Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 11(1): 5535, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33692370

ABSTRACT

Lung cancer rates are rising globally and non-small cell lung cancer (NSCLC) has a five year survival rate of only 24%. Unfortunately, the development of drugs to treat cancer is severely hampered by the inefficiency of translating pre-clinical studies into clinical benefit. Thus, we sought to apply a tumor microenvironment system (TMES) to NSCLC. Using microvascular endothelial cells, lung cancer derived fibroblasts, and NSCLC tumor cells in the presence of in vivo tumor-derived hemodynamic flow and transport, we demonstrate that the TMES generates an in-vivo like biological state and predicts drug response to EGFR inhibitors. Transcriptomic and proteomic profiling indicate that the TMES recapitulates the in vivo and patient molecular biological state providing a mechanistic rationale for the predictive nature of the TMES. This work further validates the TMES for modeling patient tumor biology and drug response indicating utility of the TMES as a predictive tool for drug discovery and development and potential for use as a system for patient avatars.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Endothelial Cells/metabolism , Lung Neoplasms/metabolism , Models, Biological , Tumor Microenvironment , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Endothelial Cells/pathology , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Mice, SCID
2.
Lab Chip ; 19(7): 1193-1204, 2019 03 27.
Article in English | MEDLINE | ID: mdl-30839006

ABSTRACT

The development of drugs to treat cancer is hampered by the inefficiency of translating pre-clinical in vitro monoculture and mouse studies into clinical benefit. There is a critical need to improve the accuracy of evaluating pre-clinical drug efficacy through the development of more physiologically relevant models. In this study, a human triculture 3D in vitro tumor microenvironment system (TMES) was engineered to accurately mimic the tumor microenvironment. The TMES recapitulates tumor hemodynamics and biological transport with co-cultured human microvascular endothelial cells, pancreatic ductal adenocarcinoma, and pancreatic stellate cells. We demonstrate that significant tumor cell transcriptomic changes occur in the TMES that correlate with the in vivo xenograft and patient transcriptome. Treatment with therapeutically relevant doses of chemotherapeutics yields responses paralleling the patients' clinical responses. Thus, this model provides a unique platform to rigorously evaluate novel therapies and is amenable to using patient tumor material directly, with applicability for patient avatars.


Subject(s)
Biomimetics/methods , Carcinoma, Pancreatic Ductal/pathology , Tumor Microenvironment , Cell Proliferation/drug effects , Humans , Tumor Microenvironment/drug effects
3.
Stem Cells Transl Med ; 6(8): 1673-1683, 2017 08.
Article in English | MEDLINE | ID: mdl-28628273

ABSTRACT

Human induced pluripotent stem cells (iPSCs) can be differentiated into vascular endothelial (iEC) and smooth muscle (iSMC) cells. However, because iECs and iSMCs are not derived from an intact blood vessel, they represent an immature phenotype. Hemodynamics and heterotypic cell:cell communication play important roles in vascular cell phenotypic modulation. Here we tested the hypothesis that hemodynamic exposure of iECs in coculture with iSMCs induces an in vivo-like phenotype. iECs and iSMCs were cocultured under vascular region-specific blood flow hemodynamics, and compared to hemodynamic cocultures of blood vessel-derived endothelial (pEC) and smooth muscle (pSMC) cells. Hemodynamic flow-induced gene expression positively correlated between pECs and iECs as well as pSMCs and iSMCs. While endothelial nitric oxide synthase 3 protein was lower in iECs than pECs, iECs were functionally mature as seen by acetylated-low-density lipoprotein (LDL) uptake. SMC contractile protein markers were also positively correlated between pSMCs and iSMCs. Exposure of iECs and pECs to atheroprone hemodynamics with oxidized-LDL induced an inflammatory response in both. Dysfunction of the transforming growth factor ß (TGFß) pathway is seen in several vascular diseases, and iECs and iSMCs exhibited a transcriptomic prolife similar to pECs and pSMCs, respectively, in their responses to LY2109761-mediated transforming growth factor ß receptor I/II (TGFßRI/II) inhibition. Although there are differences between ECs and SMCs derived from iPSCs versus blood vessels, hemodynamic coculture restores a high degree of similarity in their responses to pathological stimuli associated with vascular diseases. Thus, iPSC-derived vascular cells exposed to hemodynamics may provide a viable system for modeling rare vascular diseases and testing new therapeutic approaches. Stem Cells Translational Medicine 2017;6:1673-1683.


Subject(s)
Cell Differentiation , Endothelial Cells/cytology , Hemodynamics , Induced Pluripotent Stem Cells/cytology , Myocytes, Smooth Muscle/cytology , Phenotype , Transcriptome , Cells, Cultured , Coculture Techniques/methods , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
4.
Thromb Res ; 143: 34-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27179130

ABSTRACT

An experimental in vitro model of the hemodynamics that occur in atrial fibrillation (AFib) in the left atrial appendage (LAA) was developed to study changes in human endothelial cell thrombotic potential. We applied human-derived sinus rhythm and AFib hemodynamic shear stress patterns to primary human endothelial cells (ECs) in culture. We found that ECs exposed to AFib hemodynamics have increased thrombotic potential as measured by increased expression of pro-thrombotic gene markers and fibrin deposition on the endothelium. Treatment with the factor Xa inhibitor, apixaban, attenuated fibrin deposition thickness while increasing fibrin density at the endothelial cell surface. This study suggests that altered hemodynamics associated with AFib play a key role in driving the thrombotic potential of the LAA endothelium.


Subject(s)
Atrial Appendage/pathology , Atrial Fibrillation/blood , Atrial Fibrillation/complications , Endothelial Cells/pathology , Hemodynamics , Thrombosis/blood , Thrombosis/etiology , Atrial Fibrillation/pathology , Cells, Cultured , Fibrin/analysis , Humans , Thrombosis/pathology
5.
Mol Genet Metab ; 117(3): 355-362, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26740382

ABSTRACT

BACKGROUND: Propionic acidemia (PA) is a disorder of intermediary metabolism with defects in the alpha or beta subunits of propionyl CoA carboxylase (PCCA and PCCB respectively) enzyme. We previously described a liver culture system that uses liver-derived hemodynamic blood flow and transport parameters to restore and maintain primary human hepatocyte biology and metabolism utilizing physiologically relevant milieu concentrations. METHODS: In this study, primary hepatocytes isolated from the explanted liver of an 8-year-old PA patient were cultured in the liver system for 10 days and evaluated for retention of differentiated polarized morphology. The expression of PCCA and PCCB was assessed at a gene and protein level relative to healthy donor controls. Ammonia and urea levels were measured in the presence and absence of amino acid supplements to assess the metabolic consequences of branched-chain amino acid metabolism in this disease. RESULTS: Primary hepatocytes from the PA patient maintained a differentiated polarized morphology (peripheral actin staining) over 10 days of culture in the system. We noted lower levels of PCCA and PCCB relative to normal healthy controls at the mRNA and protein level. Supplementation of branched-chain amino acids, isoleucine (5mM) and valine (5mM) in the medium, resulted in increased ammonia and decreased urea in the PA patient hepatocyte system, but no such response was seen in healthy hepatocytes or patient-derived fibroblasts. CONCLUSIONS: We demonstrate for the first time the successful culture of PA patient-derived primary hepatocytes in a differentiated state, that stably retain the PCCA and PCCB enzyme defects at a gene and protein level. Phenotypic response of the system to an increased load of branched-chain amino acids, not possible with fibroblasts, underscores the utility of this system in the better understanding of the molecular pathophysiology of PA and examining the effectiveness of potential therapeutic agents in the most relevant tissue.


Subject(s)
Hepatocytes/cytology , Hepatocytes/metabolism , Propionic Acidemia/metabolism , Actins/analysis , Amino Acids, Branched-Chain/metabolism , Ammonia/metabolism , Carbon-Carbon Ligases/genetics , Carbon-Carbon Ligases/metabolism , Cells, Cultured , Child , Fibroblasts/drug effects , Fibroblasts/metabolism , Hemodynamics , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Isoleucine/pharmacology , Liver/enzymology , Liver/metabolism , Liver/pathology , Methylmalonyl-CoA Decarboxylase/genetics , Methylmalonyl-CoA Decarboxylase/metabolism , Mutation , Urea/metabolism , Valine/pharmacology
6.
Arterioscler Thromb Vasc Biol ; 35(10): 2185-95, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26293464

ABSTRACT

OBJECTIVES: The predictive value of animal and in vitro systems for drug development is limited, particularly for nonhuman primate studies as it is difficult to deduce the drug mechanism of action. We describe the development of an in vitro cynomolgus macaque vascular system that reflects the in vivo biology of healthy, atheroprone, or advanced inflammatory cardiovascular disease conditions. APPROACH AND RESULTS: We compare the responses of the in vitro human and cynomolgus vascular systems to 4 statins. Although statins exert beneficial pleiotropic effects on the human vasculature, the mechanism of action is difficult to investigate at the tissue level. Using RNA sequencing, we quantified the response to statins and report that most statins significantly increased the expression of genes that promote vascular health while suppressing inflammatory cytokine gene expression. Applying computational pathway analytics, we identified statin-regulated biological themes, independent of cholesterol lowering, that provide mechanisms for off-target effects, including thrombosis, cell cycle regulation, glycogen metabolism, and ethanol degradation. CONCLUSIONS: The cynomolgus vascular system described herein mimics the baseline and inflammatory regional biology of the human vasculature, including statin responsiveness, and provides mechanistic insight not achievable in vivo.


Subject(s)
Cardiovascular Diseases/drug therapy , Drug Evaluation, Preclinical/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipoproteins, LDL/drug effects , Animals , Cardiovascular Diseases/blood , Cells, Cultured , Endothelial Cells/drug effects , Humans , In Vitro Techniques , Lipoproteins, LDL/metabolism , Macaca fascicularis , Models, Cardiovascular , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Species Specificity
7.
Am J Physiol Cell Physiol ; 293(6): C1824-33, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17913848

ABSTRACT

Atherosclerosis is an inflammatory disease that preferentially forms at hemodynamically compromised regions of altered shear stress patterns. Endothelial cells (EC) and smooth muscle cells (SMC) undergo phenotypic modulation during atherosclerosis. An in vitro coculture model was developed to determine the role of hemodynamic regulation of EC and SMC phenotypes in coculture. Human ECs and SMCs were plated on a synthetic elastic lamina and human-derived atheroprone, and atheroprotective shear stresses were imposed on ECs. Atheroprone flow decreased genes associated with differentiated ECs (endothelial nitric oxide synthase, Tie2, and Kruppel-like factor 2) and SMCs (smooth muscle alpha-actin and myocardin) and induced a proinflammatory phenotype in ECs and SMCs (VCAM-1, IL-8, and monocyte chemoattractant protein-1). Atheroprone flow-induced changes in SMC differentiation markers were regulated at the chromatin level, as indicated by decreased serum response factor (SRF) binding to the smooth muscle alpha-actin-CC(a/T)(6)GG (CArG) promoter region and decreased histone H(4) acetylation. Conversely, SRF and histone H(4) acetylation were enriched at the c-fos promoter in SMCs. In the presence of atheroprotective shear stresses, ECs aligned with the direction of flow and SMCs aligned more perpendicular to flow, similar to in vivo vessel organization. These results provide a novel mechanism whereby modulation of the EC phenotype by hemodynamic shear stresses, atheroprone or atheroprotective, play a critical role in mechanical-transcriptional coupling and regulation of the SMC phenotype.


Subject(s)
Atherosclerosis/pathology , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Hemodynamics/physiology , Myocytes, Smooth Muscle/pathology , Arteries/physiopathology , Atherosclerosis/physiopathology , Cells, Cultured , Coculture Techniques , Endothelium, Vascular/physiopathology , Epigenesis, Genetic , Gene Expression , Humans , Inflammation/physiopathology , Phenotype , Proteins/isolation & purification , RNA/isolation & purification
8.
Am J Physiol Heart Circ Physiol ; 293(3): H1937-46, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17586613

ABSTRACT

Hemodynamic regulation of directional endothelial cell (EC) migration implies an essential role of shear stress in governing EC polarity. Shear stress induces reorientation of the microtubule organizing center toward the leading edge of migrating cells in a Cdc42-dependent manner. We have characterized the global patterns of EC migration in confluent monolayers as a function of shear stress direction and exogenous pleiotropic factors. Results demonstrate the presence of mitogenic factors significantly affects the flow-induced dynamics of movement by prolonging the onset of monolayer quiescence up to 4 days, but not shear stress-induced morphology. In conjunction with increased motility, exogenous growth factors contributed to the directed migration of ECs in the flow direction. ECs exposed to arterial flow in serum/growth factor-free media and then supplemented with growth factors rapidly increased directional migration to 85% of cells migrating in the direction of flow and induced an increase in the distance traveled with the flow direction. This response was modulated by the directionality of flow and inhibited by the expression of dominant-negative Par6, a major downstream effector of Cdc42-induced polarity. Shear stress-induced directed migratory polarity is modulated by exogenous growth factors and dependent on Par6 activity and shear stress direction.


Subject(s)
Cell Movement/physiology , Cell Polarity/physiology , Endothelium, Vascular/cytology , Adaptor Proteins, Signal Transducing/metabolism , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Protein Kinase C-delta/metabolism , Stress, Mechanical , cdc42 GTP-Binding Protein/metabolism
9.
J Cell Biol ; 176(5): 719-27, 2007 Feb 26.
Article in English | MEDLINE | ID: mdl-17312022

ABSTRACT

Elevated permeability of the endothelium is thought to be crucial in atherogenesis because it allows circulating lipoproteins to access subendothelial monocytes. Both local hemodynamics and cytokines may govern endothelial permeability in atherosclerotic plaque. We recently found that p21-activated kinase (PAK) regulates endothelial permeability. We now report that onset of fluid flow, atherogenic flow profiles, oxidized LDL, and proatherosclerotic cytokines all stimulate PAK phosphorylation and recruitment to cell-cell junctions. Activation of PAK is higher in cells plated on fibronectin (FN) compared to basement membrane proteins in all cases. In vivo, PAK is activated in atherosclerosis-prone regions of arteries and correlates with FN in the subendothelium. Inhibiting PAK in vivo reduces permeability in atherosclerosis-prone regions. Matrix-specific PAK activation therefore mediates elevated vascular permeability in atherogenesis.


Subject(s)
Atherosclerosis/enzymology , Capillary Permeability , Extracellular Matrix/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Cattle , Cytokines/pharmacology , Enzyme Activation , Fibronectins/metabolism , Intercellular Junctions/enzymology , Lipoproteins, LDL/pharmacology , Phosphorylation , Signal Transduction , p21-Activated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...