1
|
Kuśnierczyk P. Redundancy and absurd names in immunology. Int J Immunogenet 2024; 51:125-129. [PMID: 38403874 DOI: 10.1111/iji.12660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 02/27/2024]
Abstract
In this short review, examples of unnecessary multiple names of cell membrane molecules, for example, immune checkpoints and cytokines, are presented. Moreover, ridiculous or inaccurate names, such as 'Regulated on activation, normal T-cell expressed and secreted' and 'tissue factor', are discussed.
Collapse
Affiliation(s)
- Piotr Kuśnierczyk
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
2
|
Xu D, Liao C, Tan J. KRAS-mutant colorectal cancer cell lines cause a prothrombotic state through the upregulation of thrombin: experimental study. Ann Med Surg (Lond) 2024; 86:850-855. [PMID: 38333285 PMCID: PMC10849412 DOI: 10.1097/ms9.0000000000001645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/11/2023] [Indexed: 02/10/2024] Open
Abstract
Background The KRAS genotype status is strongly associated with a prothrombotic state in colorectal cancer, and hypercoagulability and cancer-related thrombosis are among the significant events leading to poor prognosis. However, this correlation has not been confirmed at the cellular level. This study aimed to assess the maximum platelet aggregation rate and thrombin expression induced by colorectal cancer cells under different KRAS genotypes. Materials and methods Platelet aggregation rate assay and western blotting analysis were used to detect platelet aggregation and thrombin expression induced by four colorectal cancer cells with different KRAS genotypes, including RKO, HCT116, SW480, and SW620. FVIIa/tissue factor and thrombin inhibitors were added to explore changes in platelet aggregation rates induced by colorectal cancer cells and the association between KRAS genotype status and hypercoagulable state. Results KRAS-mutant cells were more likely to increase maximal platelet aggregation, with RKO, HCT116, SW480, and SW620 inducing 34.7%, 55.4%, 44.4%, and 63.8% of platelet aggregation, respectively. The maximum platelet aggregation rate was higher in the metastatic rectal cancer tumour strain SW620 than in the primary rectal cancer strain SW480. RKO cells had lower thrombin expression than the other three cells. Furthermore, the addition of thrombin inhibitors caused a more significant decrease in the platelet aggregation rate in KRAS-mutant cell lines compared to KRAS wild-type cell lines. Conclusion Compared to KRAS wild-type colorectal cancer cells, KRAS-mutant colorectal cancer cell lines were more likely to be hypercoagulable through the upregulation of thrombin expression, which was mainly achieved through the TF-thrombin pathway.
Collapse
Affiliation(s)
- Duogang Xu
- Department of General Surgery, Yan’an Hospital Affiliated to Kunming Medical University, Kunming
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Changkang Liao
- Department of General Surgery, Yan’an Hospital Affiliated to Kunming Medical University, Kunming
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Jing Tan
- Department of General Surgery, Yan’an Hospital Affiliated to Kunming Medical University, Kunming
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| |
Collapse
|
3
|
Sachetto ATA, Mackman N. Monocyte Tissue Factor Expression: Lipopolysaccharide Induction and Roles in Pathological Activation of Coagulation. Thromb Haemost 2023; 123:1017-1033. [PMID: 37168007 PMCID: PMC10615589 DOI: 10.1055/a-2091-7006] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 05/08/2023] [Indexed: 05/13/2023]
Abstract
The coagulation system is a part of the mammalian host defense system. Pathogens and pathogen components, such as bacterial lipopolysaccharide (LPS), induce tissue factor (TF) expression in circulating monocytes that then activates the coagulation protease cascade. Formation of a clot limits dissemination of pathogens, enhances the recruitment of immune cells, and facilitates killing of pathogens. However, excessive activation of coagulation can lead to thrombosis. Here, we review studies on the mechanism of LPS induction of TF expression in monocytes and its contribution to thrombosis and disseminated intravascular coagulation. Binding of LPS to Toll-like receptor 4 on monocytes induces a transient expression of TF that involves activation of intracellular signaling pathways and binding of various transcription factors, such as c-rel/p65 and c-Fos/c-Jun, to the TF promoter. Inhibition of TF in endotoxemia and sepsis models reduces activation of coagulation and improves survival. Studies with endotoxemic mice showed that hematopoietic cells and myeloid cells play major roles in the activation of coagulation. Monocyte TF expression is also increased after surgery. Activated monocytes release TF-positive extracellular vesicles (EVs) and levels of circulating TF-positive EVs are increased in endotoxemic mice and in patients with sepsis. More recently, it was shown that inflammasomes contribute to the induction of TF expression and activation of coagulation in endotoxemic mice. Taken together, these studies indicate that monocyte TF plays a major role in activation of coagulation. Selective inhibition of monocyte TF expression may reduce pathologic activation of coagulation in sepsis and other diseases without affecting hemostasis.
Collapse
Affiliation(s)
- Ana T. A. Sachetto
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
4
|
Subramaniam S, Kothari H, Bosmann M. Tissue factor in COVID-19-associated coagulopathy. Thromb Res 2022; 220:35-47. [PMID: 36265412 PMCID: PMC9525243 DOI: 10.1016/j.thromres.2022.09.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022]
Abstract
Evidence of micro- and macro-thrombi in the arteries and veins of critically ill COVID-19 patients and in autopsies highlight the occurrence of COVID-19-associated coagulopathy (CAC). Clinical findings of critically ill COVID-19 patients point to various mechanisms for CAC; however, the definitive underlying cause is unclear. Multiple factors may contribute to the prothrombotic state in patients with COVID-19. Aberrant expression of tissue factor (TF), an initiator of the extrinsic coagulation pathway, leads to thrombotic complications during injury, inflammation, and infections. Clinical evidence suggests that TF-dependent coagulation activation likely plays a role in CAC. Multiple factors could trigger abnormal TF expression and coagulation activation in patients with severe COVID-19 infection. Proinflammatory cytokines that are highly elevated in COVID-19 (IL-1β, IL-6 and TNF-α) are known induce TF expression on leukocytes (e.g. monocytes, macrophages) and non-immune cells (e.g. endothelium, epithelium) in other conditions. Antiphospholipid antibodies, TF-positive extracellular vesicles, pattern recognition receptor (PRR) pathways and complement activation are all candidate factors that could trigger TF-dependent procoagulant activity. In addition, coagulation factors, such as thrombin, may further potentiate the induction of TF via protease-activated receptors on cells. In this systematic review, with other viral infections, we discuss potential mechanisms and cell-type-specific expressions of TF during SARS-CoV-2 infection and its role in the development of CAC.
Collapse
Affiliation(s)
- Saravanan Subramaniam
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Hema Kothari
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA; Cardiovascular Division, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Markus Bosmann
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
5
|
Subramaniam S, Liu J, Fletcher C, Ramchandran R, Weiler H. Coagulation Factor IIIa (f3a) Knockdown in Zebrafish Leads to Defective Angiogenesis and Mild Bleeding Phenotype. Front Cell Dev Biol 2022; 10:852989. [PMID: 35386206 PMCID: PMC8978257 DOI: 10.3389/fcell.2022.852989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Tissue factor (TF) is crucial for embryogenesis, as mice lacking TF are embryonically lethal (E10.5). This lethality may be attributed to defects in vascular development and circulatory failure, suggesting additional roles for TF in embryonic development beyond coagulation. In this study, we characterized the role of one of the TF paralogs (f3a) using a zebrafish model. The expression of f3a during embryonic developmental stages was determined by RT-PCR. Spatiotemporal expression pattern of f3a revealed (high expression from 28 to 36 hpf) the role of in the development of the yolk sac, circulation, and fins. Morpholinos (MO), an antisense-based oligonucleotide strategy, was used to knockdown f3a and examined for defects in morphological appearance, bleeding, and vascular patterning. f3a MO-injected embryos showed morphological abnormalities, including shorter body lengths and crooked tails. O-dianisidine staining showed f3a MO-injected embryos exhibited bleeding in the trunk (5.44%) and head (9.52%) regions. Imaging of endothelial-specific transgenic lines (flk1:egfp-NLS/kdrl:mCherry-CAAX) showed a 3-fold decreased caudal vein plexus (CVP) in f3a morphants versus controls at 48 hpf, suggesting a potential role for f3a in angiogenesis. These findings confirm that f3a is essential for angiogenesis, in addition to its involvement in hemostasis.
Collapse
Affiliation(s)
- Saravanan Subramaniam
- Department of Medicine, Pulmonary Center, Boston University School of Medicine, Boston, MA, United States
- Blood Research Institute, Blood Center of Wisconsin: Part of Versiti, Milwaukee, WI, United States
- *Correspondence: Saravanan Subramaniam,
| | - Jiandong Liu
- UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Craig Fletcher
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ramani Ramchandran
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Hartmut Weiler
- Blood Research Institute, Blood Center of Wisconsin: Part of Versiti, Milwaukee, WI, United States
| |
Collapse
|
6
|
Abstract
Cancer-associated thrombosis (including venous thromboembolism (VTE) and arterial events) is highly consequential for patients with cancer and is associated with worsened survival. Despite substantial improvements in cancer treatment, the risk of VTE has increased in recent years; VTE rates additionally depend on the type of cancer (with pancreas, stomach and primary brain tumours having the highest risk) as well as on individual patient's and cancer treatment factors. Multiple cancer-specific mechanisms of VTE have been identified and can be classified as mechanisms in which the tumour expresses proteins that alter host systems, such as levels of platelets and leukocytes, and in which the tumour expresses procoagulant proteins released into the circulation that directly activate the coagulation cascade or platelets, such as tissue factor and podoplanin, respectively. As signs and symptoms of VTE may be non-specific, diagnosis requires clinical assessment, evaluation of pre-test probability, and objective diagnostic testing with ultrasonography or CT. Risk assessment tools have been validated to identify patients at risk of VTE. Primary prevention of VTE (thromboprophylaxis) has long been recommended in the inpatient and post-surgical settings, and is now an option in the outpatient setting for individuals with high-risk cancer. Anticoagulant therapy is the cornerstone of therapy, with low molecular weight heparin or newer options such as direct oral anticoagulants. Personalized treatment incorporating risk of bleeding and patient preferences is essential, especially as a diagnosis of VTE is often considered by patients even more distressing than their cancer diagnosis, and can severely affect the quality of life. Future research should focus on current knowledge gaps including optimizing risk assessment tools, biomarker discovery, next-generation anticoagulant development and implementation science.
Collapse
|
7
|
Functional Characteristics and Regulated Expression of Alternatively Spliced Tissue Factor: An Update. Cancers (Basel) 2021; 13:cancers13184652. [PMID: 34572880 PMCID: PMC8471299 DOI: 10.3390/cancers13184652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
In human and mouse, alternative splicing of tissue factor's primary transcript yields two mRNA species: one features all six TF exons and encodes full-length tissue factor (flTF), and the other lacks exon 5 and encodes alternatively spliced tissue factor (asTF). flTF, which is oftentimes referred to as "TF", is an integral membrane glycoprotein due to the presence of an alpha-helical domain in its C-terminus, while asTF is soluble due to the frameshift resulting from the joining of exon 4 directly to exon 6. In this review, we focus on asTF-the more recently discovered isoform of TF that appears to significantly contribute to the pathobiology of several solid malignancies. There is currently a consensus in the field that asTF, while dispensable to normal hemostasis, can activate a subset of integrins on benign and malignant cells and promote outside-in signaling eliciting angiogenesis; cancer cell proliferation, migration, and invasion; and monocyte recruitment. We provide a general overview of the pioneering, as well as more recent, asTF research; discuss the current concepts of how asTF contributes to cancer progression; and open a conversation about the emerging utility of asTF as a biomarker and a therapeutic target.
Collapse
|
8
|
Zhao J, Jiang T, Li P, Dai L, Shi G, Jing X, Gao S, Jia L, Wu S, Wang Y, Peng Y, Cheng Z. Tissue factor promotes airway pathological features through epithelial-mesenchymal transition of bronchial epithelial cells in mice with house dust mite-induced asthma. Int Immunopharmacol 2021; 97:107690. [PMID: 33940323 DOI: 10.1016/j.intimp.2021.107690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/30/2021] [Accepted: 04/18/2021] [Indexed: 11/17/2022]
Abstract
It has recently been shown that expression levels of tissue factor (TF) are high in the serum and peripheral blood mononuclear cells of patients with asthma. However, whether TF impacts airway inflammation and remodelling in asthma remains unknown. The aim of this study was to investigate the effect of TF in asthma airway inflammation and remodelling using a house dust mite (HDM)-induced chronic asthma model and human bronchial epithelial (16HBE) cells. A chronic asthma model was constructed in BALB/c mice by the intranasal instillation of HDM. Mice were treated with short hairpin TF (shTF), and airway inflammation and remodelling features of asthma and epithelial-mesenchymal transition (EMT) were assessed. 16HBE cells were induced by transforming growth factor-β1 (TGF-β1) and HDM in the presence or absence of shTF; then, EMT markers and invasion and migration ability were determined. TF expression increased in the lung tissue and 16HBE cells when exposed to HDM. TF downregulation in the lung significantly reduced airway hyperresponsiveness, eosinophil inflammation, the EMT process, and levels of interleukin (IL)-4, IL-6, IL-13, and TGF-β1 in bronchoalveolar lavage fluid of asthmatic mice. Moreover, TF downregulation inhibited migration and incursion and decreased the expression levels of fibronectin 1 and TGF-β1, but increased the expression of E-cadherin in HDM- and TGF-β1-stimulated 16HBE cells. These results demonstrated that TF promoted airway pathological features by enhancing the EMT of bronchial epithelial cells both in vitro and in mice with house dust mite-induced asthma.
Collapse
Affiliation(s)
- Junwei Zhao
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, He'nan 450052, PR China.
| | - Tianci Jiang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, He'nan 450052, PR China; Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, He'nan 450052, PR China
| | - Pengfei Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, He'nan 450052, PR China; Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, He'nan 450052, PR China
| | - Lingling Dai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, He'nan 450052, PR China
| | - Guang Shi
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, He'nan 450052, PR China
| | - Xiaogang Jing
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, He'nan 450052, PR China
| | - Shuhui Gao
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, He'nan 450052, PR China
| | - Liuqun Jia
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, He'nan 450052, PR China
| | - Shujun Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, He'nan 450052, PR China
| | - Yu Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, He'nan 450052, PR China
| | - Youmei Peng
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, He'nan 450052, PR China
| | - Zhe Cheng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, He'nan 450052, PR China; Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, He'nan 450052, PR China.
| |
Collapse
|
9
|
Cañas CA, Cañas F, Bautista-Vargas M, Bonilla-Abadía F. Role of Tissue Factor in the Pathogenesis of COVID-19 and the Possible Ways to Inhibit It. Clin Appl Thromb Hemost 2021; 27:10760296211003983. [PMID: 33784877 PMCID: PMC8020089 DOI: 10.1177/10760296211003983] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
COVID-19 (Coronavirus Disease 2019) is a highly contagious infection and associated with high mortality rates, primarily in elderly; patients with heart failure; high blood pressure; diabetes mellitus; and those who are smokers. These conditions are associated to increase in the level of the pulmonary epithelium expression of angiotensin-converting enzyme 2 (ACE-2), which is a recognized receptor of the S protein of the causative agent SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2). Severe cases are manifested by parenchymal lung involvement with a significant inflammatory response and the development of microvascular thrombosis. Several factors have been involved in developing this prothrombotic state, including the inflammatory reaction itself with the participation of proinflammatory cytokines, endothelial dysfunction/endotheliitis, the presence of antiphospholipid antibodies, and possibly the tissue factor (TF) overexpression. ARS-Cov-19 ACE-2 down-regulation has been associated with an increase in angiotensin 2 (AT2). The action of proinflammatory cytokines, the increase in AT2 and the presence of antiphospholipid antibodies are known factors for TF activation and overexpression. It is very likely that the overexpression of TF in COVID-19 may be related to the pathogenesis of the disease, hence the importance of knowing the aspects related to this protein and the therapeutic strategies that can be derived. Different therapeutic strategies are being built to curb the expression of TF as a therapeutic target for various prothrombotic events; therefore, analyzing this treatment strategy for COVID-19-associated coagulopathy is rational. Medications such as celecoxib, cyclosporine or colchicine can impact on COVID-19, in addition to its anti-inflammatory effect, through inhibition of TF.
Collapse
Affiliation(s)
- Carlos A. Cañas
- Unit of Rheumatology, Fundación Valle del Lili, Universidad Icesi, Cali, Colombia
| | - Felipe Cañas
- Unit of Cardiology, Clínica Medellín, Medellín, Colombia
| | | | - Fabio Bonilla-Abadía
- Unit of Rheumatology, Fundación Valle del Lili, Universidad Icesi, Cali, Colombia
| |
Collapse
|
10
|
Mei T, Shashni B, Maeda H, Nagasaki Y. Fibrinolytic tissue plasminogen activator installed redox-active nanoparticles (t-PA@iRNP) for cancer therapy. Biomaterials 2020; 259:120290. [PMID: 32829147 DOI: 10.1016/j.biomaterials.2020.120290] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/28/2020] [Accepted: 08/01/2020] [Indexed: 12/21/2022]
Abstract
Favorable blood flow within solid tumors has become the principal strategy for drug delivery. The use of thrombolytic drugs, such as tissue plasminogen activator (t-PA), in combination with other drugs or drug carriers may increase their therapeutic effect by increasing drug delivery near the solid tumor through fibrin degradation and blood flow restoration. We, therefore, designed t-PA-installed redox-active nanoparticles (t-PA@iRNP) to improve the perfusion of antioxidant nanoparticles in tumors, via fibrin degradation to decompress tumor vessels. Additionally, antioxidant iRNP was developed for tumor inhibition by reduction of critically elevated levels of reactive oxygen species (ROS) in tumors. The t-PA@iRNP, when administered to a colon cancer model, degraded the deposited fibrin and improved the iRNP and immune cells penetration in tumor tissues via the restored blood flow, thus more effectively inhibited tumor growth. The anti-tumor effect of iRNP was attributed to ROS-reduction mediated downregulation of crucial a transcriptional factor, NF-κB. Conclusively, this study provides a new strategy to enhance the delivery of nanotherapeutics into solid tumors.
Collapse
Affiliation(s)
- Ting Mei
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8573, Japan; Present Address: School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Babita Shashni
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8573, Japan
| | - Hiroshi Maeda
- BioDynamics Research Foundation, 1-24-6 Kuwamizu, Chuo-ku, Kumamoto, 862-0954, Japan
| | - Yukio Nagasaki
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8573, Japan; Master's School in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8573, Japan; Center for Research in Isotopes and Environmental Dynamics (CRiED), University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8573, Japan.
| |
Collapse
|
11
|
Ziętek Z. Tissue Factor and Risk of Complications After Kidney Transplantation. Transplant Proc 2020; 52:2331-2336. [PMID: 32475531 DOI: 10.1016/j.transproceed.2020.01.092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/30/2019] [Accepted: 01/22/2020] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Tissue factor (TF) is a membrane component of many cells and a strong activator of blood coagulation. Damage to the cells induces an increase in its expression and concentration in blood plasma. The injury and breakdown of the cells is inseparably connected with the harvesting and preservation of the kidney. PURPOSE The aim of the study was an analysis of TF in the renal vein after of restoration of circulation in the transplanted kidney. An additional goal was to investigate the impact of warm ischemia on TF. MATERIALS AND METHODS The examined group included 61 kidney recipients. Blood was taken from the renal vein in the first minute during reperfusion. Simultaneously, blood from a peripheral vein was also drawn. Apart from tissue factor (TF), I also examined thrombin/antithrombin complexes and fragments 1+2 of prothrombin. RESULTS In blood from renal veins, I noticed higher level of TF, thrombin/antithrombin complexes and fragments 1+2 of prothrombin in comparison with blood from peripheral veins (P < .0048, P < .016, P < .046, respectively). The 29 recipients (47% of the total) with postoperative complications had much higher concentrations of TF than others (P < .019). TF showed a strong positive correlation with the time of warm ischemia (r = 0.53864, P < .05). CONCLUSIONS The donor kidney appeared to be one of the main sources of TF in the blood of recipients. Warm ischemia significantly increased its concentration in renal vein blood. This concentration of TF may be associated with damage to the kidney. TF significantly increased the risk of postoperative complications.
Collapse
Affiliation(s)
- Zbigniew Ziętek
- Department of General Surgery and Transplantology, Pomeranian Medical University, Szczecin, Poland; Department of Normal and Clinical Anatomy, Pomeranian Medical University, Szczecin, Poland.
| |
Collapse
|
12
|
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: 14.8] [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
|
13
|
Kroone C, Vos M, Rademakers T, Kuijpers M, Hoogenboezem M, van Buul J, Heemskerk JWM, Ruf W, van Hylckama Vlieg A, Versteeg HH, Goumans MJ, de Vries CJM, Kurakula K. LIM-only protein FHL2 attenuates vascular tissue factor activity, inhibits thrombus formation in mice and FHL2 genetic variation associates with human venous thrombosis. Haematologica 2019; 105:1677-1685. [PMID: 31467128 PMCID: PMC7271603 DOI: 10.3324/haematol.2018.203026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/26/2019] [Indexed: 12/21/2022] Open
Abstract
Bleeding disorders and thrombotic complications are major causes of morbidity and mortality with many cases being unexplained. Thrombus formation involves aberrant expression and activation of tissue factor (TF) in vascular endothelial and smooth muscle cells. Here, we sought to identify factors that modulate TF gene expression and activity in these vascular cells. The LIM-only protein FHL2 is a scaffolding protein that modulates signal transduction pathways with crucial functions in endothelial and smooth muscle cells. However, the role of FHL2 in TF regulation and thrombosis remains unexplored. Using a murine model of venous thrombosis in mesenteric vessels, we demonstrated that FHL2 deficiency results in exacerbated thrombus formation. Gain- and loss-of-function experiments revealed that FHL2 represses TF expression in endothelial and smooth muscle cells through inhibition of the transcription factors nuclear factor κB and activating protein-1. Furthermore, we observed that FHL2 interacts with the cytoplasmic tail of TF. In line with our in vivo observations, FHL2 decreases TF activity in endothelial and smooth muscle cells whereas FHL2 knockdown or deficiency results in enhanced TF activity. Finally, the FHL2 single nucleotide polymorphism rs4851770 was associated with the risk of venous thrombosis in a large population of venous thrombosis cases and control subjects from 12 studies (INVENT consortium). Altogether, our results highlight functional involvement of FHL2 in TF-mediated coagulation and identify FHL2 as a novel gene associated with venous thrombosis in humans.
Collapse
Affiliation(s)
- Chantal Kroone
- The Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center (UMC), Leiden, the Netherlands
| | - Mariska Vos
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Timo Rademakers
- Department of Molecular Cell Biology, Sanquin Research, Amsterdam, the Netherlands
| | - Marijke Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC, Maastricht, The Netherlands
| | - Mark Hoogenboezem
- Department of Molecular Cell Biology, Sanquin Research, Amsterdam, the Netherlands
| | - Jaap van Buul
- Department of Molecular Cell Biology, Sanquin Research, Amsterdam, the Netherlands
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC, Maastricht, The Netherlands
| | - Wolfram Ruf
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.,Center for Thrombosis and Hemostasis Mainz, Germany
| | | | - Henri H Versteeg
- The Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center (UMC), Leiden, the Netherlands
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Carlie J M de Vries
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Kondababu Kurakula
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands .,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | | |
Collapse
|
14
|
Lewis CS, Bogdanov VY. KLF11 (Krüppel-Like Factor 11) Modulates Arterial Thrombosis. Arterioscler Thromb Vasc Biol 2019; 39:309-310. [PMID: 30811250 DOI: 10.1161/atvbaha.119.312368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
Unruh D, Mirkov S, Wray B, Drumm M, Lamano J, Li YD, Haider QF, Javier R, McCortney K, Saratsis A, Scholtens DM, Sarkaria JN, James CD, Horbinski C. Methylation-dependent Tissue Factor Suppression Contributes to the Reduced Malignancy of IDH1-mutant Gliomas. Clin Cancer Res 2018; 25:747-759. [PMID: 30266764 DOI: 10.1158/1078-0432.ccr-18-1222] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/15/2018] [Accepted: 09/24/2018] [Indexed: 01/08/2023]
Abstract
PURPOSE Gliomas with isocitrate dehydrogenase 1 mutations (IDH1mut) are less aggressive than IDH1 wild-type (IDH1wt) gliomas and have global genomic hypermethylation. Yet it is unclear how specific hypermethylation events contribute to the IDH1mut phenotype. Previously, we showed that the gene encoding the procoagulant tissue factor (TF), F3, is among the most hypermethylated and downregulated genes in IDH1mut gliomas, correlating with greatly reduced thrombosis in patients with IDH1mut glioma. Because TF also increases the aggressiveness of many cancers, the current study explored the contribution of TF suppression to the reduced malignancy of IDH1mut gliomas.Experimental Design: TF expression was manipulated in patient-derived IDH1mut and IDH1wt glioma cells, followed by evaluation of in vitro and in vivo behavior and analyses of cell signaling pathways. RESULTS A demethylating agent, decitabine, increased F3 transcription and TF-dependent coagulative activity in IDH1mut cells, but not in IDH1wt cells. TF induction enhanced the proliferation, invasion, and colony formation of IDH1mut cells, and increased the intracranial engraftment of IDH1mut GBM164 from 0% to 100% (P = 0.0001). Conversely, TF knockdown doubled the median survival of mice engrafted with IDH1wt/EGFRvIIIamp GBM6, and caused complete regression of IDH1wt/EGFRamp GBM12 (P = 0.001). In vitro and in vivo effects were linked to activation of receptor tyrosine kinases (RTK) by TF through a Src-dependent intracellular pathway, even when extracellular RTK stimulation was blocked. TF stimulated invasion predominately through upregulation of β-catenin. CONCLUSIONS These data show that TF suppression is a component of IDH1mut glioma behavior, and that it may therefore be an attractive target against IDH1wt gliomas.
Collapse
Affiliation(s)
- Dusten Unruh
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Snezana Mirkov
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Brian Wray
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Michael Drumm
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Jonathan Lamano
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Yuping D Li
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Qazi F Haider
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Rodrigo Javier
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Kathleen McCortney
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Amanda Saratsis
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Denise M Scholtens
- Department of Preventive Medicine, Northwestern University, Chicago, Illinois
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - C David James
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Craig Horbinski
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois. .,Department of Pathology, Northwestern University, Chicago, Illinois
| |
Collapse
|
16
|
Grover SP, Mackman N. Tissue Factor: An Essential Mediator of Hemostasis and Trigger of Thrombosis. Arterioscler Thromb Vasc Biol 2018; 38:709-725. [PMID: 29437578 DOI: 10.1161/atvbaha.117.309846] [Citation(s) in RCA: 452] [Impact Index Per Article: 64.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/25/2018] [Indexed: 12/21/2022]
Abstract
Tissue factor (TF) is the high-affinity receptor and cofactor for factor (F)VII/VIIa. The TF-FVIIa complex is the primary initiator of blood coagulation and plays an essential role in hemostasis. TF is expressed on perivascular cells and epithelial cells at organ and body surfaces where it forms a hemostatic barrier. TF also provides additional hemostatic protection to vital organs, such as the brain, lung, and heart. Under pathological conditions, TF can trigger both arterial and venous thrombosis. For instance, atherosclerotic plaques contain high levels of TF on macrophage foam cells and microvesicles that drives thrombus formation after plaque rupture. In sepsis, inducible TF expression on monocytes leads to disseminated intravascular coagulation. In cancer patients, tumors release TF-positive microvesicles into the circulation that may contribute to venous thrombosis. TF also has nonhemostatic roles. For instance, TF-dependent activation of the coagulation cascade generates coagulation proteases, such as FVIIa, FXa, and thrombin, which induce signaling in a variety of cells by cleavage of protease-activated receptors. This review will focus on the roles of TF in protective hemostasis and pathological thrombosis.
Collapse
Affiliation(s)
- Steven P Grover
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Nigel Mackman
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill.
| |
Collapse
|
17
|
Cancer-associated pathways and biomarkers of venous thrombosis. Blood 2017; 130:1499-1506. [PMID: 28807983 DOI: 10.1182/blood-2017-03-743211] [Citation(s) in RCA: 249] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/02/2017] [Indexed: 12/14/2022] Open
Abstract
Cancer patients have an increased risk of venous thromboembolism (VTE). In this review, we summarize common and cancer type-specific pathways of VTE in cancer patients. Increased levels of leukocytes, platelets, and tissue factor-positive (TF+) microvesicles (MVs) are all potential factors that alone or in combination increase cancer-associated thrombosis. Patients with lung or colorectal cancer often exhibit leukocytosis. Neutrophils could increase VTE in cancer patients by releasing neutrophil extracellular traps whereas monocytes may express TF. Thrombocytosis is often observed in gastrointestinal, lung, breast, and ovarian cancer and this could decrease the threshold required for VTE. Soluble P-selectin has been identified as a biomarker of cancer-associated thrombosis in a general cancer population and may reflect activation of the endothelium. P-selectin expression by the endothelium may enhance VTE by increasing the recruitment of leukocytes. Studies in patients with pancreatic or brain cancer suggest that elevated levels of PAI-1 may contribute to VTE. Although elevated levels of TF+ MVs have been observed in patients with different types of cancer, an association between TF+ MVs and VTE has been observed only in pancreatic cancer. Podoplanin expression is associated with VTE in patients with brain cancer and may activate platelets. Future studies should measure multiple biomarkers in each cancer type to determine whether combinations of biomarkers can be used as predictors of VTE. A better understanding of the pathways that increase VTE in cancer patients may lead to the development of new therapies to reduce the morbidity and mortality associated with thrombosis.
Collapse
|
18
|
Giri A, Edwards TL, Hartmann KE, Torstenson ES, Wellons M, Schreiner PJ, Velez Edwards DR. African genetic ancestry interacts with body mass index to modify risk for uterine fibroids. PLoS Genet 2017; 13:e1006871. [PMID: 28715450 PMCID: PMC5536439 DOI: 10.1371/journal.pgen.1006871] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 07/31/2017] [Accepted: 06/15/2017] [Indexed: 01/20/2023] Open
Abstract
Race, specifically African ancestry, and obesity are important risk factors for uterine fibroids, and likely interact to provide the right conditions for fibroid growth. However, existing studies largely focus on the main-effects rather than their interaction. Here, we firstly provide evidence for interaction between categories of body mass index (BMI) and reported-race in relation to uterine fibroids. We then investigate whether the association between inferred local European ancestry and fibroid risk is modified by BMI in African American (AA) women in the Vanderbilt University Medical Center bio-repository (BioVU) (539 cases and 794 controls) and the Coronary Artery Risk Development in Young Adults study (CARDIA, 264 cases and 173 controls). We used multiple logistic regression to evaluate interactions between local European ancestry and BMI in relation to fibroid risk, then performed fixed effects meta-analysis. Statistical significance threshold for local-ancestry and BMI interactions was empirically estimated with 10,000 permutations (p-value = 1.18x10-4). Admixture mapping detected an association between European ancestry and fibroid risk which was modified by BMI (continuous-interaction p-value = 3.75x10-5) around ADTRP (chromosome 6p24); the strongest association was found in the obese category (ancestry odds ratio (AOR) = 0.51, p-value = 2.23x10-5). Evaluation of interaction between genotyped/imputed variants and BMI in this targeted region suggested race-specific interaction, present in AAs only; strongest evidence was found for insertion/deletion variant (6:11946435), again in the obese category (OR = 1.66, p-value = 1.72x10-6). We found nominal evidence for interaction between local ancestry and BMI at a previously reported region in chromosome 2q31-32, which includes COL5A2, and TFPI, an immediate downstream target of ADTRP. Interactions between BMI and SNPs (single nucleotide polymorphisms) found in this region in AA women were also detected in an independent European American population of 1,195 cases and 1,164 controls. Findings from our study provide an example of how modifiable and non-modifiable factors may interact to influence fibroid risk and suggest a biological role for BMI in fibroid etiology.
Collapse
Affiliation(s)
- Ayush Giri
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Todd L. Edwards
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Katherine E. Hartmann
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Eric S. Torstenson
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Melissa Wellons
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Pamela J. Schreiner
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Digna R. Velez Edwards
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
19
|
Mitchel JA, Antoniak S, Lee JH, Kim SH, McGill M, Kasahara DI, Randell SH, Israel E, Shore SA, Mackman N, Park JA. IL-13 Augments Compressive Stress-Induced Tissue Factor Expression in Human Airway Epithelial Cells. Am J Respir Cell Mol Biol 2016; 54:524-31. [PMID: 26407210 DOI: 10.1165/rcmb.2015-0252oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tissue factor (TF) is best known as a cellular initiator of coagulation, but it is also a multifunctional protein that has been implicated in multiple pathophysiologic conditions, including asthma. In the lung, airway epithelial cells express TF, but it is unknown how TF expression is regulated by asthma-associated mediators. We investigated the role of IL-13, a type 2 cytokine, alone and in combination with compressive stress, which mimics asthmatic bronchoconstriction, on TF expression and release of TF-positive extracellular vesicles from primary normal human bronchial epithelial cells. Well-differentiated normal human bronchial epithelial cells were treated with IL-13 and compressive stress, alone and in combination. TF mRNA, protein and activity were measured in the cells and conditioned media. TF was also measured in the bronchoalveolar lavage (BAL) fluid of allergen-challenged mice and patients with asthma. IL-13 and compressive stress increased TF expression, but only compressive stress induced TF-positive extracellular vesicle release. Pretreatment with IL-13 augmented compressive stress-induced TF expression and release. TF protein and activity in BAL fluid were increased in allergen-sensitized and -challenged mice. TF was elevated in the BAL fluid of patients with mild asthma after an allergen challenge. Our in vitro and in vivo data indicate close cooperation between mechanical and inflammatory stimuli on TF expression and release of TF-positive extracellular vesicles in the lungs, which may contribute to pathophysiology of asthma.
Collapse
Affiliation(s)
- Jennifer A Mitchel
- 1 Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Silvio Antoniak
- 2 Department of Medicine, Division of Hematology/Oncology, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Sae-Hoon Kim
- 1 Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts.,4 Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul, South Korea
| | - Maureen McGill
- 1 Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - David I Kasahara
- 1 Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Scott H Randell
- 5 Marsico Lung Institute/Cystic Fibrosis Center, Chapel Hill, North Carolina; and
| | - Elliot Israel
- 6 Brigham and Women's Hospital, Boston, Massachusetts
| | - Stephanie A Shore
- 1 Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Nigel Mackman
- 2 Department of Medicine, Division of Hematology/Oncology, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jin-Ah Park
- 1 Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
20
|
Wan K, Li J, Li D, Ge J, Wang Y, Li X, Guo Y, Guo J, Leng M, Wang P, An Y. Novel hydroxybutyl chitosan nanoparticles for siRNA delivery targeting tissue factor inhibits proliferation and induces apoptosis in human vascular smooth muscle cells. Mol Med Rep 2015; 12:7957-62. [PMID: 26497351 PMCID: PMC4758285 DOI: 10.3892/mmr.2015.4461] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 09/16/2015] [Indexed: 01/08/2023] Open
Abstract
Chitosan, a polysaccharide isolated from shrimp and other crustacean shells, has been widely investigated for DNA and siRNA delivery. Despite substantial effort having been made to improve chitosan as a non-viral gene delivery vector, the application is severely limited by its poor solubility under physiological conditions. Hydroxybutyl chitosan (HBC), a modified chitosan, is soluble under neutral conditions. Tissue factor (TF) is involved in the pathogenesis of cardiovascular diseases by promoting thrombus formation and inducing the migration and proliferation of vascular smooth muscle cells. Targeting TF is an attractive therapeutic strategy for cardiovascular diseases. In the present study, the use of HBC for the transfer of TF-siRNAs into human umbilical vein smooth muscle cells (HUVSMCs) was investigated, and the effects of TF knockdown on cell proliferation and apoptosis were examined. HBC/siRNA nanoparticles were produced by mixing HBC and siRNA solutions with the assistance of tripolyphosphate buffer. The transfection efficiency with these nanoparticles was 74±2.5%, which was determined using a fluorescence-labeled siRNA under fluorescence microscopy. The delivery of HBC/TF-siRNA resulted in reductions in the production of cellular and soluble TF protein in HUVMSCs, which were measured using western blotting and enzyme-linked immunosorbent assay, respectively. TF knockdown led to inhibited cell proliferation, as assessed using a Cell Counting Kit-8 assay, and increased cell apoptosis, determined using Annexin V-fluorescein isothiocyanate staining. These findings suggested that HBC may be a promising vector for siRNA delivery, and that in vivo HBC/siRNA nanoparticle delivery targeting TF may be a potential option for the treatment of cardiovascular diseases, which warrants further investigation.
Collapse
Affiliation(s)
- Kang Wan
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Jian Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Dan Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Junhua Ge
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yunlong Wang
- Department of Gout, The Affiliated Hospital of Qingdao University, Shandong Provincial Key Laboratory of Metabolic Diseases, Qingdao, Shandong 266003, P.R. China
| | - Xuexun Li
- Department of Cardiology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 266000, P.R. China
| | - Yongfang Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Junjie Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Min Leng
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Pan Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yi An
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
21
|
Åberg M, Eriksson O, Mokhtari D, Siegbahn A. Tissue factor/factor VIIa induces cell survival and gene transcription by transactivation of the insulin-like growth factor 1 receptor. Thromb Haemost 2013; 111:748-60. [PMID: 24336871 DOI: 10.1160/th13-07-0593] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Accepted: 11/19/2013] [Indexed: 11/05/2022]
Abstract
The insulin-like growth factor 1 receptor (IGF-1R) is known to promote survival and has also been implicated in the pathogenesis of several disease states, including cardiovascular disorders and cancer. Recently, we showed that binding of coagulation factor VIIa (FVIIa) to its receptor tissue factor (TF) protects cancer cells from TNF-related apoptosis inducing ligand (TRAIL)-induced apoptosis. Here we present evidence that this biological function of TF/FVIIa is dependent on the IGF-1R. IGF-1R inhibitors AG1024 and PPP as well as siRNA-mediated downregulation of IGF-1R, abolished the TF/FVIIa-mediated protection against TRAIL-induced apoptosis. Moreover, FVIIa rapidly induced a time- and concentration-dependent tyrosine phosphorylation of the IGF-1R in MDA-MB-231 breast cancer cells and in primary human monocytes, an event that was accompanied by IGF-1R chromatin binding and gene transcription. We hereby present novel evidence of a cross-talk between the coagulation and IGF-1R signalling systems, and propose that the IGF-1R is a key player in mediating TF/FVIIa-induced cell survival.
Collapse
Affiliation(s)
| | | | | | - Agneta Siegbahn
- Prof. Agneta Siegbahn, MD, PhD, FESC, Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, University Hospital, Entr. 61 3rd floor, S-751 85 Uppsala, Sweden, E-mail: , Tel.: +46 186114251, Fax: +46 18552562
| |
Collapse
|
22
|
Åberg M, Siegbahn A. Tissue factor non-coagulant signaling - molecular mechanisms and biological consequences with a focus on cell migration and apoptosis. J Thromb Haemost 2013; 11:817-25. [PMID: 23384027 DOI: 10.1111/jth.12156] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tissue factor (TF), a transmembrane glycoprotein, is the main initiator of the blood coagulation cascade. TF is also recognized as a true signaling receptor. There is accumulating evidence that the downstream signaling effects of the TF complexes are transduced by several mechanisms, including: activation of protease-activated receptor (PAR)-1 and PAR-2, and the PAR-dependent pathways, via the TF cytoplasmic domain and by transactivation of receptor tyrosine kinases. Triggering of signaling cascades such as the mitogen-activated protein kinase and phosphoinositide 3-kinase/AKT pathways couples TF to a multitude of functions within the cell, such as proliferation, cell migration, and survival. Thus, TF has a Janus face; on the one hand, it has vital life-maintaining functions, and on the other it has harmful effects, exemplified by inflammation, the acute coronary syndromes, and cancer. TF mediates a broad spectrum of signaling mechanisms. Learning more about these different mechanisms/pathways will lead to new treatment strategies, which can ultimately be personalized.
Collapse
Affiliation(s)
- M Åberg
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
23
|
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: 1.8] [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
|
24
|
Evidence for direct transfer of tissue factor from monocytes to platelets in whole blood. Blood Coagul Fibrinolysis 2012; 23:345-50. [PMID: 22343684 DOI: 10.1097/mbc.0b013e328350bf2f] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Varying specificity of anti-tissue factor (anti-TF) antibodies gives rise to erroneous conclusions on TF positivity of platelets. Although monocytes are a well established source of TF in whole blood, there is no consensus whether platelets express or acquire TF from external sources. To test whether platelets can acquire TF expressed in monocytes, we studied a transfer of TF-yellow fluorescent protein (TF-YFP) from monocytes nucleofected with TF-YFP to platelets in a whole blood model. Platelets isolated from whole blood were found positive for TF when immunostained with anti-TF antibody from one supplier, whereas no platelet TF antigen was found in whole blood immunostained with anti-TF antibody from another supplier. Both antibodies recognized TF in monocytes. Platelets isolated from whole blood reconstituted with monocytes expressing TF-YFP fusion protein were found positive for TF-YFP only after stimulation with lipopolysaccharide (LPS). Taken together, TF protein could be transferred from monocytes upon stimulation with LPS.
Collapse
|
25
|
Anand M, Brat DJ. Oncogenic regulation of tissue factor and thrombosis in cancer. Thromb Res 2012; 129 Suppl 1:S46-9. [DOI: 10.1016/s0049-3848(12)70015-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
26
|
Abstract
Tissue factor plays a primary role in both hemorrhage control and thrombosis depending upon whether its presentation is extravascular or intravascular. The molecular architecture and function of the tissue factor molecule and its role in the activations of factor IX and factor X have been elegantly elucidated but controversies prevail with respect to distinctions between tissue factor sources and tissue factor "activity." This presentation will review data on the architecture and functions of the tissue factor-factor VIIa complex and discuss the elements of the controversies associated with tissue factor presentation in both normal and pathologic milieu.
Collapse
Affiliation(s)
- Kenneth G Mann
- University of Vermont, Department of Biochemistry, Burlington, VT, [corrected] USA.
| | | | | |
Collapse
|
27
|
Basavaraj MG, Sovershaev MA, Egorina EM, Gruber FX, Bogdanov VY, Fallon JT, Østerud B, Mathiesen EB, Hansen JB. Circulating monocytes mirror the imbalance in TF and TFPI expression in carotid atherosclerotic plaques with lipid-rich and calcified morphology. Thromb Res 2011; 129:e134-41. [PMID: 22178066 DOI: 10.1016/j.thromres.2011.11.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 11/15/2011] [Accepted: 11/24/2011] [Indexed: 11/29/2022]
Abstract
BACKGROUND Thrombogenicity of atherosclerotic plaque largely depends on plaque morphology and their content of tissue factor (TF) and tissue factor pathway inhibitor (TFPI). The relationship between morphological composition of plaque (lipid-rich or calcified) and expression of TF and TFPI in circulating blood monocytes and within the plaques is not characterized. OBJECTIVE To investigate whether lipid-rich (echolucent) or calcified (echogenic) morphology of carotid atherosclerotic plaques is associated with differences in TF and TFPI expression in circulating blood monocytes and within carotid atherosclerotic plaques. METHODS We studied levels of monocyte TF and TFPI mRNA and protein expression and association with traditional risk factors for atherosclerosis in asymptomatic subjects with echolucent (n=20) or echogenic (n=20) carotid plaques, or controls without carotid atherosclerosis (n=20) determined by ultrasonography. Sections of calcified or lipid-rich carotid plaques obtained from symptomatic patients were assessed for TF and TFPI antigen expression. RESULTS TF and TFPI surface presentation, surface TF/TFPI ratio, and TF activity were higher in monocytes obtained from subjects with echolucent than with echogenic plaques or controls without carotid atherosclerosis. Multiple regression analyses revealed inverse association between serum apoA1 and monocyte surface TF antigen expression (p=0.007), and positive association between serum apoB and monocyte surface TFPI expression (p=0.028). Sections from lipid-rich carotid plaques contained 2.5-fold more TF and 1.5-fold more TFPI antigens relative to calcified lesions, also yielding a higher TF/TFPI ratio. CONCLUSIONS Our findings indicate that circulating monocytes of asymptomatic individuals with echolucent lipid-rich carotid atherosclerosis express an imbalance between TF and TFPI expression cohering with changes found within advanced carotid atherosclerotic plaques obtained from symptomatic patients.
Collapse
|
28
|
Control of RNA processing by a large non-coding RNA over-expressed in carcinomas. FEBS Lett 2011; 585:671-6. [PMID: 21266177 DOI: 10.1016/j.febslet.2011.01.030] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 01/15/2011] [Accepted: 01/17/2011] [Indexed: 12/27/2022]
Abstract
RNA processing is vital for the high fidelity and diversity of eukaryotic transcriptomes and the encoded proteomes. However, control of RNA processing is not fully established. Σ RNA is a class of conserved large non-coding RNAs (murine Hepcarcin; human MALAT-1) up-regulated in carcinomas. Using antisense technology, we identified that RNA post-transcriptional modification is the most significant global function of Σ RNA. Specifically, processing of the pre-mRNAs of genes including Tissue Factor and Endoglin was altered by hydrolysis of Σ RNA/MALAT-1. These results support the hypothesis that Σ RNA/MALAT-1 is a regulatory molecule exerting roles in RNA post-transcriptional modification.
Collapse
|
29
|
The Cerebral Microvasculature and Responses to Ischemia. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
30
|
Otero LL, Alonso DF, Castro M, Cinat G, Gabri MR, Gomez DE. Tissue factor as a novel marker for detection of circulating cancer cells. Biomarkers 2010; 16:58-64. [PMID: 21128872 DOI: 10.3109/1354750x.2010.533282] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Tissue factor (TF) is a molecular marker that is up-regulated in cancer cells and aids tumoral dissemination. Our purpose was to develop a nested RT-PCR strategy against TF for detecting blood-borne tumour cells. Our method detected TF expression in a minimum of 1.5 pg total RNA from MCF7 cells. A preliminary study in blood samples from 16 advanced breast carcinoma patients showed that 80% of patients with high TF load progressed and died, while only 18% with low TF load showed the same behaviour. Kaplan-Meier analysis confirmed worse overall survival in patients with high TF load.
Collapse
Affiliation(s)
- L L Otero
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
31
|
Mitroulis I, Kambas K, Anyfanti P, Doumas M, Ritis K. The multivalent activity of the tissue factor–thrombin pathway in thrombotic and non-thrombotic disorders as a target for therapeutic intervention. Expert Opin Ther Targets 2010; 15:75-89. [DOI: 10.1517/14728222.2011.532788] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
32
|
Abstract
Antiphospholipid syndrome (APS) is an acquired autoimmune disorder defined by the presence of an antiphospholipid antibody (aPL) and the occurrence of at least one associated clinical condition that includes venous thrombosis, arterial thrombosis or pregnancy morbidity. The aPL detected in APS have long been thought to have a direct prothrombotic effect in vivo. However, the pathophysiology underlying their coagulopathic effect has not been defined. Emerging data suggest a role for the procoagulant protein tissue factor (TF). In this review we provide an overview of TF, describe mouse models used in the evaluation of the role of TF in thrombosis, as well as summarize recent work on TF and APS.
Collapse
Affiliation(s)
- J Boles
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7005, USA
| | | |
Collapse
|
33
|
Patel N, Kalra VK. Placenta growth factor-induced early growth response 1 (Egr-1) regulates hypoxia-inducible factor-1alpha (HIF-1alpha) in endothelial cells. J Biol Chem 2010; 285:20570-9. [PMID: 20448047 DOI: 10.1074/jbc.m110.119495] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Leukotrienes, the lipid inflammatory products derived from arachidonic acid, are involved in the pathogenesis of respiratory and cardiovascular diseases and reactive airway disease in sickle cell disease. Placenta growth factor (PlGF), elaborated from erythroid cells, increased the mRNA expression of 5-lipoxygenase and 5-lipoxygenase-activating protein (FLAP) in human pulmonary microvascular endothelial cells. PlGF-induced both promoter activity and mRNA expression of hypoxia-inducible factor-1alpha (HIF-1alpha), which was abrogated by early growth response-1 (EGR-1) small interfering RNA. PlGF showed a temporal reciprocal relationship in the mRNA levels of EGR-1 and NAB2, the latter a repressor of Egr-1. Moreover, Nab2, but not mutant Nab2, significantly reduced promoter activity and mRNA expression of HIF-1alpha and also reduced expression of the HIF-1alpha target gene FLAP. Furthermore, overexpression of Egr-1 led to increased promoter activities for both HIF-1alpha and FLAP in the absence of PlGF. Additionally, the Egr-1-mediated induction of HIF-1alpha and FLAP promoters was reduced to basal levels by EGR-1 small interfering RNA. The binding of Egr-1 to HIF-1alpha promoter was corroborated by electrophoretic mobility shift assay and chromatin immunoprecipitation assay, which showed increased Egr-1 binding to the HIF-1alpha promoter in response to PlGF stimulation. These studies provide a novel mechanism for PlGF-mediated regulation of HIF-1alpha via Egr-1, which results in increased FLAP expression. This study provides a new therapeutic target, namely Egr-1, for attenuation of elevated leukotriene levels in patients with sickle cell disease and other inflammatory diseases.
Collapse
Affiliation(s)
- Nitin Patel
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | | |
Collapse
|
34
|
Circulating thrombotic and haemostatic components in patients with coronary artery disease. Indian J Clin Biochem 2010; 25:20-8. [PMID: 23105879 DOI: 10.1007/s12291-010-0005-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The study aimed to analyze the circulating levels of thrombotic and haemostatic components; tissue factor, tissue factor pathway inhibitor, tissue plasminogen activator and plasminogen activator inhibitor-1 in patients with acute myocardial infarction at presentation (Group 1, n=49), unstable angina and Non-ST elevated MI after treatment (Group 2, n=22), stable angina (Group 3, n=18) and healthy individuals (Group 4, n=31). Significant finding was increase in tissue factor not only in Group 1 (2.0 fold, P=0.001), Group 2 (2.2 fold, P=0.015) but also in Group 3 (1.8 fold, P=0.018) as compared to controls. In Group 1 Plasminogen activator inhibitor-1 increased significantly (35.8%, P=0.02). Tissue factor pathway inhibitor and tissue plasminogen activator demonstrated increase in Group 1 of age<40 years while insignificant changes in elder patients. Increased thrombotic and decreased fibrinolytic conditions in acute myocardial infarction patients were observed. Increase TF in stable angina demonstrates procoagulant status in these patients as well.
Collapse
|
35
|
Holy EW, Tanner FC. Tissue factor in cardiovascular disease pathophysiology and pharmacological intervention. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2010; 59:259-92. [PMID: 20933205 DOI: 10.1016/s1054-3589(10)59009-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Tissue factor (TF) is the major trigger of the coagulation cascade and thereby crucially involved in the maintenance of vascular hemostasis. By binding factor VIIa, the resulting TF:VIIa complex activates the coagulation factors IX and X ultimately leading to fibrin and clot formation. In the vessel wall, TF expression and activity is detectable in vascular smooth muscle cells and fibroblasts and, at a much lower level, in endothelial cells and can be induced by various stimuli including cytokines. In addition, TF is found in the bloodstream in circulating cells such as monocytes, in TF containing microparticles, and as a soluble splicing isoform. Besides its well-known extracellular role as a trigger of coagulation, TF also functions as a transmembrane receptor, and TF-dependent intracellular signaling events regulate the expression of genes involved in cellular responses such as proliferation and migration. TF indeed appears to be involved in the pathogenesis of neointima formation and tumor growth, and increased levels of TF have been detected in patients with cardiovascular risk factors or coronary artery disease as well as in those with cancer. Therefore, pharmacological or genetic inhibition of TF may be an attractive target for the treatment of cardiovascular disease and cancer. Different strategies for inhibition of TF have been developed such as inhibition of TF synthesis and blockade of TF action. Clinical applications of such strategies need to be tested in appropriate trials, in particular for evaluating the advantages of targeted versus systemic delivery of the inhibitors.
Collapse
Affiliation(s)
- Erik W Holy
- Cardiovascular Research, Physiology Institute, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
36
|
Abstract
The coagulation cascade represents a system of proteases responsible to maintain vascular integrity and to induce rapid clot formation after vessel injury. Tissue factor (TF), the key initiator of the coagulation cascade, binds to factor VIIa and thereby activates factor IX and factor X, resulting in thrombus formation. Different stimuli enhance TF gene expression in endothelial and vascular smooth muscle cells. In addition to these vascular cells, TF has recently been detected in the bloodstream in circulating cells such as leukocytes and platelets, as a component of microparticles, and as a soluble, alternatively spliced form of TF. Various cardiovascular risk factors like hypertension, diabetes, and dyslipidemia, increase levels of TF. In line with this observation, enhanced vascular TF expression occurs during atherogenesis, particularly in patients with acute coronary syndromes. (Circ J 2010; 74: 3 - 12).
Collapse
Affiliation(s)
- Alexander Breitenstein
- Cardiovascular Research, Physiology Institute, University of Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
37
|
Affiliation(s)
- Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7035, USA.
| | | |
Collapse
|
38
|
Koch L, Hofer S, Weigand MA, Frommhold D, Poeschl J. Lipopolysaccharide-induced activation of coagulation in neonatal cord and adult blood monitored by thrombelastography. Thromb Res 2009; 124:463-7. [DOI: 10.1016/j.thromres.2009.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Revised: 04/19/2009] [Accepted: 05/03/2009] [Indexed: 01/08/2023]
|
39
|
Milsom C, Magnus N, Meehan B, Al-Nedawi K, Garnier D, Rak J. Tissue factor and cancer stem cells: is there a linkage? Arterioscler Thromb Vasc Biol 2009; 29:2005-14. [PMID: 19628788 DOI: 10.1161/atvbaha.108.177444] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A common feature in the progression of multiple human malignancies is the protracted deregulation of the coagulation system, often referred to as cancer coagulopathy. Indeed, cancer cells and their vascular stroma often exhibit procoagulant properties, of which deregulation of tissue factor (TF) expression is a notable, although not the sole example. These changes can be traced to oncogenic influences affecting epidermal growth factor receptor (EGFR), EGFRvIII, K-ras, p53, PTEN, and probably many other proto-oncogenes and tumor suppressors in tumor parenchyma. Cancer stem cells (CSCs)/tumor initiating cells (TICs) are thought to represent the primary target and the main cellular effector through which oncogenic mutations exert their tumor-inducing effects. In so doing, CSCs/TICs depend on interactions with the tumor vasculature, which forms supportive niches for their clonal growth. We postulate that TF contributes to these interactions (directly or indirectly) through procoagulant and signaling effects, the latter executed in concert with juxtaposed protease activated receptors (mainly PAR-1 and PAR-2). TF/PAR system acts as a "blood sensing" mechanism, whereby cancer cells, including CSCs/TICs, may respond to plasma proteases (Factors VIIa, Xa, and IIa) and their related microenvironmental changes (fibrin deposition, activation of platelets). A growing body of still largely circumstantial evidence suggests that these events may contribute to the CSC/TIC niche, which could influence tumor initiation, metastasis, recurrence, and therapeutic intractability. Indeed, certain types of cancer cells harboring markers of CSCs (CD133) exhibit elevated TF expression and depend on this receptor to efficiently initiate tumor growth. We propose that both tumor cell-associated and host-related TF could influence the properties of CSCs, and that agents targeting the TF/PAR system may represent a hitherto unappreciated therapeutic opportunity to control cancer progression by influencing the CSC/TIC compartment.
Collapse
Affiliation(s)
- Chloe Milsom
- Montreal Children's Hospital, McGill University, QC, Canada
| | | | | | | | | | | |
Collapse
|
40
|
Sung CP, Arleth AJ, Ohlstein EH. Involvement of Protein Kinase C in Cytokine-Induced Tissue Factor Production in Human Vascular Endothelial Cells. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/10623329409053377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Milsom CC, Yu JL, Mackman N, Micallef J, Anderson GM, Guha A, Rak JW. Tissue factor regulation by epidermal growth factor receptor and epithelial-to-mesenchymal transitions: effect on tumor initiation and angiogenesis. Cancer Res 2009; 68:10068-76. [PMID: 19074872 DOI: 10.1158/0008-5472.can-08-2067] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ErbB oncogenes drive the progression of several human cancers. Our study shows that in human carcinoma (A431) and glioma (U373) cells, the oncogenic forms of epidermal growth factor receptor (EGFR; including EGFRvIII) trigger the up-regulation of tissue factor (TF), the transmembrane protein responsible for initiating blood coagulation and signaling through interaction with coagulation factor VIIa. We show that A431 cancer cells in culture exhibit a uniform TF expression profile; however, these same cells in vivo exhibit a heterogeneous TF expression and show signs of E-cadherin inactivation, which is coupled with multilineage (epithelial and mesenchymal) differentiation. Blockade of E-cadherin in vitro, leads to the acquisition of spindle morphology and de novo expression of vimentin, features consistent with epithelial-to-mesenchymal transition. These changes were associated with an increase in EGFR-dependent TF expression, and with enhanced stimulation of vascular endothelial growth factor production, particularly following cancer cell treatment with coagulation factor VIIa. In vivo, cells undergoing epithelial-to-mesenchymal transition exhibited an increased metastatic potential. Furthermore, injections of the TF-blocking antibody (CNTO 859) delayed the initiation of A431 tumors in immunodeficient mice, and reduced tumor growth, vascularization, and vascular endothelial growth factor expression. Collectively, our data suggest that TF is regulated by both oncogenic and differentiation pathways, and that it functions in tumor initiation, tumor growth, angiogenesis, and metastasis. Thus, TF could serve as a therapeutic target in EGFR-dependent malignancies.
Collapse
Affiliation(s)
- Chloe C Milsom
- Henderson Research Centre, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
42
|
Rak J, Yu J, Milsom C. Oncogene-Driven Hemostatic Changes in Cancer. Cancer Invest 2009. [DOI: 10.1080/07357900802656533] [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
|
Sovershaev MA, Lind KF, Devold H, Jørgensen TØ, Hansen JB, Østerud B, Egorina EM. No evidence for the presence of tissue factor in high-purity preparations of immunologically isolated eosinophils. J Thromb Haemost 2008; 6:1742-9. [PMID: 18665928 DOI: 10.1111/j.1538-7836.2008.03105.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND To date, there is no unequivocal opinion on whether human eosinophils express tissue factor (TF). Therefore, we studied the expression of TF protein and activity in resting or stimulated immunologically purified human eosinophils. METHODS AND RESULTS By use of immunologic isolation, we achieved over 96% purity of eosinophil preparations, and contamination by CD14-positive cells was below 0.3%. Flow cytometric [fluorescence-activated cell sorting (FACS)] analysis of eosinophils revealed no surface expression of TF antigen in resting or stimulated eosinophils. Immunoblotting of eosinophil lysates did not show any TF protein under resting or stimulated conditions. The lysates of resting or stimulated eosinophils contained no detectable levels of TF procoagulant activity. In contrast, monocytes, stimulated in plasma or medium, possessed readily detectable TF levels on the cell surface and in cell lysates as detected by FACS and immunoblotting. This was active TF antigen, as confirmed by TF activity assay (19.2 +/- 4.2 and 28.6 +/- 3.1 mU per 10(6) cells, stimulated in medium or plasma, respectively). We found no detectable TF mRNA levels in resting or stimulated eosinophils by real-time polymerase chain reaction (PCR), whereas in monocytes TF mRNA levels were significantly increased after stimulation. CONCLUSIONS Our data indicate that there is no evidence for TF expression in high-purity preparations of immunologically isolated eosinophils.
Collapse
Affiliation(s)
- M A Sovershaev
- Department of Medicine, University Hospital of North Norway, Tromsø, Norway
| | | | | | | | | | | | | |
Collapse
|
44
|
Conn EM, Madsen MA, Cravatt BF, Ruf W, Deryugina EI, Quigley JP. Cell surface proteomics identifies molecules functionally linked to tumor cell intravasation. J Biol Chem 2008; 283:26518-27. [PMID: 18658134 DOI: 10.1074/jbc.m803337200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In order to better understand the molecular and cellular determinants of tumor cell intravasation, our laboratory has generated a pair of congenic human HT-1080 fibrosarcoma variants (i.e. HT-hi/diss and HT-lo/diss) differing 50-100-fold in their ability to intravasate and disseminate. To investigate the molecular differences underlying the distinct dissemination capacities of these HT-1080 variants, we performed a comparative analysis of the cell surface proteomes of HT-hi/diss and HT-lo/diss. Cell membrane proteins were enriched by biotinylation and avidin precipitation and analyzed by tandem mass spectrometry employing multidimensional protein identification technology. By this approach, 47 cell surface-associated molecules were identified as differentially expressed between the HT-1080 intravasation variants. From these candidates, four targets (i.e. TIMP-2, NCAM-1, JAM-C, and tissue factor (TF)) were selected for further biochemical validation and in vivo functional verification. Western blot analysis of the cell surface enriched fractions confirmed the proteomic array data, demonstrating that, in vitro, TIMP-2 protein was increased in the HT-lo/diss variant, whereas NCAM-1, JAM-C, and TF levels were increased in the HT-hi/diss variant. Corresponding in vivo differences in levels of TIMP-2, JAM-C, and TF were demonstrated in primary tumors grown in the chick embryo. Finally, functional inhibition of one selected protein (i.e. TF) by small interfering RNA silencing or ligation with a function-blocking antibody significantly reduced HT-hi/diss intravasation, thus clearly implicating TF in the early steps of tumor cell dissemination. Overall, our cell surface proteomic analysis provides a powerful tool for identification of specific cell membrane molecules that contribute functionally to intravasation and metastasis in vivo.
Collapse
Affiliation(s)
- Erin M Conn
- Departments of Cell Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
45
|
Tardos JG, Eisenreich A, Deikus G, Bechhofer DH, Chandradas S, Zafar U, Rauch U, Bogdanov VY. SR proteins ASF/SF2 and SRp55 participate in tissue factor biosynthesis in human monocytic cells. J Thromb Haemost 2008; 6:877-84. [PMID: 18315555 DOI: 10.1111/j.1538-7836.2008.02946.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
BACKGROUND Human monocytes express two naturally occurring forms of circulating tissue factor (TF) - full-length TF, a membrane-spanning protein, and alternatively spliced TF, a soluble molecule. Presence of the variable exon 5 in TF mRNA determines whether the encoded TF protein is transmembrane, or soluble. Recently, an essential SR protein ASF/SF2 was implicated in TF pre-mRNA processing in human platelets. OBJECTIVE To examine molecular mechanisms governing regulated processing of TF pre-mRNA in human monocytic cells. METHODS AND RESULTS In silico analysis of the human TF exon 5, present only in full-length TF mRNA, revealed putative binding motifs termed exonic splicing enhancers (ESE) for the SR proteins ASF/SF2 and SRp55, which were found to be abundantly expressed in monocytic cell lines THP-1 and SC, as well as monocyte-enriched peripheral blood mononuclear cells (PBMC). Using a splice competent mini-gene reporter system transiently expressed in monocytic cells, it was determined that weakening of either five closely positioned ASF/SF2 ESE (bases 87-117) or a single conserved SRp55 ESE (base 39) results in severe skipping of exon 5. ASF/SF2 and SRp55 were found to physically associate with the identified ESE. CONCLUSIONS SR proteins ASF/SF2 and SRp55 appear to interact with the variable TF exon 5 through ESE at bases 39 and 87-117. Weakening of the above ESE modulates splicing of TF exon 5. This study is the first to identify and experimentally characterize cis-acting splicing elements involved in regulated biosynthesis of human TF.
Collapse
Affiliation(s)
- J G Tardos
- Division of Hematology and Medical Oncology, The Samuel Bronfman Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029-6754, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
The role of tumor-and host-related tissue factor pools in oncogene-driven tumor progression. Thromb Res 2008; 120 Suppl 2:S82-91. [PMID: 18023719 DOI: 10.1016/s0049-3848(07)70135-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Oncogenic events play an important role in cancer-related coagulopathy (Trousseau syndrome), angiogenesis and disease progression. This can, in part, be attributed to the up-regulation of tissue factor (TF) and release of TF-containing microvesicles into the pericellular milieu and the circulation. In addition, certain types of host cells (stromal cells, inflammatory cells, activated endothelium) may also express TF. At present, the relative contribution of host- vs tumor-related TF to tumor progression is not known. Our recent studies have indicated that the role of TF in tumor formation is complex and context-dependent. Genetic or pharmacological disruption of TF expression/activity in cancer cells leads to tumor growth inhibition in immunodeficient mice. This occurred even in the case of xenotransplants of human cancer cells, in which TF overexpression is driven by potent oncogenes (K-ras or EGFR). Interestingly, the expression of TF in vivo is not uniform and appears to be influenced by many factors, including the level of oncogenic transformation, tumor microenvironment, adhesion and the coexpression of markers of cancer stem cells (CSCs). Thus, minimally transformed, but tumorigenic embryonic stem (ES) cells were able to form malignant and angiogenic outgrowths in the absence of TF. However, these tumors were growth inhibited in hosts (mice) with dramatically reduced TF expression (low-TF mice). Depletion of host TF also resulted in changes affecting vascular patterning of some, but not all types of tumors. These observations suggest that TF may play different roles growth and angiogenesis of different tumors. Moreover, both tumor cell and host cell compartments may, in some circumstances, contribute to the functional TF pool. We postulate that activation of the coagulation system and TF signaling, may deliver growth-promoting stimuli (e.g. fibrin, thrombin, platelets) to dormant cancer stem cells (CSCs). Functionally, these influences may be tantamount to formation of a provisional (TF-dependent) cancer stem cell niche. As such these changes may contribute to the involvement of CSCs in tumor growth, angiogenesis and metastasis.
Collapse
|
47
|
Davis DR, Erlich JH. CARDIAC TISSUE FACTOR: ROLES IN PHYSIOLOGY AND FIBROSIS. Clin Exp Pharmacol Physiol 2008; 35:342-8. [DOI: 10.1111/j.1440-1681.2007.04872.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
48
|
Granulocytes do not express but acquire monocyte-derived tissue factor in whole blood: evidence for a direct transfer. Blood 2008; 111:1208-16. [DOI: 10.1182/blood-2007-08-107698] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
AbstractUnlike unanimous opinion on tissue factor (TF) expression in monocytes, the quest for TF presence in granulocytes has been going on for decades. To study the cell origin and track the blood-borne TF, we assessed TF activity and protein levels, knocked-down endogenous TF expression with small interfering RNA (siRNA), and overexpressed TF–yellow fluorescent protein (TF-YFP) fusion in immunologically isolated human monocytes and granulocytes. Monocytes and, to a much lesser extent, granulocytes isolated from lipopolysaccharide (LPS)/phorbol 12-myristate-13-acetate (PMA)–stimulated whole blood contained active TF antigen. However, only monocytes possessed significant TF activity and protein levels when stimulated with LPS/PMA in suspension. Reintroduction of TF-silenced monocytes to whole blood led to a profound reduction of LPS/PMA-stimulated TF activity in both mononuclear cell (MNC) and granulocyte fractions. No reduction in TF activity in MNC and granulocyte fractions was observed when TF-silenced granulocytes were reintroduced to whole blood. As shown by immunoblotting, flow cytometry, and confocal microscopy, granulocytes became positive for TF-YFP when isolated from whole blood reconstituted with TF-YFP–expressing monocytes. Together, we pinpoint monocytes as a major source of TF and provide solid experimental evidence for a direct transfer of TF protein from the monocytes to granulocytes in the blood.
Collapse
|
49
|
Sp1 is an essential transcription factor for LPS-induced tissue factor expression in THP-1 monocytic cells, and nobiletin represses the expression through inhibition of NF-kappaB, AP-1, and Sp1 activation. Biochem Pharmacol 2008; 75:1504-14. [PMID: 18261712 DOI: 10.1016/j.bcp.2007.12.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Revised: 12/27/2007] [Accepted: 12/27/2007] [Indexed: 01/20/2023]
Abstract
Nobiletin is a citrus polymethoxylated flavonoid extracted from Citrus depressa, and has several reported biological effects. In this study, we investigated the effect of nobiletin on bacterial lipopolysaccharide (LPS)-induced expression of tissue factor (TF), a trigger protein for the blood coagulation cascade, and studied the possible mechanism of TF transcriptional regulation. THP-1 monocytic cells stimulated with LPS showed an increased expression of both TF protein and mRNA levels. However, pretreatment with nobiletin resulted in inhibition of LPS-induced expression of both TF protein and mRNA in a dose-dependent manner. Electrophoretic mobility shift assays revealed that binding of nuclear proteins from LPS-stimulated THP-1 cells to the NF-kappaB or AP-1 binding motif was increased as compared to non-stimulated control cells. Such increased binding activities were significantly reduced by pretreatment with nobiletin. Binding activity of nuclear proteins to the Sp1 binding motif was observed irrespective of LPS stimulation, but Sp1 activation was inhibited by nobiletin treatment of the cells. Treatment of THP-1 cells with Sp1-specific small interfering RNA (Sp1 siRNA) abolished the ability of LPS to induce TF activity. A similar reduction in the level of TF mRNA was also observed upon treatment of cells with Sp1 siRNA. These studies reveal that constitutive Sp1 activation is an essential event for transcriptional activation of TF, and nobiletin prevents LPS-induced TF expression by inhibiting NF-kappaB, AP-1, and Sp1 activation.
Collapse
|
50
|
Yoon SZ, Kim CS, Lee YH, Heo WS, Kim SH, Lee JH, Lim YJ, Jang IJ. Association of Tissue Factor Polymorphism with Fibrinolysis and Excessive Bleeding after Open Heart Surgery - A preliminary report -. Korean J Anesthesiol 2007. [DOI: 10.4097/kjae.2007.53.6.720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Seung Zhoo Yoon
- Department of Anesthesiology, Seoul National University College of Medicine, Seoul, Korea
| | - Chong Seong Kim
- Department of Anesthesiology, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Hun Lee
- Department of Anesthesiology, Seoul National University College of Medicine, Seoul, Korea
| | - Won Seok Heo
- Department of Anesthesiology, Seoul National University College of Medicine, Seoul, Korea
| | - Soong Hyop Kim
- Department of Anesthesiology, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Hwan Lee
- Department of Anesthesiology, Seoul National University College of Medicine, Seoul, Korea
| | - Young Jin Lim
- Department of Anesthesiology, Seoul National University College of Medicine, Seoul, Korea
| | - In Jin Jang
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|