Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Platelets ; 32(3): 424-428, 2021 Apr 03.
Article En | MEDLINE | ID: mdl-32233694

The core structure of the extracellular basement membrane is made up of self-assembling networks of collagen and laminin which associate with each other through the bridging adapter proteins including the sulfated monomeric glycoprotein nidogen. While collagen and laminin are known to support platelet adhesion and activation via ß1 integrins and glycoprotein (GP) VI, respectively, whether nidogen contributes to platelet activation and hemostasis is unknown. In this study, we demonstrate that recombinant human nidogen-1 supports platelet adhesion and stimulates platelet activation in a phospholipase-C γ-2 (PLCγ2), Src and Syk kinase-dependent manner downstream. Platetet adhesion to nidogen-1 was inhibited by blocking the platelet receptors GPVI and ß1 integrins. Platelet adhesion to nidogen-1 activated the IκB kinase (IKK) complex, while pharmacological inhibition of IKK blocked platelet spreading on nidogen. Taken together our results suggest that nidogen may play a redundant role in hemostasis by activating platelets downstream of GPVI.


Membrane Glycoproteins/metabolism , Platelet Activation/physiology , Platelet Adhesiveness/physiology , Humans
2.
Blood Adv ; 3(20): 3080-3091, 2019 10 22.
Article En | MEDLINE | ID: mdl-31648335

In humans, platelet count within the normal range is required for physiological hemostasis, but, adversely, platelets also support pathological thrombosis. Moreover, by releasing growth factors, they may enhance neoplastic proliferation. We hypothesize that platelet count correlates with platelet-dependent pathologies, even within the range of hemostatic competence. Because platelet production is promoted by thrombopoietin signaling through the myeloproliferative leukemia virus oncogene (cMPL), a receptor expressed on megakaryocytes, we evaluated the feasibility of selective targeting of hepatic thrombopoietin production to test this hypothesis. We synthesized murine- and primate-specific antisense oligonucleotides (THPO-ASO) that silence hepatic thrombopoietin gene (THPO) expression without blocking extrahepatic THPO. Repeated doses of THPO-ASO were administered to mice and a baboon, causing a sustained 50% decline in plasma thrombopoietin levels and platelet count within 4 weeks in both species. To investigate whether reducing platelet count within the translationally relevant hemostatic range could alter a neoplastic process, we administered THPO-ASO to 6-week-old transgenic MMTV-PyMT mice that develop early ductal atypia that progresses into cMPL-negative fatal metastatic breast cancer within 2 to 3 months. THPO-ASO treatment increased the average time to euthanasia (primary humane endpoint) at 2 cm3 combined palpable tumor volume. Our results show that THPO-ASO reduced blood platelet count, plasma platelet factor 4, vascular endothelial growth factor, thrombopoietin levels, bone marrow megakaryocyte density, tumor growth rate, proliferation index, vascularization, platelet and macrophage content, and pulmonary metastases vs untreated controls. These findings confirm that sustained and moderate pharmacological platelet count reduction is feasible with THPO-ASO administration and can delay progression of certain platelet-dependent pathological processes within a safe hemostatic platelet count range.


Breast Neoplasms/blood , Breast Neoplasms/etiology , Gene Silencing , Liver/metabolism , Platelet Count , Thrombopoietin/genetics , Animals , Breast Neoplasms/pathology , Cell Movement , Cell Transformation, Neoplastic , Disease Models, Animal , Disease Progression , Haplorhini , Mice , Mice, Transgenic , Neoplasm Staging , Tumor Microenvironment/genetics
3.
Am J Physiol Cell Physiol ; 316(2): C264-C273, 2019 02 01.
Article En | MEDLINE | ID: mdl-30462538

Cancer-associated thrombosis is a common first presenting sign of malignancy and is currently the second leading cause of death in cancer patients after their malignancy. However, the molecular mechanisms underlying cancer-associated thrombosis remain undefined. In this study, we aimed to develop a better understanding of how cancer cells affect the coagulation cascade and platelet activation to induce a prothrombotic phenotype. Our results show that colon cancer cells trigger platelet activation in a manner dependent on cancer cell tissue factor (TF) expression, thrombin generation, activation of the protease-activated receptor 4 (PAR4) on platelets and consequent release of ADP and thromboxane A2. Platelet-colon cancer cell interactions potentiated the release of platelet-derived extracellular vesicles (EVs) rather than cancer cell-derived EVs. Our data show that single colon cancer cells were capable of recruiting and activating platelets and generating fibrin in plasma under shear flow. Finally, in a retrospective analysis of colon cancer patients, we found that the number of venous thromboembolism events was 4.5 times higher in colon cancer patients than in a control population. In conclusion, our data suggest that platelet-cancer cell interactions and perhaps platelet procoagulant EVs may contribute to the prothrombotic phenotype of colon cancer patients. Our work may provide rationale for targeting platelet-cancer cell interactions with PAR4 antagonists together with aspirin and/or ADP receptor antagonists as a potential intervention to limit cancer-associated thrombosis, balancing safety with efficacy.


Blood Coagulation/physiology , Blood Platelets/physiology , Colonic Neoplasms/blood , Thrombosis/blood , Blood Platelets/pathology , Cell Line, Tumor , Colonic Neoplasms/pathology , Cross-Sectional Studies , Humans , Retrospective Studies , Thrombosis/pathology
4.
Platelets ; 30(1): 126-135, 2019.
Article En | MEDLINE | ID: mdl-30560697

Human platelets express two protease-activated receptors (PARs), PAR1 (F2R) and PAR4 (F2RL3), which are activated by a number of serine proteases that are generated during pathological events and cause platelet activation. Recent interest has focused on PAR4 as a therapeutic target, given PAR4 seems to promote experimental thrombosis and procoagulant microparticle formation, without a broadly apparent role in hemostasis. However, it is not yet known whether PAR4 activity plays a role in platelet-leukocyte interactions, which are thought to contribute to both thrombosis and acute or chronic thrombo-inflammatory processes. We sought to determine whether PAR4 activity contributes to granule secretion from activated platelets and platelet-leukocyte interactions. We performed in vitro and ex vivo studies of platelet granule release and platelet-leukocyte interactions in the presence of PAR4 agonists including PAR4 activating peptide, thrombin, cathepsin G, and plasmin in combination with small-molecule PAR4 antagonists. Activation of human platelets with thrombin, cathepsin G, or plasmin potentiated platelet dense granule secretion that was specifically impaired by PAR4 inhibitors. Platelet-leukocyte interactions and platelet P-selectin exposure the following stimulation with PAR4 agonists were also impaired by activated PAR4 inhibition in either a purified system or in whole blood. These results indicate PAR4-specific promotion of platelet granule release and platelet-leukocyte aggregate formation and suggest that pharmacological control of PAR4 activity could potentially attenuate platelet granule release or platelet-leukocyte interaction-mediated pathological processes.


Blood Platelets/metabolism , Cell Communication , Cytoplasmic Granules/metabolism , Leukocytes/metabolism , Receptors, Thrombin/metabolism , Animals , Biomarkers , Flow Cytometry , Humans , Male , Papio , Platelet Activation , Platelet Aggregation
5.
Cardiovasc Eng Technol ; 9(3): 515-527, 2018 09.
Article En | MEDLINE | ID: mdl-29785664

Platelet apheresis units are transfused into patients to mitigate or prevent bleeding. In a hospital, platelet apheresis units are transported from the transfusion service to the healthcare teams via two methods: a pneumatic tubing system (PTS) or ambulatory transport. Whether PTS transport affects the activity and utility of platelet apheresis units is unclear. We quantified the gravitational forces and transport time associated with PTS and ambulatory transport within our hospital. Washed platelets and supernatants were prepared from platelet apheresis units prior to transport as well as following ambulatory or PTS transport. For each group, we compared resting and agonist-induced platelet activity and platelet aggregate formation on collagen or von Willebrand factor (VWF) under shear, platelet VWF-receptor expression and VWF multimer levels. Subjection of platelet apheresis units to rapid acceleration/deceleration forces during PTS transport did not pre-activate platelets or their ability to activate in response to platelet agonists as compared to ambulatory transport. Platelets within platelet apheresis units transported via PTS retained their ability to adhere to surfaces of VWF and collagen under shear, although platelet aggregation on collagen and VWF was diminished as compared to ambulatory transport. VWF multimer levels and platelet GPIb receptor expression was unaffected by PTS transport as compared to ambulatory transport. Subjection of platelet apheresis units to PTS transport did not significantly affect the baseline or agonist-induced levels of platelet activation as compared to ambulatory transport. Our case study suggests that PTS transport may not significantly affect the hemostatic potential of platelets within platelet apheresis units.


Blood Component Removal , Blood Platelets/metabolism , Hospital Units , Platelet Activation , Platelet Transfusion , Transportation/methods , Acceleration , Deceleration , Equipment Design , Gravitation , Humans , Platelet Aggregation , Platelet Function Tests , Platelet Glycoprotein GPIb-IX Complex/metabolism , von Willebrand Factor/metabolism
6.
Front Oncol ; 8: 78, 2018.
Article En | MEDLINE | ID: mdl-29619344

In this review, we discuss the interaction between cancer and markers of inflammation (such as levels of inflammatory cells and proteins) in the circulation, and the potential benefits of routinely monitoring these markers in peripheral blood measurement assays. Next, we discuss the prognostic value and limitations of using inflammatory markers such as neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios and C-reactive protein measurements. Furthermore, the review discusses the benefits of combining multiple types of measurements and longitudinal tracking to improve staging and prognosis prediction of patients with cancer, and the ability of novel in silico frameworks to leverage this high-dimensional data.

7.
Sci Rep ; 8(1): 6564, 2018 04 26.
Article En | MEDLINE | ID: mdl-29700384

Platelets, components of hemostasis, when present in excess (>400 K/µL, thrombocytosis) have also been associated with worse outcomes in lung, ovarian, breast, renal, and colorectal cancer patients. Associations between thrombocytosis and cancer outcomes have been made mostly from single-time-point studies, often at the time of diagnosis. Using laboratory data from the Department of Veterans Affairs (VA), we examined the potential benefits of using longitudinal platelet counts in improving patient prognosis predictions. Ten features (summary statistics and engineered features) were derived to describe the platelet counts of 10,000+ VA lung, prostate, and colon cancer patients and incorporated into an age-adjusted LASSO regression analysis to determine feature importance, and predict overall or relapse-free survival, which was compared to the previously used approach of monitoring for thrombocytosis near diagnosis (Postdiag AG400 model). Temporal features describing acute platelet count increases/decreases were found to be important in cancer survival and relapse-survival that helped stratify good and bad outcomes of cancer patient groups. Predictions of overall and relapse-free survival were improved by up to 30% compared to the Postdiag AG400 model. Our study indicates the association of temporally derived platelet count features with a patients' prognosis predictions.


Colonic Neoplasms/blood , Colonic Neoplasms/mortality , Lung Neoplasms/blood , Lung Neoplasms/mortality , Platelet Count , Prostatic Neoplasms/blood , Prostatic Neoplasms/mortality , Adult , Aged , Biomarkers , Colonic Neoplasms/diagnosis , Colonic Neoplasms/therapy , Comorbidity , Humans , Kaplan-Meier Estimate , Lung Neoplasms/diagnosis , Lung Neoplasms/therapy , Male , Middle Aged , Neoplasm Staging , Odds Ratio , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/therapy , Retrospective Studies
8.
Am J Physiol Cell Physiol ; 314(5): C603-C615, 2018 05 01.
Article En | MEDLINE | ID: mdl-29412690

Upon encountering physiological cues associated with damaged or inflamed endothelium, blood platelets set forth intracellular responses to ultimately support hemostatic plug formation and vascular repair. To gain insights into the molecular events underlying platelet function, we used a combination of interactome, pathway analysis, and other systems biology tools to analyze associations among proteins functionally modified by reversible phosphorylation upon platelet activation. While an interaction analysis mapped out a relative organization of intracellular mediators in platelet signaling, pathway analysis revealed directional signaling relations around protein kinase C (PKC) isoforms and mitogen-activated protein kinases (MAPKs) associated with platelet cytoskeletal dynamics, inflammatory responses, and hemostatic function. Pathway and causality analysis further suggested that platelets activate a specific p38-MK2 axis to phosphorylate RTN4 (reticulon-4, also known as Nogo), a Bcl-xl sequestration protein and critical regulator of endoplasmic reticulum (ER) physiology. In vitro, we find that platelets drive a p38-MK2-RTN4-Bcl-xl pathway associated with the regulation of the ER and platelet phosphatidylserine exposure. Together, our results support the use of pathway tools in the analysis of omics data sets as a means to help generate novel, mechanistic, and testable hypotheses for platelet studies while uncovering RTN4 as a putative regulator of platelet cell physiological responses.


Blood Coagulation , Blood Platelets/enzymology , Endoplasmic Reticulum/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Nogo Proteins/metabolism , Platelet Activation , Protein Interaction Maps , Protein Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Computational Biology , Databases, Protein , Enzyme Activation , Humans , Phenotype , Phosphatidylserines/metabolism , Phosphorylation , Signal Transduction , bcl-X Protein/metabolism
9.
Platelets ; 29(4): 383-394, 2018 Jun.
Article En | MEDLINE | ID: mdl-28523947

The release of ADP from platelet dense granules and its binding to platelet P2Y12 receptors is key to amplifying the initial hemostatic response and propagating thrombus formation. P2Y12 has thus emerged as a therapeutic target to safely and effectively prevent secondary thrombotic events in patients with acute coronary syndrome or a history of myocardial infarction. Pharmacological inhibition of P2Y12 receptors represents a useful approach to better understand the signaling mediated by these receptors and to elucidate the role of these receptors in a multitude of platelet hemostatic and thrombotic responses. The present work examined and compared the effects of four different P2Y12 inhibitors (MRS2395, ticagrelor, PSB 0739, and AR-C 66096) on platelet function in a series of in vitro studies of platelet dense granule secretion and trafficking, calcium generation, and protein phosphorylation. Our results show that in platelets activated with the PAR-1 agonist TRAP-6 (thrombin receptor-activating peptide), inhibition of P2Y12 with the antagonist MRS2395, but not ticagrelor, PSB 0739 or AR-C 66096, potentiated human platelet dense granule trafficking to the plasma membrane and release into the extracellular space, cytosolic Ca2+ influx, and phosphorylation of GSK3ß-Ser9 through a PKC-dependent pathway. These results suggest that inhibition of P2Y12 with MRS2395 may act in concert with PAR-1 signaling and result in the aberrant release of ADP by platelet dense granules, thus reducing or counteracting the anticipated anti-platelet efficacy of this inhibitor.


Adenine/analogs & derivatives , Blood Platelets/metabolism , Peptide Fragments/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/blood , Valerates/pharmacology , Adenine/pharmacology , Blood Platelets/drug effects , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Humans , Platelet Activation/drug effects , Platelet Activation/physiology , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Receptor, PAR-1/agonists
10.
Platelets ; 29(8): 773-778, 2018 Dec.
Article En | MEDLINE | ID: mdl-29265902

Cancer metastasis is a dynamic process during which cancer cells separate from a primary tumor, migrate through the vessel wall into the bloodstream, and extravasate at distant sites to form secondary colonies. During this process, circulating tumor cells are subjected to shear stress forces from blood flow, and in contact with plasma proteins and blood cells of the immune and hemostatic system, including platelets. Many studies have shown an association between high platelet count and cancer metastasis, suggesting that platelets may play an occult role in tumorigenesis. This mini-review summarizes recent and emerging discoveries of mechanisms by which cancer cells activate platelets and the role of activated platelets in promoting tumor growth and metastasis. Moreover, the review discusses how aspirin has the potential for being clinically used as an adjuvant in cancer therapy.


Antineoplastic Agents/therapeutic use , Aspirin/therapeutic use , Neoplasms , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Animals , Humans , Neoplasm Metastasis , Neoplasms/blood , Neoplasms/drug therapy , Neoplasms/pathology , Platelet Count
11.
Converg Sci Phys Oncol ; 3(2)2017 Jun.
Article En | MEDLINE | ID: mdl-29081989

Platelets are anucleate cells in the blood at concentrations of 150,000 to 400,000 cells/µL and play a key role in hemostasis. Several studies have suggested that platelets contribute to cancer progression and cancer-associated thrombosis. In this review, we provide an overview of the biochemical and biophysical mechanisms by which platelets interact with cancer cells and review the evidence supporting a role for platelet-enhanced metastasis of cancer, and venous thromboembolism (VTE) in patients with cancer. We discuss the potential for and limitations of platelet counts to discriminate cancer disease burden and prognosis. Lastly, we consider more advanced diagnostic approaches to improve studies on the interaction between the hemostatic system and cancer cells.

13.
J Biol Chem ; 292(21): 8616-8629, 2017 05 26.
Article En | MEDLINE | ID: mdl-28408624

Activated protein C (APC) is a multifunctional serine protease with anticoagulant, cytoprotective, and anti-inflammatory activities. In addition to the cytoprotective effects of APC on endothelial cells, podocytes, and neurons, APC cleaves and detoxifies extracellular histones, a major component of neutrophil extracellular traps (NETs). NETs promote pathogen clearance but also can lead to thrombosis; the pathways that negatively regulate NETosis are largely unknown. Thus, we studied whether APC is capable of directly inhibiting NETosis via receptor-mediated cell signaling mechanisms. Here, by quantifying extracellular DNA or myeloperoxidase, we demonstrate that APC binds human leukocytes and prevents activated platelet supernatant or phorbol 12-myristate 13-acetate (PMA) from inducing NETosis. Of note, APC proteolytic activity was required for inhibiting NETosis. Moreover, antibodies against the neutrophil receptors endothelial protein C receptor (EPCR), protease-activated receptor 3 (PAR3), and macrophage-1 antigen (Mac-1) blocked APC inhibition of NETosis. Select mutations in the Gla and protease domains of recombinant APC caused a loss of NETosis. Interestingly, pretreatment of neutrophils with APC prior to induction of NETosis inhibited platelet adhesion to NETs. Lastly, in a nonhuman primate model of Escherichia coli-induced sepsis, pretreatment of animals with APC abrogated release of myeloperoxidase from neutrophils, a marker of neutrophil activation. These findings suggest that the anti-inflammatory function of APC at therapeutic concentrations may include the inhibition of NETosis in an EPCR-, PAR3-, and Mac-1-dependent manner, providing additional mechanistic insight into the diverse functions of neutrophils and APC in disease states including sepsis.


Extracellular Traps/immunology , Neutrophil Activation/immunology , Neutrophils/immunology , Protein C/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Disease Models, Animal , Endothelial Protein C Receptor , Escherichia coli , Escherichia coli Infections/blood , Escherichia coli Infections/immunology , Extracellular Traps/metabolism , Female , Humans , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/metabolism , Male , Neutrophil Activation/drug effects , Neutrophils/metabolism , Papio anubis , Protein C/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Sepsis/blood , Sepsis/immunology , Tetradecanoylphorbol Acetate/pharmacology
14.
Am J Physiol Cell Physiol ; 312(4): C527-C536, 2017 Apr 01.
Article En | MEDLINE | ID: mdl-28148498

On activation at sites of vascular injury, platelets undergo morphological alterations essential to hemostasis via cytoskeletal reorganizations driven by the Rho GTPases Rac1, Cdc42, and RhoA. Here we investigate roles for Rho-specific guanine nucleotide dissociation inhibitor proteins (RhoGDIs) in platelet function. We find that platelets express two RhoGDI family members, RhoGDI and Ly-GDI. Whereas RhoGDI localizes throughout platelets in a granule-like manner, Ly-GDI shows an asymmetric, polarized localization that largely overlaps with Rac1 and Cdc42 as well as microtubules and protein kinase C (PKC) in platelets adherent to fibrinogen. Antibody interference and platelet spreading experiments suggest a specific role for Ly-GDI in platelet function. Intracellular signaling studies based on interactome and pathways analyses also support a regulatory role for Ly-GDI, which is phosphorylated at PKC substrate motifs in a PKC-dependent manner in response to the platelet collagen receptor glycoprotein (GP) VI-specific agonist collagen-related peptide. Additionally, PKC inhibition diffuses the polarized organization of Ly-GDI in spread platelets relative to its colocalization with Rac1 and Cdc42. Together, our results suggest a role for Ly-GDI in the localized regulation of Rho GTPases in platelets and hypothesize a link between the PKC and Rho GTPase signaling systems in platelet function.


Blood Coagulation/physiology , Blood Platelets/physiology , Platelet Activation/physiology , Platelet Adhesiveness/physiology , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , Cells, Cultured , Hemostasis/physiology , Humans , Signal Transduction/physiology , Subcellular Fractions/metabolism
15.
Am J Physiol Cell Physiol ; 312(2): C176-C189, 2017 Feb 01.
Article En | MEDLINE | ID: mdl-27903583

Aspirin, an anti-inflammatory and antithrombotic drug, has become the focus of intense research as a potential anticancer agent owing to its ability to reduce tumor proliferation in vitro and to prevent tumorigenesis in patients. Studies have found an anticancer effect of aspirin when used in low, antiplatelet doses. However, the mechanisms through which low-dose aspirin works are poorly understood. In this study, we aimed to determine the effect of aspirin on the cross talk between platelets and cancer cells. For our study, we used two colon cancer cell lines isolated from the same donor but characterized by different metastatic potential, SW480 (nonmetastatic) and SW620 (metastatic) cancer cells, and a pancreatic cancer cell line, PANC-1 (nonmetastatic). We found that SW480 and PANC-1 cancer cell proliferation was potentiated by human platelets in a manner dependent on the upregulation and activation of the oncoprotein c-MYC. The ability of platelets to upregulate c-MYC and cancer cell proliferation was reversed by an antiplatelet concentration of aspirin. In conclusion, we show for the first time that inhibition of platelets by aspirin can affect their ability to induce cancer cell proliferation through the modulation of the c-MYC oncoprotein.


Aspirin/administration & dosage , Blood Platelets/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Blood Platelets/metabolism , Blood Platelets/pathology , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Communication/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Humans , Oncogene Proteins/metabolism , Pancreatic Neoplasms/pathology , Platelet Aggregation Inhibitors/administration & dosage , Treatment Outcome
16.
Blood Rev ; 30(1): 11-9, 2016 Jan.
Article En | MEDLINE | ID: mdl-26219246

The hemostatic system is often subverted in patients with cancer, resulting in life-threatening venous thrombotic events. Despite the multifactorial and complex etiology of cancer-associated thrombosis, changes in the expression and activity of cancer-derived tissue factor (TF) - the principle initiator of the coagulation cascade - are considered key to malignant hypercoagulopathy and to the pathophysiology of thrombosis. However, many of the molecular and cellular mechanisms coupling the hemostatic degeneration to malignancy remain largely uncharacterized. In this review we discuss some of the tumor-intrinsic and tumor-extrinsic mechanisms that may contribute to the prothrombotic state of cancer, and we bring into focus the potential for circulating tumor cells (CTCs) in advancing our understanding of the field. We also summarize the current status of anti-coagulant therapy for the treatment of thrombosis in patients with cancer.


Gene Expression Regulation, Neoplastic , Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Thromboplastin/genetics , Thrombosis/pathology , Anticoagulants/therapeutic use , Antineoplastic Agents/adverse effects , Blood Coagulation , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/pathology , Factor VIIa/genetics , Factor VIIa/metabolism , Fibrin/metabolism , Fibrinogen/metabolism , Humans , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/genetics , Neoplastic Cells, Circulating/pathology , Platelet Activation/drug effects , Prothrombin/genetics , Prothrombin/metabolism , Thromboplastin/metabolism , Thrombosis/complications , Thrombosis/drug therapy , Thrombosis/genetics
17.
Am J Physiol Cell Physiol ; 308(10): C792-802, 2015 May 15.
Article En | MEDLINE | ID: mdl-25788574

Circulating tumor cells (CTC) have been implicated in the hematogenous spread of cancer. To investigate the fluid phase of cancer from a physical sciences perspective, the multi-institutional Physical Sciences-Oncology Center (PS-OC) Network performed multidisciplinary biophysical studies of single CTC and CTC aggregates from a patient with breast cancer. CTCs, ranging from single cells to aggregates comprised of 2-5 cells, were isolated using the high-definition CTC assay and biophysically profiled using quantitative phase microscopy. Single CTCs and aggregates were then modeled in an in vitro system comprised of multiple breast cancer cell lines and microfluidic devices used to model E-selectin mediated rolling in the vasculature. Using a numerical model coupling elastic collisions between red blood cells and CTCs, the dependence of CTC vascular margination on single CTCs and CTC aggregate morphology and stiffness was interrogated. These results provide a multifaceted characterization of single CTC and CTC aggregate dynamics in the vasculature and illustrate a framework to integrate clinical, biophysical, and mathematical approaches to enhance our understanding of the fluid phase of cancer.


Breast Neoplasms/diagnosis , Cell Movement , E-Selectin/metabolism , Neoplastic Cells, Circulating/pathology , Transcytosis/physiology , Breast Neoplasms/metabolism , Cell Count/methods , Female , Humans , Microfluidic Analytical Techniques/methods
18.
Molecules ; 19(11): 17559-77, 2014 Oct 30.
Article En | MEDLINE | ID: mdl-25361422

Polymer-peptide conjugates are a promising class of compounds, where polymers can be used to overcome some of the limitations associated with peptides intended for therapeutic and/or diagnostic applications. Linear polymers such as poly(ethylene glycol) can be conjugated through terminal moieties and have therefore limited loading capacities. In this research, functionalised linear poly(ethylene glycol)s are utilised for peptide conjugation, to increase their potential loading capacities. These poly(ethylene glycol) derivatives are conjugated to peptide sequences containing representative side-chain functionalised amino acids, using different conjugation chemistries, including copper-catalysed azide-alkyne cycloaddition, amide coupling and thiol-ene reactions. Conjugation of a sequence containing the RGD motif to poly(allyl glycidyl ether) by the thiol-ene reaction, provided a conjugate which could be used in platelet adhesion studies.


Peptides/chemistry , Peptides/pharmacology , Polyethylene Glycols/chemistry , Alkynes/chemistry , Amino Acids/chemistry , Azides/chemistry , Blood Platelets/drug effects , Epoxy Compounds/chemistry , Humans , Polymers/chemistry , Sulfhydryl Compounds/chemistry
19.
Blood ; 123(2): 249-60, 2014 Jan 09.
Article En | MEDLINE | ID: mdl-24258815

Platelets play a role in cancer by acting as a dynamic reservoir of effectors that facilitate tumor vascularization, growth, and metastasis. However, little information is available about the mechanism of tumor cell-induced platelet secretion (TCIPS) or the molecular machinery by which effector molecules are released from platelets. Here we demonstrate that tumor cells directly induce platelet secretion. Preincubation of platelets with human colon cancer (Caco-2), prostate cancer (PC3M-luc), or breast cancer cells (MDA-MB-231;MCF-7) resulted in a marked dose-dependent secretion of dense granules. Importantly, TCIPS preceded aggregation which always displayed a characteristic lag time. We investigated the role of platelet receptors and downstream molecules in TCIPS. The most potent modulators of TCIPS were the pharmacologic antagonists of Syk kinase, phospholipase C and protein kinase C, all downstream mediators of the immunoreceptor tyrosine-based activation motif (ITAM) cascade in platelets. Supporting this, we demonstrated a central role for the immune Fcγ receptor IIa (FcγRIIa) in mediating platelet-tumor cell cross-talk. In conclusion, we demonstrate that cancer cells can promote platelet dense-granule secretion, which is required to augment platelet aggregation. In addition, we show a novel essential role for FcγRIIa in prostate cancer cell-induced platelet activation opening the opportunity to develop novel antimetastatic therapies.


Blood Platelets/metabolism , Neoplasms/metabolism , Platelet Activation , Receptors, IgG/metabolism , Animals , Cell Line, Tumor , Focal Adhesion Kinase 1/metabolism , Humans , Membrane Glycoproteins/metabolism , Mice , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex , Receptor, PAR-1/metabolism , Receptors, Purinergic P2Y/metabolism , Receptors, Thromboxane/metabolism , Signal Transduction , src-Family Kinases/metabolism
...