Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nature ; 589(7841): 270-275, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33116299

RESUMEN

There is an urgent need to create novel models using human disease-relevant cells to study severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) biology and to facilitate drug screening. Here, as SARS-CoV-2 primarily infects the respiratory tract, we developed a lung organoid model using human pluripotent stem cells (hPSC-LOs). The hPSC-LOs (particularly alveolar type-II-like cells) are permissive to SARS-CoV-2 infection, and showed robust induction of chemokines following SARS-CoV-2 infection, similar to what is seen in patients with COVID-19. Nearly 25% of these patients also have gastrointestinal manifestations, which are associated with worse COVID-19 outcomes1. We therefore also generated complementary hPSC-derived colonic organoids (hPSC-COs) to explore the response of colonic cells to SARS-CoV-2 infection. We found that multiple colonic cell types, especially enterocytes, express ACE2 and are permissive to SARS-CoV-2 infection. Using hPSC-LOs, we performed a high-throughput screen of drugs approved by the FDA (US Food and Drug Administration) and identified entry inhibitors of SARS-CoV-2, including imatinib, mycophenolic acid and quinacrine dihydrochloride. Treatment at physiologically relevant levels of these drugs significantly inhibited SARS-CoV-2 infection of both hPSC-LOs and hPSC-COs. Together, these data demonstrate that hPSC-LOs and hPSC-COs infected by SARS-CoV-2 can serve as disease models to study SARS-CoV-2 infection and provide a valuable resource for drug screening to identify candidate COVID-19 therapeutics.


Asunto(s)
Antivirales/farmacología , COVID-19/virología , Colon/citología , Evaluación Preclínica de Medicamentos/métodos , Pulmón/citología , Organoides/efectos de los fármacos , Organoides/virología , SARS-CoV-2/efectos de los fármacos , Animales , COVID-19/prevención & control , Colon/efectos de los fármacos , Colon/virología , Aprobación de Drogas , Femenino , Xenoinjertos/efectos de los fármacos , Humanos , Técnicas In Vitro , Pulmón/efectos de los fármacos , Pulmón/virología , Masculino , Ratones , Organoides/citología , Organoides/metabolismo , SARS-CoV-2/genética , Estados Unidos , United States Food and Drug Administration , Tropismo Viral , Internalización del Virus/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
2.
Nature ; 585(7825): 426-432, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32908310

RESUMEN

Endothelial cells adopt tissue-specific characteristics to instruct organ development and regeneration1,2. This adaptability is lost in cultured adult endothelial cells, which do not vascularize tissues in an organotypic manner. Here, we show that transient reactivation of the embryonic-restricted ETS variant transcription factor 2 (ETV2)3 in mature human endothelial cells cultured in a serum-free three-dimensional matrix composed of a mixture of laminin, entactin and type-IV collagen (LEC matrix) 'resets' these endothelial cells to adaptable, vasculogenic cells, which form perfusable and plastic vascular plexi. Through chromatin remodelling, ETV2 induces tubulogenic pathways, including the activation of RAP1, which promotes the formation of durable lumens4,5. In three-dimensional matrices-which do not have the constraints of bioprinted scaffolds-the 'reset' vascular endothelial cells (R-VECs) self-assemble into stable, multilayered and branching vascular networks within scalable microfluidic chambers, which are capable of transporting human blood. In vivo, R-VECs implanted subcutaneously in mice self-organize into durable pericyte-coated vessels that functionally anastomose to the host circulation and exhibit long-lasting patterning, with no evidence of malformations or angiomas. R-VECs directly interact with cells within three-dimensional co-cultured organoids, removing the need for the restrictive synthetic semipermeable membranes that are required for organ-on-chip systems, therefore providing a physiological platform for vascularization, which we call 'Organ-On-VascularNet'. R-VECs enable perfusion of glucose-responsive insulin-secreting human pancreatic islets, vascularize decellularized rat intestines and arborize healthy or cancerous human colon organoids. Using single-cell RNA sequencing and epigenetic profiling, we demonstrate that R-VECs establish an adaptive vascular niche that differentially adjusts and conforms to organoids and tumoroids in a tissue-specific manner. Our Organ-On-VascularNet model will permit metabolic, immunological and physiochemical studies and screens to decipher the crosstalk between organotypic endothelial cells and parenchymal cells for identification of determinants of endothelial cell heterogeneity, and could lead to advances in therapeutic organ repair and tumour targeting.


Asunto(s)
Vasos Sanguíneos/citología , Carcinogénesis , Células Endoteliales/citología , Hemodinámica , Neoplasias/irrigación sanguínea , Organogénesis , Organoides/irrigación sanguínea , Vasos Sanguíneos/crecimiento & desarrollo , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Cromatina/metabolismo , Epigénesis Genética , Epigenómica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro , Islotes Pancreáticos/irrigación sanguínea , Modelos Biológicos , Especificidad de Órganos , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción , Transcriptoma
3.
Proc Natl Acad Sci U S A ; 120(41): e2308941120, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37782785

RESUMEN

Impaired lymphatic drainage and lymphedema are major morbidities whose mechanisms have remained obscure. To study lymphatic drainage and its impairment, we engineered a microfluidic culture model of lymphatic vessels draining interstitial fluid. This lymphatic drainage-on-chip revealed that inflammatory cytokines that are known to disrupt blood vessel junctions instead tightened lymphatic cell-cell junctions and impeded lymphatic drainage. This opposing response was further demonstrated when inhibition of rho-associated protein kinase (ROCK) was found to normalize fluid drainage under cytokine challenge by simultaneously loosening lymphatic junctions and tightening blood vessel junctions. Studies also revealed a previously undescribed shift in ROCK isoforms in lymphatic endothelial cells, wherein a ROCK2/junctional adhesion molecule-A (JAM-A) complex emerges that is responsible for the cytokine-induced lymphatic junction zippering. To validate these in vitro findings, we further demonstrated in a genetic mouse model that lymphatic-specific knockout of ROCK2 reversed lymphedema in vivo. These studies provide a unique platform to generate interstitial fluid pressure and measure the drainage of interstitial fluid into lymphatics and reveal a previously unappreciated ROCK2-mediated mechanism in regulating lymphatic drainage.


Asunto(s)
Molécula A de Adhesión de Unión , Vasos Linfáticos , Linfedema , Quinasas Asociadas a rho , Animales , Ratones , Biomimética , Citocinas/metabolismo , Células Endoteliales/metabolismo , Uniones Intercelulares , Molécula A de Adhesión de Unión/metabolismo , Vasos Linfáticos/metabolismo , Linfedema/genética , Linfedema/metabolismo , Quinasas Asociadas a rho/metabolismo
5.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33542154

RESUMEN

Cells derived from pluripotent sources in vitro must resemble those found in vivo as closely as possible at both transcriptional and functional levels in order to be a useful tool for studying diseases and developing therapeutics. Recently, differentiation of human pluripotent stem cells (hPSCs) into brain microvascular endothelial cells (ECs) with blood-brain barrier (BBB)-like properties has been reported. These cells have since been used as a robust in vitro BBB model for drug delivery and mechanistic understanding of neurological diseases. However, the precise cellular identity of these induced brain microvascular endothelial cells (iBMECs) has not been well described. Employing a comprehensive transcriptomic metaanalysis of previously published hPSC-derived cells validated by physiological assays, we demonstrate that iBMECs lack functional attributes of ECs since they are deficient in vascular lineage genes while expressing clusters of genes related to the neuroectodermal epithelial lineage (Epi-iBMEC). Overexpression of key endothelial ETS transcription factors (ETV2, ERG, and FLI1) reprograms Epi-iBMECs into authentic endothelial cells that are congruent with bona fide endothelium at both transcriptomic as well as some functional levels. This approach could eventually be used to develop a robust human BBB model in vitro that resembles the human brain EC in vivo for functional studies and drug discovery.


Asunto(s)
Endotelio Vascular/citología , Células Madre Pluripotentes/citología , Factores de Transcripción/genética , Animales , Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Encéfalo/citología , Diferenciación Celular , Línea Celular , Reprogramación Celular/fisiología , Endotelio Vascular/fisiología , Expresión Génica , Humanos , Ratones Endogámicos , Células Madre Pluripotentes/fisiología , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
6.
Annu Rev Biomed Eng ; 23: 517-546, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-33974812

RESUMEN

Liver disease is an important clinical problem, impacting 600 million people worldwide. It is the 11th-leading cause of death in the world. Despite constant improvement in treatment and diagnostics, the aging population and accumulated risk factors led to increased morbidity due to nonalcoholic fatty liver disease and steatohepatitis. Liver transplantation, first established in the 1960s, is the second-most-common solid organ transplantation and is the gold standard for the treatment of liver failure. However, less than 10% of the global need for liver transplantation is met at the current rates of transplantation due to the paucity of available organs. Cell- and tissue-based therapies present an alternative to organ transplantation. This review surveys the approaches and tools that have been developed, discusses the distinctive challenges that exist for cell- and tissue-based therapies, and examines the future directions of regenerative therapies for the treatment of liver disease.


Asunto(s)
Trasplante de Hígado , Enfermedad del Hígado Graso no Alcohólico , Anciano , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Enfermedad del Hígado Graso no Alcohólico/terapia , Factores de Riesgo
7.
Microcirculation ; 24(5)2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28376260

RESUMEN

OBJECTIVES: The morphogenetic events that occur during angiogenic sprouting involve several members of the Rho family of GTPases, including Cdc42. However, the precise roles of Cdc42 in angiogenic sprouting have been difficult to elucidate owing to the lack of models to study these events in vitro. Here, we aim to identify the roles of Cdc42 in branching morphogenesis in angiogenesis. METHODS: Using a 3D biomimetic model of angiogenesis in vitro, where endothelial cells were seeded inside a cylindrical channel within collagen gel and sprouted from the channel in response to a defined biochemical gradient of angiogenic factors, we inhibited Cdc42 activity with a small molecule inhibitor ML141 and examined the effects of Cdc42 on the morphogenetic processes of angiogenic sprouting. RESULTS: We find that partial inhibition of Cdc42 had minimal effects on directional migration of endothelial cells, but led to fewer branching events without affecting the length of these branches. We also observed that antagonizing Cdc42 reduced collective migration in favor of single cell migration. Additionally, Cdc42 also regulated the initiation of filopodial extensions in endothelial tip cells. CONCLUSIONS: Our findings suggest that Cdc42 can affect multiple morphogenetic processes during angiogenic sprouting and ultimately impact the architecture of the vasculature.


Asunto(s)
Morfogénesis , Neovascularización Fisiológica , Proteína de Unión al GTP cdc42/fisiología , Biomimética , Movimiento Celular , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Modelos Cardiovasculares , Seudópodos , Pirazoles/farmacología , Sulfonamidas/farmacología , Proteína de Unión al GTP cdc42/antagonistas & inhibidores , Proteína de Unión al GTP cdc42/farmacología
8.
Proc Natl Acad Sci U S A ; 111(22): 7968-73, 2014 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-24843171

RESUMEN

The density and architecture of capillary beds that form within a tissue depend on many factors, including local metabolic demand and blood flow. Here, using microfluidic control of local fluid mechanics, we show the existence of a previously unappreciated flow-induced shear stress threshold that triggers angiogenic sprouting. Both intraluminal shear stress over the endothelium and transmural flow through the endothelium above 10 dyn/cm(2) triggered endothelial cells to sprout and invade into the underlying matrix, and this threshold is not impacted by the maturation of cell-cell junctions or pressure gradient across the monolayer. Antagonizing VE-cadherin widened cell-cell junctions and reduced the applied shear stress for a given transmural flow rate, but did not affect the shear threshold for sprouting. Furthermore, both transmural and luminal flow induced expression of matrix metalloproteinase 1, and this up-regulation was required for the flow-induced sprouting. Once sprouting was initiated, continuous flow was needed to both sustain sprouting and prevent retraction. To explore the potential ramifications of a shear threshold on the spatial patterning of new sprouts, we used finite-element modeling to predict fluid shear in a variety of geometric settings and then experimentally demonstrated that transmural flow guided preferential sprouting toward paths of draining interstitial fluid flow as might occur to connect capillary beds to venules or lymphatics. In addition, we show that luminal shear increases in local narrowings of vessels to trigger sprouting, perhaps ultimately to normalize shear stress across the vasculature. Together, these studies highlight the role of shear stress in controlling angiogenic sprouting and offer a potential homeostatic mechanism for regulating vascular density.


Asunto(s)
Células Endoteliales/fisiología , Metaloproteinasa 1 de la Matriz/fisiología , Microfluídica/instrumentación , Microfluídica/métodos , Modelos Biológicos , Neovascularización Fisiológica/fisiología , Adenoviridae/genética , Capilares/citología , Capilares/fisiología , Movimiento Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/ultraestructura , Análisis de Elementos Finitos , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Metaloproteinasa 1 de la Matriz/genética , Mecanotransducción Celular/fisiología , Microscopía Electrónica de Transmisión , Estrés Mecánico
9.
Proc Natl Acad Sci U S A ; 110(17): 6712-7, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23569284

RESUMEN

Angiogenesis is a complex morphogenetic process whereby endothelial cells from existing vessels invade as multicellular sprouts to form new vessels. Here, we have engineered a unique organotypic model of angiogenic sprouting and neovessel formation that originates from preformed artificial vessels fully encapsulated within a 3D extracellular matrix. Using this model, we screened the effects of angiogenic factors and identified two distinct cocktails that promoted robust multicellular endothelial sprouting. The angiogenic sprouts in our system exhibited hallmark structural features of in vivo angiogenesis, including directed invasion of leading cells that developed filopodia-like protrusions characteristic of tip cells, following stalk cells exhibiting apical-basal polarity, and lumens and branches connecting back to the parent vessels. Ultimately, sprouts bridged between preformed channels and formed perfusable neovessels. Using this model, we investigated the effects of angiogenic inhibitors on sprouting morphogenesis. Interestingly, the ability of VEGF receptor 2 inhibition to antagonize filopodia formation in tip cells was context-dependent, suggesting a mechanism by which vessels might be able to toggle between VEGF-dependent and VEGF-independent modes of angiogenesis. Like VEGF, sphingosine-1-phosphate also seemed to exert its proangiogenic effects by stimulating directional filopodial extension, whereas matrix metalloproteinase inhibitors prevented sprout extension but had no impact on filopodial formation. Together, these results demonstrate an in vitro 3D biomimetic model that reconstitutes the morphogenetic steps of angiogenic sprouting and highlight the potential utility of the model to elucidate the molecular mechanisms that coordinate the complex series of events involved in neovascularization.


Asunto(s)
Biomimética/métodos , Microfluídica/métodos , Modelos Biológicos , Morfogénesis/fisiología , Neovascularización Fisiológica/fisiología , Polaridad Celular/fisiología , Dimetilpolisiloxanos , Clorhidrato de Fingolimod , Técnica del Anticuerpo Fluorescente , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indoles/farmacología , Lisofosfolípidos/metabolismo , Morfogénesis/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Glicoles de Propileno/farmacología , Seudópodos/fisiología , Pirroles/farmacología , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
10.
Bio Protoc ; 14(2): e4925, 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38268979

