Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Blood ; 127(7): 921-6, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26647394

ABSTRACT

In times of physiological stress, platelet count can transiently rise. What initiates this reactive thrombocytosis is poorly understood. Intriguingly, we found that treating megakaryocytes (MKs) with the releasate from activated platelets increased proplatelet production by 47%. Platelets store inflammatory cytokines, including the chemokine ligand 5 (CCL5, RANTES); after TRAP activation, platelets release over 25 ng/mL CCL5. We hypothesized that CCL5 could regulate platelet production by binding to its receptor, CCR5, on MKs. Maraviroc (CCR5 antagonist) or CCL5 immunodepletion diminished 95% and 70% of the effect of platelet releasate, respectively, suggesting CCL5 derived from platelets is sufficient to drive increased platelet production through MK CCR5. MKs cultured with recombinant CCL5 increased proplatelet production by 50% and had significantly higher ploidy. Pretreating the MK cultures with maraviroc prior to exposure to CCL5 reversed the augmented proplatelet formation and ploidy, suggesting that CCL5 increases MK ploidy and proplatelet formation in a CCR5-dependent manner. Interrogation of the Akt signaling pathway suggested that CCL5/CCR5 may influence proplatelet production by suppressing apoptosis. In an in vivo murine acute colitis model, platelet count significantly correlated with inflammation whereas maraviroc treatment abolished this correlation. We propose that CCL5 signaling through CCR5 may increase platelet counts during physiological stress.


Subject(s)
Blood Platelets/metabolism , Chemokine CCL5/metabolism , Megakaryocytes/pathology , Signal Transduction/physiology , Animals , Blood Platelets/cytology , Chemokine CCL5/genetics , Cyclohexanes/pharmacology , Humans , Maraviroc , Megakaryocytes/cytology , Mice , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Signal Transduction/drug effects , Triazoles/pharmacology
2.
Arterioscler Thromb Vasc Biol ; 37(4): 664-674, 2017 04.
Article in English | MEDLINE | ID: mdl-28153880

ABSTRACT

OBJECTIVE: Platelets, which are mainly known for their role in hemostasis, are now known to play a crucial role in metastasis. Tamoxifen is a selective estrogen receptor modulator that is widely used for the treatment of breast cancer. Tamoxifen and its metabolites have been shown to directly impact platelet function, suggesting that this drug has additional mechanisms of action. The purpose of this study was to determine whether tamoxifen exerts antitumor effects through direct platelet inhibition. APPROACH AND RESULTS: This study found that pretreatment with tamoxifen leads to a significant inhibition of platelet activation. Platelets exposed to tamoxifen released significantly lower amounts of proangiogenic regulator vascular endothelial growth factor. In vitro angiogenesis assays confirmed that tamoxifen pretreatment led to diminished capillary tube formation and decreased endothelial migration. Tamoxifen and its metabolite, 4-hydroxytamoxifen, also significantly inhibited the ability of platelets to promote metastasis in vitro. Using a membrane-based array, we identified several proteins associated with angiogenesis metastasis that were lower in activated releasate from tamoxifen-treated platelets, including angiogenin, chemokine (C-X-C motif) ligand 1, chemokine (C-C motif) ligand 5, epidermal growth factor, chemokine (C-X-C motif) ligand 5, platelet-derived growth factor dimeric isoform BB, whereas antiangiogenic angiopoietin-1 was elevated. Platelets isolated from patients on tamoxifen maintenance therapy were also found to have decreased activation responses, diminished vascular endothelial growth factor release, and lower angiogenic and metastatic potential. CONCLUSIONS: We demonstrate that tamoxifen and its metabolite 4-hydroxytamoxifen directly alter platelet function leading to decreased angiogenic and metastatic potential. Furthermore, this study supports the idea of utilizing targeted platelet therapies to inhibit the platelet's role in angiogenesis and malignancy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Blood Platelets/drug effects , Breast Neoplasms/drug therapy , Cell Movement/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Tamoxifen/analogs & derivatives , Blood Platelets/metabolism , Breast Neoplasms/blood , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Coculture Techniques , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MCF-7 Cells , Neoplasm Metastasis , Signal Transduction/drug effects , Tamoxifen/pharmacology , Vascular Endothelial Growth Factor A/metabolism
3.
Blood Adv ; 3(2): 198-211, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670536

ABSTRACT

It is now recognized that compounds released from tumor cells can activate platelets, causing the release of platelet-derived factors into the tumor microenvironment. Several of these factors have been shown to directly promote neovascularization and metastasis, yet how the feedback between platelet releasate and the tumor cell affects metastatic phenotype remains largely unstudied. Here, we identify that breast tumor cells secrete high levels of interleukin 8 (IL-8, CXCL8) in response to platelet releasate, which promotes their invasive capacity. Furthermore, we found that platelets activate the Akt pathway in breast tumor cells, and inhibition of this pathway eliminated IL-8 production. We therefore hypothesized inhibiting platelets with aspirin could reverse the prometastatic effects of platelets on tumor cell signaling. Platelets treated with aspirin did not activate the Akt pathway, resulting in reduced IL-8 secretion and impaired tumor cell invasion. Of note, patients with breast cancer receiving aspirin had lower circulating IL-8, and their platelets did not increase tumor cell invasion compared with patients not receiving aspirin. Our data suggest platelets support breast tumor metastasis by inducing tumor cells to secrete IL-8. Our data further support that aspirin acts as an anticancer agent by disrupting the communication between platelets and breast tumor cells.


Subject(s)
Aspirin/pharmacology , Blood Platelets/drug effects , Breast Neoplasms/blood , Breast Neoplasms/pathology , Platelet Aggregation Inhibitors/pharmacology , Blood Platelets/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cytokines/metabolism , Female , Humans , Neoplasm Metastasis , Neoplasm Staging , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL