1
|
Drygiannakis I, Valatas V, Filidou E, Tzenaki N, Archontoulaki E, Dovrolis N, Kandilogiannakis L, Kefalogiannis G, Sidiropoulos P, Kolios G, Koutroubakis IE. Low-Grade Activation of the Extrinsic Coagulation Pathway in Patients with Ulcerative Colitis. Dig Dis Sci 2024; 69:3773-3785. [PMID: 39322807 DOI: 10.1007/s10620-024-08640-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) increases the risk for venous thromboembolism. Tissue factor (TF) initiates the extrinsic coagulation pathway (ECP). AIMS To investigate the correlation of UC severity with latent ECP activation and TF expression in primary colonic stromal cells (PCSC). METHODS In plasma of 38 UC patients (31 males, disease duration 151 ± 25 months) and 28 healthy controls, exosomes and microparticles (EM) were counted. Moreover, TF protein concentration, activities of EM-bound TF (EM-TFa) and coagulation factor VII (FVIIa) were assessed. In PCSC in culture, TF mRNA (F3) from 12 patients with active UC and 7 controls was evaluated. RESULTS UC patients had 4- and 3.7- times more exosomes and microparticles, respectively, than controls. TF protein in UC was correlated with several disease severity indices, such as partial Mayo score (pMs; r 0.443), albumin (- 0.362), ESR (0.353), PLT (0.575), and endoscopic Ms (eMs 0.468). EM-TFa was also significantly higher in UC and was correlated to SIBDQ (- 0.64), albumin (- 0.624), disease extent and eMs (0.422). Refractory-to-treatment patients had significantly higher TF protein, EM-TFa and FVIIa. Even within responders, the need for steroids or biologics correlated with a 2.2-times higher EM-TFa. PCSC from active UC maintained higher F3 than controls, which was correlated to pMs (0.56), albumin (- 0.543) and eMs. Treatment with cytokines further upregulated F3. P for all comparisons was < 0.05. CONCLUSION Low-grade activation of the ECP associates with clinical, endoscopic UC activity and response to treatment. TF in PCSC mirrors its systemic activity and points to them as a source.
Collapse
Affiliation(s)
- Ioannis Drygiannakis
- Gastroenterology Research Laboratory, School of Medicine, University of Crete, 71500, Heraklion, Crete, Greece
- Department of Gastroenterology, University Hospital, P.O. BOX 1352, 71110, Heraklion, Crete, Greece
| | - Vassilis Valatas
- Gastroenterology Research Laboratory, School of Medicine, University of Crete, 71500, Heraklion, Crete, Greece
- Department of Gastroenterology, University Hospital, P.O. BOX 1352, 71110, Heraklion, Crete, Greece
| | - Eirini Filidou
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece
| | - Niki Tzenaki
- Gastroenterology Research Laboratory, School of Medicine, University of Crete, 71500, Heraklion, Crete, Greece
| | - Evangelia Archontoulaki
- Gastroenterology Research Laboratory, School of Medicine, University of Crete, 71500, Heraklion, Crete, Greece
| | - Nikolas Dovrolis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece
| | - Leonidas Kandilogiannakis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece
| | | | - Prodromos Sidiropoulos
- Laboratory of Rheumatology, Autoimmunity and Inflammation, School of Medicine, University of Crete, 71500, Heraklion, Crete, Greece
| | - George Kolios
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece
| | - Ioannis E Koutroubakis
- Gastroenterology Research Laboratory, School of Medicine, University of Crete, 71500, Heraklion, Crete, Greece.
- Department of Gastroenterology, University Hospital, P.O. BOX 1352, 71110, Heraklion, Crete, Greece.
| |
Collapse
|
2
|
Li J, Yan S, Zhang X, Xiang M, Zhang C, Gu L, Wei X, You C, Chen S, Zeng D, Jiang J. Circulating D-Dimers Increase the Risk of Mortality and Venous Thromboembolism in Patients With Lung Cancer: A Systematic Analysis Combined With External Validation. Front Med (Lausanne) 2022; 9:853941. [PMID: 35308559 PMCID: PMC8924589 DOI: 10.3389/fmed.2022.853941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/04/2022] [Indexed: 12/14/2022] Open
Abstract
Background D-dimer is a fibrin-degrading substance that is soluble and whose degradation is produced by plasma protein-mediated degradation of cross-linked fibrin. Previous investigations have shown a link between D-dimer and the mortality in lung cancer patients. However, different investigations varied whether D-dimer could predict prognosis in these patients. Methods A meta-analysis and systematic review of all available cohort studies were performed on the link between circulating D-dimer levels and survival of lung cancer patients. Relevant studies were searched in Embase, Cochrane Library, and PubMed databases. Data from 540 lung cancer patients from the First Hospital of Soochow University and Sichuan Cancer Hospital were used for external validation. Results We finally obtained 19 eligible cohort studies with pooled HR showing that high D-dimer levels contribute to death in tumor group (HR 1.62, 95% CI: 1.39-1.88, I2 = 75.0%). Further stratified analysis showed that higher circulating D-dimer in the advanced lung cancer group was linked to a 1.91-fold risk (HR = 2.91, 95% CI: 2.24-3.78, I2 = 6.0%). Incorporation of other variables, including days of follow-up, country, design, public year, population, disease status, and quality score, into the meta-regression model, indicated that disease status was an additional source of heterogeneity (p < 0.001). External validation of 540 patients also showed that high levels of D-dimer showed a higher risk of overall mortality (HR 1.39, 95% CI: 1.13-1.72, p = 0.002) and VTE events (HR 3.98, 95% CI: 1.99-8.70, p = 0.002) in lung cancer patients. Conclusions High circulating plasma D-dimer levels independently predict long-term prognosis and the risk of venous thromboembolism in lung cancer.
Collapse
Affiliation(s)
- Jing Li
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The Affiliated Dushu Lake Hospital of Soochow University, Suzhou, China
| | - Shanle Yan
- Department of Rheumatology and Immunology, The Affiliated Dushu Lake Hospital of Soochow University, Suzhou, China
| | - Xiaohui Zhang
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mengqi Xiang
- Department of Medical Oncology, Sichuan Cancer Hospital, Medical School, University of Electronic Science and Technology of China, Chengdu, China
| | - Chuanhua Zhang
- Department of Thoracic Surgery, Sichuan Cancer Hospital, Medical School, University of Electronic Science and Technology of China, Chengdu, China
| | - Ling Gu
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoying Wei
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The Affiliated Dushu Lake Hospital of Soochow University, Suzhou, China
| | - Chuanyun You
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shenhua Chen
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Daxiong Zeng
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The Affiliated Dushu Lake Hospital of Soochow University, Suzhou, China
| | - Junhong Jiang
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The Affiliated Dushu Lake Hospital of Soochow University, Suzhou, China
| |
Collapse
|
3
|
Beleva EA, Deneva TI, Stoencheva SS, Grudeva-Popova ZG. Longitudinal Dynamics of Coagulation and Angiogenesis Markers in Cancer Patients During and After Chemotherapy. Clin Appl Thromb Hemost 2021; 27:10760296211056637. [PMID: 34918975 PMCID: PMC8728769 DOI: 10.1177/10760296211056637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hemostatic parameters have been investigated as molecular determinants of tumor
progression. To analyze the dynamics of microparticle-associated tissue factor
activity (MPTF), tissue factor antigen (TF-Ag), and angiopоietin-2 (ANG-2) in
cancer patients before, during, and after active treatment and to explore their
potential as biomarkers for metastatic occurrence and death. Blood for the
analysis of MPTF, TF-Ag, ANG-2, and conventional hemostatic tests was sampled in
111 patients with various cancers at 4 consecutive visits: before first
chemotherapy cycle, after 3 courses, at the sixth course, and 3 months after
chemotherapy cessation. Patients were followed up until metastatic
progression/death or the end of the study. MPTF did not change during
chemotherapy, but increased significantly after treatment cessation. Total TF-Ag
and ANG-2 decreased throughout active treatment. Significant drop of their
levels was observed 3 months post therapy cessation. Progressive disease was
significantly associated with higher pre-chemotherapy TF-Ag and fibrinogen.
Elevated baseline levels of fibrinogen were associated with increased risk of
shortened progression free survival. Cessation of chemotherapy is associated
with significant change of hemostatic parameters. Pre-chemotherapy levels of
TF-Ag and fibrinogen may be informative of disease state and prognosis.
Collapse
Affiliation(s)
- Elina A Beleva
- 118870Medical University of Plovdiv, Plovdiv, Bulgaria.,564825University Multiprofile Hospital for Active Treatment "Sveti Georgi" EAD-Plovdiv, Plovdiv, Bulgaria
| | - Tanya I Deneva
- 118870Medical University of Plovdiv, Plovdiv, Bulgaria.,564825University Multiprofile Hospital for Active Treatment "Sveti Georgi" EAD-Plovdiv, Plovdiv, Bulgaria
| | - Snezhana S Stoencheva
- 118870Medical University of Plovdiv, Plovdiv, Bulgaria.,564825University Multiprofile Hospital for Active Treatment "Sveti Georgi" EAD-Plovdiv, Plovdiv, Bulgaria
| | - Zhanet G Grudeva-Popova
- 118870Medical University of Plovdiv, Plovdiv, Bulgaria.,564825University Multiprofile Hospital for Active Treatment "Sveti Georgi" EAD-Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
4
|
Lewis CS, Karve A, Matiash K, Stone T, Li J, Wang JK, Versteeg HH, Aronow BJ, Ahmad SA, Desai PB, Bogdanov VY. A First-In-Class, Humanized Antibody Targeting Alternatively Spliced Tissue Factor: Preclinical Evaluation in an Orthotopic Model of Pancreatic Ductal Adenocarcinoma. Front Oncol 2021; 11:691685. [PMID: 34395257 PMCID: PMC8358774 DOI: 10.3389/fonc.2021.691685] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/28/2021] [Indexed: 01/22/2023] Open
Abstract
In 2021, pancreatic ductal adenocarcinoma (PDAC) is the 3rd leading cause of cancer deaths in the United States. This is largely due to a lack of symptoms and limited treatment options, which extend survival by only a few weeks. There is thus an urgent need to develop new therapies effective against PDAC. Previously, we have shown that the growth of PDAC cells is suppressed when they are co-implanted with RabMab1, a rabbit monoclonal antibody specific for human alternatively spliced tissue factor (asTF). Here, we report on humanization of RabMab1, evaluation of its binding characteristics, and assessment of its in vivo properties. hRabMab1 binds asTF with a KD in the picomolar range; suppresses the migration of high-grade Pt45.P1 cells in Boyden chamber assays; has a long half-life in circulation (~ 5 weeks); and significantly slows the growth of pre-formed orthotopic Pt45.P1 tumors in athymic nude mice when administered intravenously. Immunohistochemical analysis of tumor tissue demonstrates the suppression of i) PDAC cell proliferation, ii) macrophage infiltration, and iii) neovascularization, whereas RNAseq analysis of tumor tissue reveals the suppression of pathways that promote cell division and focal adhesion. This is the first proof-of-concept study whereby a novel biologic targeting asTF has been investigated as a systemically administered single agent, with encouraging results. Given that hRabMab1 has a favorable PK profile and is able to suppress the growth of human PDAC cells in vivo, it comprises a promising candidate for further clinical development.
Collapse
Affiliation(s)
- Clayton S Lewis
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Aniruddha Karve
- Division of Pharmaceutical Sciences, University of Cincinnati College of Pharmacy, Cincinnati, OH, United States
| | - Kateryna Matiash
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Timothy Stone
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Jingxing Li
- Technology Development, LakePharma, Inc., Belmont, CA, United States
| | - Jordon K Wang
- Technology Development, LakePharma, Inc., Belmont, CA, United States
| | - Henri H Versteeg
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Bruce J Aronow
- Department of Biomedical Informatics, Cincinnati Children's Hospital and Medical Center, Cincinnati, OH, United States
| | - Syed A Ahmad
- Division of Surgical Oncology, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Pankaj B Desai
- Division of Pharmaceutical Sciences, University of Cincinnati College of Pharmacy, Cincinnati, OH, United States
| | - Vladimir Y Bogdanov
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
5
|
Li H, Yu Y, Shi Q, Chen X, Zheng P, Wang D, Tao P, Gu B, Li X, Zhang T, Xiang L, Xi D, Gao L, Maswikiti Ewetse P, Chen H. Prognostic significance of tissue factor in patients with pancreatic cancer: a systematic review protocol. BMJ Open 2020; 10:e037431. [PMID: 32928856 PMCID: PMC7490930 DOI: 10.1136/bmjopen-2020-037431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Pancreatic cancer is a highly aggressive digestive system tumour with poor prognosis. Venous thromboembolism (VTE) is a well-known complication of pancreatic cancer, and tissue factor (TF) contributes to the generation of a hypercoagulable state and thrombotic disease in pancreatic cancer. Several studies showed that an elevated TF level was related to the development of VTE and influenced the survival of patients with pancreatic cancer. Thus, we wish to conduct a systematic review of literature to clarify the prognostic significance of TF in pancreatic cancer. METHODS AND ANALYSIS Studies comparing the circulating microparticle-associated TF (MP TF) level between patients who had pancreatic cancer with and without VTE will be included to evaluate the roles of TF in VTE development. Studies comparing the survival data between patients with high TF expression and low TF expression will also be included to explore the association of TF expression with patient survival. The outcomes are plasma MP TF level and survival endpoints (overall and progression-free survival), respectively. Primary studies of any type published in English will be included. Two reviewers will search Medline, EMBASE and Cochrane databases from inception to June 2020, retrieve relevant studies, and independently select the literatures and extract data from the included studies. The quality of each included study will be assessed by the Newcastle-Ottawa Scale score. The HR and 95% CI of each study will be pooled for survival outcome, and the standardised mean difference (SMD) with 95% CIs will be used for continuous outcomes. If meta-analysis is inappropriate, the result will only be reported qualitatively. Subgroup and sensitivity analyses will be considered to identify sources of heterogeneity. The Grades of Recommendation, Assessment, Development and Evaluation method will be applied to assess the level of evidence of this systematic review. ETHICS AND DISSEMINATION There are no concerning ethical issues. The results will be published. PROSPERO REGISTRATION NUMBER CRD42019133665.
Collapse
Affiliation(s)
- Haiyuan Li
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yang Yu
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Qianling Shi
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xueping Chen
- Sleep Medicine Center, Gansu Provincial Hospital, Lanzhou, China
| | - Peng Zheng
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dengfeng Wang
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Pengxian Tao
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Baohong Gu
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xuemei Li
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Tao Zhang
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Lin Xiang
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dayong Xi
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Lei Gao
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Paul Maswikiti Ewetse
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Hao Chen
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
6
|
Xu J, Lu S, Wu P, Kong L, Ning C, Li H. Molecular mechanism whereby paraoxonase-2 regulates coagulation activation through endothelial tissue factor in rat haemorrhagic shock model. Int Wound J 2020; 17:735-741. [PMID: 32090497 PMCID: PMC7949017 DOI: 10.1111/iwj.13329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/05/2020] [Accepted: 02/09/2020] [Indexed: 01/17/2023] Open
Abstract
We investigated the molecular mechanism of paraoxonase-2 (PON-2) in regulating blood coagulation activation in rats with haemorrhagic shock through endothelial tissue factor (TF). Thirty adult Sprague Dawley rats were randomly divided into three groups: healthy control group (group A), the haemorrhagic shock PON-2 treatment group (group B), and the haemorrhagic shock group (group C). After the model was established, blood was withdrawn from the inferior vena cava of all rats. The difference in plasma thrombomodulin (TM) levels of the three groups was determined by Western blotting. The expression of transcription factors Egr-1 and Sp1 was detected by Western blotting assays. reverse transcription-polymerase chain Reaction (RT-PCR) was used to determine the mRNA expression of t-PA, PAI-1, TM, and PON-2 in the serum of three groups of rats. Endothelial TF was measured by enzyme linked immunosorbent assay (ELISA), and coagulation assay was used to detect the activity of coagulation factor VIII. Histopathological examination of the arteries of the rats was performed. The molecular mechanism of PON-2 in regulating blood coagulation activation in haemorrhagic shock model rats by endothelial tissue factor was analysed. The expression of thrombin was determined by electrophoresis. Compared with the healthy control group, the expression of TM in groups B and C decreased, both 188.64 ± 12.47 and 137.48 ± 9.72, respectively, with a significant difference. The mRNA expression of TM and PON was determined by RT-PCR. The mRNA expression of TM and PON in group B was 0.97 ± 0.07 and 1.14 ± 0.09, compared with the control group, and the mRNA expression of TM and PON in group C was 0.86 ± 0.38 and 1.12 ± 0.41, both of which increased, and there were significant differences. By measuring the expression of endothelial TF, the expression of TF in groups B and C was elevated to 12.69 ± 1.07 and 11.59 ± 0.87, with significant differences. The enzyme activities of PON-2 in groups B and C, which were 110.34 ± 14.37 and 52.37 ± 8.06, respectively, were increased compared with the healthy control group and there were significant differences. PON-2 regulates the activation of coagulation in rats with haemorrhagic shock by regulating the expression of endothelial tissue-related genes such as plasma TM and endothelial TF under hypoxic and ischaemic conditions.
Collapse
Affiliation(s)
- Jian‐Hua Xu
- Intensive Care UnitLinyi Central HospitalLinyiShandongChina
| | - Shi‐Jun Lu
- Intensive Care UnitLinyi Central HospitalLinyiShandongChina
| | - Peng Wu
- Intensive Care UnitLinyi Central HospitalLinyiShandongChina
| | - Ling‐Chen Kong
- Intensive Care UnitLinyi Central HospitalLinyiShandongChina
| | - Chao Ning
- Intensive Care UnitLinyi Central HospitalLinyiShandongChina
| | - Hai‐Yan Li
- Intensive Care UnitLinyi Central HospitalLinyiShandongChina
| |
Collapse
|
7
|
Hisada Y, Mackman N. Update from the laboratory: mechanistic studies of pathways of cancer-associated venous thrombosis using mouse models. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:182-186. [PMID: 31808871 PMCID: PMC6913477 DOI: 10.1182/hematology.2019000025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Cancer patients have an increased risk of venous thromboembolism (VTE). The rate of VTE varies with cancer type, with pancreatic cancer having one of the highest rates, suggesting that there are cancer type-specific mechanisms of VTE. Risk assessment scores, such as the Khorana score, have been developed to identify ambulatory cancer patients at high risk of VTE. However, the Khorana score performed poorly in discriminating pancreatic cancer patients at risk of VTE. Currently, thromboprophylaxis is not recommended for cancer outpatients. Recent clinical trials showed that factor Xa (FXa) inhibitors reduced VTE in high-risk cancer patients but also increased major bleeding. Understanding the mechanisms of cancer-associated thrombosis should lead to the development of safer antithrombotic drugs. Mouse models can be used to study the role of different prothrombotic pathways in cancer-associated thrombosis. Human and mouse studies support the notion that 2 prothrombotic pathways contribute to VTE in pancreatic cancer patients: tumor-derived, tissue factor-positive (TF+) extracellular vesicles (EVs), and neutrophils and neutrophil extracellular traps (NETs). In pancreatic cancer patients, elevated levels of plasma EVTF activity and citrullinated histone H3 (H3Cit), a NET biomarker, are independently associated with VTE. We observed increased levels of circulating tumor-derived TF+ EVs, neutrophils, cell-free DNA, and H3Cit in nude mice bearing human pancreatic tumors. Importantly, inhibition of tumor-derived human TF, depletion of neutrophils, or administration of DNAse I to degrade cell-free DNA (including NETs) reduced venous thrombosis in tumor-bearing mice. These studies demonstrate that tumor-derived TF+ EVs, neutrophils, and cell-free DNA contribute to venous thrombosis in a mouse model of pancreatic cancer.
Collapse
Affiliation(s)
- Yohei Hisada
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nigel Mackman
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
8
|
Elevated blood plasma levels of tissue factor-bearing extracellular vesicles in patients with atrial fibrillation. Thromb Res 2019; 173:141-150. [DOI: 10.1016/j.thromres.2018.11.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/05/2018] [Accepted: 11/27/2018] [Indexed: 02/06/2023]
|
9
|
Lewis CS, Thomas HE, Orr-Asman M, Green LC, Boody RE, Matiash K, Karve A, Hisada YM, Davis HW, Qi X, Mercer C, Lucas FV, Aronow BJ, Mackman N, Versteeg HH, Bogdanov VY. mTOR kinase inhibition reduces tissue factor expression and growth of pancreatic neuroendocrine tumors. J Thromb Haemost 2019; 17:169-182. [PMID: 30472780 PMCID: PMC6345540 DOI: 10.1111/jth.14342] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Indexed: 12/22/2022]
Abstract
Essentials Tissue factor (TF) isoforms are expressed in pancreatic neuroendocrine tumors (pNET). TF knockdown inhibits proliferation of human pNET cells in vitro. mTOR kinase inhibitor sapanisertib/MLN0128 suppresses TF expression in human pNET cells. Sapanisertib suppresses TF expression and activity and reduces the growth of pNET tumors in vivo. SUMMARY: Background Full-length tissue factor (flTF) and alternatively spliced TF (asTF) contribute to growth and spread of pancreatic ductal adenocarcinoma. It is unknown, however, if flTF and/or asTF contribute to the pathobiology of pancreatic neuroendocrine tumors (pNETs). Objective To assess TF expression in pNETs and the effects of mTOR complex 1/2 (mTORC1/2) inhibition on pNET growth. Methods Human pNET specimens were immunostained for TF. Human pNET cell lines QGP1 and BON were evaluated for TF expression and responsiveness to mTOR inhibition. shRNA were used to knock down TF in BON. TF cofactor activity was assessed using a two-step FXa generation assay. TF promoter activity was assessed using transient transfection of human TF promoter-driven reporter constructs into cells. Mice bearing orthotopic BON tumors were treated with the mTORC1/2 ATP site competitive inhibitor sapanisertib/MLN0128 (3 mg kg-1 , oral gavage) for 34 days. Results Immunostaining of pNET tissue revealed flTF and asTF expression. BON and QGP1 expressed both TF isoforms, with BON exhibiting higher levels. shRNA directed against TF suppressed BON proliferation in vitro. Treatment of BON with sapanisertib inhibited mTOR signaling and suppressed TF levels. BON tumors grown in mice treated with sapanisertib had significantly less TF protein and cofactor activity, and were smaller compared with tumors grown in control mice. Conclusions TF isoforms are expressed in pNETs. Sapanisertib suppresses TF mRNA and protein expression as well as TF cofactor activity in vitro and in vivo. Thus, further studies are warranted to evaluate the clinical utility of TF-suppressing mTORC1/2 inhibitor sapanisertib in pNET management.
Collapse
Affiliation(s)
- Clayton S Lewis
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Hala Elnakat Thomas
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Melissa Orr-Asman
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Lisa C Green
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Rachel E Boody
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Kateryna Matiash
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Aniruddha Karve
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Yohei M. Hisada
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Harold W Davis
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Xiaoyang Qi
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Carol Mercer
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Fred V Lucas
- Department of Pathology & Laboratory Medicine, University of Cincinnati College of Medicine
| | - Bruce J. Aronow
- Computational Medicine and Division of Biomedical Informatics, Cincinnati Children’s Hospital and Medical Center
| | - Nigel Mackman
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Henri H Versteeg
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center
| | - Vladimir Y Bogdanov
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine
| |
Collapse
|
10
|
Alternatively spliced tissue factor levels are elevated in the plasma of patients with chronic liver diseases. Eur J Gastroenterol Hepatol 2018; 30:1470-1475. [PMID: 30113368 DOI: 10.1097/meg.0000000000001236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES In patients with chronic liver diseases, hypercoagulability can contribute to the progression of fibrosis and complications of cirrhosis. Tissue factor (TF) is a transmembrane glycoprotein that initiates the extrinsic pathway of blood coagulation. Recent investigations have established that TF is elevated in patients with pancreatic cancer, blood disorders, diabetes, and cardiovascular disease. Alternatively spliced tissue factor (asTF), a secreted form of TF, induces angiogenesis and exhibits low-level procoagulant activity. The aim of this study was to investigate whether the circulating levels of asTF are elevated in the plasma of patients with liver disease. MATERIALS AND METHODS In a single-center study, we retrospectively analyzed asTF plasma levels in healthy participants and patients having stage F0-F3 liver fibrosis, liver cirrhosis, as well as hepatocellular carcinoma (HCC). AsTF plasma levels were measured using a sandwich enzyme-linked immunosorbent assay. Values were expressed as median with interquartile range (IQR). RESULTS The lowest median plasma asTF concentration (94 pg/ml, IQR: 33-275) was found in the healthy control group. The patients with low-grade liver fibrosis (F0-F1 group) displayed the highest median asTF concentration (404 pg/ml, IQR: 277-789). Significant differences between the asTF levels in the plasma of healthy participants and those in patients with grade F0-F1 fibrosis (P<0.001), patients with grade F2-F3 fibrosis (P=0.019), patients with cirrhosis (P=0.004), and patients with HCC (P<0.001) were found using a Wilcoxon rank-sum test. Treatment-naive patients with HCC had significantly higher asTF levels (P=0.018) than those receiving treatment. AsTF levels were found to increase with worsening Child-Pugh scores and heightened liver disease activity. CONCLUSION AsTF levels are elevated in patients with chronic liver diseases, which increase with worsening Child-Pugh scores and decrease following HCC therapy.
Collapse
|
11
|
Mørk M, Nielsen MH, Bæk R, Jørgensen MM, Pedersen S, Kristensen SR. Postprandial Increase in Blood Plasma Levels of Tissue Factor-Bearing (and Other) Microvesicles Measured by Flow Cytometry: Fact or Artifact? TH OPEN 2018; 2:e147-e157. [PMID: 31249938 PMCID: PMC6524869 DOI: 10.1055/s-0038-1642021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/01/2018] [Indexed: 11/23/2022] Open
Abstract
Tissue factor (TF)–bearing microvesicles (MVs) and exosomes may play a role in hemostasis and thrombosis. MVs may be quantified by flow cytometry (FC)–based detection of phosphatidylserine (PS)-positive submicron particles carrying specific antigens, although interference from lipoproteins complicates this approach. In this study, we evaluated the effect of food intake on blood levels of TF-bearing particles measured by FC and small extracellular vesicles (EVs) measured by a protein microarray–based test termed EV Array. Platelet-free plasma (PFP) was obtained from 20 healthy persons in the fasting state and 75 minutes after consumption of a meal. Postprandial changes in the concentration of PS-positive particles, including subgroups binding labeled antibodies against TF, CD41, CD146, and CD62E, respectively (FC), small EVs (EV Array), and TF antigen and procoagulant phospholipids (PPLs) were measured. Furthermore, we tested the effect on FC results of in vitro addition of lipoproteins to fasting PFP. We found significantly increased plasma concentrations of PS-positive particles and all examined subgroups postprandially, while no changes in small EVs, PPL, or TF antigen levels were found. Levels of all types of particles measured by FC were also elevated by lipoprotein spiking. In conclusion, meal consumption as well as in vitro addition of lipoproteins to fasting plasma induces increased levels of PS-positive particles as measured by FC, including TF-positive subtypes and subtypes exposing other antigens. While the observed postprandial increase may to some extent reflect elevated MV levels, our results indicate a substantial interference from lipoproteins.
Collapse
Affiliation(s)
- Morten Mørk
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.,Aalborg AF Study Group, Aalborg University Hospital, Aalborg, Denmark.,EVsearch.dk, Aalborg, Denmark
| | - Morten H Nielsen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.,EVsearch.dk, Aalborg, Denmark
| | - Rikke Bæk
- EVsearch.dk, Aalborg, Denmark.,Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Malene M Jørgensen
- EVsearch.dk, Aalborg, Denmark.,Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Shona Pedersen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.,EVsearch.dk, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Søren R Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.,Aalborg AF Study Group, Aalborg University Hospital, Aalborg, Denmark.,EVsearch.dk, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
12
|
Grover SP, Mackman N. Tissue Factor: An Essential Mediator of Hemostasis and Trigger of Thrombosis. Arterioscler Thromb Vasc Biol 2018; 38:709-725. [PMID: 29437578 DOI: 10.1161/atvbaha.117.309846] [Citation(s) in RCA: 465] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/25/2018] [Indexed: 12/21/2022]
Abstract
Tissue factor (TF) is the high-affinity receptor and cofactor for factor (F)VII/VIIa. The TF-FVIIa complex is the primary initiator of blood coagulation and plays an essential role in hemostasis. TF is expressed on perivascular cells and epithelial cells at organ and body surfaces where it forms a hemostatic barrier. TF also provides additional hemostatic protection to vital organs, such as the brain, lung, and heart. Under pathological conditions, TF can trigger both arterial and venous thrombosis. For instance, atherosclerotic plaques contain high levels of TF on macrophage foam cells and microvesicles that drives thrombus formation after plaque rupture. In sepsis, inducible TF expression on monocytes leads to disseminated intravascular coagulation. In cancer patients, tumors release TF-positive microvesicles into the circulation that may contribute to venous thrombosis. TF also has nonhemostatic roles. For instance, TF-dependent activation of the coagulation cascade generates coagulation proteases, such as FVIIa, FXa, and thrombin, which induce signaling in a variety of cells by cleavage of protease-activated receptors. This review will focus on the roles of TF in protective hemostasis and pathological thrombosis.
Collapse
Affiliation(s)
- Steven P Grover
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Nigel Mackman
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill.
| |
Collapse
|
13
|
Graf C, Ruf W. Tissue factor as a mediator of coagulation and signaling in cancer and chronic inflammation. Thromb Res 2018; 164 Suppl 1:S143-S147. [PMID: 29703473 DOI: 10.1016/j.thromres.2018.01.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/23/2022]
Abstract
Thrombosis is frequently diagnosed as a first symptom in tumor patients and the clinical management of hypercoagulability in cancer patients remains challenging due to concomitant changes in risk factors for severe bleeding. It therefore remains a priority to better understand interactions of the hemostatic system with cancer biology. Specifically, further research is needed to elucidate the details and effects of new anticoagulants on extravascular coagulation and the interplay between cancer progression and chronic inflammation. In addition, it will be important to identify subgroups of cancer patients benefiting from specific modulations of the coagulation system without increasing the bleeding risk. Here, we review recent findings on tissue factor (TF) regulation, its procoagulant activity and TF signaling in the various cell types of the tumor microenvironment.
Collapse
Affiliation(s)
- Claudine Graf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany.
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
14
|
Unruh D, Ünlü B, Lewis CS, Qi X, Chu Z, Sturm R, Keil R, Ahmad SA, Sovershaev T, Adam M, Van Dreden P, Woodhams BJ, Ramchandani D, Weber GF, Rak JW, Wolberg AS, Mackman N, Versteeg HH, Bogdanov VY. Antibody-based targeting of alternatively spliced tissue factor: a new approach to impede the primary growth and spread of pancreatic ductal adenocarcinoma. Oncotarget 2018; 7:25264-75. [PMID: 26967388 PMCID: PMC5041902 DOI: 10.18632/oncotarget.7955] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 02/13/2016] [Indexed: 01/08/2023] Open
Abstract
Alternatively spliced Tissue Factor (asTF) is a secreted form of Tissue Factor (TF), the trigger of blood coagulation whose expression levels are heightened in several forms of solid cancer, including pancreatic ductal adenocarcinoma (PDAC). asTF binds to β1 integrins on PDAC cells, whereby it promotes tumor growth, metastatic spread, and monocyte recruitment to the stroma. In this study, we determined if targeting asTF in PDAC would significantly impact tumor progression. We here report that a novel inhibitory anti-asTF monoclonal antibody curtails experimental PDAC progression. Moreover, we show that tumor-derived asTF is able to promote PDAC primary growth and spread during early as well as later stages of the disease. This raises the likelihood that asTF may comprise a viable target in early- and late-stage PDAC. In addition, we show that TF expressed by host cells plays a significant role in PDAC spread. Together, our data demonstrate that targeting asTF in PDAC is a novel strategy to stem PDAC progression and spread.
Collapse
Affiliation(s)
- Dusten Unruh
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Betül Ünlü
- Leiden University Medical Center, Leiden, The Netherlands
| | - Clayton S Lewis
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Xiaoyang Qi
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Zhengtao Chu
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Robert Sturm
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Ryan Keil
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Syed A Ahmad
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | | | | | | | | | | | - Georg F Weber
- College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Janusz W Rak
- McGill University Health Centre, Montreal Children's Hospital, Montreal, Canada
| | - Alisa S Wolberg
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nigel Mackman
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | |
Collapse
|
15
|
Kanikarla-Marie P, Lam M, Menter DG, Kopetz S. Platelets, circulating tumor cells, and the circulome. Cancer Metastasis Rev 2017; 36:235-248. [PMID: 28667367 DOI: 10.1007/s10555-017-9681-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Mørk M, Handberg A, Pedersen S, Jørgensen MM, Bæk R, Nielsen MK, Kristensen SR. Prospects and limitations of antibody-mediated clearing of lipoproteins from blood plasma prior to nanoparticle tracking analysis of extracellular vesicles. J Extracell Vesicles 2017; 6:1308779. [PMID: 28473885 PMCID: PMC5405563 DOI: 10.1080/20013078.2017.1308779] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/31/2017] [Indexed: 01/12/2023] Open
Abstract
Introduction: Nanoparticle tracking analysis (NTA) enables measurement of extracellular vesicles (EVs) but lacks the ability to distinct between EVs and lipoproteins which are abundantly present in blood plasma. Limitations in ultracentrifugation and size exclusion chromatography applied for EV isolation may result in inadequate EV purification and preservation. In this proof of concept study, we aimed to evaluate the potential of antibody-mediated removal of lipoproteins from plasma prior to extracellular vesicle (EV) analysis by nanoparticle tracking analysis (NTA). Methods: Ten platelet-free plasma (PFP) samples from healthy fasting subjects were incubated with magnetic beads coated with antibodies against apolipoprotein B-48 and B-100 (ApoB). Plasma samples were analysed with NTA before and after application of the bead procedure. Four fasting PFP samples were analysed with an ELISA specific for human ApoB to estimate the degree of removal of lipoproteins and EV array analysis was used for identification of possible EV loss. Results: The magnetic bead separation procedure resulted in a median reduction of the particle concentration in plasma by 62% (interquartile range 32-72%). The mean size of the remaining particles generally increased. ApoB concentration was reduced to a level close to the background signal, whereas a median reduction of the EV content by 21% (range 8-43%) was observed. Conclusion: Anti-ApoB antibody coated magnetic beads may hold potential for removal of lipoproteins from human PFP prior to EV measurement by NTA but some artefactual effect and EV loss may have to be endured.
Collapse
Affiliation(s)
- Morten Mørk
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.,AF Study Group, Aalborg University Hospital, Aalborg, Denmark
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Shona Pedersen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.,AF Study Group, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Malene M Jørgensen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark, part of EVsearch.dk
| | - Rikke Bæk
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark, part of EVsearch.dk
| | - Morten K Nielsen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Søren R Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.,AF Study Group, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
17
|
Detection of tissue factor-positive extracellular vesicles by laser scanning confocal microscopy. Thromb Res 2016; 150:65-72. [PMID: 28043041 DOI: 10.1016/j.thromres.2016.12.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Increased levels of tissue factor-positive extracellular vesicles (TF+EVs) have been detected in the plasma of patients with various diseases, including cancer and endotoxemia. Levels of TF+EVs in plasma samples can be measured by antigen and activity assays. The aim of the present study was to visualize TF+EVs by laser scanning confocal microscopy (LSCM). METHODS EVs were isolated from the supernatant of two cultured human pancreatic cancer cell lines (Panc-1 and BxPc-3), from untreated or lipopolysaccharide (LPS) treated whole blood, and from plasma of pancreatic cancer patients. EV-TF activity was determined using an in-house assay. The EVs were labeled with 5(6)-carboxyfluorescein diacetate N-succinimidyl ester, which is converted to the impermeant green fluorescent molecule carboxyfluorescein inside the EVs. EVs were either captured using annexin V and detected using a fluorescent-labeled anti-TF antibody, or captured using an anti-TF antibody and detected using fluorescent-labeled annexin V. EVs were visualized by LSCM. RESULTS TF+EVs were easily detected from high TF-expressing BxPc-3 cells using annexin V capture, whereas the addition of tyramide amplification was required to detect TF+EVs from low TF-expressing Panc-1 cells. Visualization of TF+EVs in plasma from LPS treated whole human blood and in plasma from pancreatic cancer patients required either capture with annexin V and detection with a fluorescent-labeled anti-TF antibody with tyramide signal amplification, or capture with an anti-TF antibody and detection with a fluorescent-labeled annexin V. CONCLUSION LSCM enables visualization of TF+EVs in the supernatant from cultured cells and in clinical samples.
Collapse
|
18
|
Santilli F, Boccatonda A, Davì G. Aspirin, platelets, and cancer: The point of view of the internist. Eur J Intern Med 2016; 34:11-20. [PMID: 27344083 DOI: 10.1016/j.ejim.2016.06.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 01/18/2023]
Abstract
Growing evidence suggests the beneficial effect of aspirin against some types of cancer, particularly of the gastrointestinal tract, and it has been provided for an effect both in cancer prevention as well as in survival improvement of cancer patients. Aspirin benefits increase with duration of treatment, especially after 10years of treatment. The inhibition of platelet activation at sites of gastrointestinal mucosal lesions could be the primary mechanism of action of low-dose aspirin. Indeed, the formation of tumor cell-induced platelet aggregates may favor immune evasion, by releasing angiogenic and growth factors, and also by promoting cancer cell dissemination. Moreover, platelets may contribute to aberrant COX-2 expression in colon carcinoma cells, thereby contributing to downregulation of oncosuppressor genes and upregulation of oncogenes, such as cyclin B1. Platelet adhesion to cancer cells leads also to an increased expression of genes involved in the EMT, such as the EMT-inducing transcription factors ZEB1 and TWIST1 and the mesenchymal marker vimentin. The aspirin-mediated inactivation of platelets may restore antitumor reactivity by blocking the release of paracrine lipid and protein mediators that induce COX-2 expression in adjacent nucleated cells at sites of mucosal injury. Thus, recent findings suggest interesting perspectives on "old" aspirin and NSAID treatment and/or "new" specific drugs to target the "evil" interactions between platelets and cancer for chemoprevention.
Collapse
Affiliation(s)
- F Santilli
- Center for Aging Science (Ce.S.I.), Università G. d'Annunzio" Foundation, Italy; Department of Internal Medicine, "G. d'Annunzio" University of Chieti, Italy
| | - A Boccatonda
- Center for Aging Science (Ce.S.I.), Università G. d'Annunzio" Foundation, Italy; Department of Internal Medicine, "G. d'Annunzio" University of Chieti, Italy
| | - G Davì
- Center for Aging Science (Ce.S.I.), Università G. d'Annunzio" Foundation, Italy; Department of Internal Medicine, "G. d'Annunzio" University of Chieti, Italy.
| |
Collapse
|
19
|
Key NS, Khorana AA, Mackman N, McCarty OJT, White GC, Francis CW, McCrae KR, Palumbo JS, Raskob GE, Chan AT, Sood AK. Thrombosis in Cancer: Research Priorities Identified by a National Cancer Institute/National Heart, Lung, and Blood Institute Strategic Working Group. Cancer Res 2016; 76:3671-5. [PMID: 27527638 DOI: 10.1158/0008-5472.can-15-3100] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 04/25/2016] [Indexed: 11/16/2022]
Abstract
The risk for venous thromboembolism (VTE) is increased in cancer and particularly with chemotherapy, and it portends poorer survival among patients with cancer. However, many fundamental questions about cancer-associated VTE, or Trousseau syndrome, remain unanswered. This report summarizes the proceedings of a working group assembled by the NCI and NHLBI in August 2014 to explore the state of the science in cancer-associated VTE, identify clinically important research gaps, and develop consensus on priorities for future research. Representing a convergence of research priorities between the two NIH Institutes, the workshop addressed epidemiologic, basic science, clinical, and translational issues in cancer-associated VTE. Cancer Res; 76(13); 3671-5. ©2016 AACR.
Collapse
Affiliation(s)
- Nigel S Key
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alok A Khorana
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Nigel Mackman
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - Owen J T McCarty
- Department of Biomedical Engineering and Cell and Development Biology, Oregon Health and Science University, Portland, Orlando
| | - Gilbert C White
- Blood Research Institute, Blood Center of Wisconsin, Department of Medicine, Biochemistry, and Pharmacology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Charles W Francis
- James P. Wilmot Cancer Center and Department of Medicine, University of Rochester, Rochester, New York
| | - Keith R McCrae
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio. Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Joseph S Palumbo
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Gary E Raskob
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Anil K Sood
- Department of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
20
|
Mørk M, Pedersen S, Botha J, Lund SM, Kristensen SR. Preanalytical, analytical, and biological variation of blood plasma submicron particle levels measured with nanoparticle tracking analysis and tunable resistive pulse sensing. Scandinavian Journal of Clinical and Laboratory Investigation 2016; 76:349-60. [PMID: 27195974 DOI: 10.1080/00365513.2016.1178801] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Nanoparticle tracking analysis (NTA) and tunable resistive pulse sensing (TRPS) enable measurement of extracellular vesicles (EVs) in blood plasma but also measure other particles present in plasma. Complete isolation of EVs from similarly sized particles with full EV recovery is currently not possible due to limitations in existing isolation techniques. AIM This study aimed to evaluate preanalytical, analytical, and biological variation of particle measurements with NTA and TRPS on blood plasma. METHODS Blood from 20 healthy subjects was sampled in the fasting and postprandial state. Platelet free plasma (PFP) was analyzed immediately and after a freeze-thaw cycle. Additionally, the effect of prandial state and a freeze-thaw cycle on EV-enriched particle fractions obtained via size-exclusion chromatography (SEC) was examined. RESULTS We observed analytical linearity in the range of 1.0-10.0 × 10(8) particles/mL for NTA and 1.0 × 10(8)-1.8 × 10(9) particles/mL for TRPS. The analytical variation was generally below 10%. A considerable intra- and inter-individual variation was demonstrated with estimated reference intervals of 1.4 × 10(11)-1.2 × 10(12) particles/mL for NTA and 1.8 × 10(8)-1.6 × 10(9) particles/mL for TRPS. Food intake and to a lesser extent a freeze-thaw cycle affected particle populations in PFP and, similarly, in EV-enriched fractions. CONCLUSION In this study NTA and TRPS enabled acceptably precise concentration and size measurement of submicron particles in PFP. An appreciable intra- and inter-individual biological variation was observed. In studies on particle populations in PFP or EV-enriched fractions, we recommend analysis of fresh, fasting samples.
Collapse
Affiliation(s)
- Morten Mørk
- a Department of Clinical Biochemistry , Aalborg University Hospital , Denmark ;,b AF Study Group , Aalborg University Hospital , Denmark
| | - Shona Pedersen
- a Department of Clinical Biochemistry , Aalborg University Hospital , Denmark ;,b AF Study Group , Aalborg University Hospital , Denmark ;,c Department of Clinical Medicine , Aalborg University , Aalborg , Denmark
| | - Jaco Botha
- a Department of Clinical Biochemistry , Aalborg University Hospital , Denmark
| | - Sigrid Marie Lund
- a Department of Clinical Biochemistry , Aalborg University Hospital , Denmark
| | - Søren Risom Kristensen
- a Department of Clinical Biochemistry , Aalborg University Hospital , Denmark ;,b AF Study Group , Aalborg University Hospital , Denmark ;,c Department of Clinical Medicine , Aalborg University , Aalborg , Denmark
| |
Collapse
|
21
|
Witkowski M, Landmesser U, Rauch U. Tissue factor as a link between inflammation and coagulation. Trends Cardiovasc Med 2016; 26:297-303. [DOI: 10.1016/j.tcm.2015.12.001] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 12/08/2015] [Accepted: 12/08/2015] [Indexed: 10/22/2022]
|
22
|
Eisenreich A, Bolbrinker J, Leppert U. Tissue Factor: A Conventional or Alternative Target in Cancer Therapy. Clin Chem 2016; 62:563-70. [DOI: 10.1373/clinchem.2015.241521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 01/14/2016] [Indexed: 11/06/2022]
Abstract
Abstract
BACKGROUND
Tissue factor (TF) is an evolutionary conserved glycoprotein that plays an important role in the pathogenesis of cancer. TF is expressed in 2 naturally occurring protein isoforms, membrane-bound full-length (fl)TF and soluble alternatively spliced (as)TF. Both isoforms have been shown to affect a variety of pathophysiologically relevant functions, such as tumor-associated angiogenesis, thrombogenicity, tumor growth, and metastasis. Therefore, targeting TF either by direct inhibition or indirectly, i.e., on a posttranscriptional level, offers a novel therapeutic option for cancer treatment.
CONTENT
In this review we summarize the latest findings regarding the role of TF and its isoforms in cancer biology. Moreover, we briefly depict and discuss the therapeutic potential of direct and/or indirect inhibition of TF activity and expression for the treatment of cancer.
SUMMARY
asTF and flTF play important and often distinct roles in cancer biology, i.e., in thrombogenicity and angiogenesis, which is mediated by isoform-specific signal transduction pathways. Therefore, both TF isoforms and downstream signaling are promising novel therapeutic targets in malignant diseases.
Collapse
Affiliation(s)
- Andreas Eisenreich
- Charité-Universitätsmedizin Berlin, CC04, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany
| | - Juliane Bolbrinker
- Charité-Universitätsmedizin Berlin, CC04, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany
| | - Ulrike Leppert
- Charité-Universitätsmedizin Berlin, CC02, Institut für Physiologie, Berlin, Germany
| |
Collapse
|
23
|
Nomura S, Niki M, Nisizawa T, Tamaki T, Shimizu M. Microparticles as Biomarkers of Blood Coagulation in Cancer. BIOMARKERS IN CANCER 2015; 7:51-6. [PMID: 26462252 PMCID: PMC4592056 DOI: 10.4137/bic.s30347] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 12/20/2022]
Abstract
Cancer is associated with hypercoagulopathy and increased risk of thrombosis. This negatively influences patient morbidity and mortality. Cancer is also frequently complicated by the development of venous thromboembolism (VTE). Tumor-derived tissue factor (TF)-bearing microparticles (MPs) are associated with VTE events in malignancy. MPs are small membrane vesicles released from many different cell types by exocytic budding of the plasma membrane in response to cellular activation or apoptosis. MPs may also be involved in clinical diseases through expression of procoagulative phospholipids. The detection of TF-expressing MPs in cancer patients may be clinically useful. In lung and breast cancer patients, MPs induce metastasis and angiogenesis and may be indicators of vascular complications. Additionally, MPs in patients with various types of cancer possess adhesion proteins and bind target cells to promoting cancer progression or metastasis. Overexpression of TF by cancer cells is closely associated with tumor progression, and shedding of TF-expressing MPs by cancer cells correlates with the genetic status of cancer. Consequently, TF-expressing MPs represent important markers to consider in the prevention of and therapy for VTE complications in cancer patients.
Collapse
Affiliation(s)
- Shosaku Nomura
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Maiko Niki
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Tohru Nisizawa
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Takeshi Tamaki
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Michiomi Shimizu
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
24
|
Bogdanov VY, Versteeg HH. "Soluble Tissue Factor" in the 21st Century: Definitions, Biochemistry, and Pathophysiological Role in Thrombus Formation. Semin Thromb Hemost 2015; 41:700-7. [PMID: 26408917 DOI: 10.1055/s-0035-1556049] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tissue factor (TF), the main trigger of blood coagulation, is essential for normal hemostasis. Over the past 20 years, heightened intravascular levels and activity of TF have been increasingly perceived as an entity that significantly contributes to venous as well as arterial thrombosis. Various forms of the TF protein in the circulation have been described and proposed to be thrombogenic. Aside from cell and vessel wall-associated TF, several forms of non-cell-associated TF circulate in plasma and may serve as a causative factor in thrombosis. At the present time, no firm consensus exists regarding the extent, the vascular setting(s), and/or the mechanisms by which such TF forms contribute to thrombus initiation and propagation. Here, we summarize the existing paradigms and recent, sometimes paradigm-shifting findings elucidating the structural, mechanistic, and pathophysiological characteristics of plasma-borne TF.
Collapse
Affiliation(s)
- Vladimir Y Bogdanov
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Henri H Versteeg
- Department of Internal Medicine, Section of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
25
|
PML-RARa modulates the vascular signature of extracellular vesicles released by acute promyelocytic leukemia cells. Angiogenesis 2015; 19:25-38. [PMID: 26374632 DOI: 10.1007/s10456-015-9486-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/10/2015] [Indexed: 01/13/2023]
Abstract
Oncogenic transformation is believed to impact the vascular phenotype and microenvironment in cancer, at least in part, through mechanisms involving extracellular vesicles (EVs). We explored these questions in the context of acute promyelocytic leukemia cells (NB4) expressing oncogenic fusion protein, PML-RARa and exquisitely sensitive to its clinically used antagonist, the all-trans retinoic acid (ATRA). We report that NB4 cells produce considerable numbers of EVs, which are readily taken up by cultured endothelial cells triggering their increased survival. NB4 EVs contain PML-RARa transcript, but no detectable protein, which is also absent in endothelial cells upon the vesicle uptake, thereby precluding an active intercellular trafficking of this oncogene in this setting. ATRA treatment changes the emission profile of NB4-related EVs resulting in preponderance of smaller vesicles, an effect that occurs in parallel with the onset of cellular differentiation. ATRA also increases IL-8 mRNA and protein content in NB4 cells and their EVs, while decreasing the levels of VEGF and tissue factor (TF). Endothelial cell uptake of NB4-derived EVs renders these cells more TF-positive and procoagulant, and this effect is diminished by pre-treatment of EV donor cells with ATRA. Profiling angiogenesis-related transcripts in intact and ATRA-treated APL cells and their EVs reveals multiple differences attributable to cellular responses and EV molecular packaging. These observations point to the potential significance of changes in the angiogenic signature and activity associated with EVs released from tumor cells subjected to targeted therapy.
Collapse
|
26
|
Thomas GM, Brill A, Mezouar S, Crescence L, Gallant M, Dubois C, Wagner DD. Tissue factor expressed by circulating cancer cell-derived microparticles drastically increases the incidence of deep vein thrombosis in mice. J Thromb Haemost 2015; 13:1310-9. [PMID: 25955268 PMCID: PMC4496280 DOI: 10.1111/jth.13002] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 04/13/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND The risk of thrombotic complications such as deep vein thrombosis (DVT) during tumor development is well known. Tumors release into the circulation procoagulant microparticles (MPs) that can participate in thrombus formation following vessel injury. The importance of this MP tissue factor (TF) in the initiation of cancer-associated DVT remains uncertain. OBJECTIVE To investigate how pancreatic cancer MPs promote DVT in vivo. METHODS We combined a DVT mouse model in which thrombosis is induced by flow restriction in the inferior vena cava with one of subcutaneous pancreatic cancer in C57BL/6J mice. We infused high-TF and low-TF tumor MPs to determine the importance of TF in experimental cancer-associated DVT. RESULTS Both tumor-bearing mice and mice infused with tumor MPs subjected to 3 h of partial flow restriction developed an occlusive thrombus; fewer than one-third of the control mice did. We observed that MPs adhered to neutrophil extracellular traps (NETs), which are functionally important players during DVT, whereas neither P-selectin nor glycoprotein Ib were required for MP recruitment in DVT. The thrombotic phenotype induced by MP infusion was suppressed by hirudin, suggesting the importance of thrombin generation. TF carried by tumor MPs was essential to promote DVT, as mice infused with low-TF tumor MPs had less thrombosis than mice infused with high-TF tumor MPs. CONCLUSIONS TF expressed on tumor MPs contributes to the increased incidence of cancer-associated venous thrombosis in mice in vivo. These MPs may adhere to NETs formed at the site of thrombosis.
Collapse
Affiliation(s)
- G M Thomas
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- VRCM, Aix Marseille Université, Inserm UMR-S 1076, Marseille, France
| | - A Brill
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - S Mezouar
- VRCM, Aix Marseille Université, Inserm UMR-S 1076, Marseille, France
| | - L Crescence
- VRCM, Aix Marseille Université, Inserm UMR-S 1076, Marseille, France
| | - M Gallant
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - C Dubois
- VRCM, Aix Marseille Université, Inserm UMR-S 1076, Marseille, France
| | - D D Wagner
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
27
|
Unruh D, Sagin F, Adam M, Van Dreden P, Woodhams BJ, Hart K, Lindsell CJ, Ahmad SA, Bogdanov VY. Levels of Alternatively Spliced Tissue Factor in the Plasma of Patients with Pancreatic Cancer May Help Predict Aggressive Tumor Phenotype. Ann Surg Oncol 2015; 22 Suppl 3:S1206-11. [DOI: 10.1245/s10434-015-4592-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Indexed: 12/30/2022]
|
28
|
Dicke C, Langer F. Pathophysiology of Trousseau's syndrome. Hamostaseologie 2014; 35:52-9. [PMID: 25403091 DOI: 10.5482/hamo-14-08-0037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 11/06/2014] [Indexed: 12/26/2022] Open
Abstract
Clinically relevant clotting abnormalities in cancer patients are referred to as Trousseau's syndrome. While thrombotic complications such as venous thromboembolism are most frequent in every day's practice, cancer patients may also experience severe bleeding symptoms due to complex systemic coagulopathies, including disseminated intravascular coagulation, haemolytic thrombotic microangiopathy, and hyperfibrinolysis. The pathophysiology of Trousseau's syndrome involves all aspects of Virchow's triad, but previous basic research has mainly focused on the cellular and molecular mechanisms underlying blood hypercoagulability in solid cancers and haematological malignancies. In this regard, over-expression of tissue factor (TF), the principal initiator of the extrinsic coagulation pathway, by primary tumour cells and increased shedding of TF-bearing plasma microparticles are critical to both thrombus formation and cancer progression. However, novel findings on intrinsic contact activation in vivo, such as the release of polyphosphates or DNA by activated platelets and neutrophils, respectively, have pointed to additional pathways in the complex pathophysiology of Trousseau's syndrome.
Collapse
Affiliation(s)
| | - F Langer
- Priv.-Doz. Dr. med. Florian Langer, II. Medizinische Klinik und Poliklinik, Hubertus Wald Tumorzentrum - Universitäres Cancer Center Hamburg (UCCH), Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany, Tel. +49/(0)40/741 05-24 53, -06 64; Fax -51 93, E-mail:
| |
Collapse
|
29
|
Ettelaie C, Collier MEW, Maraveyas A, Ettelaie R. Characterization of physical properties of tissue factor-containing microvesicles and a comparison of ultracentrifuge-based recovery procedures. J Extracell Vesicles 2014; 3. [PMID: 25206957 PMCID: PMC4134674 DOI: 10.3402/jev.v3.23592] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 07/21/2014] [Accepted: 07/21/2014] [Indexed: 12/11/2022] Open
Abstract
Microvesicles were isolated from the conditioned media of 3 cell lines (MDA-MB-231, AsPC-1 and A375) by ultracentrifugation at a range of relative centrifugal forces, and the tissue factor (TF) protein and activity, microvesicle number, size distribution and relative density compared. Also, by expressing TF-tGFP in cells and isolating the microvesicles, the relative density of TF-containing microvesicles was established. Nanoparticle tracking analysis (NTA) indicated that the larger-diameter microvesicles (>200 nm) were primarily sedimented at 100,000g and possessed TF-dependent thrombin and factor Xa generation potential, while in the absence of factor VII, all microvesicles possessed some thrombin generation capacity. Immuno-precipitation of TF-containing microvesicles followed by NTA also indicated the range of these microvesicles to be 200–400 nm. Analysis of the microvesicles by gradient density centrifugation showed that lower-density (<1.1 g/ml) microvesicles were mainly present in the samples recovered at 100,000g and were associated with TF antigen and activity. Analysis of these fractions by NTA confirmed that these fractions were principally composed of the larger-diameter microvesicles. Similar analysis of microvesicles from healthy or patient plasma supported those obtained from conditioned media indicating that TF activity was mainly associated with lower-density microvesicles. Furthermore, centrifugation of healthy plasma, supplemented with TF-tGFP-containing microvesicles, resulted in 67% retrieval of the fluorescent microvesicles at 100,000g, but only 26% could be recovered at 20,000g. Pre-centrifugation of conditioned media or plasma at 10,000g improved the speed and yield of recovered TF-containing microvesicles by subsequent centrifugation at either 20,000g or 100,000g. In conclusion, TF appears to be associated with low-density (1.03–1.08 g/ml), larger-diameter (200–350 nm) microvesicles.
Collapse
Affiliation(s)
- Camille Ettelaie
- Biomedical Section, Department of Biological Sciences, University of Hull, Hull, UK
| | - Mary E W Collier
- Biomedical Section, Department of Biological Sciences, University of Hull, Hull, UK
| | - Anthony Maraveyas
- Division of Cancer-Hull York Medical School, University of Hull, Hull, UK
| | - Rammile Ettelaie
- Food Colloids Group, School of Food Science and Nutrition, University of Leeds, Leeds, UK
| |
Collapse
|
30
|
Leppert U, Eisenreich A. The role of tissue factor isoforms in cancer biology. Int J Cancer 2014; 137:497-503. [PMID: 24806794 DOI: 10.1002/ijc.28959] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 05/02/2014] [Indexed: 12/17/2022]
Abstract
Tissue Factor (TF) is an evolutionary conserved glycoprotein, which is of immense importance for a variety of biologic processes. TF is expressed in two naturally occurring protein isoforms, membrane-bound "full-length" (fl)TF and soluble alternatively spliced (as)TF. The TF isoform expression is differentially modulated on post-transcriptional level via regulatory factors, such as serine/arginine-rich (SR) proteins, SR protein kinases and micro (mi)RNAs. Both isoforms mediate a variety of physiologic- and pathophysiologic-relevant functions, such as thrombogenicity, angiogenesis, cell signaling, tumor cell proliferation and metastasis. In this review, we will depict the main mechanisms regulating the TF isoform expression in cancer and under other pathophysiologic-relevant conditions. Moreover, we will summarize and discuss the latest findings regarding the role of TF and its isoforms in cancer biology.
Collapse
Affiliation(s)
- Ulrike Leppert
- Charité - Universitätsmedizin Berlin, Campus Mitte, Charite Centrum 04/13, Berlin, Germany
| | - Andreas Eisenreich
- Charité - Universitätsmedizin Berlin, Campus Mitte, Charite Centrum 04/13, Berlin, Germany
| |
Collapse
|
31
|
Devalet B, Mullier F, Chatelain B, Dogné JM, Chatelain C. The central role of extracellular vesicles in the mechanisms of thrombosis in paroxysmal nocturnal haemoglobinuria: a review. J Extracell Vesicles 2014; 3:23304. [PMID: 24672668 PMCID: PMC3965713 DOI: 10.3402/jev.v3.23304] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/17/2014] [Accepted: 02/20/2014] [Indexed: 12/22/2022] Open
Abstract
Paroxysmal nocturnal haemoglobinuria (PNH) is an acquired disorder of the haematopoietic stem cell that makes blood cells more sensitive to the action of complement. PNH patients experience an increased risk of arterial and venous thrombosis – major causes of death due to this disease. Though many potential interlaced mechanisms are suspected, extracellular vesicles (EVs) of various origins may play a central role. The processes possibly involved are haemolysis, platelet activation, injured endothelial cells and monocyte activation. The impact of transfusion should be evaluated. A better understanding of the mechanisms involved may help to propose guidelines for the prophylaxis and treatment of thrombosis in PNH. In this paper, we propose an updated review of the pathophysiology of the underlying mechanisms of thrombosis associated with PNH, with specific focus on the prominent role of EVs.
Collapse
Affiliation(s)
- Bérangère Devalet
- Department of Hematology, Namur Thrombosis and Hemostasis Center (NTHC), CHU Dinant-Godinne UCL Namur, Yvoir, Belgium
| | - François Mullier
- Hematology Laboratory, Namur Thrombosis and Hemostasis Center (NTHC), CHU Dinant-Godinne UCL Namur, Yvoir, Belgium ; Department of Pharmacy, Namur Thrombosis and Hemostasis Center (NTHC), University of Namur, Belgium
| | - Bernard Chatelain
- Hematology Laboratory, Namur Thrombosis and Hemostasis Center (NTHC), CHU Dinant-Godinne UCL Namur, Yvoir, Belgium
| | - Jean-Michel Dogné
- Department of Pharmacy, Namur Thrombosis and Hemostasis Center (NTHC), University of Namur, Belgium
| | - Christian Chatelain
- Department of Hematology, Namur Thrombosis and Hemostasis Center (NTHC), CHU Dinant-Godinne UCL Namur, Yvoir, Belgium
| |
Collapse
|