RESUMEN

Cell-based liver therapies utilizing functionally stabilized engineered hepatic tissue hold promise in improving host liver functions and are emerging as a potential alternative to whole-organ transplantation. Owing to the ability to accommodate a large ex vivo engineered hepatocyte mass and dense vascularization, the mesenteric parametrial fat pad in female nude mice forms an ideal anatomic microenvironment for ectopic hepatocyte transplantation. However, the lack of any reported protocol detailing the presurgical preparation and construction of the engineered hepatic hydrogel, fat pad surgery, and postsurgical care and bioluminescence imaging to confirm in vivo hepatocyte implantation makes it challenging to reliably perform and test engraftment and integration with the host. In this report, we provide a step-by-step protocol for in vivo hepatocyte implantation, including preparation of hepatic tissue for implantation, the surgery process, and bioluminescence imaging to assess survival of functional hepatocytes. This will be a valuable protocol for researchers in the fields of tissue engineering, transplantation, and regenerative medicine. Key features • Primary human hepatocytes transduced ex vivo with a lentiviral vector carrying firefly luciferase are surgically implanted onto the fat pad. • Bioluminescence helps monitor survival of transplanted hepatic tissue over time. • Applicable for assessment of graft survival, graft-host integration, and liver regeneration.

11.
Nat Mater ; 11(9): 768-74, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22751181

RESUMEN

In the absence of perfusable vascular networks, three-dimensional (3D) engineered tissues densely populated with cells quickly develop a necrotic core. Yet the lack of a general approach to rapidly construct such networks remains a major challenge for 3D tissue culture. Here, we printed rigid 3D filament networks of carbohydrate glass, and used them as a cytocompatible sacrificial template in engineered tissues containing living cells to generate cylindrical networks that could be lined with endothelial cells and perfused with blood under high-pressure pulsatile flow. Because this simple vascular casting approach allows independent control of network geometry, endothelialization and extravascular tissue, it is compatible with a wide variety of cell types, synthetic and natural extracellular matrices, and crosslinking strategies. We also demonstrated that the perfused vascular channels sustained the metabolic function of primary rat hepatocytes in engineered tissue constructs that otherwise exhibited suppressed function in their core.


Asunto(s)
Vasos Sanguíneos/citología , Perfusión , Impresión/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/metabolismo , Materiales Biocompatibles/farmacología , Carbohidratos/química , Vidrio/química , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Ratas , Factores de Tiempo
12.
Cell Rep ; 37(4): 109897, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34706243

RESUMEN

Elevated circulating activity of adenosine deaminase 2 (ADA2) is associated with liver fibrosis in nonalcoholic fatty liver disease (NAFLD). In the liver of NAFLD patients, ADA2-positive portal macrophages are significantly associated with the degree of liver fibrosis. These liver macrophages are CD14- and CD16-positive and co-express chemokine receptors CCR2, CCR5, and CXCR3, indicating infiltrative monocyte origin. Human circulatory monocytes release ADA2 upon macrophage differentiation in vitro. When stimulated by recombinant human ADA2 (rhADA2), human monocyte-derived macrophages demonstrate upregulation of pro-inflammatory and pro-fibrotic genes, including PDGF-B, a key pro-fibrotic cytokine. This PDGF-B upregulation is reproduced by inosine, the enzymatic product of ADA2, but not adenosine, and is abolished by E359N, a loss-of-function mutation in ADA2. Finally, rhADA2 also stimulates PDGF-B production from Kupffer cells in primary human liver spheroids. Together, these data suggest that infiltrative monocytes promote fibrogenesis in NAFLD via ADA2-mediated autocrine/paracrine signaling culminating in enhanced PDGF-B production.


Asunto(s)
Adenosina Desaminasa/metabolismo , Comunicación Autocrina , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Macrófagos del Hígado/enzimología , Hígado/enzimología , Monocitos/enzimología , Enfermedad del Hígado Graso no Alcohólico/enzimología , Comunicación Paracrina , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-sis/metabolismo
13.
Cell Stem Cell ; 27(1): 125-136.e7, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32579880

RESUMEN

SARS-CoV-2 has caused the COVID-19 pandemic. There is an urgent need for physiological models to study SARS-CoV-2 infection using human disease-relevant cells. COVID-19 pathophysiology includes respiratory failure but involves other organ systems including gut, liver, heart, and pancreas. We present an experimental platform comprised of cell and organoid derivatives from human pluripotent stem cells (hPSCs). A Spike-enabled pseudo-entry virus infects pancreatic endocrine cells, liver organoids, cardiomyocytes, and dopaminergic neurons. Recent clinical studies show a strong association with COVID-19 and diabetes. We find that human pancreatic beta cells and liver organoids are highly permissive to SARS-CoV-2 infection, further validated using adult primary human islets and adult hepatocyte and cholangiocyte organoids. SARS-CoV-2 infection caused striking expression of chemokines, as also seen in primary human COVID-19 pulmonary autopsy samples. hPSC-derived cells/organoids provide valuable models for understanding the cellular responses of human tissues to SARS-CoV-2 infection and for disease modeling of COVID-19.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/virología , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Organoides/virología , Neumonía Viral/virología , Tropismo , Enzima Convertidora de Angiotensina 2 , Animales , Autopsia , COVID-19 , Línea Celular , Infecciones por Coronavirus/patología , Hepatocitos/patología , Hepatocitos/virología , Humanos , Células Madre Pluripotentes Inducidas/virología , Hígado/patología , Ratones , Páncreas/patología , Páncreas/virología , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/patología , SARS-CoV-2 , Internalización del Virus
14.
Sci Adv ; 5(8): eaav6789, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31489365

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, lethal malignancy that invades adjacent vasculatures and spreads to distant sites before clinical detection. Although invasion into the peripancreatic vasculature is one of the hallmarks of PDAC, paradoxically, PDAC tumors also exhibit hypovascularity. How PDAC tumors become hypovascular is poorly understood. We describe an organotypic PDAC-on-a-chip culture model that emulates vascular invasion and tumor-blood vessel interactions to better understand PDAC-vascular interactions. The model features a 3D matrix containing juxtaposed PDAC and perfusable endothelial lumens. PDAC cells invaded through intervening matrix, into vessel lumen, and ablated the endothelial cells, leaving behind tumor-filled luminal structures. Endothelial ablation was also observed in in vivo PDAC models. We also identified the activin-ALK7 pathway as a mediator of endothelial ablation by PDAC. This tumor-on-a-chip model provides an important in vitro platform for investigating the process of PDAC-driven endothelial ablation and may provide a mechanism for tumor hypovascularity.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Células Endoteliales/metabolismo , Neoplasias Pancreáticas/metabolismo , Transducción de Señal/fisiología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Biomimética/métodos , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular , Línea Celular Tumoral , Células Endoteliales/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Invasividad Neoplásica/patología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
15.
J Biomed Opt ; 19(1): 16006, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24395588

RESUMEN

Understanding the mechanism of angiogenesis could help to decipher wound healing and embryonic development and to develop better treatment for diseases such as cancer. Microengineered devices were developed to reveal the mechanisms of angiogenesis, but monitoring the angiogenic process nondestructively in these devices is a challenge. In this study, we utilized a label-free imaging technique, ultrahigh-resolution optical coherence microscopy (OCM), to evaluate angiogenic sprouting in a microengineered device. The OCM system was capable of providing ∼1.5-µm axial resolution and ∼2.3-µm transverse resolution. Three-dimensional (3-D) distribution of the sprouting vessels in the microengineered device was imaged over 0.6×0.6×0.5 mm3, and details such as vessel lumens and branching points were clearly visualized. An algorithm based on stretching open active contours was developed for tracking and segmenting the sprouting vessels in 3-D-OCM images. The lengths for the first-, second-, and third-order vessels were measured as 127.8±48.8 µm (n=8), 67.3±25.9 µm (n=9), and 62.5±34.7 µm (n=10), respectively. The outer diameters for the first-, second-, and third-order vessels were 13.2±1.0, 8.0±2.1, and 4.4±0.8 µm, respectively. These results demonstrate OCM as a promising tool for nondestructive and label-free evaluation of angiogenic sprouting in microengineered devices.


Asunto(s)
Neovascularización Fisiológica , Tomografía de Coherencia Óptica/métodos , Algoritmos , Animales , Colágeno/química , Humanos , Imagenología Tridimensional , Luz , Microscopía Confocal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA