high spatiotemporal resolution and with minimal photo-bleaching or photo-toxicity. We demonstrated the capacity of this novel light-sheet approach for scanning a region of interest with specific fluorescence contrast, thereby providing axial and temporal resolution at the cellular level without stitching image columns or pivoting illumination beams during one-time imaging. This cutting-edge imaging technique allows for elucidating the differentiation of stem cells in cardiac regeneration, providing an entry point to discover novel micro-circulation phenomenon with clinical significance for injury and repair. These findings demonstrate the multi-scale applications of this novel light-sheet imaging strategy to advance research in cardiovascular development and regeneration.


Asunto(s)
Enfermedades Cardiovasculares/diagnóstico por imagen , Lesiones Cardíacas/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional , Miocardio/patología , Regeneración/fisiología , Pez Cebra/embriología , Animales , Enfermedades Cardiovasculares/patología , Lesiones Cardíacas/patología , Microscopía Fluorescente , Modelos Animales , Modelos Cardiovasculares , Roedores
18.
Sensors (Basel) ; 18(1)2017 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-29283402

RESUMEN

Heart disease is the leading cause of mortality in the U.S. with approximately 610,000 people dying every year. Effective therapies for many cardiac diseases are lacking, largely due to an incomplete understanding of their genetic basis and underlying molecular mechanisms. Zebrafish (Danio rerio) are an excellent model system for studying heart disease as they enable a forward genetic approach to tackle this unmet medical need. In recent years, our team has been employing electrocardiogram (ECG) as an efficient tool to study the zebrafish heart along with conventional approaches, such as immunohistochemistry, DNA and protein analyses. We have overcome various challenges in the small size and aquatic environment of zebrafish in order to obtain ECG signals with favorable signal-to-noise ratio (SNR), and high spatial and temporal resolution. In this paper, we highlight our recent efforts in zebrafish ECG acquisition with a cost-effective simplified microelectrode array (MEA) membrane providing multi-channel recording, a novel multi-chamber apparatus for simultaneous screening, and a LabVIEW program to facilitate recording and processing. We also demonstrate the use of machine learning-based programs to recognize specific ECG patterns, yielding promising results with our current limited amount of zebrafish data. Our solutions hold promise to carry out numerous studies of heart diseases, drug screening, stem cell-based therapy validation, and regenerative medicine.


Asunto(s)
Electrocardiografía , Animales , Corazón , Microelectrodos , Relación Señal-Ruido , Pez Cebra
19.
Dev Biol ; 404(2): 49-60, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26004360

RESUMEN

Angiogenesis relies on specialized endothelial tip cells to extend toward guidance cues in order to direct growing blood vessels. Although many of the signaling pathways that control this directional endothelial sprouting are well known, the specific cellular mechanisms that mediate this process remain to be fully elucidated. Here, we show that Polo-like kinase 2 (PLK2) regulates Rap1 activity to guide endothelial tip cell lamellipodia formation and subsequent angiogenic sprouting. Using a combination of high-resolution in vivo imaging of zebrafish vascular development and a human umbilical vein endothelial cell (HUVEC) in vitro cell culture system, we observed that loss of PLK2 function resulted in a reduction in endothelial cell sprouting and migration, whereas overexpression of PLK2 promoted angiogenesis. Furthermore, we discovered that PLK2 may control angiogenic sprouting by binding to PDZ-GEF to regulate RAP1 activity during endothelial cell lamellipodia formation and extracellular matrix attachment. Consistent with these findings, constitutively active RAP1 could rescue the endothelial cell sprouting defects observed in zebrafish and HUVEC PLK2 knockdowns. Overall, these findings reveal a conserved PLK2-RAP1 pathway that is crucial to regulate endothelial tip cell behavior in order to ensure proper vascular development and patterning in vertebrates.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica/genética , Proteínas Serina-Treonina Quinasas/genética , Pez Cebra/embriología , Proteínas de Unión al GTP rap1/metabolismo , Animales , Línea Celular , Movimiento Celular/genética , Proliferación Celular/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/metabolismo , Seudópodos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal
20.
Sens Actuators B Chem ; 235: 154-161, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27773967

RESUMEN

Plaque rupture causes acute coronary syndromes and stroke. Intraplaque oxidized low density lipoprotein (oxLDL) is metabolically unstable and prone to induce rupture. We designed an intravascular ultrasound (IVUS)-guided electrochemical impedance spectroscopy (EIS) sensor to enhance the detection reproducibility of oxLDL-laden plaques. The flexible 2-point micro-electrode array for EIS was affixed to an inflatable balloon anchored onto a co-axial double layer catheter (outer diameter = 2 mm). The mechanically scanning-driven IVUS transducer (45 MHz) was deployed through the inner catheter (diameter = 1.3 mm) to the acoustic impedance matched-imaging window. Water filled the inner catheter to match acoustic impedance and air was pumped between the inner and outer catheters to inflate the balloon. The integrated EIS and IVUS sensor was deployed into the ex vivo aortas dissected from the fat-fed New Zealand White (NZW) rabbits (n=3 for fat-fed, n= 5 normal diet). IVUS imaging was able to guide the 2-point electrode to align with the plaque for EIS measurement upon balloon inflation. IVUS-guided EIS signal demonstrated reduced variability and increased reproducibility (p < 0.0001 for magnitude, p < 0.05 for phase at < 15 kHz) as compared to EIS sensor alone (p < 0.07 for impedance, p < 0.4 for phase at < 15 kHz). Thus, we enhanced topographic and EIS detection of oxLDL-laden plaques via a catheter-based integrated sensor design to enhance clinical assessment for unstable plaque.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA