GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    American Society of Hematology ; 2021
    In:  Blood Vol. 138, No. Supplement 1 ( 2021-11-05), p. 1006-1006
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 1006-1006
    Abstract: Introduction: Programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) are important immune checkpoint proteins in cancer immunotherapy and targeted therapies against PD-L1 have significantly prolonged many patients' lives. Recently, high baseline platelet to lymphocyte ratio was reported to be associated with decreased patient response rate to immune checkpoint inhibition (ICI) therapies, including anti-PD-L1 therapy, suggesting the potential role of platelets in tumor immunity. Platelets express PD-L1 on their surface, and platelets binding to PD-L1 negative tumor cells can "decorate" tumor cells with PD-L1 and protect against T cell-mediated cytotoxicity. However, whether platelet can affect PD-L1 expression on tumor cells is still unknown. Methods: In this study, we designed platelet-tumor cell co-culture systems to investigate whether direct or indirect exposure to platelets affects tumor cell PD-L1 surface expression. Considering platelets can be artificially activated by commonly used cell culture medium, the co-culture was performed in platelet resuspension buffer (HEPES, NaCl, KCl, MgCl2, NaHCO3, Glucose, pH7.4) supplied with fetal bovine serum and L-glutamine. After 24 hours of co-culture, platelets were washed out and fresh culturing medium was added to tumor cells and cultured for another 24 hours. At the end of the experiments, tumor cells were harvested and the PD-L1 expression analyzed by flow cytometry and RT-qPCR. Results and discussion: Here we report that direct co-culture of platelets with either breast cancer cell line MDA-MB-468 or lung cancer cell line A549 increased tumor cell PD-L1 surface expression by up-regulating PD-L1 transcription. This platelet-induced tumor cell PD-L1 up-regulation can be partly reduced by pre-treating platelets with antiplatelet agents such as aspirin and ticagrelor, suggesting platelet activation contributes to platelet induced tumor cell PD-L1 up-regulation. The up-regulation of tumor cell PD-L1 by platelets was not due to abundant platelet cytokines such as C-C Motif Chemokine Ligand 5 (CCL5) and C-X-C motif chemokine 5 (CXCL5). However, both an epidermal growth factor (EGF) neutralizing antibody and cetuximab (EGFR neutralizing monoclonal antibody) decreased the platelet-induced increase in tumor cell PD-L1, suggesting that platelets initiate tumor cell PD-L1 transcription through the EGF signaling pathway. Our data indicate a novel function of platelets in tumor immunity and warrant further investigation to determine if targeting platelets offers a novel adjuvant approach to improve ICI therapy. Disclosures Italiano: Sierra Oncology: Consultancy; PlateletBio: Current holder of individual stocks in a privately-held company, Membership on an entity's Board of Directors or advisory committees; Carrick Therapeutics: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2018
    In:  Blood Vol. 132, No. Supplement 1 ( 2018-11-29), p. 522-522
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 522-522
    Abstract: The importance of platelets in clot formation is well understood, but the role of platelets in the inflammatory response is becoming increasingly apparent. Platelet count increases during acute inflammatory stress; platelets also facilitate inflammation by releasing chemokines and cytokines from their granules. Our lab has previously shown that platelet derived chemokine ligand 5 (CCL5, RANTES), an inflammatory chemokine that is stored in platelets and released upon activation, acts to increase megakaryocyte maturation and proplatelet production in vitro. Furthermore, our lab found that CCL5 treated megakaryocytes have enhanced phosphorylation of proteins in the AKT pathway, such as BAD, which when phosphorylated induces pro-survival signaling. This indicates that CCL5-induced increase of platelet production is mediated by suppression of apoptosis, but the exact mechanism of CCL5 on megakaryocytes is not understood. To more completely asses the global impact of CCL5 on megakaryocytes, we performed proteomic analysis of CCL5-treated megakaryocytes and found that, as expected, CCL5 increases the expression of several pro-survival proteins in the AKT pathway. We have now confirmed increased phosphorylation of BAD by western blot in megakaryocytes treated with CCL5. In addition, we found that CCL5 treatment protects megakaryocytes from etoposide-induced apoptosis. Together, our data support the model of CCL5-induced inhibition of mitochondrial apoptosis via BAD phosphorylation. Interestingly, proteomic analysis also revealed an increase in proteins involved in quantity and differentiation of hematopoietic progenitor cells, as well as differentiation of megakaryocytes. To test whether CCL5 can also affect the differentiation of hematopoietic progenitor cells, we cultured mouse bone marrow with 100 ng/mL CCL5 and TPO and found a small but sustained increase in the number of megakaryocytes in vitro. To better understand the effect of CCL5 on skewing of hematopoietic cells in a physiological setting, we serially injected mice with 0.025 mg/kg or 0.05 mg/kg CCL5 for one week. Flow cytometry of bone marrow from these mice showed an increase in the number of Pre-MK E (Lin-/cKit+/Sca-1-/CD41-/FcgR-/CD105-/CD150+) and Pre-CFU E (Lin-/cKit+/Sca-1-/CD41-/FcgR-/CD105+/CD150+) populations and a decrease in the number of Pre GM (Lin-/cKit+/Sca-1-/CD41-/FcgR-/CD105-/CD150-) cells in CCL5 treated mice. Interestingly, the CCL5 treated mice did not have a significant inflammatory response, as was measured by complete blood count, indicating that these hematopoietic changes are independent from inflammation. These data suggest that CCL5 may act to skew hematopoiesis toward a megakaryocyte-erythroid lineage and away from a granulocyte lineage in non-inflamed mice. Our proteomics data and in vitro live-dead assay suggest that CCL5 treatment protects against apoptosis through increased BAD phosphorylation, potentially leading to increased megakaryocyte maturation and proplatelet formation. Furthermore, CCL5 may have a dual action in both enhancing platelet production via pro-survival signaling and skewing hematopoiesis toward the megakaryocyte-erythroid lineage. Hematopoietic skewing was seen in CCL5 treated mice despite not having increased inflammation, suggesting that CCL5 itself does not cause a significant inflammatory response. Nonetheless, CCL5 has previously been shown to increase in an inflammatory setting, indicating that the effects seen here could be triggered by systemic inflammation in a different model. Further investigation of the mechanisms leading to these phenotypes may reveal novel therapeutic options to enhance megakaryocyte development and increase platelet counts in patients with thrombocytopenia. Disclosures Battinelli: Sanofi: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 123, No. 1 ( 2014-01-02), p. 101-112
    Abstract: Anticoagulants inhibit release of angiogenic proteins from platelets.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1793-1793
    Abstract: Platelets play important roles in both physiological and pathological conditions. For instance, platelets may facilitate cancer metastasis by protecting circulating tumor cells (CTCs) from shear stress and immunological assault during their intravascular phase and by supporting CTCs extravasation. Moreover, soluble factors released from activated platelets enhance proliferation and migration of endothelial cells, thereby promoting tumor angiogenesis. Correlations between increased platelet counts and shorter survival time have been described for many solid tumors. However, the role of platelets/megakaryocytes (MKs) in regulating tumor progression and dissemination in Multiple myeloma (MM) has not been previously examined. We measured the platelet aggregation-inducing abilities of MM cell lines and found MM cell lines (MM.1S, OPM-2, KMS-11, U266, and H929) induced platelet aggregation. This was not observed using leukemia cells (K562) and primary healthy donor-derived peripheral blood mononuclear cells. We next investigated whether platelets interact with MM cells within the bone marrow (BM) niche, in vivo. In order to identify platelets/MKs, femurs were harvested from MM.1S GFP+-harboring mice and stained with a DyLight649-conjugated anti-GPIb-beta antibody. The femurs were rendered transparent by the CUBIC method as previously described (Cell 159, 911-24 (2014)) and examined using confocal microscopy; and found that platelets/MKs co-localized within MM.1S GFP+-infiltrated BM niches. Further confirmation of the co-localization of MM cells and MKs was performed using immunohistochemistry (CD138+ and GPIb-alpha+). We next investigated the effect of platelets on MM cell proliferation: MM cells, where co-cultured with platelets and found that platelets enhanced the proliferation rate of MM cells in a platelet number-dependent manner, as shown by using BrdU (p 〈 0.001). The observed platelet-dependent induction of MM cells proliferation was also demonstrated by using platelet aggregation releasate. We next performed RNA sequencing studies using MM cells and platelet-interacting MM cells, and demonstrated that platelet-interacting MM cells presented with enrichment for signaling pathways such as TGF-beta as compared to MM cell control. We performed cytokine array analysis and confirmed that several cytokines, including TGF-beta, ENA-78, SDF-1, and TARC, were released from platelets activated by co-culture with MM cells. We next investigated the effect of platelets on MM tumor growth in vivo, by using bioluminescence imaging (BLI). MM cells were pre-cultured with platelets for 2 days, washed and then intravenously injected into SCID/Beige mice (n=6). We found a significantly higher MM tumor burden in mice injected with MM cell pre-cultured with platelets, as compared to control mice where buffer treated MM cells were used (p 〈 0.01), together with reduced survival (p=0.0011). These findings indicate the ability of platelets to enhance MM cell growth in vivo. To further confirm the role of platelets in mediating MM tumor growth, we performed platelet depletion by treating mice with an anti-platelet antibody (R300, 4 ug/g IP injection), and subsequently injected MM cells intravenously. We found that platelet-depleted mice presented with decreased MM tumor growth (p 〈 0.05) and improved survival rate compared to control mice treated with IgG isotype control (p=0.0044). These findings indicate that MM cells induce platelet aggregation and interact with platelets/MKs within the BM. Importantly, MM cell-platelet interaction is responsible for the activation of pro-survival signaling pathways in MM cells at transcriptome level, leading to enhanced MM cell proliferation, as shown both in vitro and in vivo. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2012
    In:  Blood Vol. 120, No. 21 ( 2012-11-16), p. SCI-35-SCI-35
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. SCI-35-SCI-35
    Abstract: Abstract SCI-35 In addition to their “classic” role in hemostasis, platelets are now known to be major contributors in wound healing, tumor growth, and angiogenesis. Stored within the α-granules is an array of angiogenic regulatory proteins, which are deposited by the secretion reaction of surface-activated platelets into the local environment of a tumor or wound. Despite the recognized importance of platelets in regulating new blood vessel growth, our understanding of how platelets modulate the angiogenic response remains unclear. Here, we consider how new α-granule biology may provide insights into how platelets regulate angiogenesis. First, we will present data on a new form of α-granule movement. We will show that actin filament assembly can power the propulsion of α-granule movement, and this may be a novel mechanism underlying the secretion reaction. Second, we show that one mechanism by which anticoagulants may impact malignancy is by disrupting the tumor cells' ability to hijack the angiogenic potential of platelets. Third, we will review the molecular basis of transport and delivery of α-granules to assembling platelets. Finally, we will discuss the concept of using “designer platelets” as a drug delivery system for targeted agents. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Elsevier BV ; 2021
    In:  Research and Practice in Thrombosis and Haemostasis Vol. 5, No. 7 ( 2021-10), p. e12594-
    In: Research and Practice in Thrombosis and Haemostasis, Elsevier BV, Vol. 5, No. 7 ( 2021-10), p. e12594-
    Type of Medium: Online Resource
    ISSN: 2475-0379
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 2901840-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 15_Supplement ( 2016-08-01), p. C12-C12
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 15_Supplement ( 2016-08-01), p. C12-C12
    Abstract: Platelets, primarily known for their role in hemostasis, are now recognized to play an integral role in cancer progression and metastasis. Recent evidence has established that platelets are activated by tumor cells, including breast cancer cells, leading to the release of hundreds of growth factors, cytokines, chemokines and angiogenesis mediators that could influence tumor growth and metastasis. Indeed, work from our group has demonstrated that factors released from activated platelets promote both metastasis and angiogenesis. However, little is known about the specific factors and signaling pathways that mediate this critical platelet-tumor cell crosstalk. To address this question, we performed an angiogenesis array (Ray Biotech) to identify specific pro-angiogenic and pro-metastatic factors released by tumor cells during platelet-tumor cell interactions. We identified several factors that were secreted by MCF-7 breast tumor cells in response to activated platelet releasate, including high levels of interleukin 8 (IL-8, CXCL8). IL-8 is a cytokine known to play a critical role in metastasis and angiogenesis and is elevated in the serum and tumor tissue of breast cancer patients. We confirmed that exposure to platelets strongly induced the production of IL-8 in several human breast cancer cell lines (MDA-MB-231, BT-20, SKBR-3 and MCF-7) by ELISA and found that platelets themselves do not contain detectable levels of IL-8. Furthermore, IL-8 production was highest in the more aggressive, triple negative MDA-MB-231 and BT-20 lines, suggesting a link between platelet-induced IL-8 and tumor subtype. Next we sought to determine the role of platelet-induced IL-8 in metastasis. We performed standard invasion assays using MDA-MB-231 cells transfected with IL-8shRNA or control cells. Platelets were able to increase the invasion of control MDA-MD-231 cells by 5 fold, while IL-8 knockdown reduced the effect by 50%. Furthermore, the ability of platelets to promote tumor cell migration across an endothelialized membrane was reduced 87% in IL-8 knockdown MDA-MB-231s compared to controls in standard transendothelial migration assays. These results suggest that platelets promote metastasis, in part, by driving tumor cell IL-8. To identify the specific component or components of platelet releasate responsible for driving tumor cell IL-8, we first characterized the contents of activated platelet releasate by array (Ray Biotech) and found an abundance of both chemokine (C-C motif) ligand 5 (CCL5, RANTES) and epidermal growth factor (EGF). Next, we treated breast tumor cell lines directly with recombinant CCL5 or EGF and observed an increase in IL-8 production; however, sensitivity to CCL5, EGF or the combination varied among the cell lines tested. We found that cell lines MCF-7 and MDA-MB-231, which express the CCL5 receptor CCR5, produced IL-8 in response to CCL5 while BT-20 and SKBR-3 cells produce IL-8 in response to EGF and express high levels of EGFR. To determine if platelet-derived CCL5 drives tumor cell IL-8 in MDA-MD-231 and MCF-7 cells, tumor cells were pretreated with the CCR5 blocker maraviroc and then exposed to platelets. CCR5 blockade abrogated the induction of IL-8 in response to platelets and decreased platelet-induced invasion. Similarly EGFR blockage with AG-1478 reduced IL-8 production in platelet-treated BT-20 and SKBR-3 tumor cells. Furthermore, pre-treatment of platelets with aspirin, an irreversible platelet inhibitor, diminished their ability to drive tumor cell IL-8 and to enhance invasion. Taken together, these results suggest that platelets, through release of soluble factors, drive tumor cells to produce IL-8 and that blocking this communication can disrupt the pro-metastatic potential of platelets. Ultimately, these studies support targeting specific platelet-tumor cell interactions as a novel means of limiting disease progression in breast cancer. Citation Format: Kelly E. Johnson, Kellie R. Machlus, Jodi A. Forward, Mason D. Tippy, Saleh A. El-Husayni, Joseph E. Italiano, Jr., Elisabeth M. Battinelli. Platelets promote breast cancer metastasis by reprogramming tumor cells to produce IL-8. [abstract]. In: Proceedings of the AACR Special Conference: Function of Tumor Microenvironment in Cancer Progression; 2016 Jan 7–10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2016;76(15 Suppl):Abstract nr C12.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    Cold Spring Harbor Laboratory ; 2023
    In:  Cold Spring Harbor Perspectives in Medicine Vol. 13, No. 1 ( 2023-01), p. a041174-
    In: Cold Spring Harbor Perspectives in Medicine, Cold Spring Harbor Laboratory, Vol. 13, No. 1 ( 2023-01), p. a041174-
    Type of Medium: Online Resource
    ISSN: 2157-1422
    Language: English
    Publisher: Cold Spring Harbor Laboratory
    Publication Date: 2023
    detail.hit.zdb_id: 2628603-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Arteriosclerosis, Thrombosis, and Vascular Biology, Ovid Technologies (Wolters Kluwer Health), Vol. 37, No. 4 ( 2017-04), p. 664-674
    Abstract: 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.
    Type of Medium: Online Resource
    ISSN: 1079-5642 , 1524-4636
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2017
    detail.hit.zdb_id: 1494427-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 2011
    In:  Circulation Vol. 124, No. 14 ( 2011-10-04), p. 1508-1510
    In: Circulation, Ovid Technologies (Wolters Kluwer Health), Vol. 124, No. 14 ( 2011-10-04), p. 1508-1510
    Type of Medium: Online Resource
    ISSN: 0009-7322 , 1524-4539
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2011
    detail.hit.zdb_id: 1466401-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...