Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Thromb Res ; 193: 180-189, 2020 09.
Article in English | MEDLINE | ID: mdl-32717643

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) is associated with a 5-fold increased risk of thromboembolic stroke. Extracellular vesicles (EVs) convey pathophysiological information and are possible biomarkers for risk of stroke. METHODS: EVs were measured in 836 patients with AF (of which 280 were stroke cases) selected from the ARISTOTLE trial and in a cohort of unselected 70 year old individuals (n = 1007, reference material). EVs from platelets, leukocytes, erythrocytes and inflammatory endothelial cells were measured using flow cytometry and a solid-phase proximity ligation assay. RESULTS: Concentrations of EVs were higher in the ARISTOTLE patients than in the PIVUS cohort for all the EV groups except EVs from endothelial cells (p < 0.0001). The distributions of the concentrations of the EVs were similar among the control group and the stroke cases for all of the sources of EVs in the ARISTOTLE study. EVs were modestly correlated with the levels of NT-ProBNP, Cystatin C, GDF-15 and D-dimer. Stronger correlations were found for platelet EVs as well as phosphatidyl serine positive EVs that were correlated with CD40 ligand in the ARISTOTLE study. Leukocyte EVs were correlated with IL-6 in both the ARISTOTLE and the PIVUS study, implicating them in different physiological processes. CONCLUSIONS: Higher levels of EVs were found in anticoagulated patients with AF and a higher risk of stroke than in a general population of similar age, possibly due to the high disease burden in AF patients. Our data with EVs representing a broad repertoire of activated blood cells in AF patients suggest that EVs are likely not a key mediator of occurrence of stroke in this population.


Subject(s)
Atrial Fibrillation , Extracellular Vesicles , Stroke , Aged , Biomarkers , Endothelial Cells , Humans
2.
Cancer Res ; 80(16): 3345-3358, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32586981

ABSTRACT

Platelet-derived growth factor B (PDGFB) plays a crucial role in recruitment of PDGF receptor ß-positive pericytes to blood vessels. The endothelium is an essential source of PDGFB in this process. Platelets constitute a major reservoir of PDGFB and are continuously activated in the tumor microenvironment, exposing tumors to the plethora of growth factors contained in platelet granules. Here, we show that tumor vascular function, as well as pericyte coverage is significantly impaired in mice with conditional knockout of PDGFB in platelets. A lack of PDGFB in platelets led to enhanced hypoxia and epithelial-to-mesenchymal transition in the primary tumors, elevated levels of circulating tumor cells, and increased spontaneous metastasis to the liver or lungs in two mouse models. These findings establish a previously unknown role for platelet-derived PDGFB, whereby it promotes and maintains vascular integrity in the tumor microenvironment by contributing to the recruitment of pericytes. SIGNIFICANCE: Conditional knockout of PDGFB in platelets demonstrates its previously unknown role in the maintenance of tumor vascular integrity and host protection against metastasis.


Subject(s)
Cell Movement , Endothelium, Vascular/metabolism , Pericytes/physiology , Proto-Oncogene Proteins c-sis/physiology , Animals , Blood Vessels , Colonic Neoplasms/blood supply , Epithelial-Mesenchymal Transition , Extracellular Matrix , Gene Knockout Techniques , Hybridization, Genetic , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Melanoma/blood supply , Melanoma/secondary , Mice , Neoplastic Cells, Circulating , Pancreatic Neoplasms , Pericytes/metabolism , Platelet Activation/physiology , Proto-Oncogene Proteins c-sis/deficiency , Proto-Oncogene Proteins c-sis/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Thrombocytopenia , Tumor Hypoxia , Tumor Microenvironment
3.
TH Open ; 2(3): e250-e260, 2018 Jul.
Article in English | MEDLINE | ID: mdl-31276087

ABSTRACT

Extracellular vesicles (EVs) derived from blood cells are promising biomarkers for various diseases. However, they are difficult to measure accurately in plasma due to their small size. Here, we demonstrate that platelet-derived EVs in plasma can be measured using solid-phase proximity ligation assay with high sensitivity and specificity using very small sample volume of biological materials. The results correlate well with high-sensitivity flow cytometry with the difference that the smallest EVs are detected. Briefly, the EVs are first captured on a solid phase, using lactadherin binding, and detection requires recognition with two antibodies followed by qPCR. The assay, using cholera toxin subunit-B or lactadherin as capture agents, also allowed detection of the more rare population of tissue factor (TF)-positive EVs at a concentration similar to sensitive TF activity assays. Thus, this assay can detect different types of EVs with high specificity and sensitivity, and has the potential to be an attractive alternative to flow cytometric analysis of preclinical and clinical samples. Improved techniques for measuring EVs in plasma will hopefully contribute to the understanding of their role in several diseases.

4.
Thromb Haemost ; 117(7): 1379-1390, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28492698

ABSTRACT

Monocytes and monocyte-derived microvesicles (MVs) are the main source of circulating tissue factor (TF). Increased monocyte TF expression and increased circulating levels of procoagulant MVs contribute to the formation of a prothrombotic state in patients with cardiovascular disease. Interleukin (IL)-33 is a pro-inflammatory cytokine involved in atherosclerosis and other inflammatory diseases, but its role in regulating thrombosis is still unclear. The aim of the present study was to investigate the effects of IL-33 on the procoagulant properties of human monocytes and monocyte-derived MVs. IL-33 induced a time- and concentration-dependent increase of monocyte TF mRNA and protein levels via binding to the ST2-receptor and activation of the NF-κB-pathway. The IL-33 treated monocytes also released CD14+TF+ MVs and IL-33 was found to increase the TF activity of both the isolated monocytes and monocyte-derived MVs. The monocytes were classified into subsets according to their CD14 and CD16 expression. Intermediate monocytes (IM) showed the highest ST2 receptor expression, followed by non-classical monocytes (NCM), and classical monocytes (CM). IL-33 induced a significant increase of TF only in the IM (p<0.01), with a tendency in NCM (p=0.06), but no increase was observed in CM. Finally, plasma levels of IL-33 were positively correlated with CD14+TF+ MVs in patients undergoing carotid endarterectomy (r=0.480; p=0.032; n=20). We hereby provide novel evidence that the proinflammatory cytokine IL-33 induces differential TF expression and activity in monocyte subsets, as well as the release of procoagulant MVs. In this manner, IL-33 may contribute to the formation of a prothrombotic state characteristic for cardiovascular disease.


Subject(s)
Cell-Derived Microparticles/metabolism , Interleukin-33/physiology , Monocytes/physiology , Thromboplastin/physiology , Aged , Carotid Stenosis/blood , Carotid Stenosis/immunology , Cells, Cultured , Female , Humans , Interleukin-1 Receptor-Like 1 Protein/blood , Interleukin-33/pharmacology , Lipopolysaccharide Receptors/blood , Male , Middle Aged , Monocytes/classification , Monocytes/immunology , NF-kappa B/blood , RNA, Messenger/blood , RNA, Messenger/genetics , Recombinant Proteins/pharmacology , Thromboplastin/genetics , Thrombosis/etiology
5.
Thromb Haemost ; 117(8): 1571-1581, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28424820

ABSTRACT

Microparticles (MPs) are formed from platelets (PMPs), endothelial cells (EMPs) and monocytes (MMPs), and in acute myocardial infarction (MI), there is an increase of MPs in the culprit artery. In this study MPs were evaluated in whole blood in 105 patients with MI at five time-points during a two-year follow-up (FU). Patients with non-ST-elevated MI had higher concentrations of CD41+MPs compared to ST-elevated MI patients (p=0.024). The concentrations of PMPs in whole blood increased during the time period (p<0.001), but no significant change over time was found for EMPs and MMPs. CD62P+MP counts were higher in MI patients with diabetes (p=0.020), and patients with hypertension had increased levels of CD14+MPs (p=0.004). The amount of CD62P+TF+MPs increased significantly during FU (p<0.001). Patients with atherosclerosis in three arterial beds, i. e. coronary, carotid and peripheral arteries, had lower concentrations of CD62P+TF+MPs (p=0.035) and CD144+TF+MPs (p=0.004) compared to patients with atherosclerosis in one or two arterial beds. Higher concentrations of CD62P+MPs early after MI were associated with an increased risk of cardiovascular events during FU, hazard ratio 3.32 (95 %CI1.20-9.31). Only small variations in PMP, EMP and MMP concentrations were found during long-term FU after MI and their levels seem to reflect the underlying cardiovascular disease rather than the acute MI. PMPs expressing P-selectin might be a promising biomarker for predicting future cardiovascular events, but further studies are needed to confirm these results.


Subject(s)
Blood Platelets/metabolism , Carotid Artery Diseases/blood , Cell-Derived Microparticles/metabolism , Coronary Artery Disease/blood , Endothelial Cells/metabolism , Monocytes/metabolism , Myocardial Infarction/blood , Peripheral Arterial Disease/blood , Aged , Annexin A5/blood , Antigens, CD/blood , Biomarkers/blood , Cadherins/blood , Carotid Artery Diseases/diagnosis , Carotid Artery Diseases/epidemiology , Comorbidity , Coronary Artery Disease/diagnosis , Coronary Artery Disease/epidemiology , Female , Follow-Up Studies , Humans , Lipopolysaccharide Receptors/blood , Male , Middle Aged , Myocardial Infarction/diagnosis , Myocardial Infarction/epidemiology , P-Selectin/blood , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/epidemiology , Prognosis , Risk Assessment , Risk Factors , Sweden/epidemiology , Thromboplastin/metabolism , Time Factors
6.
Biomark Med ; 10(9): 1009-22, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27586235

ABSTRACT

Cardiovascular diseases (CVDs) are a common cause of death, and a search for biomarkers for risk stratification is warranted. Elevated levels of cell-derived microparticles (MPs) are found in patients with CVD and in groups with risk factors for CVD. Subpopulations of MPs are promising biomarkers for improving risk prediction, as well as monitoring treatment. However, the field has been hampered by technical difficulties, and the ongoing development of sensitive standardized techniques is crucial for implementing MP analyses in the clinic. Large prospective studies are required to establish which MPs are of prognostic value in different patient groups. In this review, we discuss methodological challenges and progress in the field, as well as MP populations that are of interest for further clinical evaluation.


Subject(s)
Biomarkers/blood , Cardiovascular Diseases/diagnosis , Cell-Derived Microparticles/metabolism , Antigens, CD/metabolism , Atherosclerosis/diagnosis , Atherosclerosis/pathology , Atrial Fibrillation/diagnosis , Atrial Fibrillation/pathology , Cardiovascular Diseases/pathology , Heart Failure/diagnosis , Humans , Prognosis , Risk Factors , Shear Strength
7.
BMC Cancer ; 16: 341, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27246245

ABSTRACT

BACKGROUND: Tissue Factor (TF) forms a proteolytically active complex together with coagulation factor VIIa (FVIIa) and functions as the trigger of blood coagulation or alternatively activates cell signaling. We recently described that EphA2 of the Eph tyrosine kinase receptor family is cleaved directly by the TF/FVIIa complex. The aim of the present study was to further characterize the cross-talk between TF/FVIIa and EphA2 using in vitro model systems and human cancer specimens. METHODS: Cleavage and phosphorylation of EphA2 was studied by Western blot. Subcellular localization of TF and EphA2 was investigated by a proximity ligation assay and confocal microscopy. Phalloidin staining of the actin cytoskeleton was used to study cell rounding and retraction fiber formation. Expression of TF and EphA2 in human colorectal cancer specimens was examined by immunohistochemistry. RESULTS: TF and EphA2 co-localized constitutively in MDA-MB-231 cells, and addition of FVIIa resulted in cleavage of EphA2 by a PAR2-independent mechanism. Overexpression of TF in U251 glioblastoma cells lead to co-localization with EphA2 at the leading edge and FVIIa-dependent cleavage of EphA2. FVIIa potentiated ephrin-A1-induced cell rounding and retraction fiber formation in MDA-MB-231 cells through a RhoA/ROCK-dependent pathway that did not require PAR2-activation. TF and EphA2 were expressed in colorectal cancer specimens, and were significantly correlated. CONCLUSIONS: These results suggest that TF/FVIIa-EphA2 cross-talk might potentiate ligand-dependent EphA2 signaling in human cancers, and provide initial evidence that it is possible for this interaction to occur in vivo.


Subject(s)
Colorectal Neoplasms/metabolism , Factor VIIa/metabolism , Receptor Cross-Talk/physiology , Receptor, EphA2/metabolism , Thromboplastin/metabolism , Blotting, Western , Gene Knockdown Techniques , Humans , Immunohistochemistry , Microscopy, Confocal , Tissue Array Analysis
8.
Angiogenesis ; 16(4): 889-902, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23793459

ABSTRACT

Mice lacking histidine-rich glycoprotein (HRG) display an accelerated angiogenic switch and larger tumors-a phenotype caused by enhanced platelet activation in the HRG-deficient mice. Here we show that platelets induce molecular changes in the pre-tumorigenic environment in HRG-deficient mice, promoting cell survival, angiogenesis and epithelial-to-mesenchymal transition (EMT) and that these effects involved signaling via TBK1, Akt2 and PDGFRß. These early events subsequently translate into an enhanced rate of spontaneous metastasis to distant organs in mice lacking HRG. Later in tumor development characteristic features of pathological angiogenesis, such as decreased perfusion and pericyte coverage, are more pronounced in HRG-deficient mice. At this stage, platelets are essential to support the larger tumor volumes formed in mice lacking HRG by keeping their tumor vasculature sufficiently functional. We conclude that HRG-deficiency promotes tumor progression via enhanced platelet activity and that platelets play a dual role in this process. During early stages of transformation, activated platelets promote tumor cell survival, the angiogenic switch and invasiveness. In the more progressed tumor, platelets support the enhanced pathological angiogenesis and hence increased tumor growth seen in the absence of HRG. Altogether, our findings strengthen the notion of HRG as a potent tumor suppressor, with capacity to attenuate the angiogenic switch, tumor growth, EMT and subsequent metastatic spread, by regulating platelet activity.


Subject(s)
Blood Platelets/physiology , Carcinoma/blood supply , Epithelial-Mesenchymal Transition/physiology , Neoplasm Metastasis/physiopathology , Neovascularization, Pathologic/physiopathology , Pancreatic Neoplasms/blood supply , Proteins/physiology , Signal Transduction/physiology , Tumor Suppressor Proteins/deficiency , Animals , Carcinoma/genetics , Carcinoma/physiopathology , Carcinoma/secondary , Cell Transformation, Neoplastic , Cellular Microenvironment , Disease Progression , Insulinoma/genetics , Insulinoma/pathology , Insulinoma/physiopathology , Islets of Langerhans/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Metastasis/genetics , Neovascularization, Pathologic/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/physiopathology , Protein Serine-Threonine Kinases/physiology , Proteins/genetics , Proto-Oncogene Proteins c-akt/physiology , Receptor, Platelet-Derived Growth Factor beta/physiology , Tumor Burden , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
9.
PLoS One ; 6(1): e14526, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21264222

ABSTRACT

BACKGROUND: The heparin-binding plasma protein histidine-rich glycoprotein (HRG; alternatively, HRGP/HPRG) can suppress tumor angiogenesis and growth in vitro and in vivo. Mice lacking the HRG gene are viable and fertile, but have an enhanced coagulation resulting in decreased bleeding times. In addition, the angiogenic switch is significantly enhanced in HRG-deficient mice. METHODOLOGY/PRINCIPAL FINDINGS: To address whether HRG deficiency affects tumor development, we have crossed HRG knockout mice with the RIP1-Tag2 mouse, a well established orthotopic model of multistage carcinogenesis. RIP1-Tag2 HRG(-/-) mice display significantly larger tumor volume compared to their RIP1-Tag2 HRG(+/+) littermates, supporting a role for HRG as an endogenous regulator of tumor growth. In the present study we also demonstrate that platelet activation is increased in mice lacking HRG. To address whether this elevated platelet activation contributes to the increased pathological angiogenesis in HRG-deficient mice, they were rendered thrombocytopenic before the onset of the angiogenic switch by injection of the anti-platelet antibody GP1bα. Interestingly, this treatment suppressed the increase in angiogenic neoplasias seen in HRG knockout mice. However, if GP1bα treatment was initiated at a later stage, after the onset of the angiogenic switch, no suppression of tumor growth was detected in HRG-deficient mice. CONCLUSIONS: Our data show that increased platelet activation mediates the accelerated angiogenic switch in HRG-deficient mice. Moreover, we conclude that platelets play a crucial role in the early stages of tumor development but are of less significance for tumor growth once angiogenesis has been initiated.


Subject(s)
Neovascularization, Pathologic , Platelet Activation , Proteins/genetics , Animals , Blood Coagulation , Glycoproteins , Mice , Mice, Knockout , Neoplasms/etiology , Tumor Burden/genetics
10.
Mol Cancer Res ; 7(11): 1792-802, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19903770

ABSTRACT

The angiogenesis inhibitor histidine-rich glycoprotein (HRG) constitutes one of several examples of molecules regulating both angiogenesis and hemostasis. The antiangiogenic properties of HRG are mediated via its proteolytically released histidine- and proline-rich (His/Pro-rich) domain. Using a combination of immunohistochemistry and mass spectrometry, we here provide biochemical evidence for the presence of a proteolytic peptide, corresponding to the antiangiogenic domain of HRG, in vivo in human tissue. This finding supports a role for HRG as an endogenous regulator of angiogenesis. Interestingly, the His/Pro-rich peptide bound to the vessel wall in tissue from cancer patients but not to the vasculature in tissue from healthy persons. Moreover, the His/Pro-rich peptide was found in close association with platelets. Relesate from in vitro-activated platelets promoted binding of the His/Pro-rich domain of HRG to endothelial cells, an effect mediated by Zn(2+). Previous studies have shown that zinc-dependent binding of the His/Pro-rich domain of HRG to heparan sulfate on endothelial cells is required for inhibition of angiogenesis. We describe a novel mechanism to increase the local concentration and activity of an angiogenesis inhibitor, which may reflect a host response to counteract angiogenesis during pathologic conditions. Our finding that tumor angiogenesis is elevated in HRG-deficient mice supports this conclusion.


Subject(s)
Blood Platelets/metabolism , Neoplasms/blood supply , Neoplasms/metabolism , Proteins/metabolism , Amino Acid Sequence , Angiogenesis Inhibitors/metabolism , Animals , Endothelial Cells/metabolism , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Neoplasms/blood , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/pathology , Peptide Fragments/blood , Peptide Fragments/metabolism , Platelet Activation , Repetitive Sequences, Amino Acid
11.
Exp Cell Res ; 312(13): 2547-56, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16769050

ABSTRACT

Histidine-rich glycoprotein (HRGP) is an abundant heparin-binding plasma protein. We have shown that a fragment released from the central histidine/proline-rich (His/Pro-rich) domain of HRGP blocks endothelial cell migration in vitro and vascularization and growth of murine fibrosarcoma in vivo. The minimal active HRGP domain exerting the anti-angiogenic effect was recently narrowed down to a 35 amino acid peptide, HRGP330, derived from the His/Pro-rich domain of HRGP. By use of a signal transduction antibody array representing 400 different signal transduction molecules, we now show that HRGP and the synthetic peptide HRGP330 specifically induce tyrosine phosphorylation of focal adhesion kinase and its downstream substrate paxillin in endothelial cells. HRGP/HRGP330 treatment of endothelial cells induced disruption of actin stress fibers, a process reversed by treatment of cells with the FAK inhibitor geldanamycin. In addition, VEGF-mediated endothelial cell tubular morphogenesis in a three-dimensional collagen matrix was inhibited by HRGP and HRGP330. In contrast, VEGF-induced proliferation was not affected by HRGP or HRGP330, demonstrating the central role of cell migration during tube formation. In conclusion, our data show that HRGP targets focal adhesions in endothelial cells, thereby disrupting the cytoskeletal organization and the ability of endothelial cells to assemble into vessel structures.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Focal Adhesions/drug effects , Proteins/pharmacology , Signal Transduction/drug effects , Actins/metabolism , Animals , Benzoquinones , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Lactams, Macrocyclic , Mice , Peptides/pharmacology , Phosphoproteins/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Array Analysis , Quinones/pharmacology , Stress Fibers/drug effects , Vascular Endothelial Growth Factor A/pharmacology
12.
Cancer Res ; 66(4): 2089-97, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16489009

ABSTRACT

Histidine-rich glycoprotein (HRGP) is an abundant heparin-binding plasma protein that efficiently arrests growth and vascularization of mouse tumor models. We have shown that the antiangiogenic effect of HRGP is dependent on its histidine/proline-rich domain, which needs to be released from the mother protein to exert its effects. Here we identify a 35-amino-acid peptide, HRGP330, derived from the histidine/proline-rich domain as endowed with antiangiogenic properties in vitro and in vivo. The mechanism of action of HRGP330 involves subversion of focal adhesion function by disruption of integrin-linked kinase (ILK) and focal adhesion kinase (FAK) functions, inhibition of vascular endothelial growth factor (VEGF)-induced tyrosine phosphorylation of the FAK substrate alpha-actinin, and, as a consequence, an arrest in endothelial cell motility. The disturbed focal adhesion function is reflected in the ability of HRGP as well as of HRGP330 to prevent endothelial cell adhesion to vitronectin in a manner involving alpha(v)beta3 integrin. In conclusion, HRGP330, which we define as the minimal antiangiogenic domain of HRGP, exerts its effects through signal transduction targeting focal adhesions, thereby interrupting VEGF-induced endothelial cell motility.


Subject(s)
Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Proteins/pharmacology , Actinin/metabolism , Amino Acid Sequence , Animals , Cattle , Cell Movement/drug effects , Cell Movement/physiology , Endothelial Cells/cytology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin alphaVbeta3/metabolism , Molecular Sequence Data , Paxillin/antagonists & inhibitors , Paxillin/biosynthesis , Peptide Fragments/chemistry , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proteins/chemistry , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...