1
|
Heidari Z, Naeimzadeh Y, Fallahi J, Savardashtaki A, Razban V, Khajeh S. The Role of Tissue Factor In Signaling Pathways of Pathological Conditions and Angiogenesis. Curr Mol Med 2024; 24:1135-1151. [PMID: 37817529 DOI: 10.2174/0115665240258746230919165935] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/10/2023] [Accepted: 07/27/2023] [Indexed: 10/12/2023]
Abstract
Tissue factor (TF) is an integral transmembrane protein associated with the extrinsic coagulation pathway. TF gene expression is regulated in response to inflammatory cytokines, bacterial lipopolysaccharides, and mechanical injuries. TF activity may be affected by phosphorylation of its cytoplasmic domain and alternative splicing. TF acts as the primary initiator of physiological hemostasis, which prevents local bleeding at the injury site. However, aberrant expression of TF, accompanied by the severity of diseases and infections under various pathological conditions, triggers multiple signaling pathways that support thrombosis, angiogenesis, inflammation, and metastasis. Protease-activated receptors (PARs) are central in the downstream signaling pathways of TF. In this study, we have reviewed the TF signaling pathways in different pathological conditions, such as wound injury, asthma, cardiovascular diseases (CVDs), viral infections, cancer and pathological angiogenesis. Angiogenic activities of TF are critical in the repair of wound injuries and aggressive behavior of tumors, which are mainly performed by the actions of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1 (HIF1-α). Pro-inflammatory effects of TF have been reported in asthma, CVDs and viral infections, including COVID-19, which result in tissue hypertrophy, inflammation, and thrombosis. TF-FVII induces angiogenesis via clotting-dependent and -independent mechanisms. Clottingdependent angiogenesis is induced via the generation of thrombin and cross-linked fibrin network, which facilitate vessel infiltration and also act as a reservoir for endothelial cells (ECs) growth factors. Expression of TF in tumor cells and ECs triggers clotting-independent angiogenesis through induction of VEGF, urokinase-type plasminogen activator (uPAR), early growth response 1 (EGR1), IL8, and cysteine-rich angiogenic inducer 61 (Cyr61).
Collapse
Affiliation(s)
- Zahra Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Khajeh
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Ahmadi SE, Shabannezhad A, Kahrizi A, Akbar A, Safdari SM, Hoseinnezhad T, Zahedi M, Sadeghi S, Mojarrad MG, Safa M. Tissue factor (coagulation factor III): a potential double-edge molecule to be targeted and re-targeted toward cancer. Biomark Res 2023; 11:60. [PMID: 37280670 DOI: 10.1186/s40364-023-00504-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/19/2023] [Indexed: 06/08/2023] Open
Abstract
Tissue factor (TF) is a protein that plays a critical role in blood clotting, but recent research has also shown its involvement in cancer development and progression. Herein, we provide an overview of the structure of TF and its involvement in signaling pathways that promote cancer cell proliferation and survival, such as the PI3K/AKT and MAPK pathways. TF overexpression is associated with increased tumor aggressiveness and poor prognosis in various cancers. The review also explores TF's role in promoting cancer cell metastasis, angiogenesis, and venous thromboembolism (VTE). Of note, various TF-targeted therapies, including monoclonal antibodies, small molecule inhibitors, and immunotherapies have been developed, and preclinical and clinical studies demonstrating the efficacy of these therapies in various cancer types are now being evaluated. The potential for re-targeting TF toward cancer cells using TF-conjugated nanoparticles, which have shown promising results in preclinical studies is another intriguing approach in the path of cancer treatment. Although there are still many challenges, TF could possibly be a potential molecule to be used for further cancer therapy as some TF-targeted therapies like Seagen and Genmab's tisotumab vedotin have gained FDA approval for treatment of cervical cancer. Overall, based on the overviewed studies, this review article provides an in-depth overview of the crucial role that TF plays in cancer development and progression, and emphasizes the potential of TF-targeted and re-targeted therapies as potential approaches for the treatment of cancer.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Departments of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ashkan Shabannezhad
- Departments of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Kahrizi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Armin Akbar
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mehrab Safdari
- Departments of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Taraneh Hoseinnezhad
- Department of Hematolog, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soroush Sadeghi
- Faculty of Science, Engineering and Computing, Kingston University, London, UK
| | - Mahsa Golizadeh Mojarrad
- Shahid Beheshti Educational and Medical Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Majid Safa
- Departments of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Ghalehbandi S, Yuzugulen J, Pranjol MZI, Pourgholami MH. The role of VEGF in cancer-induced angiogenesis and research progress of drugs targeting VEGF. Eur J Pharmacol 2023; 949:175586. [PMID: 36906141 DOI: 10.1016/j.ejphar.2023.175586] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/16/2023] [Accepted: 02/08/2023] [Indexed: 03/11/2023]
Abstract
Angiogenesis is a double-edged sword; it is a mechanism that defines the boundary between health and disease. In spite of its central role in physiological homeostasis, it provides the oxygen and nutrition needed by tumor cells to proceed from dormancy if pro-angiogenic factors tip the balance in favor of tumor angiogenesis. Among pro-angiogenic factors, vascular endothelial growth factor (VEGF) is a prominent target in therapeutic methods due to its strategic involvement in the formation of anomalous tumor vasculature. In addition, VEGF exhibits immune-regulatory properties which suppress immune cell antitumor activity. VEGF signaling through its receptors is an integral part of tumoral angiogenic approaches. A wide variety of medicines have been designed to target the ligands and receptors of this pro-angiogenic superfamily. Herein, we summarize the direct and indirect molecular mechanisms of VEGF to demonstrate its versatile role in the context of cancer angiogenesis and current transformative VEGF-targeted strategies interfering with tumor growth.
Collapse
Affiliation(s)
| | - Jale Yuzugulen
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus via Mersin 10, Turkey
| | | | | |
Collapse
|
4
|
D’Andrea G, Margaglione M. Rare Defects: Looking at the Dark Face of the Thrombosis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18179146. [PMID: 34501736 PMCID: PMC8430787 DOI: 10.3390/ijerph18179146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/16/2022]
Abstract
Venous thromboembolism (VTE) constitutes a serious and potentially fatal disease, often complicated by pulmonary embolism and is associated with inherited or acquired factors risk. A series of risk factors are known to predispose to venous thrombosis, and these include mutations in the genes that encode anticoagulant proteins as antithrombin, protein C and protein S, and variants in genes that encode instead pro-coagulant factors as factor V (FV Leiden) and factor II (FII G20210A). However, the molecular causes responsible for thrombotic events in some individuals with evident inherited thrombosis remain unknown. An improved knowledge of risk factors, as well as a clear understanding of their role in the pathophysiology of VTE, are crucial to achieve a better identification of patients at higher risk. Moreover, the identification of genes with rare variants but a large effect size may pave the way for studies addressing new antithrombotic agents in order to improve the management of VTE patients. Over the past 20 years, qualitative or quantitative genetic risk factors such as inhibitor proteins of the hemostasis and of the fibrinolytic system, including fibrinogen, thrombomodulin, plasminogen activator inhibitor-1, and elevated concentrations of factors II, FV, VIII, IX, XI, have been associated with thrombotic events, often with conflicting results. The aim of this review is to evaluate available data in literature on these genetic variations to give a contribution to our understanding of the complex molecular mechanisms involved in physiologic and pathophysiologic clot formation and their role in clinical practice.
Collapse
|
5
|
Association between short-term heart rate variability and blood coagulation in patients with breast cancer. Sci Rep 2021; 11:15414. [PMID: 34326419 PMCID: PMC8322388 DOI: 10.1038/s41598-021-94931-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/19/2021] [Indexed: 11/30/2022] Open
Abstract
The purpose of this study was to investigate the relationship between heart rate variability (HRV), a non-invasive tool for evaluating autonomic function, and routine coagulation indices (RCIs) in patients with breast cancer (BC). Forty-six BC patients were enrolled in this study. Blood biochemistry tests were performed to extract RCIs, including prothrombin time (PT), activated partial thromboplastin time (APTT), and thrombin time (TT). Five-minute electrocardiograms were collected for analysis of HRV parameters (SDNN, RMSSD, LF, HF, LF n.u., HF n.u., LF/HF). Multiple linear regression models examined the relationship of HRV parameters with RCIs. RMSSD, LF n.u., HF n.u., LF/HF were significantly associated with PT. Specifically, the value of PT increased by 0.192 ± 0.091 or 0.231 ± 0.088 s, respectively for each 1 standard deviation (SD) increase in RMSSD or HF n.u.; it increased by 0.230 ± 0.088 or 0.215 ± 0.088 s, respectively for each 1 − SD decrease in LF n.u. or ln (LF/HF) (all P < 0.05). RMSSD was significantly associated with APTT, i.e., the value of APTT increased by 1.032 ± 0.470 s for each 1 − SD increase in RMSSD (P < 0.05). HRV parameters were associated with RCIs in patients with BC. These observations suggest that the autonomic nervous system and coagulation indices in BC patients are linked, potentially explaining the reason that they are both associated with the prognosis.
Collapse
|
6
|
Gomez S, Tsung A, Hu Z. Current Targets and Bioconjugation Strategies in Photodynamic Diagnosis and Therapy of Cancer. Molecules 2020; 25:E4964. [PMID: 33121022 PMCID: PMC7662882 DOI: 10.3390/molecules25214964] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/18/2020] [Accepted: 10/25/2020] [Indexed: 02/07/2023] Open
Abstract
Photodynamic diagnosis (PDD) and therapy (PDT) are emerging, non/minimally invasive techniques for cancer diagnosis and treatment. Both techniques require a photosensitizer and light to visualize or destroy cancer cells. However, a limitation of conventional, non-targeted PDT is poor selectivity, causing side effects. The bioconjugation of a photosensitizer to a tumor-targeting molecule, such as an antibody or a ligand peptide, is a way to improve selectivity. The bioconjugation strategy can generate a tumor-targeting photosensitizer conjugate specific for cancer cells, or ideally, for multiple tumor compartments to improve selectivity and efficacy, such as cancer stem cells and tumor neovasculature within the tumor microenvironment. If successful, such targeted photosensitizer conjugates can also be used for specific visualization and detection of cancer cells and/or tumor angiogenesis (an early event in tumorigenesis) with the hope of an early diagnosis of cancer. The purpose of this review is to summarize some current promising target molecules, e.g., tissue factor (also known as CD142), and the currently used bioconjugation strategies in PDT and PDD, with a focus on newly developed protein photosensitizers. These are genetically engineered photosensitizers, with the possibility of generating a fusion protein photosensitizer by recombinant DNA technology for both PDT and PDD without the need of chemical conjugation. We believe that providing an overview of promising targets and bioconjugation strategies will aid in driving research in this field forward towards more effective, less toxic, and non- or minimally invasive treatment and diagnosis options for cancer patients.
Collapse
Affiliation(s)
- Salvador Gomez
- The James-Comprehensive Cancer Center, Division of Surgical Oncology Department of Surgery, College of Medicine, The Ohio State University, 460 W 12th Ave, Columbus, OH 43210, USA; (S.G.); (A.T.)
- College of Medicine, The Ohio State University, 370 W 9th Ave, Columbus, OH 43210, USA
| | - Allan Tsung
- The James-Comprehensive Cancer Center, Division of Surgical Oncology Department of Surgery, College of Medicine, The Ohio State University, 460 W 12th Ave, Columbus, OH 43210, USA; (S.G.); (A.T.)
| | - Zhiwei Hu
- The James-Comprehensive Cancer Center, Division of Surgical Oncology Department of Surgery, College of Medicine, The Ohio State University, 460 W 12th Ave, Columbus, OH 43210, USA; (S.G.); (A.T.)
| |
Collapse
|
7
|
Rytelewski M, Harutyunyan K, Baran N, Mallampati S, Zal MA, Cavazos A, Butler JM, Konoplev S, El Khatib M, Plunkett S, Marszalek JR, Andreeff M, Zal T, Konopleva M. Inhibition of Oxidative Phosphorylation Reverses Bone Marrow Hypoxia Visualized in Imageable Syngeneic B-ALL Mouse Model. Front Oncol 2020; 10:991. [PMID: 32695673 PMCID: PMC7339962 DOI: 10.3389/fonc.2020.00991] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Abnormally low level of interstitial oxygen, or hypoxia, is a hallmark of tumor microenvironment and a known promoter of cancer chemoresistance. Inside a solid tumor mass, the hypoxia stems largely from inadequate supply of oxygenated blood through sparse or misshapen tumor vasculature whilst oxygen utilization rates are low in typical tumor's glycolytic metabolism. In acute leukemias, however, markers of intracellular hypoxia such as increased pimonidazole adduct staining and HIF-1α stabilization are observed in advanced leukemic bone marrows (BM) despite an increase in BM vasculogenesis. We utilized intravital fast scanning two-photon phosphorescence lifetime imaging microscopy (FaST-PLIM) in a BCR-ABL B-ALL mouse model to image the extracellular oxygen concentrations (pO2) in leukemic BM, and we related the extracellular oxygen levels to intracellular hypoxia, vascular markers and local leukemia burden. We observed a transient increase in BM pO2 in initial disease stages with intermediate leukemia BM burden, which correlated with an expansion of blood-carrying vascular network in the BM. Yet, we also observed increased formation of intracellular pimonidazole adducts in leukemic BM at the same time. This intermediate stage was followed by a significant decrease of extracellular pO2 and further increase of intracellular hypoxia as leukemia cellularity overwhelmed BM in disease end-stage. Remarkably, treatment of leukemic mice with IACS-010759, a pharmacological inhibitor of mitochondrial Complex I, substantially increased pO2 in the BM with advanced B-ALL, and it alleviated intracellular hypoxia reported by pimonidazole staining. High rates of oxygen consumption by B-ALL cells were confirmed by Seahorse assay including in ex vivo cells. Our results suggest that B-ALL expansion in BM is associated with intense oxidative phosphorylation (OxPhos) leading to the onset of metabolic BM hypoxia despite increased BM vascularization. Targeting mitochondrial respiration may be a novel approach to counteract BM hypoxia in B-ALL and, possibly, tumor hypoxia in other OxPhos-reliant malignancies.
Collapse
Affiliation(s)
- Mateusz Rytelewski
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Karine Harutyunyan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Saradhi Mallampati
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - M Anna Zal
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Antonio Cavazos
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jason M Butler
- Weill Cornell Medicine, Medical School of Biological Sciences, Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ, United States
| | - Sergej Konoplev
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mirna El Khatib
- Department of Biochemistry and Biophysics, The University of Pennsylvania, Philadelphia, PA, United States
| | - Shane Plunkett
- Department of Biochemistry and Biophysics, The University of Pennsylvania, Philadelphia, PA, United States
| | - Joseph R Marszalek
- TRACTION, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Tomasz Zal
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
8
|
Gangaraju R, Song J, Kim SJ, Tashi T, Reeves BN, Sundar KM, Thiagarajan P, Prchal JT. Thrombotic, inflammatory, and HIF-regulated genes and thrombosis risk in polycythemia vera and essential thrombocythemia. Blood Adv 2020; 4:1115-1130. [PMID: 32203583 PMCID: PMC7094018 DOI: 10.1182/bloodadvances.2019001379] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/19/2020] [Indexed: 02/07/2023] Open
Abstract
Thrombosis is a major cause of morbidity and mortality in polycythemia vera (PV) and essential thrombocythemia (ET). The pathophysiology of thrombosis in these disorders remains unclear, and we hypothesized that upregulation of thrombotic, inflammatory, and hypoxia-inducible factor (HIF)-regulated genes may play a role in it. We performed unbiased RNA sequencing in granulocytes and platelets of PV patients and found differential expression of several thrombotic, inflammatory, and HIF-regulated genes. The expression of many of these genes positively correlated with JAK2 expression and JAK2V617F allelic burden. We then validated these findings by quantitative polymerase chain reaction analyses of selected gene transcripts in a larger number of PV and ET granulocytes and platelets (58 patients) and in 28 controls, and we compared these findings in patients with and without thrombosis. The study included 29 females and 29 males; of these, 28 had a history of thrombosis. We found that transcripts of several selected genes were upregulated in patients with PV or ET compared with controls. In granulocytes, the expression levels of F3, SELP, VEGFA, and SLC2A1 were significantly higher in patients with a history of thrombosis compared with those who did not have thrombosis. Patients with a history of thrombosis have significantly higher expression of IL1RAP (P < .05) in platelets compared with those without thrombosis. Our study confirms the presence of a thrombo-inflammatory state and augmented HIF activity in PV and ET and its role in thrombosis. These data may provide the background for targeted therapies in PV and ET.
Collapse
Affiliation(s)
- Radhika Gangaraju
- Division of Hematology-Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Jihyun Song
- Division of Hematology, School of Medicine, University of Utah, Salt Lake City, UT
| | - Soo Jin Kim
- Division of Hematology, School of Medicine, University of Utah, Salt Lake City, UT
| | - Tsewang Tashi
- Division of Hematology, School of Medicine, University of Utah, Salt Lake City, UT
| | - Brandi N Reeves
- Division of Hematology-Oncology, University of North Carolina, Chapel Hill, NC
| | - Krishna M Sundar
- Division of Pulmonology, Critical Care and Sleep Medicine, School of Medicine, University of Utah, Salt Lake City, UT
| | - Perumal Thiagarajan
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine and Pathology, Baylor College of Medicine, Houston, TX; and
| | - Josef T Prchal
- Division of Hematology, School of Medicine, University of Utah, Salt Lake City, UT
- Veterans Administration Medical Center, Salt Lake City, UT
| |
Collapse
|
9
|
Hu Z. Tissue factor as a new target for CAR-NK cell immunotherapy of triple-negative breast cancer. Sci Rep 2020; 10:2815. [PMID: 32071339 PMCID: PMC7028910 DOI: 10.1038/s41598-020-59736-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 02/03/2020] [Indexed: 02/06/2023] Open
Abstract
Triple-negative breast cancer (TNBC), representing ~15% of globally diagnosed breast cancer, is typically an incurable malignancy due to the lack of targetable surface targets for development of effective therapy. To address the unmet need for TNBC treatment, we recently determined that tissue factor (TF) is a useful surface target in 50–85% of patients with TNBC and developed a second-generation TF-targeting antibody-like immunoconjugate (called L-ICON) for preclinical treatment of TNBC. Using the chimeric antigen receptor (CAR) approach, here we develop and test TF-targeting CAR-engineered natural killer (TF-CAR-NK) cells that co-express CD16, the Fc receptor (FcγIII) to mediate antibody-dependent cellular toxicity (ADCC), for a preclinical assessment of immunotherapy of TNBC using TF-CAR-NK cell as single agent therapy and in combination with L-ICON. Our preclinical results demonstrate that TF-CAR-NK cells alone could kill TNBC cells and its efficacy was enhanced with L-ICON ADCC in vitro. Moreover, TF-CAR-NK cells were effective in vivo for the treatment of TNBC in cell line- and patient’s tumor-derived xenograft mouse models. Thus, this study established the proof of concept of targeting TF as a new target in CAR-NK immunotherapy for effective treatment of TNBC and may warrant further preclinical study and potentially future investigation in TNBC patients.
Collapse
Affiliation(s)
- Zhiwei Hu
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and The OSU James Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| |
Collapse
|
10
|
Gadomska G, Ziołkowska K, Boinska J, Filipiak J, Rość D. Activation of TF-Dependent Blood Coagulation Pathway and VEGF-A in Patients with Essential Thrombocythemia. ACTA ACUST UNITED AC 2019; 55:medicina55020054. [PMID: 30781507 PMCID: PMC6409549 DOI: 10.3390/medicina55020054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 01/05/2023]
Abstract
Background and objectives: Recent studies suggest that a vascular endothelial growth factor (VEGF-A) may be involved in the thrombotic process by stimulating the expression of tissue factor in vascular endothelial cells. Tissue factor (TF) can also stimulate the transcription of the gene encoding VEGF-A. The relationship between coagulation and angiogenesis in myeloproliferative neoplasms is not fully understood. The aim of this study was to evaluate the concentration of TF in relation to VEGF-A in the blood of patients with essential thrombocythemia (ET). Patients and methods: The study group consisted of 130, newly diagnosed patients with ET (mean age 61 years). The control group consisted of 35 healthy volunteers (mean age 51 years). Concentrations of VEGF-A, TF, and tissue factor pathway inhibitor (TFPI) were analysed using immunoenzymatic methods. TF and TFPI activities were performed using chromogenic assays. Results: The median concentration of TF Ag was 3-fold higher and the TF activity was more than 15-fold higher in ET patients than in normal individuals. There were no statistically significant differences in the TFPI concentration and activity between groups. VEGF-A was significantly increased in patients with ET (p < 0.000001). Analysis of correlations revealed a positive correlation between VEGF-A and TF Ag as well as a positive correlation between VEGF-A and TFPI activity. Conclusions: The simultaneous increase of TF concentration and activity, VEGF-A in the blood of patients with ET, as well as a positive correlation between the concentration of TF and VEGF-A demonstrates the coexistence of TF-dependent coagulation and activation of angiogenesis.
Collapse
Affiliation(s)
- Grażyna Gadomska
- Department of Hematology and Malignant Diseases of Hematopoietic System, Faculty of Medicine, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-168 Bydgoszcz, Poland.
| | - Katarzyna Ziołkowska
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland.
| | - Joanna Boinska
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland.
| | - Jan Filipiak
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland.
| | - Danuta Rość
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland.
| |
Collapse
|
11
|
Hu Z, Shen R, Campbell A, McMichael E, Yu L, Ramaswamy B, London CA, Xu T, Carson WE. Targeting Tissue Factor for Immunotherapy of Triple-Negative Breast Cancer Using a Second-Generation ICON. Cancer Immunol Res 2018; 6:671-684. [PMID: 29622581 PMCID: PMC5984705 DOI: 10.1158/2326-6066.cir-17-0343] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 01/17/2018] [Accepted: 03/27/2018] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is a leading cause of breast cancer death and is often associated with BRCA1 and BRCA2 mutation. Due to the lack of validated target molecules, no targeted therapy for TNBC is approved. Tissue factor (TF) is a common yet specific surface target receptor for cancer cells, tumor vascular endothelial cells, and cancer stem cells in several types of solid cancers, including breast cancer. Here, we report evidence supporting the idea that TF is a surface target in TNBC. We used in vitro cancer lines and in vivo tumor xenografts in mice, all with BRCA1 or BRCA2 mutations, derived from patients' tumors. We showed that TF is overexpressed on TNBC cells and tumor neovasculature in 50% to 85% of TNBC patients (n = 161) and in TNBC cell line-derived xenografts (CDX) and patient-derived xenografts (PDX) from mice, but was not detected in adjacent normal breast tissue. We then describe the development of a second-generation TF-targeting immunoconjugate (called L-ICON1, for lighter or light chain ICON) with improved efficacy and safety profiles compared with the original ICON. We showed that L-ICON1 kills TNBC cells in vitro via antibody-dependent cell-mediated cytotoxicity and can be used to treat human and murine TNBC CDX as well as PDX in vivo in orthotopic mouse models. Thus, TF could be a useful target for the development of immunotherapeutics for TNBC patients, with or without BRCA1 and BRCA2 mutations. Cancer Immunol Res; 6(6); 671-84. ©2018 AACR.
Collapse
Affiliation(s)
- Zhiwei Hu
- Department of Surgery Division of Surgical Oncology, The Ohio State University Wexner Medical Center and The OSU James Comprehensive Cancer Center, Columbus, Ohio.
| | - Rulong Shen
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Amanda Campbell
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Elizabeth McMichael
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Bhuvaneswari Ramaswamy
- Department of Medical Oncology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Cheryl A London
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Tian Xu
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut
| | - William E Carson
- Department of Surgery Division of Surgical Oncology, The Ohio State University Wexner Medical Center and The OSU James Comprehensive Cancer Center, Columbus, Ohio
| |
Collapse
|
12
|
Hu Z. Therapeutic Antibody-Like Immunoconjugates against Tissue Factor with the Potential to Treat Angiogenesis-Dependent as Well as Macrophage-Associated Human Diseases. Antibodies (Basel) 2018; 7:8. [PMID: 31105982 PMCID: PMC6519474 DOI: 10.3390/antib7010008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/18/2018] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence suggests that tissue factor (TF) is selectively expressed in pathological angiogenesis-dependent as well as macrophage-associated human diseases. Pathological angiogenesis, the formation of neovasculature, is involved in many clinically significant human diseases, notably cancer, age-related macular degeneration (AMD), endometriosis and rheumatoid arthritis (RA). Macrophage is involved in the progression of a variety of human diseases, such as atherosclerosis and viral infections (human immunodeficiency virus, HIV and Ebola). It is well documented that TF is selectively expressed on angiogenic vascular endothelial cells (VECs) in these pathological angiogenesis-dependent human diseases and on disease-associated macrophages. Under physiology condition, TF is not expressed by quiescent VECs and monocytes but is solely restricted on some cells (such as pericytes) that are located outside of blood circulation and the inner layer of blood vessel walls. Here, we summarize TF expression on angiogenic VECs, macrophages and other diseased cell types in these human diseases. In cancer, for example, the cancer cells also overexpress TF in solid cancers and leukemia. Moreover, our group recently reported that TF is also expressed by cancer-initiating stem cells (CSCs) and can serve as a novel oncotarget for eradication of CSCs without drug resistance. Furthermore, we review and discuss two generations of TF-targeting therapeutic antibody-like immunoconjugates (ICON and L-ICON1) and antibody-drug conjugates that are currently being tested in preclinical and clinical studies for the treatment of some of these human diseases. If efficacy and safety are proven in current and future clinical trials, TF-targeting immunoconjugates may provide novel therapeutic approaches with potential to broadly impact the treatment regimen of these significant angiogenesis-dependent, as well as macrophage-associated, human diseases.
Collapse
Affiliation(s)
- Zhiwei Hu
- Department of Surgery Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
13
|
Seo Y, Jung Y, Kim SH. Decellularized heart ECM hydrogel using supercritical carbon dioxide for improved angiogenesis. Acta Biomater 2018; 67:270-281. [PMID: 29223704 DOI: 10.1016/j.actbio.2017.11.046] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 11/08/2017] [Accepted: 11/28/2017] [Indexed: 12/30/2022]
Abstract
Initial angiogenesis within the first 3 days is critical for healing ischemic diseases such as myocardial infarction. Recently, decellularized extracellular matrix (dECM) has been reported to provide tissue-derived ECM components and can be used as a scaffold for cell delivery for angiogenesis in tissue engineering. Decellularization by various detergents such as sodium dodecyl sulfate (SDS) and triton X-100 can remove the cell nuclei in tissue organs. However, this leads to ECM structure denaturation, decreased presence of various ECM proteins and cytokines, and loss of mechanical properties. To overcome these limitations, in this study, we developed a supercritical carbon dioxide and ethanol co-solvent (scCO2-EtOH) decellularization method, which is a detergent-free system that prevents ECM structure disruption and retains various angiogenic proteins in the heart dECM, and tested on rat heart tissues. The heart tissue was placed into the scCO2 reactor and decellularized at 37 °C and 350 bar. After scCO2-EtOH treatment, the effects were evaluated by DNA, collagen, and glycosaminoglycan (GAG) quantification and hematoxylin and eosin and immunofluorescence staining to determine the absence of nucleic acids and preservation of heart ECM components. Similar to the native group, the scCO2-EtOH group contained more ECM components such as collagen, GAGs, collagen I, laminin, and fibronectin and angiogenic factors including vascular endothelial growth factor, fibroblast growth factor, and platelet-derived growth factor and others in comparison to the detergent group. In addition, to estimate angiogenesis of the dECM hydrogels, the neutralized dECM solution was injected in a rat subcutaneous layer (n = 6 in each group: collagen, scCO2-EOH, and detergent group), after which the solution naturally formed gelation in the subcutaneous layer. After 3 days, the gels were harvested and estimated by immunofluorescence staining and the ImageJ program for angiogenesis analysis. Consequently, blood vessel formation and density of vWF and α-SMA in the scCO2-EtOH group were significantly greater than that in the collagen group. Here we suggest that heart-derived decellularized extracellular matrix (dECM) with scCO2-EtOH treatment is a highly promising angiogenic material for healing in ischemic disease. STATEMENT OF SIGNIFICANCE Supercritical carbon dioxide (scCO2) in a supercritical phase has low viscosity and high diffusivity between gas and liquid properties and is known to be affordable, non-toxic, and eco-friendly. Therefore, scCO2 extraction technology has been extensively used in commercial and industrial fields. Recently, decellularized extracellular matrix (dECM) was applied to tissue engineering and regenerative medicine as a scaffold, therapeutic material, and bio-ink for 3D printing. Moreover, the general decellularization method using detergents has limitations including eliminating tissue-derived ECM components and disrupting their structures after decellularization. To overcome these limitations, heart tissues were treated with scCO2-EtOH for decellularization, resulting in preserving of tissue due to the various ECM and angiogenic factors derived. In addition, initiation of angiogenesis was highly induced even after 3 days of injection.
Collapse
Affiliation(s)
- Yoojin Seo
- NBIT, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Youngmee Jung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea.
| | - Soo Hyun Kim
- NBIT, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea.
| |
Collapse
|
14
|
Date K, Ettelaie C, Maraveyas A. Tissue factor-bearing microparticles and inflammation: a potential mechanism for the development of venous thromboembolism in cancer. J Thromb Haemost 2017; 15:2289-2299. [PMID: 29028284 DOI: 10.1111/jth.13871] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Indexed: 12/31/2022]
Abstract
Cancer is associated with an increased risk of venous thromboembolism (VTE); the exact mechanisms for the induction of VTE remain to be fully elucidated, but it is widely acknowledged that tissue factor (TF)-bearing microparticles (TF-MPs) may play a significant role. However, TF-MPs have yet to be accepted as a genuine biomarker for cancer-associated VTE, as the presence of elevated TF-MP levels is not always accompanied by thrombosis; interestingly, in certain cases, particularly in pancreatic cancer, VTE seems to be more likely in the context of acute inflammation. Although several potential mechanisms for the development of VTE in cancer have been postulated, this review explores the homeostatic disruption of TF-MPs, as the main reservoir of bloodborne TF, in the context of cancer and inflammation, and considers the abrogated responses of the activated endothelium and mononuclear phagocyte system in mediating this disruption.
Collapse
Affiliation(s)
- K Date
- Hull York Medical School, University of Hull, Hull, UK
| | - C Ettelaie
- School of Life Sciences, University of Hull, Hull, UK
| | - A Maraveyas
- Hull York Medical School, University of Hull, Hull, UK
- Queen's Centre for Oncology and Haematology, Castle Hill Hospital, Cottingham, UK
| |
Collapse
|
15
|
Kotla S, Singh NK, Kirchhofer D, Rao GN. Heterodimers of the transcriptional factors NFATc3 and FosB mediate tissue factor expression for 15( S)-hydroxyeicosatetraenoic acid-induced monocyte trafficking. J Biol Chem 2017; 292:14885-14901. [PMID: 28724635 PMCID: PMC5592668 DOI: 10.1074/jbc.m117.804344] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
Tissue factor (TF) is expressed in vascular and nonvascular tissues and functions in several pathways, including embryonic development, inflammation, and cell migration. Many risk factors for atherosclerosis, including hypertension, diabetes, obesity, and smoking, increase TF expression. To better understand the TF-related mechanisms in atherosclerosis, here we investigated the role of 12/15-lipoxygenase (12/15-LOX) in TF expression. 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE), the major product of human 15-LOXs 1 and 2, induced TF expression and activity in a time-dependent manner in the human monocytic cell line THP1. Moreover, TF suppression with neutralizing antibodies blocked 15(S)-HETE-induced monocyte migration. We also found that NADPH- and xanthine oxidase-dependent reactive oxygen species (ROS) production, calcium/calmodulin-dependent protein kinase IV (CaMKIV) activation, and interactions between nuclear factor of activated T cells 3 (NFATc3) and FosB proto-oncogene, AP-1 transcription factor subunit (FosB) are involved in 15(S)-HETE-induced TF expression. Interestingly, NFATc3 first induced the expression of its interaction partner FosB before forming the heterodimeric NFATc3-FosB transcription factor complex, which bound the proximal AP-1 site in the TF gene promoter and activated TF expression. We also observed that macrophages from 12/15-LOX-/- mice exhibit diminished migratory response to monocyte chemotactic protein 1 (MCP-1) and lipopolysaccharide compared with WT mouse macrophages. Similarly, compared with WT macrophages, monocytes from 12/15-LOX-/- mice displayed diminished trafficking, which was rescued by prior treatment with 12(S)-HETE, in a peritonitis model. These observations indicate that 15(S)-HETE-induced monocyte/macrophage migration and trafficking require ROS-mediated CaMKIV activation leading to formation of NFATc3 and FosB heterodimer, which binds and activates the TF promoter.
Collapse
Affiliation(s)
- Sivareddy Kotla
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Nikhlesh K Singh
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, California 94080
| | - Gadiparthi N Rao
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| |
Collapse
|
16
|
Arderiu G, Espinosa S, Peña E, Crespo J, Aledo R, Bogdanov VY, Badimon L. Tissue factor variants induce monocyte transformation and transdifferentiation into endothelial cell-like cells. J Thromb Haemost 2017; 15:1689-1703. [PMID: 28585414 DOI: 10.1111/jth.13751] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Indexed: 11/29/2022]
Abstract
Essentials Monocytes (Mo) transdifferentiate into endothelial cell-like (ECL) cells. Mo induce tissue factor (TF) expression and secretion in microvascular endothelial cells (mECs). TF interacts with Mo in a paracrine fashion, inducing their transdifferentiation into ECL cells. TF generates a positive feedback crosstalk between Mo and mECs that promotes angiogenesis. SUMMARY Background Monocytes (Mo) increase neovascularization by releasing proangiogenic mediators and/or transdifferentiating into endothelial cell-like (ECL) cells. Recently, we have reported that Mo-microvascular endothelial cells (mECs) crosstalk induces mEC-tissue factor (TF) expression and promotes angiogenesis. However, the effect of TF on Mo remains unknown. Objective Here, we analyzed whether TF might exert angiogenic effects by inducing transdifferentiation of Mo. Methods Full-length TF (flTF) and alternatively spliced TF (asTF) were overexpressed in mECs, and their supernatants were added to Mo cultures. CD16 positivity and expression of vascular endothelial cell (VEC) markers in Mo were analyzed by fluorescence activated cell sorting. The capacity to form tube-like structures were visualized in three-dimensional cultures. Results In mECs flTF and asTF expression and release were increased in cultures with Mo-conditioned media. TF variants induced expansion of a CD16+ Mo subset and Mo transdifferentiation into ECL-cells expressing VEC markers that can form new microvessels. CD16+ Mo exposed to TF showed an increased expression of VE-cadherin, von Willebrand factor (VWF) and eNOS. Mo cultured with supernatants obtained from TF-silenced mECs did not transdifferentiate to ECL-cells or expressed VEC markers. Blocking β1-integrin in Mo significantly blocked the effects of the TF variants. Conclusions Mo induce mECs to express and release TF, which drives CD16- Mo to transform into CD16+ Mo and to transdifferentiate into ECL-cells that can form new microvessels. Our results reveal a TF-mediated positive feedback between mECs and Mo that stimulates Mo differentiation and induces angiogenesis.
Collapse
Affiliation(s)
- G Arderiu
- Cardiovascular Science Institute-ICCC, Hospital de Sant Pau (UAB) and IIB-Sant Pau, Barcelona, Spain
| | - S Espinosa
- Cardiovascular Science Institute-ICCC, Hospital de Sant Pau (UAB) and IIB-Sant Pau, Barcelona, Spain
| | - E Peña
- Cardiovascular Science Institute-ICCC, Hospital de Sant Pau (UAB) and IIB-Sant Pau, Barcelona, Spain
- Ciber CV, Instituto Carlos III, Madrid, Spain
| | - J Crespo
- Cardiovascular Science Institute-ICCC, Hospital de Sant Pau (UAB) and IIB-Sant Pau, Barcelona, Spain
| | - R Aledo
- Cardiovascular Science Institute-ICCC, Hospital de Sant Pau (UAB) and IIB-Sant Pau, Barcelona, Spain
| | - V Y Bogdanov
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - L Badimon
- Cardiovascular Science Institute-ICCC, Hospital de Sant Pau (UAB) and IIB-Sant Pau, Barcelona, Spain
- Ciber CV, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
17
|
Leiba M, Malkiel S, Budnik I, Rozic G, Avigdor A, Duek A, Nagler A, Kenet G, Livnat T. Thrombin generation as a predictor of thromboembolic events in multiple myeloma patients. Blood Cells Mol Dis 2017; 65:1-7. [PMID: 28365523 DOI: 10.1016/j.bcmd.2017.03.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 03/21/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND Multiple myeloma (MM) is characterized by an increased incidence of thromboembolic events, especially when immunomodulatory drugs are used. Currently, our ability to predict these thrombotic events is limited. We hypothesized that global coagulation tests may be predictive of thrombotic events in MM patients. METHODS Blood samples were taken from 36 MM patients before and during routine treatment. Thrombin generation (TG) tests including endogenous thrombin potential (ETP) and peak height were analyzed. RESULTS Patients were followed for a median of 2.5years. Those who developed thrombotic events were characterized by significantly higher ETP and peak height values compared to those who did not (P=0.001). In these patients, we identified a gradual increase in TG parameters that preceded the thrombotic event. Anticoagulation therapy was associated with a significant decrease in ETP and peak height values (P<0.001). There was no statistically significant difference in TG parameters between newly diagnosed MM patients and healthy subjects, as well as between MM patients prior to and during chemotherapy. CONCLUSIONS TG tests might predict thrombotic events in MM patients. Thus, TG tests may be incorporated into decision-making protocols of prophylactic anticoagulant therapy in MM patients.
Collapse
Affiliation(s)
- Merav Leiba
- Division of Hematology and Bone Marrow Transplantation, Cancer Research Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sarah Malkiel
- Division of Pediatrics B, Edmond and Lily Safra Children's Hospital, The Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Ivan Budnik
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Gabriela Rozic
- Division of Hematology and Bone Marrow Transplantation, Cancer Research Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Abraham Avigdor
- Division of Hematology and Bone Marrow Transplantation, Cancer Research Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adrian Duek
- Division of Hematology and Bone Marrow Transplantation, Cancer Research Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arnon Nagler
- Division of Hematology and Bone Marrow Transplantation, Cancer Research Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gili Kenet
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; National Hemophilia Center and Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer, Israel
| | - Tami Livnat
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; National Hemophilia Center and Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer, Israel.
| |
Collapse
|
18
|
Barrett A, Kevane B, Hall P, Ní Áinle F, Breslin T. Screening for Malignancy in Patients With Unprovoked Venous Thromboembolism: A Single-Center Retrospective Case Series. Clin Appl Thromb Hemost 2016; 23:735-739. [PMID: 27443696 DOI: 10.1177/1076029616659694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In this prospective review of cancer screening in unselected patients with unprovoked venous thromboembolism (VTE) presenting to a large teaching hospital in the Republic of Ireland, we aimed to determine the effects of the implementation of the National Institute for Health and Care Excellence screening policy in a "real-world" population. Within our institution, 64 individuals presented with unprovoked VTE during the study period, of whom 47 underwent a screening computed tomography (CT) scan. Two cases of previously undiagnosed cancer were identified. However, in both cases, the clinical history provided by the affected individuals would have prompted a CT scan regardless of the recommendations of the screening policy. The screening CT scans identified 18 incidental lesions within the cohort, which required further diagnostic studies. None of the additional investigations completed to date have detected any lesion of clinical significance. These findings support the view that cancer screening with CT imaging in unselected individuals with unprovoked VTE is not justified or cost-effective.
Collapse
Affiliation(s)
- Aisling Barrett
- 1 Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Barry Kevane
- 2 Department of Haematology, Rotunda Hospital, Dublin, Ireland
| | - Patricia Hall
- 3 School of Medicine, University College Dublin (UCD), Dublin, Ireland
| | - Fionnuala Ní Áinle
- 1 Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland.,2 Department of Haematology, Rotunda Hospital, Dublin, Ireland.,4 UCD Conway Institute SPHERE Research Group, UCD, Dublin, Ireland.,5 Irish Heart Foundation, Dublin, Ireland
| | - Tomás Breslin
- 6 Department of Emergency Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| |
Collapse
|
19
|
Crowley MP, Kevane B, O’Shea SI, Quinn S, Egan K, Gilligan OM, Ní. Áinle F. Plasma Thrombin Generation and Sensitivity to Activated Protein C Among Patients With Myeloma and Monoclonal Gammopathy of Undetermined Significance. Clin Appl Thromb Hemost 2016; 22:554-62. [DOI: 10.1177/1076029615625825] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The etiology of the prothrombotic state in myeloma has yet to be definitively characterized. Similarly, while recent evidence suggests that patients with monoclonal gammopathy of undetermined significance (MGUS) may also be at increased risk of thrombosis, the magnitude and the etiology of this risk have also yet to be defined. The present study aims to characterize patterns of plasma thrombin generation and sensitivity to the anticoagulant activity of activated protein C (APC) at the time of initial diagnosis of myeloma and in response to therapy in comparison to that observed among patients with MGUS and matched, healthy volunteers. Patients presenting with newly diagnosed/newly relapsed myeloma (n = 8), MGUS (n = 8), and matched healthy volunteers (n = 8) were recruited. Plasma thrombin generation was determined by calibrated automated thrombography. Peak thrombin generation was significantly higher in patients with myeloma (383.4 ± 33.4 nmol/L) and MGUS (353.4 ± 16.5 nmol/L) compared to healthy volunteers (276.7 ± 20.8 nmol/L; P < .05). In the presence of APC, endogenous thrombin potential was significantly lower in control plasma (228.6 ± 44.5 nmol/L × min) than in either myeloma (866.2 ± 241.3 nmol/L × min, P = .01) or MGUS plasma (627 ± 91.5 nmol/L × min, P = .003). Within the myeloma cohort, peak thrombin generation was significantly higher at diagnosis (353.2 ± 15.9 nmol/L) than following completion of the third cycle of therapy (282.1 ± 15.2 nmol/L; P < .005). Moreover, sensitivity to APC increased progressively with each cycle of chemotherapy. Further study of the etiology and evolving patterns of hypercoagulability among patients with these conditions is warranted and may have future implications for thromboprophylaxis strategies.
Collapse
Affiliation(s)
- Maeve P. Crowley
- Department of Haematology, Cork University Hospital, Cork, Ireland
- Clinical Research Facility, University College Cork, Ireland
- Maeve P. Crowley and Barry Kevane are joint first authors
| | - Barry Kevane
- Department of Haematology, Rotunda Hospital, Dublin, Ireland
- School of Medicine & Medical Science, University College Dublin, Dublin, Ireland
- SPHERE Research Group, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
- Maeve P. Crowley and Barry Kevane are joint first authors
| | - Susan I. O’Shea
- Department of Haematology, Cork University Hospital, Cork, Ireland
| | - Shane Quinn
- Department of Haematology, Cork University Hospital, Cork, Ireland
| | - Karl Egan
- School of Medicine & Medical Science, University College Dublin, Dublin, Ireland
- SPHERE Research Group, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | | | - Fionnuala Ní. Áinle
- Department of Haematology, Rotunda Hospital, Dublin, Ireland
- School of Medicine & Medical Science, University College Dublin, Dublin, Ireland
- SPHERE Research Group, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| |
Collapse
|
20
|
Dicke C, Amirkhosravi A, Spath B, Jiménez-Alcázar M, Fuchs T, Davila M, Francis JL, Bokemeyer C, Langer F. Tissue factor-dependent and -independent pathways of systemic coagulation activation in acute myeloid leukemia: a single-center cohort study. Exp Hematol Oncol 2015; 4:22. [PMID: 26251762 PMCID: PMC4527228 DOI: 10.1186/s40164-015-0018-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 07/29/2015] [Indexed: 11/28/2022] Open
Abstract
Background In acute myeloid leukemia (AML), disseminated intravascular coagulation (DIC) contributes to morbidity and mortality, but the underlying pathomechanisms remain incompletely understood. Methods We conducted a prospective study on 69 patients with newly diagnosed AML to further define the correlates of systemic coagulation activation in this hematological malignancy. Tissue factor procoagulant activity (TF PCA) of isolated peripheral blood mononuclear cells (PBMCs) and TF expression by circulating microparticles (MPs) were assessed by single-stage clotting and thrombin generation assay, respectively. Soluble plasma TF antigen and secretion of vascular endothelial growth factor (VEGF) by cultured PBMCs were measured by ELISA. Cell-free plasma DNA was quantified by staining with a fluorescent dye. Result TF PCA of PBMCs was significantly increased in AML patients as compared to healthy controls. Furthermore, TF PCA was significantly associated with decompensated DIC at presentation, as defined by a plasma fibrinogen level of ≤1 g/L (n = 11). In addition to TF PCA and circulating blasts, serum lactate dehydrogenase, a surrogate marker for leukemic cell turnover, correlated with plasma D-Dimer in the total patient cohort and was significantly increased in DIC patients, suggesting a role for myeloblast apoptosis/necrosis in activation of the TF-dependent coagulation pathway. Consistently, TF-bearing plasma MPs were more frequently detected and levels of soluble TF antigen were significantly higher in DIC vs. non-DIC patients. No association was found between TF PCA expression and VEGF secretion by isolated PBMCs, but significantly increased levels of cell-free plasma DNA pointed to a contribution of the intrinsic contact pathway to systemic coagulation activation in the total patient cohort and in patients with lower TF PCA expression. While PBMC-associated TF PCA had no effect on long-term survival, DIC occurrence at presentation increased the risk of early mortality. Conclusion In newly diagnosed AML, TF expression by PBMCs and shedding of TF-bearing plasma MPs are central to the pathogenesis of DIC, but additional pathways, such as DNA liberation, may contribute to systemic coagulation activation.
Collapse
Affiliation(s)
- Christina Dicke
- II. Medizinische Klinik und Poliklinik, Hubertus Wald Tumorzentrum, Universitäres Cancer Center Hamburg (UCCH), Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Ali Amirkhosravi
- Florida Hospital Center for Thrombosis Research, 2566 Lee Road, Winter Park, FL 32789 USA
| | - Brigitte Spath
- II. Medizinische Klinik und Poliklinik, Hubertus Wald Tumorzentrum, Universitäres Cancer Center Hamburg (UCCH), Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Miguel Jiménez-Alcázar
- Institut für Klinische Chemie und Laboratoriumsmedizin, Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Tobias Fuchs
- Institut für Klinische Chemie und Laboratoriumsmedizin, Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Monica Davila
- Florida Hospital Center for Thrombosis Research, 2566 Lee Road, Winter Park, FL 32789 USA
| | - John L Francis
- Florida Hospital Center for Thrombosis Research, 2566 Lee Road, Winter Park, FL 32789 USA
| | - Carsten Bokemeyer
- II. Medizinische Klinik und Poliklinik, Hubertus Wald Tumorzentrum, Universitäres Cancer Center Hamburg (UCCH), Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Florian Langer
- II. Medizinische Klinik und Poliklinik, Hubertus Wald Tumorzentrum, Universitäres Cancer Center Hamburg (UCCH), Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| |
Collapse
|
21
|
Differing coagulation profiles of patients with monoclonal gammopathy of undetermined significance and multiple myeloma. J Thromb Thrombolysis 2014; 39:245-9. [DOI: 10.1007/s11239-014-1140-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
22
|
Bäumer N, Krause A, Köhler G, Lettermann S, Evers G, Hascher A, Bäumer S, Berdel WE, Müller-Tidow C, Tickenbrock L. Proteinase-Activated Receptor 1 (PAR1) regulates leukemic stem cell functions. PLoS One 2014; 9:e94993. [PMID: 24740120 PMCID: PMC3989293 DOI: 10.1371/journal.pone.0094993] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/21/2014] [Indexed: 12/30/2022] Open
Abstract
External signals that are mediated by specific receptors determine stem cell fate. The thrombin receptor PAR1 plays an important role in haemostasis, thrombosis and vascular biology, but also in tumor biology and angiogenesis. Its expression and function in hematopoietic stem cells is largely unknown. Here, we analyzed expression and function of PAR1 in primary hematopoietic cells and their leukemic counterparts. AML patients' blast cells expressed much lower levels of PAR1 mRNA and protein than CD34+ progenitor cells. Constitutive Par1-deficiency in adult mice did not affect engraftment or stem cell potential of hematopoietic cells. To model an AML with Par1-deficiency, we retrovirally introduced the oncogene MLL-AF9 in wild type and Par1−/− hematopoietic progenitor cells. Par1-deficiency did not alter initial leukemia development. However, the loss of Par1 enhanced leukemic stem cell function in vitro and in vivo. Re-expression of PAR1 in Par1−/− leukemic stem cells delayed leukemogenesis in vivo. These data indicate that Par1 contributes to leukemic stem cell maintenance.
Collapse
Affiliation(s)
- Nicole Bäumer
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Annika Krause
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Gabriele Köhler
- Gerhard Domagk Institute for Pathology, University of Muenster, Muenster, Germany
| | - Stephanie Lettermann
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Georg Evers
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Antje Hascher
- Hochschule Hamm-Lippstadt, University of Applied Science, Hamm, Germany
| | - Sebastian Bäumer
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Wolfgang E. Berdel
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Carsten Müller-Tidow
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
- Interdisciplinary Center for Clinical Research IZKF, University of Muenster, Muenster, Germany
- Dept. of Medicine IV, Hematology and Oncology, University of Halle, Halle, Germany
- * E-mail: (CMT); (LT)
| | - Lara Tickenbrock
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
- Hochschule Hamm-Lippstadt, University of Applied Science, Hamm, Germany
- * E-mail: (CMT); (LT)
| |
Collapse
|
23
|
Crowley MP, Eustace JA, O’Shea SI, Gilligan OM. Venous Thromboembolism in Patients With Myeloma. Clin Appl Thromb Hemost 2014; 20:600-6. [DOI: 10.1177/1076029614521280] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Myeloma has a well-described association with venous thromboembolism (VTE). There are few dedicated studies investigating the incidence and risk factors. Many assessment scores have been suggested to estimate the risk of VTE in patients with cancer but these have been validated in solid organ tumors. The records of patients with myeloma attending a university hospital between January 2007 and December 2012 were reviewed to investigate the incidence of VTE and the associated risk factors. In all, 217 patients with a mean (standard deviation) age at diagnosis of 65 (12) years were included. Of 217 patients, 12% had an episode of VTE, 69% received at least 1 immunomodulatory agent, and 95% had low or intermediate risk of VTE according to the Khorana score. Venous thromboembolism was a frequent occurrence in this cohort. Patients had many risk factors for VTE but no one was predictive. As myeloma outcomes continue to improve, a dedicated prospective study is warranted to investigate the most appropriate thromboprophylaxis strategy.
Collapse
Affiliation(s)
- Maeve P. Crowley
- Clinical Research Facility, University College Cork, Cork, Ireland
- Department of Haematology, Cork University Hospital, Cork, Ireland
| | | | - Susan I. O’Shea
- Department of Haematology, Cork University Hospital, Cork Ireland
| | | |
Collapse
|
24
|
Cesarman-Maus G, Braggio E, Lome-Maldonado C, Morales-Leyte AL, Fonseca R. Absence of tissue factor is characteristic of lymphoid malignancies of both T- and B-cell origin. Thromb Res 2014; 133:606-9. [PMID: 24491425 DOI: 10.1016/j.thromres.2014.01.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/10/2014] [Accepted: 01/14/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Thrombosis is a marker of poor prognosis in individuals with solid tumors. The expression of tissue factor (TF) on the cell surface membrane of malignant cells is a pivotal molecular link between activation of coagulation, angiogenesis, metastasis, aggressive tumor behavior and poor survival. Interestingly, thrombosis is associated with shortened survival in solid, but not in lymphoid neoplasias. OBJECTIVES We sought to study whether the lack of impact of thrombosis on survival in lymphoid neoplasias could be due to a lack of tumor-derived TF expression. METHODS We analyzed TF gene (F3) expression in lymphoid (N=114), myeloid (N=49) and solid tumor (N=856) cell lines using the publicly available dataset from the Broad-Novartis Cancer Cell Line Encyclopedia (http://www.broadinstitute.org/ccle/home), and in 90 patient-derived lymphoma samples. TF protein expression was studied by immunohistochemistry (IHC). RESULTS In sharp contrast to wide F3 expression in solid tumors (74.2%), F3 was absent in all low and high grade T- and B-cell lymphomas, and in most myeloid tumors, except for select acute myeloid leukemias with monocytic component. IHC confirmed the absence of TF protein in all indolent and high-grade B-cell (0/90) and T-cell (0/20) lymphomas, and acute leukemias (0/11). CONCLUSIONS We show that TF in lymphomas does not derive from the malignant cells, since these do not express either F3 or TF protein. Therefore, it is unlikely that thrombosis in patients with lymphoid neoplasms is secondary to tumor-derived tissue factor.
Collapse
Affiliation(s)
| | - Esteban Braggio
- Department of Hematology and Oncology, Mayo Clinic in Arizona, USA
| | - Carmen Lome-Maldonado
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, México City, México
| | - Ana Lilia Morales-Leyte
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, México City, México
| | - Rafael Fonseca
- Department of Hematology and Oncology, Mayo Clinic in Arizona, USA
| |
Collapse
|
25
|
Butenas S. Comparison of natural and recombinant tissue factor proteins: new insights. Biol Chem 2013; 394:819-29. [PMID: 23412875 DOI: 10.1515/hsz-2012-0350] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 02/12/2013] [Indexed: 11/15/2022]
Abstract
Tissue factor (TF), an initiator of blood coagulation in vivo, is expressed in a variety of cells. Sufficient natural TF has been isolated to clone and express recombinant proteins ranging from full-length TF to its extracellular domain. Because of the limited availability of natural TF, recombinant proteins have been used as surrogates. Despite the differences in their post-translational modifications, it has been accepted that membrane-anchored recombinant TFs are quite similar to the natural TF. Recent studies, however, have shown that post-translational modifications play an important role in TF-triggered thrombin generation.
Collapse
Affiliation(s)
- Saulius Butenas
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA.
| |
Collapse
|
26
|
Tormoen GW, Recht O, Gruber A, Levine RL, McCarty OJT. Phosphatidylserine index as a marker of the procoagulant phenotype of acute myelogenous leukemia cells. Phys Biol 2013; 10:056010. [PMID: 24104188 DOI: 10.1088/1478-3975/10/5/056010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Patients with acute myelogenous leukemia (AML) are at risk for thrombotic complications. Risk to develop thrombosis is closely tied to leukemia subtype, and studies have shown an association between leukocytosis and thrombosis in AML M3. We evaluated the relative roles of cell count and the surface expression of tissue factor (TF) and phosphatidylserine (PS) in the procoagulant phenotype of AML cell lines. The TF-positive AML M3 cell lines, NB4 and HL60, and AML M2 cell line, AML14, exhibited both extrinsic tenase and prothrombinase activity in a purified system and promoted experimental thrombus formation. In contrast, the TF-negative AML cell line, HEL, exhibited only prothrombinase activity and did not affect the rate of occlusive thrombus formation. In plasma, NB4, HL60 and AML14 shortened clotting times in a cell-count, PS- and TF-dependent manner. Exposure of cultured NB4, HL60, and AML14 cells to the chemotherapeutic agent daunorubicin increased their extrinsic tenase activity and PS expression. Clot initiation time inversely correlated with logarithm of PS index, defined as the product of multiplying leukocyte count with cell surface PS exposure. We propose that leukemia cell PS index may serve as a biomarker for procoagulant activity.
Collapse
Affiliation(s)
- Garth W Tormoen
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | | | | | | | | |
Collapse
|
27
|
Butenas S. Tissue factor structure and function. SCIENTIFICA 2012; 2012:964862. [PMID: 24278763 PMCID: PMC3820524 DOI: 10.6064/2012/964862] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 11/19/2012] [Indexed: 06/02/2023]
Abstract
Tissue factor (TF) is an integral membrane protein that is essential to life. It is a component of the factor VIIa-TF complex enzyme and plays a primary role in both normal hemostasis and thrombosis. With a vascular injury, TF becomes exposed to blood and binds plasma factor VIIa, and the resulting complex initiates a series of enzymatic reactions leading to clot formation and vascular sealing. Many cells, both healthy, and tumor cells, produce detectable amounts of TF, especially when they are stimulated by various agents. Despite the relative simplicity and small size of TF, there are numerous contradictory reports about the synthesis and presentation of TF on blood cells and circulation in normal blood either on microparticles or as a soluble protein. Another subject of controversy is related to the structure/function of TF. It has been almost commonly accepted that cell-surface-associated TF has low (if any) activity, that is, is "encrypted" and requires specific conditions/reagents to become active, that is, "decrypted." However there is a lack of agreement related to the mechanism and processes leading to alterations in TF function. In this paper TF structure, presentation, and function, and controversies concerning these features are discussed.
Collapse
Affiliation(s)
- Saulius Butenas
- Department of Biochemistry, University of Vermont, 208 South Park Drive, Room 235A, Colchester, VT 05446, USA
| |
Collapse
|
28
|
Ettelaie C, Elkeeb AM, Maraveyas A, Collier MEW. p38α phosphorylates serine 258 within the cytoplasmic domain of tissue factor and prevents its incorporation into cell-derived microparticles. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012. [PMID: 23195225 DOI: 10.1016/j.bbamcr.2012.11.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We previously showed that the phosphorylation of Ser253 within the cytoplasmic domain of human tissue factor (TF) initiates the incorporation and release of this protein into cell-derived microparticles. Furthermore, subsequent phosphorylation of Ser258 terminates this process. However, the identity of the kinase responsible for the phosphorylation of Ser258 and mode of action of this enzyme remain unknown. In this study, p38α was identified as the proline-directed kinase capable of phosphorylating Ser258 specifically, and without any detectable activity towards Ser253. Furthermore, using synthetic peptides, it was shown that the Km for the reaction decreased by approximately 10 fold on substitution of Ser253 with phospho-Ser253. Either inhibition of p38 using SB202190 or knockdown of p38α expression in coronary artery endothelial cells overexpressing wild-type TF, resulted in decreased phosphorylation of Ser258, following activation of cells with PAR2-agonist peptide (PAR2-AP). In agreement with our previous data, inhibition of phosphorylation of this residue maintained the release of TF. Activation of PAR2 in cells transfected to overexpress TF, resulted in two separate peaks of p38 activity at approximately 40 and 120 min post-activation. Furthermore, overexpression of Ala253-substituted TF enhanced the second p38 activation peak. However, the second peak was absent in cells devoid of TF or in cells overexpressing the Asp253-substituted TF. Our data clearly identifies p38α as a kinase capable of phosphorylating Ser258 within the cytoplasmic domain of TF. Moreover, it appears that the presence of TF within the cells regulates the late activation of p38 and consequently the termination of TF release into microparticles.
Collapse
Affiliation(s)
- Camille Ettelaie
- Biomedical Section, Department of Biological Sciences, University of Hull, Hull, HU6 7RX, UK.
| | | | | | | |
Collapse
|
29
|
Song HB, Park KD, Kim JH, Kim DH, Yu YS, Kim JH. Tissue factor regulates tumor angiogenesis of retinoblastoma via the extracellular signal-regulated kinase pathway. Oncol Rep 2012; 28:2057-62. [PMID: 23007470 DOI: 10.3892/or.2012.2048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 07/25/2012] [Indexed: 11/05/2022] Open
Abstract
Retinoblastoma, a well-vascularized tumor that is dependent on a very robust angiogenic response, is the most common intraocular malignancy in children. Tissue factor (TF) is known to regulate tumor progression and in the present study we demonstrated that TF regulates tumor angiogenesis of retinoblastoma. In an orthotopic transplantation model of retinoblastoma, TF was selectively expressed in the proliferative area of retinoblastoma including tumor vessels as well as tumor cells, where TF expression was co-localized with endothelial cells of tumor vessels. TF expression progressively increased with fibroblast growth factor-2 (FGF-2)-induced proliferation of human umbilical vein endothelial cells (HUVECs), which was effectively inhibited by blockade of the TF pathway by TF pathway inhibitor (TFPI). In addition, FGF-2-induced angiogenic processes of migration and tube formation of vascular endothelial cells were also effectively suppressed by TFPI, which would be mediated by inhibition of extracellular signal-regulated kinase activation. Therefore, further to our previous report that TF is involved in tumor cell proliferation of retinoblastoma, our current data suggest that blockade of the TF pathway by TFPI could effectively inhibit tumor growth by suppressing tumor cell proliferation and tumor angiogenesis at the same time.
Collapse
Affiliation(s)
- Hyun Beom Song
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
30
|
Arderiu G, Peña E, Aledo R, Espinosa S, Badimon L. Ets-1 transcription is required in tissue factor driven microvessel formation and stabilization. Angiogenesis 2012; 15:657-69. [PMID: 22869003 DOI: 10.1007/s10456-012-9293-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 07/28/2012] [Indexed: 12/19/2022]
Abstract
Tissue factor (TF) has well-recognized roles as initiator of blood coagulation as well as an intracellular signaling receptor. TF signaling regulates gene transcription and protein translation. Recently, we have shown that TF-induced mature neovessel formation is ultimately driven by CCL2 expression. However, the signaling process induced by TF to promote microvessel formation remains to be determined. This study was designed with the objective to investigate the mechanisms involved in TF-induced neovessel formation. Here, we have identified that Ets-1 expression is a downstream effector of TF signaling. TF-siRNA induced a highly significant reduction in Ets-1 expression levels and in Ets-1/DNA binding while inducing abrogation of microvessel formation. Activation of Ets-1 rescued the effect of TF inhibition and restored microvessel formation confirming the critical role of Ets-1 in TF-induced angiogenesis. VE-cadherin expression, a key regulator of endothelial intercellular junctions, and an Ets-1 target molecule was dependent of TF-inhibition. We show that TF signals through ERK1/2 to activate Ets-1 and induce CCL2 gene expression by binding to its promoter region. We conclude that endothelial cell TF signals through ERK1/2 and Ets-1 to trigger microvessel formation.
Collapse
Affiliation(s)
- Gemma Arderiu
- Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau (UAB), IIB-Sant Pau. CiberOBN, Instituto de Salut Carlos III, C/ Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
| | | | | | | | | |
Collapse
|
31
|
Karvunidis T, Chvojka J, Lysak D, Sykora R, Krouzecky A, Radej J, Novak I, Matejovic M. Septic shock and chemotherapy-induced cytopenia: effects on microcirculation. Intensive Care Med 2012; 38:1336-44. [PMID: 22584795 DOI: 10.1007/s00134-012-2582-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 04/08/2012] [Indexed: 12/17/2022]
Abstract
PURPOSE Neutrophil and platelet activation and their interactions with endothelial cells are considered central features of sepsis-induced microcirculatory alterations. However, no study has evaluated the microvascular pattern of septic shock patients with chemotherapy-induced severe cytopenia. METHODS Demographic and hemodynamic variables together with sublingual microcirculation recording [orthogonal polarization spectral imaging enhanced by sidestream dark-field technology (OPS-SDF) videomicroscopy] were collected in four groups of subjects: septic shock (SS, N = 9), septic shock in cytopenic patients (NSS, N = 8), cytopenia without infection (NEUTR, N = 7), and healthy controls (CTRL, N = 13). Except for controls, all measurements were repeated after complete resolution of septic shock and/or neutropenia. Video files were processed using appropriate software tool and semiquantitatively evaluated [total vascular density (TVD, mm/mm(2)), perfused vessel density (PVD, mm/mm(2)), proportion of perfused vessels (PPV, %), mean flow index (MFI), and flow heterogeneity index (FHI)]. RESULTS Compared with controls, there were statistically significant microcirculatory alterations within all tested groups of patients (TVD: SS = 8.8, NSS = 8.8, NEUTR = 9.1 versus CTRL = 12.6, p < 0.001; PVD: SS = 6.3, NSS = 6.1, NEUTR = 6.9 versus CTRL = 12.5, p < 0.001; PPV: SS = 71.6, NSS = 68.9, NEUTR = 73.3 versus CTRL = 98.7, p < 0.001; MFI: SS = 2.1, NSS = 1.9, NEUTR = 2.1 versus CTRL = 3.0, p < 0.05; FHI: SS = 1.0, NSS = 0.9, NEUTR = 0.6 versus CTRL = 0.0, p < 0.001). No significant differences were detected between SS, NSS, and NEUTR groups at baseline. Incomplete restoration of microcirculatory perfusion was observed after septic shock and/or neutropenia resolution with a trend towards better recovery in MFI and FHI variables in NSS as compared with SS patients. CONCLUSIONS Microvascular derangements in septic shock did not differ between noncytopenic and cytopenic patients. Our data might suggest that profound neutropenia and thrombocytopenia do not render microcirculation more resistant to sepsis-induced microvascular alterations. The role and mechanisms of microvascular alterations associated with chemotherapy-induced cytopenia warrant further investigation.
Collapse
Affiliation(s)
- Thomas Karvunidis
- 1st Department of Medicine, Intensive Care Unit, Charles University in Prague, Medical School and Teaching Hospital in Pilsen, Alej Svobody 80, 304 60 Pilsen, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Arderiu G, Peña E, Aledo R, Juan-Babot O, Badimon L. Tissue Factor Regulates Microvessel Formation and Stabilization by Induction of Chemokine (C-C motif) Ligand 2 Expression. Arterioscler Thromb Vasc Biol 2011; 31:2607-15. [DOI: 10.1161/atvbaha.111.233536] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Tissue factor (TF) triggers arterial thrombosis. TF is also able to initiate cellular signaling mechanisms leading to angiogenesis. Because high cardiovascular risk atherosclerotic plaques show significant angiogenesis, our objective was to investigate whether TF is able to trigger and stabilize atherosclerotic plaque neovessel formation.
Methods and Results—
In this study, we showed, by real-time confocal microscopy in 3-dimensional basement membrane cocultures, that TF in human microvascular endothelial cells (HMEC-1) and in human vascular smooth muscle cells (HVSMCs) plays an important role in the formation of capillary-like networks. TF silencing in endothelial cells and smooth muscle cells inhibits the formation of tube-like structures with stable phenotype. Using an in vivo model, we observed that TF inhibition in either HMEC-1 or HVSMCs reduced their shared ability to form new capillaries. The phenotypic changes induced by TF silencing were linked to reduced chemokine (C-C motif) ligand 2 (CCL2) expression in endothelial cells. Wound healing and chemotactic assays demonstrated that TF-induced release of CCL2 stimulated HVSMC migration to HMEC-1.
Conclusion—
Endogenous TF regulates CCL2 production in endothelial cells. Secreted CCL2 mediates the angiogenic effect of TF by recruiting smooth muscle cells toward endothelial cells and facilitates the maturation of newly formed microvessels.
Collapse
Affiliation(s)
- Gemma Arderiu
- From the Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau, IIB-Sant Pau and Universitat Autonoma de Barcelona, CiberOBN-Instituto de Salud Carlos III, Barcelona, Spain
| | - Esther Peña
- From the Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau, IIB-Sant Pau and Universitat Autonoma de Barcelona, CiberOBN-Instituto de Salud Carlos III, Barcelona, Spain
| | - Rosa Aledo
- From the Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau, IIB-Sant Pau and Universitat Autonoma de Barcelona, CiberOBN-Instituto de Salud Carlos III, Barcelona, Spain
| | - Oriol Juan-Babot
- From the Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau, IIB-Sant Pau and Universitat Autonoma de Barcelona, CiberOBN-Instituto de Salud Carlos III, Barcelona, Spain
| | - Lina Badimon
- From the Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau, IIB-Sant Pau and Universitat Autonoma de Barcelona, CiberOBN-Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
33
|
Association study of selected genetic polymorphisms and occurrence of venous thromboembolism in patients with multiple myeloma who were treated with thalidomide. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2011; 11:414-20. [PMID: 21859556 DOI: 10.1016/j.clml.2011.03.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 03/04/2011] [Accepted: 03/07/2011] [Indexed: 11/23/2022]
Abstract
INTRODUCTION/BACKGROUND Venous thromboembolism (VTE), with the subsequent risk of pulmonary embolism, is a common adverse effect of thalidomide treatment in patients with multiple myeloma (MM). In our retrospective study, we analyzed candidate single-nucleotide polymorphisms (SNP), CINP (rs7011), CETP (rs289747), ALDH1A1 (rs610529), CDKN1A (rs3829963), GAN (rs2608555), vascular endothelial growth factor (rs699947), and ALDH1A1 (rs168351), previously identified in a large association study based on the hypothesis-driven candidate gene approach nominated by the International Myeloma Foundation "Bank On A Cure" (3404 SNPs). In that study, the researchers built a classification tree that enables prediction of individual risk of VTE in patients with MM. PATIENTS AND METHODS Genotypes of these SNPs were determined in an independent cohort of 111 patients with MM through TaqMan real-time polymerase chain reaction (PCR) allelic discrimination and were used for prediction of individual VTE risk. RESULTS The results of this study did not confirm the ability of this classification tree to predict VTE risk in patients with MM from the Czech Republic; of these patients, 21 (19%) developed high-grade VTE. However, in patients with VTE, we found higher frequency of the AC genotype in the CDKN1A gene (42.9% vs. 16.7%; odds ratio 3.64) in comparison with the CC genotype (P = .015). SNPs of other genes as well as age and sex of the patients had no statistically significant influence on the risk of VTE. CONCLUSION Further studies are needed to confirm the initial analysis that provided predictive information of genetic variations in patients with myeloma that may influence risk of VTE.
Collapse
|
34
|
Rodrigues CA, Ferrarotto R, Kalil Filho R, Novis YAS, Hoff PMG. Venous thromboembolism and cancer: a systematic review. J Thromb Thrombolysis 2010; 30:67-78. [PMID: 20140479 DOI: 10.1007/s11239-010-0441-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Venous thromboembolism (VTE) is a serious and potentially fatal disorder, which is often associated with a significant impact on the quality of life and on the clinical outcome of cancer patients. The pathophysiology of the association between thrombosis and cancer is complex: malignancy is associated with a baseline hypercoagulable state due to many factors including release of inflammatory cytokines, activation of the clotting system, expression of hemostatic proteins on tumor cells, inhibition of natural anticoagulants, and impaired fibrinolysis. Several risk factors, related to the patient, the disease, and the therapeutic interventions, have been identified as contributing to the occurrence of VTE. There is convincing evidence to recommend the use of heparins or fondaparinux for prevention of VTE in selected cancer patients, and, especially in some particular types of malignancies and cancer treatments. Management of VTE in patients with cancer is more challenging and bleeding complications associated with the use of anticoagulants are significantly higher in cancer patients than in those without malignancy. Important issues that need to be considered in all cases are interference with anticancer therapy, inconvenience of treatment, and impact on quality of life.
Collapse
|
35
|
Kim JE, Kim KJ, Ahn W, Han KS, Kim HK. Local Anesthetics Inhibit Tissue Factor Expression in Activated Monocytes via Inhibition of Tissue Factor mRNA Synthesis. Clin Appl Thromb Hemost 2010; 17:E4-9. [DOI: 10.1177/1076029610378500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Local anesthetics have been reported to have anticoagulant properties, but the mechanisms responsible for this action are poorly understood. Here, we evaluated the in vitro effects of 3 local anesthetics—lidocaine, ropivacaine, and bupivacaine—on the tissue factor expression by monocytes. Monocytes from peripheral blood were stimulated with lipopolysaccharide (LPS) in the presence or absence of local anesthetics. All 3 local anesthetics inhibited the expression of tissue factor antigen and tissue factor activity in LPS-stimulated monocytes in a dose- and time-dependent manner and reduced tissue factor messenger RNA (mRNA) expression in endothelial cells and a monocytic cell line. None of the 3 drugs induced apoptosis or affected the viability of monocytes. Our findings that local anesthetics inhibited the tissue factor induction in activated monocytes by inhibiting tissue factor mRNA level may demonstrate the feasibility of using local anesthetics in hypercoagulable and inflammatory conditions.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Laboratory Medicine and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ki Jun Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Wonsik Ahn
- Department of Anesthesiology and Pain Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyou-Sup Han
- Department of Laboratory Medicine and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Kyung Kim
- Department of Laboratory Medicine and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
36
|
DelGiudice LA, White GA. The role of tissue factor and tissue factor pathway inhibitor in health and disease states. J Vet Emerg Crit Care (San Antonio) 2009; 19:23-9. [PMID: 19691583 DOI: 10.1111/j.1476-4431.2008.00380.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To review the veterinary and human literature on the role of tissue factor (TF) and tissue factor pathway inhibitor (TFPI) in health and disease states. DATA SOURCES Original research articles and scientific reviews from both human and veterinary literature were searched for relevance to TF and TFPI. HUMAN DATA SYNTHESIS Interest in both TF and TFPI has grown widely over the last several years. The impact TF plays in coagulation, inflammation, angiogenesis, tumor metastasis, and cellular signaling has become apparent. Treatment with TFPI for severe sepsis has been examined and is still currently under investigation. Inhibition of the TF pathway is being studied as an aid in the treatment of neoplasia. The important physiologic and pathophysiologic role these molecules play has only begun to be understood. VETERINARY DATA SYNTHESIS There is a paucity of publications that discuss the importance of TF and TFPI in veterinary medicine. An enhanced understanding of the TF pathway in human medicine, in experimental animal models treating sepsis with TFPI, and in animal models demonstrating the proangiogenic properties of TF provides relevance to veterinary medicine. CONCLUSION It is apparent that TF and TFPI are important in health and disease. An enhanced understanding of the physiologic and pathophysiologic roles of these factors provides better insight into coagulation, inflammation, angiogenesis, disseminated intravascular coagulation, and tumor metastasis. This greater understanding may provide for the development of therapeutics for sepsis, disseminated intravascular coagulation, and neoplasia.
Collapse
|
37
|
Yao JL, Ryan CK, Francis CW, Kohli M, Taubman MB, Khorana AA. Tissue Factor and VEGF Expression in Prostate Carcinoma: A Tissue Microarray Study. Cancer Invest 2009; 27:430-4. [DOI: 10.1080/07357900802527247] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
38
|
Krikun G, Lockwood CJ, Paidas MJ. Tissue factor and the endometrium: from physiology to pathology. Thromb Res 2009; 124:393-6. [PMID: 19619892 DOI: 10.1016/j.thromres.2009.06.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 05/11/2009] [Accepted: 06/02/2009] [Indexed: 12/31/2022]
Abstract
Tissue factor (TF), is a transmembrane protein whose role was first identified as that of the initiator of hemostasis via a series of complicated protein cascades. It is now known however, that TF participates in angiogenesis as well as several processes that contribute to disease progression. Over the last 20 years, our laboratory has studied the expression and function of this molecule in both the pregnant and non-pregnant human endometrium. We demonstrated that TF is particularly upregulated at the time of implantation making it a critical factor to protect against excessive bleeding during trophoblast invasion. We have subsequently demonstrated the altered expression of this factor after long term contraception as well as in endometriosis. We proposed that any changes in this well regulated process can result in various pathologies of the endometrium including, infertility, bleeding, endometriosis, preeclampsia, preterm labor or thrombosis. Below we describe the latest findings of the expression and function of TF as well as its specific role in physiologic or pathologic conditions of the human endometrium.
Collapse
Affiliation(s)
- Graciela Krikun
- Department of Ob/Gyn, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.
| | | | | |
Collapse
|
39
|
Butenas S, Orfeo T, Mann KG. Tissue factor in coagulation: Which? Where? When? Arterioscler Thromb Vasc Biol 2009; 29:1989-96. [PMID: 19592470 DOI: 10.1161/atvbaha.108.177402] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Tissue factor (TF) is an integral membrane protein, normally separated from the blood by the vascular endothelium, which plays a key role in the initiation of blood coagulation. With a perforating vascular injury, TF becomes exposed to blood and binds plasma factor VIIa. The resulting complex initiates a series of enzymatic reactions leading to clot formation and vascular sealing. In some pathological states, circulating blood cells express TF as a result of exposure to an inflammatory stimulus leading to intravascular clotting, vessel occlusion, and thrombotic pathology. Numerous controversies have arisen related to the influence of structural features of TF, its presentation, and its function. There are contradictory reports about the synthesis and presentation of TF on blood cells and the presence (or absence) of functionally active TF circulating in normal blood either on microparticles or as a soluble protein. In this review we discuss TF structure-function relationships and the role of TF during various phases of the blood coagulation process. We also highlight controversies concerning the expression/presence of TF on various cells and in blood in normal and pathological states.
Collapse
Affiliation(s)
- Saulius Butenas
- Department of Biochemistry, University of Vermont, 208 South Park Drive, Suite 2, Room 235A, Colchester, VT 05446, USA.
| | | | | |
Collapse
|
40
|
Connolly GC, Khorana AA. Risk stratification for cancer-associated venous thromboembolism. Best Pract Res Clin Haematol 2009; 22:35-47. [DOI: 10.1016/j.beha.2008.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
41
|
Borensztajn K, Aberson H, Peppelenbosch MP, Spek CA. FXa-induced intracellular signaling links coagulation to neoangiogenesis: potential implications for fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:798-805. [PMID: 19339215 DOI: 10.1016/j.bbamcr.2009.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 01/14/2009] [Accepted: 01/15/2009] [Indexed: 02/06/2023]
Abstract
Fibrosis represents the end-stage of a broad range of disorders affecting organ function. These disorders are often associated with aberrant angiogenesis, but whether vascular abnormalities during fibrosis are characterized by excessive or diminished neo-vascularization remains questionable. Strikingly, activation of the coagulation cascade is frequently observed in association with the progression of fibroproliferative disorders. As we recently showed that coagulation factor (F)Xa induced fibrotic responses in fibroblasts, we hypothesized that FXa might indirectly induce angiogenesis by triggering fibroblasts to secrete proangiogenic factors. In the present study, we show that although FXa induces p42/44 MAP Kinase phosphorylation in endothelial cells, it has no direct effect on endothelial cell proliferation, protein synthesis and tube formation. In contrast, conditioned medium of fibroblasts stimulated with FXa enhanced endothelial cell proliferation, extra cellular matrix synthesis, wound healing and endothelial tube formation. FXa induced VEGF production by fibroblasts and a VEGF neutralizing antibody blocked the indirect effect of FXa on proliferation and realignment of endothelial cells identifying VEGF as a crucial player in angiogenesis during coagulation factor-induced fibrosis. Overall, our results establish a link between the coagulation cascade and angiogenesis during fibrosis.
Collapse
Affiliation(s)
- Keren Borensztajn
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, NL-9713 AV Groningen, The Netherlands.
| | | | | | | |
Collapse
|
42
|
Menzies KE, Mackman N, Taubman MB. Role of Tissue Factor in Cancer. Cancer Invest 2009. [DOI: 10.1080/07357900802656665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
43
|
Genetic associations with thalidomide mediated venous thrombotic events in myeloma identified using targeted genotyping. Blood 2008; 112:4924-34. [PMID: 18805967 DOI: 10.1182/blood-2008-02-140434] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A venous thromboembolism (VTE) with the subsequent risk of pulmonary embolism is a major concern in the treatment of patients with multiple myeloma with thalidomide. The susceptibility to developing a VTE in response to thalidomide therapy is likely to be influenced by both genetic and environmental factors. To test genetic variation associated with treatment related VTE in patient peripheral blood DNA, we used a custom-built molecular inversion probe (MIP)-based single nucleotide polymorphism (SNP) chip containing 3404 SNPs. SNPs on the chip were selected in "functional regions" within 964 genes spanning 67 molecular pathways thought to be involved in the pathogenesis, treatment response, and side effects associated with myeloma therapy. Patients and controls were taken from 3 large clinical trials: Medical Research Council (MRC) Myeloma IX, Hovon-50, and Eastern Cooperative Oncology Group (ECOG) EA100, which compared conventional treatments with thalidomide in patients with myeloma. Our analysis showed that the set of SNPs associated with thalidomide-related VTE were enriched in genes and pathways important in drug transport/metabolism, DNA repair, and cytokine balance. The effects of the SNPs associated with thalidomide-related VTE may be functional at the level of the tumor cell, the tumor-related microenvironment, and the endothelium. The clinical trials described in this paper have been registered as follows: MRC Myeloma IX: ISRCTN68454111; Hovon-50: NCT00028886; and ECOG EA100: NCT00033332.
Collapse
|
44
|
Abstract
Tissue factor (TF), is a cellular receptor that binds the ligand factor VII/VIIa to initiate the blood coagulation cascade. In addition to its role as the initiator of the hemostatic cascade, TF is known to be involved in angiogenesis via an interaction with factor VIIa and protease-activated receptor-2 (PAR-2). In this article we review previous studies from our laboratory demonstrating that the pattern and level of TF expression is altered in multiple cell types derived from eutopic and ectopic endometrium from women with endometriosis compared with normal endometrium. We posit that the inflammatory environment that occurs in ectopic and eutopic endometrium from patients with disease results in high TF expression that in turn, signals via PAR-2 to further produce inflammatory cytokine or chemokine production and macrophage recruitment. Thus, our studies suggest that TF might be an ideal target for therapeutic intervention in endometriosis.
Collapse
Affiliation(s)
- Graciela Krikun
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, School of Medicine. 333 Cedar St., P.O. Box 208063, New Haven, CT, 06520-8063, USA.
| | | | | | | |
Collapse
|
45
|
Khorana AA, Rao MV. Approaches to risk-stratifying cancer patients for venous thromboembolism. Thromb Res 2008; 120 Suppl 2:S41-50. [PMID: 18023712 DOI: 10.1016/s0049-3848(07)70129-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer patients are clearly at increased risk for venous thromboembolism (VTE). However, the risk is not equal for all cancer patients or even in the same patient over the course of the natural history of cancer. VTE prophylaxis has been shown to be beneficial in certain high-risk populations such as post-surgical or hospitalized cancer patients but data in the ambulatory setting are conflicting. A majority of solid tumor patients are currently treated in the ambulatory setting and studying risk factors for VTE and their interaction is essential to identifying a population of ambulatory cancer patients that could benefit from prophylaxis. In this review, we evaluate the literature on risk factors for cancer-associated VTE and discuss data from a large observational study of cancer patients receiving chemotherapy which has revealed novel risk factors for chemotherapy-associated VTE. Finally, we discuss model-based approaches to risk-stratifying cancer patients that incorporate the known multiple risk factors for VTE and can help identify high-risk subgroups in the cancer population.
Collapse
Affiliation(s)
- Alok A Khorana
- James P Wilmot Cancer Center, and the Department of Medicine, University of Rochester, Rochester, NY, USA.
| | | |
Collapse
|
46
|
Abstract
The research aims of our laboratory are to provide a realistic description of biologic processes involved in protection from hemorrhage and the evolution of thrombosis. To evaluate these processes, we use 4 models of coagulation ranging from 1) studies of blood exiting from microvascular wounds in humans through 2) minimally altered whole blood induced to clot by tissue factor (TF) to 3) reconstitution of the blood coagulation proteome with purified components and to 4) mathematical descriptions of the chemical processes and dynamics that occur. The integration of these 4 models permits comprehensive analyses of the blood coagulation system and predictions of its behavior under normal and pathologic conditions. Data accumulated thus far have led to advances in our understanding of 1) the processes occurring during the initiation and propagation phases of thrombin generation, 2) the roles for individual proteins involved in blood coagulation and its regulation, 3) defects in thrombin generation and clot formation in hemophilia, 4) actions and limitations of pharmacologic agents used to control hemorrhage, thrombosis, and chronic cardiovascular disease, and 5) the relationship between genotypic and phenotypic features of an individual's plasma proteome and his/her immediate and long-term thrombotic risk.
Collapse
|
47
|
Cerhan JR, Ansell SM, Fredericksen ZS, Kay NE, Liebow M, Call TG, Dogan A, Cunningham JM, Wang AH, Liu-Mares W, Macon WR, Jelinek D, Witzig TE, Habermann TM, Slager SL. Genetic variation in 1253 immune and inflammation genes and risk of non-Hodgkin lymphoma. Blood 2007; 110:4455-63. [PMID: 17827388 PMCID: PMC2234796 DOI: 10.1182/blood-2007-05-088682] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Smaller-scale evaluations suggest that common genetic variation in candidate genes related to immune function may predispose to the development of non-Hodgkin lymphoma (NHL). We report an analysis of variants within genes associated with immunity and inflammation and risk of NHL using a panel of 9412 single-nucleotide polymorphisms (SNPs) from 1253 genes in a study of 458 patients with NHL and 484 frequency-matched controls. We modeled haplotypes and risk of NHL, as well as the main effects for all independent SNPs from a gene in multivariate logistic regression models; we separately report results for nonsynonymous (ns) SNPs. In gene-level analyses, the strongest findings (P < or = .001) were for CREB1, FGG, MAP3K5, RIPK3, LSP1, TRAF1, DUSP2, and ITGB3. In nsSNP analyses, the strongest findings (P < or = .01) were for ITGB3 L59P (odds ratio [OR] = 0.66; 95% confidence interval [CI] 0.52-0.85), TLR6 V427A (OR = 5.20; CI 1.77-15.3), SELPLG M264V (OR = 3.20; CI 1.48-6.91), UNC84B G671S (OR = 1.50; CI 1.12-2.00), B3GNT3 H328R (OR = 0.74; CI 0.59-0.93), and BAT2 V1883L (OR = 0.64; CI 0.45-0.90). Our results suggest that genetic variation in genes associated with immune response (TRAF1, RIPK3, BAT2, and TLR6), mitogen-activated protein kinase (MAPK) signaling (MAP3K5, DUSP2, and CREB1), lymphocyte trafficking and migration (B3GNT3, SELPLG, and LSP1), and coagulation pathways (FGG and ITGB3) may be important in the etiology of NHL, and should be prioritized in replication studies.
Collapse
Affiliation(s)
- James R Cerhan
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Khorana AA, Ahrendt SA, Ryan CK, Francis CW, Hruban RH, Hu YC, Hostetter G, Harvey J, Taubman MB. Tissue factor expression, angiogenesis, and thrombosis in pancreatic cancer. Clin Cancer Res 2007; 13:2870-5. [PMID: 17504985 DOI: 10.1158/1078-0432.ccr-06-2351] [Citation(s) in RCA: 281] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE Hemostatic activation is common in pancreatic cancer and may be linked to angiogenesis and venous thromboembolism. We investigated expression of tissue factor (TF), the prime initiator of coagulation, in noninvasive and invasive pancreatic neoplasia. We correlated TF expression with vascular endothelial growth factor (VEGF) expression, microvessel density, and venous thromboembolism in resected pancreatic cancer. EXPERIMENTAL DESIGN Tissue cores from a tri-institutional retrospective series of patients were used to build tissue microarrays. TF expression was graded semiquantitatively using immunohistochemistry in normal pancreas (n=10), intraductal papillary mucinous neoplasms (n=70), pancreatic intraepithelial neoplasia (n=40), and resected or metastatic pancreatic adenocarcinomas (n=130). RESULTS TF expression was observed in a majority of noninvasive and invasive pancreatic neoplasia, including 77% of pancreatic intraepithelial neoplasias, 91% of intraductal papillary mucinous neoplasms, and 89% of pancreatic cancers, but not in normal pancreas. Sixty-six of 122 resected pancreatic cancers (54%) were found to have high TF expression (defined as grade >or=2, the median score). Carcinomas with high TF expression were more likely to also express VEGF (80% versus 27% with low TF expression, P<0.0001) and had a higher median MVD (8 versus 5 per tissue core with low TF expression, P=0.01). Pancreatic cancer patients with high TF expression had a venous thromboembolism rate of 26.3% compared with 4.5% in patients with low TF expression (P=0.04). CONCLUSIONS TF expression occurs early in pancreatic neoplastic transformation and is associated with VEGF expression, increased microvessel density, and possibly clinical venous thromboembolism in pancreatic cancer. Prospective studies evaluating the role of TF in pancreatic cancer outcomes are warranted.
Collapse
Affiliation(s)
- Alok A Khorana
- James P. Wilmot Cancer Center, Department of Medicine, University of Rochester, Rochester, New York 14642, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Méndez-Cruz AR, Paez A, Jiménez-Flores R, Reyes-Reali J, Varela E, Cerbulo-Vazquez A, Rodriguez E, López-Marure R, Masso FA, Flores-Romo L, Montaño LF. Increased expression of inflammation-related co-stimulatory molecules by HUVECs from newborns with a strong family history of myocardial infarction stimulated with TNF-alpha and oxLDL. Immunol Lett 2007; 111:116-23. [PMID: 17675167 DOI: 10.1016/j.imlet.2007.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 06/13/2007] [Accepted: 06/14/2007] [Indexed: 11/21/2022]
Abstract
BACKGROUND Recent findings indicate that atherosclerosis, a chronic inflammatory process, might start during childhood. Nevertheless, the expression of inflammation-related molecules of endothelial cell isolated from healthy neonates with a strong family history of myocardial infarction (SFHMI) has been rarely analyzed. METHODS Human umbilical vein endothelial cells (HUVECs) from children with SFHMI were assessed for the expression of CD40 and CD40L, in the presence of TNF-alpha and oxLDL. The intracellular content of CD80, CXCL8 and tissue factor by HUVECs stimulated with a CD40 agonist monoclonal antibody as well as monocytes/lymphocyte adhesion to TNF-alpha-stimulated HUVECs was also evaluated. RESULTS The basal expression of CD40 and CD40L was higher in SFHMI-positive HUVECs in comparison to controls. TNF-alpha and oxLDL upregulated the expression of CD40 and CD40L in SFHMI versus control HUVECs (p<0.001). The intracellular expression of CXCL8, tissue factor and CD80 was also higher than in controls, and the adhesion of lymphocyte- and monocyte-like cells augmented upon TNF-alpha stimulation. CONCLUSIONS It is possible that the modifications observed in the SFHMI-positive HUVECs, all of them relevant to the atherosclerosis process, may lead to early inflammatory reactions, thus contributing to the premature initiation of atherosclerotic lesions in these children.
Collapse
|
50
|
Franchini M, Montagnana M, Targher G, Manzato F, Lippi G. Pathogenesis, clinical and laboratory aspects of thrombosis in cancer. J Thromb Thrombolysis 2007; 24:29-38. [PMID: 17396228 DOI: 10.1007/s11239-007-0028-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Accepted: 03/02/2007] [Indexed: 10/23/2022]
Abstract
The relationship between increased clotting and malignancy is well recognized, though the bidirectional development of this association is often overlooked. In the challenging cancer biology, transforming genes often act in concert with numerous epigenetic factors, including hypoxia, inflammation, contact between blood and cancer cells, and emission of procoagulant vesicles from tumors, to determine a net imbalance of the hemostatic potential which is detectable by a variety of laboratory tests. Procoagulant factors, in particular, are intimately involved in all aspects of hemostatic, cell proliferation and cellular signalling systems. However, the biggest as yet unresolved question is why cancer patients develop thrombosis? Since the thrombus itself does not apparently contributes directly to the tumor biology, enhanced hemostasis activation in cancer patients may be interpreted according to the most recent biological evidences. Coagulation and cancer biology interact bidirectionally in a "vicious cycle", in which greater tumor burden supplies greater procoagulants (tissue factor, cancer procoagulant) and thrombin, which would in turn act as strong promoters of cancer growth and spread. In this perspective, thrombosis may be interpreted as a epiphenomenon of an intricate an effective biological feedback to maintain or promote cancer progression. In this review article, we briefly analyze the pathogenesis, laboratory, clinical and therapeutic features of cancer and thrombosis.
Collapse
Affiliation(s)
- Massimo Franchini
- Servizio di Immunoematologia e Trasfusione - Centro Emofilia, Azinda ospedaliera de Verona, Ospedale Policlinico, Piazzale Ludovico Scuro, 37134, Verona, Italy,
| | | | | | | | | |
Collapse
|