Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Integr Biol (Camb) ; 12(2): 21-33, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32118264

ABSTRACT

Accumulating evidence suggests that our ability to predict chemical effects on breast cancer is limited by a lack of physiologically relevant in vitro models; the typical in vitro breast cancer model consists of the cancer cell and excludes the mammary microenvironment. As the effects of the microenvironment on cancer cell behavior becomes more understood, researchers have called for the integration of the microenvironment into in vitro chemical testing systems. However, given the complexity of the microenvironment and the variety of platforms to choose from, identifying the essential parameters to include in a chemical testing platform is challenging. This review discusses the need for more complex in vitro breast cancer models and outlines different approaches used to model breast cancer in vitro. We provide examples of the microenvironment modulating breast cancer cell responses to chemicals and discuss strategies to help pinpoint what components should be included in a model.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Carcinoma, Intraductal, Noninfiltrating/diagnosis , Cell Line, Tumor , Disease Progression , Drug Screening Assays, Antitumor , Epithelium/pathology , Extracellular Matrix/metabolism , Female , Humans , Inflammation , Models, Statistical , Neoplasms , Phenotype , Tissue Engineering
2.
Nat Biomed Eng ; 2: 453-463, 2018.
Article in English | MEDLINE | ID: mdl-30533277

ABSTRACT

Alterations in the mechanical properties of erythrocytes occurring in inflammatory and hematologic disorders such as sickle cell disease (SCD) and malaria often lead to increased endothelial permeability, haemolysis, and microvascular obstruction. However, the associations among these pathological phenomena remain unknown. Here, we report a perfusable, endothelialized microvasculature-on-a-chip featuring an interpenetrating-polymer-network hydrogel that recapitulates the stiffness of blood-vessel intima, basement membrane self-deposition and self-healing endothelial barrier function for longer than 1 month. The microsystem enables the real-time visualization, with high spatiotemporal resolution, of microvascular obstruction and endothelial permeability under physiological flow conditions. We found how extracellular heme, a hemolytic byproduct, induces delayed but reversible endothelial permeability in a dose-dependent manner, and demonstrate that endothelial interactions with SCD or malaria-infected erythrocytes cause reversible microchannel occlusion and increased in situ endothelial permeability. The microvasculature-on-a-chip enables mechanistic insight into the endothelial barrier dysfunction associated with SCD, malaria and other inflammatory and haematological diseases.

3.
Nat Commun ; 9(1): 509, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29410404

ABSTRACT

Hemostasis encompasses an ensemble of interactions among platelets, coagulation factors, blood cells, endothelium, and hemodynamic forces, but current assays assess only isolated aspects of this complex process. Accordingly, here we develop a comprehensive in vitro mechanical injury bleeding model comprising an "endothelialized" microfluidic system coupled with a microengineered pneumatic valve that induces a vascular "injury". With perfusion of whole blood, hemostatic plug formation is visualized and "in vitro bleeding time" is measured. We investigate the interaction of different components of hemostasis, gaining insight into several unresolved hematologic issues. Specifically, we visualize and quantitatively demonstrate: the effect of anti-platelet agent on clot contraction and hemostatic plug formation, that von Willebrand factor is essential for hemostasis at high shear, that hemophilia A blood confers unstable hemostatic plug formation and altered fibrin architecture, and the importance of endothelial phosphatidylserine in hemostasis. These results establish the versatility and clinical utility of our microfluidic bleeding model.


Subject(s)
Bleeding Time , Blood Coagulation Tests , Hemorrhage , Hemostasis , Microfluidics , Blood Coagulation , Blood Platelets/metabolism , Cell Membrane/metabolism , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Ligands , Platelet Adhesiveness , Shear Strength , Stress, Mechanical
4.
Lab Chip ; 17(22): 3804-3816, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29052682

ABSTRACT

Blood cells circulate in a dynamic fluidic environment, and during hematologic processes such as hemostasis, thrombosis, and inflammation, blood cells interact biophysically with a myriad of vascular matrices-blood clots and the subendothelial matrix. While it is known that adherent cells physiologically respond to the mechanical properties of their underlying matrices, how blood cells interact with their mechanical microenvironment of vascular matrices remains poorly understood. To that end, we developed microfluidic systems that achieve high fidelity, high resolution, single-micron PDMS features that mimic the physical geometries of vascular matrices. With these electron beam lithography (EBL)-based microsystems, the physical interactions of individual blood cells with the mechanical properties of the matrices can be directly visualized. We observe that the physical presence of the matrix, in and of itself, mediates hematologic processes of the three major blood cell types: platelets, erythrocytes, and leukocytes. First, we find that the physical presence of single micron micropillars creates a shear microgradient that is sufficient to cause rapid, localized platelet adhesion and aggregation that leads to complete microchannel occlusion; this response is enhanced with the presence of fibrinogen or collagen on the micropillar surface. Second, we begin to describe the heretofore unknown biophysical parameters for the formation of schistocytes, pathologic erythrocyte fragments associated with various thrombotic microangiopathies (poorly understood, yet life-threatening blood disorders associated with microvascular thrombosis). Finally, we observe that the physical interactions with a vascular matrix is sufficient to cause neutrophils to form procoagulant neutrophil extracellular trap (NET)-like structures. By combining electron beam lithography (EBL), photolithography, and soft lithography, we thus create microfluidic devices that provide novel insight into the response of blood cells to the mechanical microenvironment of vascular matrices and have promise as research-enabling and diagnostic platforms.


Subject(s)
Blood Cells , Cellular Microenvironment/physiology , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Blood Cells/cytology , Blood Cells/physiology , Cells, Cultured , Dimethylpolysiloxanes , Equipment Design , Humans , Models, Biological , Nylons , Platelet Activation/physiology , Thrombosis/physiopathology , Thrombotic Microangiopathies/physiopathology
5.
Blood ; 130(24): 2654-2663, 2017 12 14.
Article in English | MEDLINE | ID: mdl-28978568

ABSTRACT

Abnormal sickle red blood cell (sRBC) biomechanics, including pathological deformability and adhesion, correlate with clinical severity in sickle cell disease (SCD). Clinical intravenous fluids (IVFs) of various tonicities are often used during treatment of vaso-occlusive pain episodes (VOE), the major cause of morbidity in SCD. However, evidence-based guidelines are lacking, and there is no consensus regarding which IVFs to use during VOE. Further, it is unknown how altering extracellular fluid tonicity with IVFs affects sRBC biomechanics in the microcirculation, where vaso-occlusion takes place. Here, we report how altering extracellular fluid tonicity with admixtures of clinical IVFs affects sRBC biomechanical properties by leveraging novel in vitro microfluidic models of the microcirculation, including 1 capable of deoxygenating the sRBC environment to monitor changes in microchannel occlusion risk and an "endothelialized" microvascular model that measures alterations in sRBC/endothelium adhesion under postcapillary venular conditions. Admixtures with higher tonicities (sodium = 141 mEq/L) affected sRBC biomechanics by decreasing sRBC deformability, increasing sRBC occlusion under normoxic and hypoxic conditions, and increasing sRBC adhesion in our microfluidic human microvasculature models. Admixtures with excessive hypotonicity (sodium = 103 mEq/L), in contrast, decreased sRBC adhesion, but overswelling prolonged sRBC transit times in capillary-sized microchannels. Admixtures with intermediate tonicities (sodium = 111-122 mEq/L) resulted in optimal changes in sRBC biomechanics, thereby reducing the risk for vaso-occlusion in our models. These results have significant translational implications for patients with SCD and warrant a large-scale prospective clinical study addressing optimal IVF management during VOE in SCD.


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/physiopathology , Erythrocyte Deformability/physiology , Extracellular Fluid/physiology , Biomechanical Phenomena , Cell Adhesion/physiology , Cells, Cultured , Erythrocytes, Abnormal/physiology , Extracellular Fluid/chemistry , Hemorheology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Osmolar Concentration
6.
Nat Mater ; 16(2): 230-235, 2017 02.
Article in English | MEDLINE | ID: mdl-27723740

ABSTRACT

Haemostasis occurs at sites of vascular injury, where flowing blood forms a clot, a dynamic and heterogeneous fibrin-based biomaterial. Paramount in the clot's capability to stem haemorrhage are its changing mechanical properties, the major drivers of which are the contractile forces exerted by platelets against the fibrin scaffold. However, how platelets transduce microenvironmental cues to mediate contraction and alter clot mechanics is unknown. This is clinically relevant, as overly softened and stiffened clots are associated with bleeding and thrombotic disorders. Here, we report a high-throughput hydrogel-based platelet-contraction cytometer that quantifies single-platelet contraction forces in different clot microenvironments. We also show that platelets, via the Rho/ROCK pathway, synergistically couple mechanical and biochemical inputs to mediate contraction. Moreover, highly contractile platelet subpopulations present in healthy controls are conspicuously absent in a subset of patients with undiagnosed bleeding disorders, and therefore may function as a clinical diagnostic biophysical biomarker.


Subject(s)
Blood Coagulation/physiology , Blood Flow Velocity/physiology , Blood Platelets/physiology , Flow Cytometry/methods , Mechanotransduction, Cellular/physiology , Platelet Activation/physiology , Platelet Adhesiveness/physiology , Cells, Cultured , Elastic Modulus/physiology , Hardness/physiology , Humans , Nanoparticles/chemistry
7.
Blood ; 126(6): 817-24, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-25931587

ABSTRACT

The mechanism of action of the widely used in vivo ferric chloride (FeCl3) thrombosis model remains poorly understood; although endothelial cell denudation is historically cited, a recent study refutes this and implicates a role for erythrocytes. Given the complexity of the in vivo environment, an in vitro reductionist approach is required to systematically isolate and analyze the biochemical, mass transfer, and biological phenomena that govern the system. To this end, we designed an "endothelial-ized" microfluidic device to introduce controlled FeCl3 concentrations to the molecular and cellular components of blood and vasculature. FeCl3 induces aggregation of all plasma proteins and blood cells, independent of endothelial cells, by colloidal chemistry principles: initial aggregation is due to binding of negatively charged blood components to positively charged iron, independent of biological receptor/ligand interactions. Full occlusion of the microchannel proceeds by conventional pathways, and can be attenuated by antithrombotic agents and loss-of-function proteins (as in IL4-R/Iba mice). As elevated FeCl3 concentrations overcome protective effects, the overlap between charge-based aggregation and clotting is a function of mass transfer. Our physiologically relevant in vitro system allows us to discern the multifaceted mechanism of FeCl3-induced thrombosis, thereby reconciling literature findings and cautioning researchers in using the FeCl3 model.


Subject(s)
Blood Platelets/drug effects , Chlorides/pharmacology , Erythrocytes/drug effects , Ferric Compounds/pharmacology , Protein Aggregates/drug effects , Aspirin/pharmacology , Biomechanical Phenomena , Blood Platelets/chemistry , Blood Platelets/cytology , Cell Aggregation/drug effects , Chlorides/antagonists & inhibitors , Chlorides/chemistry , Erythrocytes/chemistry , Erythrocytes/cytology , Ferric Compounds/antagonists & inhibitors , Ferric Compounds/chemistry , Fibrinolytic Agents/pharmacology , Heparin/pharmacology , Humans , Microfluidic Analytical Techniques , Models, Biological , Platelet-Rich Plasma/chemistry , Primary Cell Culture , Protein Binding , Static Electricity , Thrombosis/metabolism , Thrombosis/pathology
8.
Cell Rep ; 8(6): 1905-1918, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25242334

ABSTRACT

Circulating tumor cells (CTCs) are shed from primary tumors into the bloodstream, mediating the hematogenous spread of cancer to distant organs. To define their composition, we compared genome-wide expression profiles of CTCs with matched primary tumors in a mouse model of pancreatic cancer, isolating individual CTCs using epitope-independent microfluidic capture, followed by single-cell RNA sequencing. CTCs clustered separately from primary tumors and tumor-derived cell lines, showing low-proliferative signatures, enrichment for the stem-cell-associated gene Aldh1a2, biphenotypic expression of epithelial and mesenchymal markers, and expression of Igfbp5, a gene transcript enriched at the epithelial-stromal interface. Mouse as well as human pancreatic CTCs exhibit a very high expression of stromal-derived extracellular matrix (ECM) proteins, including SPARC, whose knockdown in cancer cells suppresses cell migration and invasiveness. The aberrant expression by CTCs of stromal ECM genes points to their contribution of microenvironmental signals for the spread of cancer to distant organs.


Subject(s)
Extracellular Matrix/genetics , Gene Expression Regulation, Neoplastic , Neoplastic Cells, Circulating/metabolism , Pancreatic Neoplasms/pathology , Aldehyde Dehydrogenase 1 Family , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Movement , Extracellular Matrix/metabolism , Humans , Mice , Osteonectin/antagonists & inhibitors , Osteonectin/genetics , Osteonectin/metabolism , Pancreatic Neoplasms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Sequence Analysis, RNA , Tumor Cells, Cultured
9.
Thromb Res ; 133(4): 532-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24440140

ABSTRACT

While the role of platelets in hemostasis is well characterized from a biological perspective, the biophysical interactions between platelets and their mechanical microenvironment are relatively unstudied. The field of cellular mechanics has developed a number of approaches to study the effects of extracellular matrix (ECM)-derived mechanical forces on various cells, and has elucidated that integrin-cytoskeleton-mediated force transduction governs many cellular processes. As platelets adhere and spread via molecular machinery that is similar to that which enables other cells to mechanosense and mechanotransduce forces from their biophysical microenvironment, platelets too are likely governed by the same overarching mechanisms. Indeed, recent platelet mechanobiology studies have revealed that key aspects of platelet physiology and activation are regulated by the mechanical and spatial properties of the ECM microenvironment. At the same time, there are also key differences that make platelets unique in the world of cells-- their size, origin as megakaryocyte fragments, and unique αIIbß3 integrin-- render their mechanosensing activities particularly interesting. The structurally "simple," anucleate nature of platelets coupled with their high actin concentration (20% of total protein) and integrin density [1] seem to make them ideal for mechanical force generation and transmission. Further studies will enhance our understanding of the role of platelet mechanobiology in hemostasis and thrombosis, potentially leading to new categories of diagnostics that investigate the mechanical properties of clots to determine bleeding risk, as well as therapies that target the mechanotransduction signaling pathway to alter the stability of clots.


Subject(s)
Blood Platelets/physiology , Mechanotransduction, Cellular/physiology , Biophysical Phenomena , Blood Platelets/cytology , Hemostasis , Humans , Microscopy, Atomic Force
10.
Sci Transl Med ; 5(179): 179ra47, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23552373

ABSTRACT

Circulating tumor cells (CTCs) are shed into the bloodstream from primary and metastatic tumor deposits. Their isolation and analysis hold great promise for the early detection of invasive cancer and the management of advanced disease, but technological hurdles have limited their broad clinical utility. We describe an inertial focusing-enhanced microfluidic CTC capture platform, termed "CTC-iChip," that is capable of sorting rare CTCs from whole blood at 10(7) cells/s. Most importantly, the iChip is capable of isolating CTCs using strategies that are either dependent or independent of tumor membrane epitopes, and thus applicable to virtually all cancers. We specifically demonstrate the use of the iChip in an expanded set of both epithelial and nonepithelial cancers including lung, prostate, pancreas, breast, and melanoma. The sorting of CTCs as unfixed cells in solution allows for the application of high-quality clinically standardized morphological and immunohistochemical analyses, as well as RNA-based single-cell molecular characterization. The combination of an unbiased, broadly applicable, high-throughput, and automatable rare cell sorting technology with generally accepted molecular assays and cytology standards will enable the integration of CTC-based diagnostics into the clinical management of cancer.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Separation/methods , Microfluidics/methods , Neoplastic Cells, Circulating/pathology , Cell Line, Tumor , Cell Shape , Cell Size , Female , Humans , Magnetic Phenomena , Male , RNA, Neoplasm/metabolism
11.
Science ; 339(6119): 580-4, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23372014

ABSTRACT

Epithelial-mesenchymal transition (EMT) of adherent epithelial cells to a migratory mesenchymal state has been implicated in tumor metastasis in preclinical models. To investigate its role in human cancer, we characterized EMT in circulating tumor cells (CTCs) from breast cancer patients. Rare primary tumor cells simultaneously expressed mesenchymal and epithelial markers, but mesenchymal cells were highly enriched in CTCs. Serial CTC monitoring in 11 patients suggested an association of mesenchymal CTCs with disease progression. In an index patient, reversible shifts between these cell fates accompanied each cycle of response to therapy and disease progression. Mesenchymal CTCs occurred as both single cells and multicellular clusters, expressing known EMT regulators, including transforming growth factor (TGF)-ß pathway components and the FOXC1 transcription factor. These data support a role for EMT in the blood-borne dissemination of human breast cancer.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Neoplastic Cells, Circulating/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/blood , Breast Neoplasms/genetics , Cell Count , Cell Movement , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mesoderm/pathology , Mice , Neoplasm Transplantation , Neoplastic Cells, Circulating/metabolism , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , Transcription, Genetic , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
Nature ; 487(7408): 510-3, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22763454

ABSTRACT

Circulating tumour cells (CTCs) shed into blood from primary cancers include putative precursors that initiate distal metastases. Although these cells are extraordinarily rare, they may identify cellular pathways contributing to the blood-borne dissemination of cancer. Here, we adapted a microfluidic device for efficient capture of CTCs from an endogenous mouse pancreatic cancer model and subjected CTCs to single-molecule RNA sequencing, identifying Wnt2 as a candidate gene enriched in CTCs. Expression of WNT2 in pancreatic cancer cells suppresses anoikis, enhances anchorage-independent sphere formation, and increases metastatic propensity in vivo. This effect is correlated with fibronectin upregulation and suppressed by inhibition of MAP3K7 (also known as TAK1) kinase. In humans, formation of non-adherent tumour spheres by pancreatic cancer cells is associated with upregulation of multiple WNT genes, and pancreatic CTCs revealed enrichment for WNT signalling in 5 out of 11 cases. Thus, molecular analysis of CTCs may identify candidate therapeutic targets to prevent the distal spread of cancer.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Neoplasm Metastasis/genetics , Neoplastic Cells, Circulating/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , Animals , Cell Survival , Contact Inhibition , Disease Models, Animal , Genes, Neoplasm/genetics , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mice , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Sequence Analysis, RNA , Wnt Proteins/genetics , Wnt2 Protein/genetics , Wnt2 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...