601 results on '"Nigel Mackman"'
Search Results
2. Commentary: Tissue factor as a potential coagulative/vascular marker in relapsing-remitting multiple sclerosis
- Author
-
Nigel Mackman and Ana T.A. Sachetto
- Subjects
multiple sclerosis ,tissue factor ,inflammation ,biomarker ,coagulation ,Immunologic diseases. Allergy ,RC581-607 - Published
- 2023
- Full Text
- View/download PDF
3. Tissue factor pathway inhibitor is associated with risk of venous thromboembolism and all-cause mortality in patients with cancer
- Author
-
Cornelia Englisch, Florian Moik, Johannes Thaler, Silvia Koder, Nigel Mackman, Matthias Preusser, Ingrid Pabinger, and Cihan Ay
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Venous thromboembolism (VTE) is a common complication in patients with cancer. Data on the role of natural inhibitors of coagulation for occurrence of cancer-associated VTE are limited, thus, we investigated the association of tissue factor pathway inhibitor (TFPI) with risk of VTE and all-cause mortality in patients with cancer. Total TFPI antigen levels were measured with a commercially available enzyme-linked immunosorbant assay in patients included in the Vienna Cancer and Thrombosis Study, a prospective observational cohort study with the primary outcome VTE. Competing risk analysis and Cox regression analysis were performed to explore the association of TFPI levels with VTE and all-cause mortality. TFPI was analyzed in 898 patients (median age 62 years; interquartile range [IQR], 53-68; 407 (45%) women). Sixty-seven patients developed VTE and 387 died (24-month cumulative risk 7.5% and 42.1%, respectively). Patients had median TFPI levels at study inclusion of 56.4 ng/mL (IQR, 45.7-70.0), with highest levels in tumor types known to have a high risk of VTE (gastroesophageal, pancreatic and brain cancer: 62.0 ng/mL; IQR, 52.0-75.0). In multivariable analysis adjusting for age, sex, cancer type and stage, TFPI levels were associated with VTE risk (subdistribution hazard ratio per doubling =1.63, 95% confidence interval [CI]: 1.03-2.57). When patients with high and intermediate/low VTE risk were analyzed separately, the association remained independently associated in the high risk group only (subdistribution hazard ratio =2.63, 95% CI: 1.40-4.94). TFPI levels were independently associated with all-cause mortality (hazard ratio =2.36, 95% CI: 1.85-3.00). In cancer patients increased TFPI levels are associated with VTE risk, specifically in patients with high-risk tumor types, and with all-cause mortality.
- Published
- 2023
- Full Text
- View/download PDF
4. Cell type-specific roles of PAR1 in Coxsackievirus B3 infection
- Author
-
Michael F. Bode, Clare M. Schmedes, Grant J. Egnatz, Vanthana Bharathi, Yohei M. Hisada, David Martinez, Tomohiro Kawano, Alice Weithauser, Leah Rosenfeldt, Ursula Rauch, Joseph S. Palumbo, Silvio Antoniak, and Nigel Mackman
- Subjects
Medicine ,Science - Abstract
Abstract Protease-activated receptor 1 (PAR1) is widely expressed in humans and mice, and is activated by a variety of proteases, including thrombin. Recently, we showed that PAR1 contributes to the innate immune response to viral infection. Mice with a global deficiency of PAR1 expressed lower levels of CXCL10 and had increased Coxsackievirus B3 (CVB3)-induced myocarditis compared with control mice. In this study, we determined the effect of cell type-specific deletion of PAR1 in cardiac myocytes (CMs) and cardiac fibroblasts (CFs) on CVB3-induced myocarditis. Mice lacking PAR1 in either CMs or CFs exhibited increased CVB3 genomes, inflammatory infiltrates, macrophages and inflammatory mediators in the heart and increased CVB3-induced myocarditis compared with wild-type controls. Interestingly, PAR1 enhanced poly I:C induction of CXCL10 in rat CFs but not in rat neonatal CMs. Importantly, activation of PAR1 reduced CVB3 replication in murine embryonic fibroblasts and murine embryonic cardiac myocytes. In addition, we showed that PAR1 reduced autophagy in murine embryonic fibroblasts and rat H9c2 cells, which may explain how PAR1 reduces CVB3 replication. These data suggest that PAR1 on CFs protects against CVB3-induced myocarditis by enhancing the anti-viral response whereas PAR1 on both CMs and fibroblasts inhibits viral replication.
- Published
- 2021
- Full Text
- View/download PDF
5. Myeloid cell-derived coagulation tissue factor is associated with renal tubular damage in mice fed an adenine diet
- Author
-
Shu Yamakage, Yuji Oe, Emiko Sato, Koji Okamoto, Akiyo Sekimoto, Satoshi Kumakura, Hiroshi Sato, Mai Yoshida, Tasuku Nagasawa, Mariko Miyazaki, Sadayoshi Ito, Nigel Mackman, and Nobuyuki Takahashi
- Subjects
Medicine ,Science - Abstract
Abstract Patients with chronic kidney disease (CKD) commonly exhibit hypercoagulability. Increased levels of uremic toxins cause thrombogenicity by increasing tissue factor (TF) expression and activating the extrinsic coagulation cascade. TF is induced in monocytes and macrophages under pathological conditions, such as inflammatory diseases. However, the role of monocyte myeloid cell TF in CKD progression remains unclear. We aimed to clarify this issue, and the present study found that patients with CKD had elevated levels of D-dimer, a marker of fibrin degradation, which was associated with decreased estimated glomerular filtration rate and increased serum levels of uremic toxins, such as indoxyl sulfate. In vitro studies showed that several uremic toxins increased cellular TF levels in monocytic THP-1 cells. Mice with TF specifically deleted in myeloid cells were fed an adenine diet to cause uremic kidney injury. Myeloid TF deletion reduced tubular injury and pro-inflammatory gene expression in the kidneys of adenine-induced CKD but did not improve renal function as measured by plasma creatinine or blood urea nitrogen. Collectively, our findings suggest a novel concept of pathogenesis of coagulation-mediated kidney injury, in which elevated TF levels in monocytes under uremic conditions is partly involved in the development of CKD.
- Published
- 2021
- Full Text
- View/download PDF
6. Illustrated State‐of‐the‐Art Capsules of the ISTH 2022 Congress
- Author
-
Robert A. Ariëns, Beverley J. Hunt, Ejaife O. Agbani, Josefin Ahnström, Robert Ahrends, Raza Alikhan, Alice Assinger, Zsuzsa Bagoly, Alessandra Balduini, Elena Barbon, Christopher D. Barrett, Paul Batty, Jorge David Aivazoglou Carneiro, Wee Shian Chan, Moniek deMaat, Kerstin deWit, Cécile Denis, Martin H. Ellis, Renee Eslick, Hongxia Fu, Catherine P. M. Hayward, Benoit Ho‐Tin‐Noé, Frederikus A. Klok, Riten Kumar, Karin Leiderman, Rustem I. Litvinov, Nigel Mackman, Zoe McQuilten, Matthew D. Neal, William A. E. Parker, Roger J. S. Preston, Julie Rayes, Alireza R. Rezaie, Lara N. Roberts, Bianca Rocca, Susan Shapiro, Deborah M. Siegal, Lirlândia P. Sousa, Katsue Suzuki‐Inoue, Tahira Zafar, and Jiaxi Zhou
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract The ISTH London 2022 Congress is the first held (mostly) face‐to‐face again since the COVID‐19 pandemic took the world by surprise in 2020. For 2 years we met virtually, but this year’s in‐person format will allow the ever‐so‐important and quintessential creativity and networking to flow again. What a pleasure and joy to be able to see everyone! Importantly, all conference proceedings are also streamed (and available recorded) online for those unable to travel on this occasion. This ensures no one misses out. The 2022 scientific program highlights new developments in hemophilia and its treatment, acquired and other inherited bleeding disorders, thromboinflammation, platelets and coagulation, clot structure and composition, fibrinolysis, vascular biology, venous thromboembolism, women’s health, arterial thrombosis, pediatrics, COVID‐related thrombosis, vaccine‐induced thrombocytopenia with thrombosis, and omics and diagnostics. These areas are elegantly reviewed in this Illustrated Review article. The Illustrated Review is a highlight of the ISTH Congress. The format lends itself very well to explaining the science, and the collection of beautiful graphical summaries of recent developments in the field are stunning and self‐explanatory. This clever and effective way to communicate research is revolutionary and different from traditional formats. We hope you enjoy this article and will be inspired by its content to generate new research ideas.
- Published
- 2022
- Full Text
- View/download PDF
7. Platelets and viruses
- Author
-
Silvio Antoniak and Nigel Mackman
- Subjects
platelets ,myocarditis ,influenza a virus ,coronavirus ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Platelets play an essential role in maintaining vascular integrity after injury. In addition, platelets contribute to the immune response to pathogens. For instance, they express receptors that mediate binding of viruses, and toll-like receptors that activate the cell in response to pathogen-associated molecular patterns. Platelets can be beneficial and/or detrimental during viral infections. They reduce blood-borne viruses by engulfing the free virus and presenting the virus to neutrophils. However, platelets can also enhance inflammation and tissue injury during viral infections. Here, we discuss the roles of platelets in viral infection.
- Published
- 2021
- Full Text
- View/download PDF
8. Prognostic value of circulating markers of neutrophil activation, neutrophil extracellular traps, coagulation and fibrinolysis in patients with terminal cancer
- Author
-
Axel Rosell, Katherina Aguilera, Yohei Hisada, Clare Schmedes, Nigel Mackman, Håkan Wallén, Staffan Lundström, and Charlotte Thålin
- Subjects
Medicine ,Science - Abstract
Abstract Predicting survival accurately in patients with advanced cancer is important in guiding interventions and planning future care. Objective tools are therefore needed. Blood biomarkers are appealing due to their rapid measurement and objective nature. Thrombosis is a common complication in cancer. Recent data indicate that tumor-induced neutrophil extracellular traps (NETs) are pro-thrombotic. We therefore performed a comprehensive investigation of circulating markers of neutrophil activation, NET formation, coagulation and fibrinolysis in 106 patients with terminal cancer. We found that neutrophil activation and NET markers were prognostic in terminal cancer patients. Interestingly, markers of coagulation and fibrinolysis did not have a prognostic value in this patient group, and there were weak or no correlations between these markers and markers of neutrophil activation and NETs. This suggest that NETs are linked to a poor prognosis through pathways independent of coagulation. Additional studies are needed to determine the utility of circulating neutrophil activation and NET markers, alone or in concert with established clinical parameters, as objective and reliable prognostic tools in advanced cancer.
- Published
- 2021
- Full Text
- View/download PDF
9. Prothrombotic changes in patients with COVID‐19 are associated with disease severity and mortality
- Author
-
Fien A. vonMeijenfeldt, Sebastian Havervall, Jelle Adelmeijer, Annika Lundström, Ann‐Sofie Rudberg, Maria Magnusson, Nigel Mackman, Charlotte Thalin, and Ton Lisman
- Subjects
coagulation ,COVID‐19 ,fibrinolysis ,hemostasis ,thrombosis ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Background and Aims Patients with severe coronavirus disease 2019 (COVID‐19) are at significant risk of thrombotic complications. However, their prothrombotic state is incompletely understood. Therefore, we measured in vivo activation markers of hemostasis, plasma levels of hemostatic proteins, and functional assays of coagulation and fibrinolysis in plasma from patients with COVID‐19 and determined their association with disease severity and 30‐day mortality. Methods We included 102 patients with COVID‐19 receiving various levels of respiratory support admitted to general wards, intermediate units, or intensive care units and collected plasma samples shortly after hospital admission. Results Patients with COVID‐19 with higher respiratory support had increased in vivo activation of coagulation and fibrinolysis, as reflected by higher plasma levels of d‐dimer, thrombin‐antithrombin, and plasmin‐antiplasmin complexes as compared to patients with no to minimal respiratory support and healthy controls. Moreover, the patients with COVID‐19 with higher respiratory support exhibited substantial ex vivo thrombin generation and lower ex vivo fibrinolytic capacity, despite higher doses of anticoagulant therapy compared to less severely ill patients. Fibrinogen, factor VIII, and von Willebrand factor levels increased, and ADAMTS13 levels decreased with increasing respiratory support in patients with COVID‐19. Low platelet count; low levels of prothrombin, antithrombin, and ADAMTS13; and high levels of von Willebrand factor were associated with short‐term mortality. Conclusions Severe COVID‐19 is associated with prothrombotic changes with increased in vivo activation of coagulation and fibrinolysis, despite anticoagulant therapy.
- Published
- 2021
- Full Text
- View/download PDF
10. Anticoagulant SERPINs: Endogenous Regulators of Hemostasis and Thrombosis
- Author
-
Steven P. Grover and Nigel Mackman
- Subjects
anticoagulants ,coagulation factors ,hemostasis ,serine protease inhibitors ,thrombosis ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Appropriate activation of coagulation requires a balance between procoagulant and anticoagulant proteins in blood. Loss in this balance leads to hemorrhage and thrombosis. A number of endogenous anticoagulant proteins, such as antithrombin and heparin cofactor II, are members of the serine protease inhibitor (SERPIN) family. These SERPIN anticoagulants function by forming irreversible inhibitory complexes with target coagulation proteases. Mutations in SERPIN family members, such as antithrombin, can cause hereditary thrombophilias. In addition, low plasma levels of SERPINs have been associated with an increased risk of thrombosis. Here, we review the biological activities of the different anticoagulant SERPINs. We further consider the clinical consequences of SERPIN deficiencies and insights gained from preclinical disease models. Finally, we discuss the potential utility of engineered SERPINs as novel therapies for the treatment of thrombotic pathologies.
- Published
- 2022
- Full Text
- View/download PDF
11. Evaluation of different commercial antibodies for their ability to detect human and mouse tissue factor by western blotting
- Author
-
Axel Rosell, Bernhard Moser, Yohei Hisada, Rukesh Chinthapatla, Grace Lian, Yi Yang, Matthew J. Flick, and Nigel Mackman
- Subjects
antibody ,glycosylation ,tissue factor ,validation ,western blotting ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Background Western blotting is used to measure protein expression in cells and tissues. Appropriate interpretation of resulting data is contingent upon antibody validation. Objectives We assessed several commercial anti‐human and anti‐mouse tissue factor (TF) antibodies for their ability to detect TF by western blotting. Material and Methods We used human pancreatic cancer cell lines expressing different levels of TF and a mouse pancreatic cancer cell line expressing TF with a matched knockout derivative. Results Human and mouse TF protein detected by western blotting correlated with levels of TF mRNA in these cell lines. The apparent molecular weight of TF is increased by N‐linked glycosylation and, as expected, deglycosylation decreased the size of TF based on western blotting. We found that four commercial anti‐human TF antibodies detected TF in a TF‐positive cell line HPAF‐II whereas no signal was observed in a TF‐negative cell line MIA PaCa‐2. More variability was observed in detecting mouse TF. Two anti‐mouse TF antibodies detected mouse TF in a TF‐positive cell line and no signal was observed in a TF knockout cell line. However, a third anti‐mouse TF antibody detected a nonspecific protein in both the mouse TF‐positive and TF‐negative cell lines. Two anti‐human TF antibodies that are claimed to cross react with mouse TF either recognized a nonspecific band or did not detect mouse TF. Discussion Our results indicate that there is a range in quality of commercial anti‐TF antibodies. Conclusion We recommend that all commercial antibodies should be validated to ensure that they detect TF.
- Published
- 2020
- Full Text
- View/download PDF
12. How useful are ferric chloride models of arterial thrombosis?
- Author
-
Steven P. Grover and Nigel Mackman
- Subjects
animal models ,erythrocytes ,ferric chloride ,platelets ,thrombosis ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
The ferric chloride models of arterial thrombosis are useful tools with which to investigate the cellular and molecular mechanisms that contribute to arterial thrombosis. Recent insights have, however, revealed the complex and multifaceted mechanism by which ferric chloride induces thrombus formation. Here, we discuss the strengths and weaknesses of the ferric chloride models of arterial thrombosis. Particular focus is given to the phenotypes of different knockout mice in the ferric chloride models and how these compare to other models with independent modes of initiation. Further, we discuss the relevance of the ferric chloride models to the human pathology of atherothrombotic disease.
- Published
- 2020
- Full Text
- View/download PDF
13. Effect of chemotherapy and longitudinal analysis of circulating extracellular vesicle tissue factor activity in patients with pancreatic and colorectal cancer
- Author
-
Raj S. Kasthuri, Yohei Hisada, Anton Ilich, Nigel S. Key, and Nigel Mackman
- Subjects
cancer ,chemotherapy ,extracellular vesicles ,tissue factor ,venous thromboembolism ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Introduction We conducted a longitudinal study in patients with pancreatic and colorectal cancer. We determined the effect of chemotherapy on extracellular vesicle tissue factor (EVTF) activity and the association of plasma EVTF activity with venous thromboembolism (VTE) and survival. Material and Methods We enrolled 13 patients with pancreatic and 22 patients with colorectal cancer. Plasma samples were collected during the 85‐day study period. Patients were followed for 3 months after the study period. We recorded symptomatic VTE during the study period (3 months) or asymptomatic deep vein thrombosis detected by ultrasound at day 85. We measured EVTF activity before and after chemotherapy. Results and Conclusions In the pancreatic cancer group, 2 patients had elevated levels of EVTF activity. One of these patients developed symptomatic VTE and died, and the second patient did not have a VTE but died. Chemotherapy decreased EVTF activity in 2 pancreatic patients with high levels. In the colorectal cancer group, 4 patients developed VTE, but EVTF activity was not elevated in any patient and no patient died. We observed a borderline significant correlation between EVTF activity and D‐dimer in the patients with pancreatic but not colorectal cancer. In this small descriptive study, 2 patients with pancreatic cancer had an elevated level of EVTF activity. Both patients died during the study period, and one had a VTE. Chemotherapy decreased EVTF activity in these patients. In contrast, elevated levels of EVTF activity were not observed in patients with colorectal cancer with or without VTE.
- Published
- 2020
- Full Text
- View/download PDF
14. Differential roles of factors IX and XI in murine placenta and hemostasis under conditions of low tissue factor
- Author
-
Steven P. Grover, Clare M. Schmedes, Alyson C. Auriemma, Emily Butler, Molly L. Parrish, Adam Miszta, Audrey C. Cleuren, Mayken Visser, Stefan Heitmeier, Jens J. Posma, Henri M. Spronk, Silvio Antoniak, Alisa S. Wolberg, Rafal Pawlinski, David Gailani, and Nigel Mackman
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: The intrinsic tenase complex (FIXa-FVIIIa) of the intrinsic coagulation pathway and, to a lesser extent, thrombin-mediated activation of FXI, are necessary to amplify tissue factor (TF)-FVIIa–initiated thrombin generation. In this study, we determined the contribution of murine FIX and FXI to TF-dependent thrombin generation in vitro. We further investigated TF-dependent FIX activation in mice and the contribution of this pathway to hemostasis. Thrombin generation was decreased in FIX- but not in FXI-deficient mouse plasma. Furthermore, injection of TF increased levels of FIXa-antithrombin complexes in both wild-type and FXI−/− mice. Genetic studies were used to determine the effect of complete deficiencies of either FIX or FXI on the survival of mice expressing low levels of TF. Low-TF;FIX−/y male mice were born at the expected frequency, but none survived to wean. In contrast, low-TF;FXI−/− mice were generated at the expected frequency at wean and had a 6-month survival equivalent to that of low-TF mice. Surprisingly, a deficiency of FXI, but not FIX, exacerbated the size of blood pools in low-TF placentas and led to acute hemorrhage and death of some pregnant dams. Our data indicate that FIX, but not FXI, is essential for survival of low-TF mice after birth. This finding suggests that TF-FVIIa–mediated activation of FIX plays a critical role in murine hemostasis. In contrast, FXI deficiency, but not FIX deficiency, exacerbated blood pooling in low-TF placentas, indicating a tissue-specific requirement for FXI in the murine placenta under conditions of low TF.
- Published
- 2020
- Full Text
- View/download PDF
15. Evaluation of a new bead‐based assay to measure levels of human tissue factor antigen in extracellular vesicles in plasma
- Author
-
Sierra J. Archibald, Yohei Hisada, Victoria L. Bae‐Jump, and Nigel Mackman
- Subjects
biomarker ,COVID‐19 ,extracellular vesicle ,thrombosis ,tissue factor ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Background Circulating tissue factor (TF)‐expressing extracellular vesicles (EVs) are associated with thrombosis in several diseases, such as coronavirus disease 2019 (COVID‐19). Activity assays have higher sensitivity and specificity compared to antigen assays for measuring TF+ EVs in plasma. The MACSPlex Exosome Kit is designed to detect 37 exosomal surface epitopes, including TF, on EVs in plasma using various fluorescently labeled beads. The different EV‐bead complexes are detected by flow cytometry. A recent study used the MACSPlex Exosome Kit to measure levels of TF+ EVs in serum from patients with COVID‐19. Objectives To evaluate the ability of the MACSPlex Exosome Kit to detect TF on EVs in plasma. Methods We measured levels of TF+ EVs isolated from plasma with or without TF detected using our in‐house EVTF activity assay and the MACSPlex Exosome Kit. Results The MACSPlex Exosome Kit gave a very low TF antigen signal (TF bead signal) compared to platelet‐derived CD41b+ EVs, which was used as a control. Lipopolysaccharide (LPS) increased levels of EVTF activity but not TF bead signal in four donors. Inhibition of TF reduced levels of EVTF activity but did not affect the TF bead signal in EVs isolated from plasma from LPS‐treated blood. Finally, we found no correlation between levels of EVTF activity and TF bead signal in EVs isolated from plasma from ovarian cancer patients (r = .16, P = .62). Conclusion Our data suggest that the MACSPlex Exosome Kit gives a nonspecific signal for TF and does not have the sensitivity to detect TF+ EVs in plasma.
- Published
- 2022
- Full Text
- View/download PDF
16. Markers of neutrophil activation and neutrophil extracellular traps in diagnosing patients with acute venous thromboembolism: A feasibility study based on two VTE cohorts.
- Author
-
Philip Smith, Axel Rosell, Maria Farm, Maria Bruzelius, Katherina Aguilera Gatica, Nigel Mackman, Jacob Odeberg, and Charlotte Thålin
- Subjects
Medicine ,Science - Abstract
BackgroundVenous thromboembolism (VTE) diagnosis would greatly benefit from the identification of novel biomarkers to complement D-dimer, a marker limited by low specificity. Neutrophil extracellular traps (NETs) have been shown to promote thrombosis and could hypothetically be used for diagnosis of acute VTE.ObjectivesTo assess the levels of specific markers of neutrophil activation and NETs and compare their diagnostic accuracy to D-dimer.MethodsWe measured plasma levels of neutrophil activation marker neutrophil elastase (NE), the NET marker nucleosomal citrullinated histone H3 (H3Cit-DNA) and cell-free DNA in patients (n = 294) with suspected VTE (pulmonary embolism and deep vein thrombosis) as well as healthy controls (n = 30). A total of 112 VTE positive and 182 VTE negative patients from two prospective cohort studies were included.ResultsHigher levels of H3Cit-DNA and NE, but not cell-free DNA, were associated with VTE. Area under receiver operating curves (AUC) were 0.90 and 0.93 for D-dimer, 0.65 and 0.68 for NE and 0.60 and 0.67 for H3Cit-DNA in the respective cohorts. Adding NE and H3Cit-DNA to a D-dimer based risk model did not improve AUC.ConclusionsOur study demonstrates the presence of neutrophil activation and NET formation in VTE using specific markers. However, the addition of NE or H3Cit-DNA to D-dimer did not improve the discrimination compared to D-dimer alone. This study provides information on the feasibility of using markers of NETs as diagnostic tools in acute VTE. Based on our findings, we believe the potential of these markers are limited in this setting.
- Published
- 2022
- Full Text
- View/download PDF
17. Myeloid Protease-Activated Receptor-2 Contributes to Influenza A Virus Pathology in Mice
- Author
-
Randall C. Gunther, Vanthana Bharathi, Stephen D. Miles, Lauryn R. Tumey, Clare M. Schmedes, Kohei Tatsumi, Meagan D. Bridges, David Martinez, Stephanie A. Montgomery, Melinda A. Beck, Eric Camerer, Nigel Mackman, and Silvio Antoniak
- Subjects
toll-like receptor 3 ,influenza A virus ,innate immune response ,macrophage ,lung epithelial cell ,protease-activated receptor 2 (PAR2) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
BackgroundInnate immune responses to influenza A virus (IAV) infection are initiated in part by toll-like receptor 3 (TLR3). TLR3-dependent signaling induces an antiviral immune response and an NFκB-dependent inflammatory response. Protease-activated receptor 2 (PAR2) inhibits the antiviral response and enhances the inflammatory response. PAR2 deficiency protected mice during IAV infection. However, the PAR2 expressing cell-types contributing to IAV pathology in mice and the mechanism by which PAR2 contributes to IAV infection is unknown.MethodsIAV infection was analyzed in global (Par2-/-), myeloid (Par2fl/fl;LysMCre+) and lung epithelial cell (EpC) Par2 deficient (Par2fl/fl;SPCCre+) mice and their respective controls (Par2+/+ and Par2fl/fl). In addition, the effect of PAR2 activation on polyinosinic-polycytidylic acid (poly I:C) activation of TLR3 was analyzed in bone marrow-derived macrophages (BMDM). Lastly, we determined the effect of PAR2 inhibition in wild-type (WT) mice.ResultsAfter IAV infection, Par2-/- and mice with myeloid Par2 deficiency exhibited increased survival compared to infected controls. The improved survival was associated with reduced proinflammatory mediators and reduced cellular infiltration in bronchoalveolar lavage fluid (BALF) of Par2-/- and Par2fl/fl;LysMCre+ 3 days post infection (dpi) compared to infected control mice. Interestingly, Par2fl/fl;SPCCre+ mice showed no survival benefit compared to Par2fl/fl. In vitro studies showed that Par2-/- BMDM produced less IL6 and IL12p40 than Par2+/+ BMDM after poly I:C stimulation. In addition, activation of PAR2 on Par2+/+ BMDM increased poly I:C induction of IL6 and IL12p40 compared to poly I:C stimulation alone. Importantly, PAR2 inhibition prior to IAV infection protect WT mice.ConclusionGlobal Par2 or myeloid cell but not lung EpC Par2 deficiency was associated with reduced BALF inflammatory markers and reduced IAV-induced mortality. Our study suggests that PAR2 may be a therapeutic target to reduce IAV pathology.
- Published
- 2021
- Full Text
- View/download PDF
18. Platelet-Mediated NET Release Amplifies Coagulopathy and Drives Lung Pathology During Severe Influenza Infection
- Author
-
Seok-Joo Kim, Agostina Carestia, Braedon McDonald, Amanda Z. Zucoloto, Heidi Grosjean, Rachelle P. Davis, Madison Turk, Victor Naumenko, Silvio Antoniak, Nigel Mackman, Mohamed Sarjoon Abdul-Cader, Mohamed Faizal Abdul-Careem, Morley D. Hollenberg, and Craig N. Jenne
- Subjects
influenza A virus ,platelets ,PAR4 ,thrombin ,neutrophils ,NETs ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The influenza A virus (IAV) causes a respiratory tract infection with approximately 10% of the population infected by the virus each year. Severe IAV infection is characterized by excessive inflammation and tissue pathology in the lungs. Platelet and neutrophil recruitment to the lung are involved in the pathogenesis of IAV, but the specific mechanisms involved have not been clarified. Using confocal intravital microscopy in a mouse model of IAV infection, we observed profound neutrophil recruitment, platelet aggregation, neutrophil extracellular trap (NET) production and thrombin activation within the lung microvasculature in vivo. Importantly, deficiency or antagonism of the protease-activated receptor 4 (PAR4) reduced platelet aggregation, NET production, and neutrophil recruitment. Critically, inhibition of thrombin or PAR4 protected mice from virus-induced lung tissue damage and edema. Together, these data imply thrombin-stimulated platelets play a critical role in the activation/recruitment of neutrophils, NET release and directly contribute to IAV pathogenesis in the lung.
- Published
- 2021
- Full Text
- View/download PDF
19. Comparison of the coagulopathies associated with COVID‐19 and sepsis
- Author
-
Robert A. Campbell, Yohei Hisada, Frederik Denorme, Steven P. Grover, Emma G. Bouck, Elizabeth A. Middleton, Alisa S. Wolberg, Matthew T. Rondina, and Nigel Mackman
- Subjects
coagulation ,COVID‐19 ,PAI‐1 ,sepsis ,tissue factor ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Background Coronavirus disease 2019 (COVID‐19) is associated with activation of coagulation that mainly presents as thrombosis. Sepsis is also associated with activation of coagulation that mainly presents as disseminated intravascular coagulation. Many studies have reported increased levels of plasma D‐dimer in patients with COVID‐19 that is associated with severity, thrombosis, and mortality. Objectives The aim of this study was to compare levels of circulating extracellular vesicle tissue factor (EVTF) activity and active plasminogen activator inhibitor 1 (PAI‐1) in plasma from patients with COVID‐19 or sepsis. Methods We measured levels of D‐dimer, EVTF activity, and active PAI‐1 in plasma samples from patients with COVID‐19 (intensive care unit [ICU], N = 15; and non‐ICU, N = 20) and patients with sepsis (N = 35). Results Patients with COVID‐19 had significantly higher levels of D‐dimer, EVTF activity, and active PAI‐1 compared with healthy controls. Patients with sepsis had significantly higher levels of D‐dimer and EVTF activity compared with healthy controls. Levels of D‐dimer were significantly lower in patients with COVID‐19 compared with patients with sepsis. Levels of EVTF activity were significantly higher in ICU patients with COVID‐19 compared with patients with sepsis. Levels of active PAI‐1 were significantly higher in patients with COVID‐19 compared with patients with sepsis. Conclusions High levels of both EVTF activity and active PAI‐1 may promote thrombosis in patients with COVID‐19 due to simultaneous activation of coagulation and inhibition of fibrinolysis. The high levels of active PAI‐1 in patients with COVID‐19 may limit plasmin degradation of crosslinked fibrin and the release of D‐dimer. This may explain the lower levels of D‐dimer in patients with COVID‐19 compared with patients with sepsis.
- Published
- 2021
- Full Text
- View/download PDF
20. In memoriam Dr Linda 'Kirt' Curtiss
- Author
-
Adam E. Mullick, William A. Boisvert, and Nigel Mackman
- Subjects
Biochemistry ,QD415-436 - Published
- 2021
- Full Text
- View/download PDF
21. Role of tissue factor in delayed bone repair induced by diabetic state in mice.
- Author
-
Hiroki Ehara, Kohei Tatsumi, Yoshimasa Takafuji, Naoyuki Kawao, Masayoshi Ishida, Kiyotaka Okada, Nigel Mackman, and Hiroshi Kaji
- Subjects
Medicine ,Science - Abstract
BackgroundTissue factor (TF) is the primary activator of the extrinsic coagulation protease cascade. Although TF plays roles in various pathological states, such as thrombosis, inflammatory diseases, cancer, and atherosclerosis, its involvement in bone metabolism remains unknown.Materials and methodsThe present study examined the roles of TF in delayed bone repair induced by a diabetic state in mice using wild-type (WT) and low TF-expressing (LTF) male mice. A diabetic state was induced by intraperitoneal injections of streptozotocin (STZ).ResultsA prolonged diabetic state significantly reduced total and trabecular bone mineral densities (BMD) as well as cortical bone thickness in WT and LTF mice; these BMD parameters were similar between WT and LTF mice treated with or without STZ. The diabetic state induced in WT mice delayed the repair of the femur following injury. The diabetic state induced in LTF mice was associated with further delays in bone repair. In in vitro experiments, TF significantly decreased receptor activator of nuclear factor-κB ligand-induced osteoclast formation and osteoclastogenic gene expression in RAW264.7 cells. However, it did not affect the gene expression levels of runt-related transcription factor 2 and osterix as well as alkaline phosphatase activity in mouse primary osteoblasts.ConclusionLow TF state was associated with enhanced bone repair delay induced by diabetic state in mice. The TF-induced suppression of bone remodeling may be a contributing factor to the protective effects of TF against delayed bone repair in a diabetic state.
- Published
- 2021
- Full Text
- View/download PDF
22. Neutrophils and neutrophil extracellular traps enhance venous thrombosis in mice bearing human pancreatic tumors
- Author
-
Yohei Hisada, Steven P. Grover, Anaum Maqsood, Reaves Houston, Cihan Ay, Denis F. Noubouossie, Brian C. Cooley, Håkan Wallén, Nigel S. Key, Charlotte Thålin, Ádám Z. Farkas, Veronika J. Farkas, Kiril Tenekedjiev, Krasimir Kolev, and Nigel Mackman
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Pancreatic cancer is associated with a high incidence of venous thromboembolism. Neutrophils have been shown to contribute to thrombosis in part by releasing neutrophil extracellular traps (NET). A recent study showed that increased plasma levels of the NET biomarker, citrullinated histone H3 (H3Cit), are associated with venous thromboembolism in patients with pancreatic and lung cancer but not in those with other types of cancer, including breast cancer. In this study, we examined the contribution of neutrophils and NET to venous thrombosis in nude mice bearing human pancreatic tumors. We found that tumor-bearing mice had increased circulating neutrophil counts and levels of granulocyte-colony stimulating factor, neutrophil elastase, H3Cit and cell-free DNA compared with controls. In addition, thrombi from tumor-bearing mice contained increased levels of the neutrophil marker Ly6G, as well as higher levels of H3Cit and cell-free DNA. Thrombi from tumor-bearing mice also had denser fibrin with thinner fibers consistent with increased thrombin generation. Importantly, either neutrophil depletion or administration of DNase I reduced the thrombus size in tumor-bearing but not in control mice. Our results, together with clinical data, suggest that neutrophils and NET contribute to venous thrombosis in patients with pancreatic cancer.
- Published
- 2020
- Full Text
- View/download PDF
23. Pathogenesis of cancer-associated thrombosis
- Author
-
Nigel Mackman
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2018
- Full Text
- View/download PDF
24. Extracellular vesicles, tissue factor, cancer and thrombosis – discussion themes of the ISEV 2014 Educational Day
- Author
-
Chris Gardiner, Paul Harrison, Mattias Belting, Anita Böing, Elena Campello, Bob S. Carter, Mary E. Collier, Frank Coumans, Camille Ettelaie, Nick van Es, Fred H. Hochberg, Nigel Mackman, Robert C. Rennert, Johannes Thaler, Janusz Rak, and Rienk Nieuwland
- Subjects
extracellular vesicles ,thrombosis ,cancer ,tissue factor ,Cytology ,QH573-671 - Abstract
Although the association between cancer and venous thromboembolism (VTE) has long been known, the mechanisms are poorly understood. Circulating tissue factor–bearing extracellular vesicles have been proposed as a possible explanation for the increased risk of VTE observed in some types of cancer. The International Society for Extracellular Vesicles (ISEV) and International Society on Thrombosis and Haemostasis (ISTH) held a joint Educational Day in April 2014 to discuss the latest developments in this field. This review discusses the themes of that event and the ISEV 2014 meeting that followed.
- Published
- 2015
- Full Text
- View/download PDF
25. Excess of heme induces tissue factor-dependent activation of coagulation in mice
- Author
-
Erica M. Sparkenbaugh, Pichika Chantrathammachart, Shaobin Wang, Will Jonas, Daniel Kirchhofer, David Gailani, Andras Gruber, Raj Kasthuri, Nigel S. Key, Nigel Mackman, and Rafal Pawlinski
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
An excess of free heme is present in the blood during many types of hemolytic anemia. This has been linked to organ damage caused by heme-mediated oxidative stress and vascular inflammation. We investigated the mechanism of heme-induced coagulation activation in vivo. Heme caused coagulation activation in wild-type mice that was attenuated by an anti-tissue factor antibody and in mice expressing low levels of tissue factor. In contrast, neither factor XI deletion nor inhibition of factor XIIa-mediated factor XI activation reduced heme-induced coagulation activation, suggesting that the intrinsic coagulation pathway is not involved. We investigated the source of tissue factor in heme-induced coagulation activation. Heme increased the procoagulant activity of mouse macrophages and human PBMCs. Tissue factor-positive staining was observed on leukocytes isolated from the blood of heme-treated mice but not on endothelial cells in the lungs. Furthermore, heme increased vascular permeability in the mouse lungs, kidney and heart. Deletion of tissue factor from either myeloid cells, hematopoietic or endothelial cells, or inhibition of tissue factor expressed by non-hematopoietic cells did not reduce heme-induced coagulation activation. However, heme-induced activation of coagulation was abolished when both non-hematopoietic and hematopoietic cell tissue factor was inhibited. Finally, we demonstrated that coagulation activation was partially attenuated in sickle cell mice treated with recombinant hemopexin to neutralize free heme. Our results indicate that heme promotes tissue factor-dependent coagulation activation and induces tissue factor expression on leukocytes in vivo. We also demonstrated that free heme may contribute to thrombin generation in a mouse model of sickle cell disease.
- Published
- 2015
- Full Text
- View/download PDF
26. Inactivation of factor VIIa by antithrombin in vitro, ex vivo and in vivo: role of tissue factor and endothelial cell protein C receptor.
- Author
-
Rit Vatsyayan, Hema Kothari, Nigel Mackman, Usha R Pendurthi, and L Vijaya Mohan Rao
- Subjects
Medicine ,Science - Abstract
Recent studies have suggested that antithrombin (AT) could act as a significant physiologic regulator of FVIIa. However, in vitro studies showed that AT could inhibit FVIIa effectively only when it was bound to tissue factor (TF). Circulating blood is known to contain only traces of TF, at best. FVIIa also binds endothelial cell protein C receptor (EPCR), but the role of EPCR on FVIIa inactivation by AT is unknown. The present study was designed to investigate the role of TF and EPCR in inactivation of FVIIa by AT in vivo. Low human TF mice (low TF, ∼ 1% expression of the mouse TF level) and high human TF mice (HTF, ∼ 100% of the mouse TF level) were injected with human rFVIIa (120 µg kg(-1) body weight) via the tail vein. At varying time intervals following rFVIIa administration, blood was collected to measure FVIIa-AT complex and rFVIIa antigen levels in the plasma. Despite the large difference in TF expression in the mice, HTF mice generated only 40-50% more of FVIIa-AT complex as compared to low TF mice. Increasing the concentration of TF in vivo in HTF mice by LPS injection increased the levels of FVIIa-AT complexes by about 25%. No significant differences were found in FVIIa-AT levels among wild-type, EPCR-deficient, and EPCR-overexpressing mice. The levels of FVIIa-AT complex formed in vitro and ex vivo were much lower than that was found in vivo. In summary, our results suggest that traces of TF that may be present in circulating blood or extravascular TF that is transiently exposed during normal vessel damage contributes to inactivation of FVIIa by AT in circulation. However, TF's role in AT inactivation of FVIIa appears to be minor and other factor(s) present in plasma, on blood cells or vascular endothelium may play a predominant role in this process.
- Published
- 2014
- Full Text
- View/download PDF
27. Role of tissue factor in Mycobacterium tuberculosis-induced inflammation and disease pathogenesis.
- Author
-
Hema Kothari, Shiva Keshava, Rit Vatsyayan, Nigel Mackman, L Vijaya Mohan Rao, and Usha R Pendurthi
- Subjects
Medicine ,Science - Abstract
Tuberculosis (TB) is a chronic lung infectious disease characterized by severe inflammation and lung granulomatous lesion formation. Clinical manifestations of TB include hypercoagulable states and thrombotic complications. We previously showed that Mycobacterium tuberculosis (M.tb) infection induces tissue factor (TF) expression in macrophages in vitro. TF plays a key role in coagulation and inflammation. In the present study, we investigated the role of TF in M.tb-induced inflammatory responses, mycobacterial growth in the lung and dissemination to other organs. Wild-type C57BL/6 and transgenic mice expressing human TF, either very low levels (low TF) or near to the level of wild-type (HTF), in place of murine TF were infected with M.tb via aerosol exposure. Levels of TF expression, proinflammatory cytokines and thrombin-antithrombin complexes were measured post M.tb infection and mycobacterial burden in the tissue homogenates were evaluated. Our results showed that M.tb infection did not increase the overall TF expression in lungs. However, macrophages in the granulomatous lung lesions in all M.tb-infected mice, including low TF mice, showed increased levels of TF expression. Conspicuous fibrin deposition in the granuloma was detected in wild-type and HTF mice but not in low TF mice. M.tb infection significantly increased expression levels of cytokines IFN-γ, TNF-α, IL-6 and IL-1ß in lung tissues. However, no significant differences were found in proinflammatory cytokines among the three experimental groups. Mycobacterial burden in lungs and dissemination into spleen and liver were essentially similar in all three genotypes. Our data indicate, in contrast to that observed in acute bacterial infections, that TF-mediated coagulation and/or signaling does not appear to contribute to the host-defense in experimental tuberculosis.
- Published
- 2014
- Full Text
- View/download PDF
28. Protease activated receptor-2 contributes to heart failure.
- Author
-
Silvio Antoniak, Erica M Sparkenbaugh, Michael Tencati, Mauricio Rojas, Nigel Mackman, and Rafal Pawlinski
- Subjects
Medicine ,Science - Abstract
Heart failure is a major clinical problem worldwide. Previous studies have demonstrated an important role for G protein-coupled receptors, including protease-activated receptors (PARs), in the pathology of heart hypertrophy and failure. Activation of PAR-2 on cardiomyocytes has been shown to induce hypertrophic growth in vitro. PAR-2 also contributes to myocardial infarction and heart remodeling after ischemia/reperfusion injury. In this study, we found that PAR-2 induced hypertrophic growth of cultured rat neonatal cardiomyocytes in a MEK1/2 and p38 dependent manner. In addition, PAR-2 activation on mouse cardiomyocytes increased expression of the pro-fibrotic chemokine MCP-1. Furthermore, cardiomyocyte-specific overexpression of PAR-2 in mice induced heart hypertrophy, cardiac fibrosis, inflammation and heart failure. Finally, in a mouse model of myocardial infarction induced by permanent ligation of the left anterior descending coronary artery, PAR-2 deficiency attenuated heart remodeling and improved heart function independently of its contribution to the size of the initial infarct. Taken together, our data indicate that PAR-2 signaling contributes to the pathogenesis of hypertrophy and heart failure.
- Published
- 2013
- Full Text
- View/download PDF
29. Cardiovascular Disease and Thrombosis in HIV Infection
- Author
-
Megan V. Perkins, Sarah B. Joseph, Dirk P. Dittmer, and Nigel Mackman
- Subjects
Cardiology and Cardiovascular Medicine - Abstract
HIV infection has transitioned from an acute, fatal disease to a chronic one managed by antiretroviral therapy. Thus, the aging population of people living with HIV (PLWH) continues to expand. HIV infection results in a dysregulated immune system, wherein CD4 + T cells are depleted, particularly in the gastrointestinal tract, disrupting the gut epithelial barrier. Long-term HIV infection is associated with chronic inflammation through potentially direct mechanisms caused by viral replication or exposure to viral proteins and indirect mechanisms resulting from increased translocation of microbial products from the intestine or exposure to antiretroviral therapy. Chronic inflammation (as marked by IL [interleukin]-6 and CRP [C-reactive protein]) in PLWH promotes endothelial cell dysfunction and atherosclerosis. PLWH show significantly increased rates of cardiovascular disease, such as myocardial infarction (risk ratio, 1.79 [95% CI, 1.54–2.08]) and stroke (risk ratio, 2.56 [95% CI, 1.43–4.61]). In addition, PLWH have increased levels of the coagulation biomarker D-dimer and have a two to ten-fold increased risk of venous thromboembolism compared with the general population. Several small clinical trials analyzed the effect of different antithrombotic agents on platelet activation, coagulation, inflammation, and immune cell activation. Although some markers for coagulation were reduced, most agents failed to reduce inflammatory markers in PLWH. More studies are needed to understand the underlying mechanisms driving inflammation in PLWH to create better therapies for lowering chronic inflammation in PLWH. Such therapies can potentially reduce atherosclerosis, cardiovascular disease, and thrombosis rates in PLWH and thus overall mortality in this population.
- Published
- 2023
30. Decreased Platelet Reactivity and Function in a Mouse Model of Human Pancreatic Cancer
- Author
-
Tomohiro Kawano, Yohei Hisada, Steven P. Grover, Wyatt J. Schug, David S. Paul, Wolfgang Bergmeier, and Nigel Mackman
- Subjects
Hematology - Abstract
Cancer patients have increased thrombosis and bleeding compared with the general population. Cancer is associated with activation of both platelets and coagulation. Mouse models have been used to study the dysregulation of platelets and coagulation in cancer. We established a mouse model of pancreatic cancer in which tissue factor-expressing human pancreatic tumors (BxPC-3) are grown in nude mice. Tumor-bearing mice have an activated coagulation system and increased venous thrombosis compared to control mice. We also showed that tumor-derived, tissue factor-positive extracellular vesicles activated platelets ex vivo and in vivo. In this study, we determined the effect of tumors on a platelet-dependent arterial thrombosis model. Unexpectedly, we observed significantly reduced carotid artery thrombosis in tumor-bearing mice compared to controls. In addition, we observed significantly increased tail bleeding in tumor-bearing mice compared to controls. These results suggested that the presence of the tumor affected platelets. Indeed, tumor-bearing mice exhibited a significant decrease in platelet count and an increase in mean platelet volume and percentage of reticulated platelets, findings that are consistent with increased platelet turnover. Levels of the platelet activation marker platelet factor 4 were also increased in tumor-bearing mice. We also observed decreased platelet receptor expression in tumor-bearing mice and reduced levels of active αIIb/β3 integrin in response to PAR4 agonist peptide and convulxin in platelets from tumor-bearing mice compared with platelets from control mice. In summary, our study suggests that in tumor-bearing mice there is chronic platelet activation, leading to thrombocytopenia, decreased receptor expression, and impaired platelet adhesive function.
- Published
- 2023
31. Neutrophil extracellular traps in the pathology of cancer and other inflammatory diseases
- Author
-
Melanie Herre, Jessica Cedervall, Nigel Mackman, and Anna-Karin Olsson
- Subjects
Neutrophils ,Physiology ,Neoplasms ,Physiology (medical) ,Humans ,Thrombosis ,General Medicine ,Extracellular Traps ,Molecular Biology ,Chromatin - Abstract
Neutrophil extracellular trap (NET) formation, first described in 2004 as a previously unknown strategy of neutrophils to fight microbes, has attracted an increasing interest in the research community. NETs are formed when neutrophils externalize their decondensed chromatin together with content from their azurophilic granules. In addition to their role in defense against microbes, NETs have been implicated as mediators of pathology in sterile inflammation, such as cancer and autoimmunity, and their potential as therapeutic targets is actively explored. However, targeting of NETs is challenging since the beneficial effects of their removal need to be balanced against the potential harmful loss of their function in microbial defense. Moreover, depending on the stimuli or species, NETs can be formed via distinct mechanisms and are not always made up of the same components, making direct comparisons between various studies challenging. This review focuses on the role of NETs in cancer-associated pathology, such as thrombosis, organ dysfunction, and metastasis. Different strategies to target NETs, by either preventing their formation or degrading existing ones, are also discussed.
- Published
- 2023
32. Tissue Factor and COVID-19: An Update
- Author
-
Nigel Mackman and Ana Teresa Azevedo Sachetto
- Subjects
Pharmacology ,SARS-CoV-2 ,Clinical Biochemistry ,Drug Discovery ,Humans ,Endothelial Cells ,COVID-19 ,Molecular Medicine ,Thromboplastin - Abstract
Abstract: The coronavirus 2019 (COVID-19) pandemic is caused by severe acute respiratory syn-drome coronavirus 2 (SARS-CoV-2) virus. Infection with SARS-CoV-2 is associated with acute respiratory distress syndrome, thrombosis and a high rate of mortality. Thrombotic events increase with severity. Tissue factor (TF) expression is increased during viral and bacterial infections. This review summarizes studies that have examined TF expression in response to SARS-CoV-2 infec-tion. SARS-CoV-2 virus and its proteins upregulate TF mRNA, protein and activity in a variety of cells, including bronchial epithelial cells, neutrophils, monocytes, macrophages, endothelial cells and adventitial fibroblasts. COVID-19 patients have increased TF expression in lungs, bron-choalveolar lavage fluid and circulating extracellular vesicles. The increase in TF was associated with coagulation activation markers, thrombosis, inflammatory markers, severity of disease and mortality. Taken together, the studies suggest that TF plays a central role in thrombosis in COVID-19. TF may be a useful prognostic marker and therapeutic target to reduce thrombosis and inflam-mation.
- Published
- 2022
33. 'A rapid, sensitive, and specific assay to measure tissue factor activity based on chromogenic determination of thrombin generation': comment from Tanratana et al
- Author
-
Pansakorn Tanratana, Ana Teresa Azevedo Sachetto, and Nigel Mackman
- Subjects
Hematology - Published
- 2023
34. 'Hereditary angioedema is associated with an increased risk of venous thromboembolism': reply
- Author
-
Steven P. Grover, Linda Sundler Björkman, Arne Egesten, Stephan Moll, and Nigel Mackman
- Subjects
Hematology - Published
- 2023
35. Measurement of tissue factor-positive extracellular vesicles in plasma: strengths and weaknesses of current methods
- Author
-
Nigel, Mackman, Ana Teresa Azevedo, Sachetto, and Yohei, Hisada
- Subjects
Extracellular Vesicles ,Plasma ,Humans ,Thrombosis ,Hematology ,Thromboplastin - Abstract
This review evaluates the different methods used to measure levels of tissue factor (TF) in plasma and on extracellular vesicles (EVs). Levels of TF-positive (TF+) EVs in blood are increased in a variety of diseases, such as cancer, sepsis, and viral infection, and are associated with thrombosis. Highly sensitive assays are required to measure the low levels of TF+ EVs in blood.TF antigen levels in plasma have been measured using standard ELISAs, SimpleStep ELISA technology, and solid-phase proximity ligation assay. Some studies reported the detection of TF+ EVs in plasma by flow cytometry. In addition, TF+ EVs can be captured onto beads and chips using anti-TF antibodies. Several assays have been developed to measure TF activity in EVs isolated from plasma. Importantly, activity-based assays are more sensitive than antigen-based assays as a single TF/FVIIa complex can generate large amounts of FXa.We recommend isolating EVs from plasma and measuring TF activity using a functional assay in the presence and absence of an anti-TF antibody. We do not recommend using antigen-based assays as these are not sensitive enough to detect the low levels of TF in plasma.
- Published
- 2022
36. Circulating tissue factor‐positive extracellular vesicles and their association with thrombosis in different diseases
- Author
-
Yohei Hisada, Ana Teresa Azevedo Sachetto, and Nigel Mackman
- Subjects
Pancreatic Neoplasms ,Extracellular Vesicles ,SARS-CoV-2 ,Sepsis ,Immunology ,COVID-19 ,Humans ,Immunology and Allergy ,Thrombosis ,Biomarkers ,Thromboplastin - Abstract
Tissue factor (TF) is a procoagulant protein released from activated host cells, such as monocytes, and tumor cells on extracellular vesicles (EVs). TF + EVs are observed in the circulation of patients with various types of diseases. In this review, we will summarize the association between TF + EVs and activation of coagulation and survival in different types of diseases, including cancer, sepsis, and infections with different viruses, such as human immunodeficiency virus (HIV), influenza A virus (IAV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We will also discuss the source of TF + EVs in various diseases. EVTF activity is associated with thrombosis in pancreatic cancer patients and coronavirus disease 2019 patients (COVID-19) and with disseminated intravascular coagulation in cancer patients. EVTF activity is also associated with worse survival in patients with cancer and COVID-19. Monocytes are the major sources of TF + EVs in sepsis, and viral infections, such as HIV, Ebola virus, and SARS-CoV-2. In contrast, alveolar epithelial cells are the major source of TF + EVs in bronchoalveolar lavage fluid in COVID-19 and influenza A patients. These studies indicate that EVTF activity could be used as a biomarker to identify patients that have an increased risk of coagulopathy and mortality.
- Published
- 2022
37. Monocyte tissue factor expression: LPS induction and roles in pathological activation of coagulation
- Author
-
Ana Teresa Azevedo Sachetto and Nigel Mackman
- Subjects
Hematology - 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 TF expression 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 the LPS-induced activation of coagulation. Selective inhibition of monocyte TF expression may reduce pathologic activation of coagulation in sepsis and other diseases.
- Published
- 2023
38. Epidemiology, biology, and management of venous thromboembolism in gliomas: An interdisciplinary review
- Author
-
Jasmin Jo, Maria Diaz, Craig Horbinski, Nigel Mackman, Stephen Bagley, Marika Broekman, Janusz Rak, James Perry, Ingrid Pabinger, Nigel S Key, and David Schiff
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Patients with diffuse glioma are at high risk of developing venous thromboembolism (VTE) over the course of the disease, with up to 30% incidence in patients with glioblastoma (GBM) and a lower but nonnegligible risk in lower-grade gliomas. Recent and ongoing efforts to identify clinical and laboratory biomarkers of patients at increased risk offer promise, but to date, there is no proven role for prophylaxis outside of the perioperative period. Emerging data suggest a higher risk of VTE in patients with isocitrate dehydrogenase (IDH) wild-type glioma and the potential mechanistic role of IDH mutation in the suppression of production of the procoagulants tissue factor and podoplanin. According to published guidelines, therapeutic anticoagulation with low molecular weight heparin (LMWH) or alternatively, direct oral anticoagulants (DOACs) in patients without increased risk of gastrointestinal or genitourinary bleeding is recommended for VTE treatment. Due to the elevated risk of intracranial hemorrhage (ICH) in GBM, anticoagulation treatment remains challenging and at times fraught. There are conflicting data on the risk of ICH with LMWH in patients with glioma; small retrospective studies suggest DOACs may convey lower ICH risk than LMWH. Investigational anticoagulants that prevent thrombosis without impairing hemostasis, such as factor XI inhibitors, may carry a better therapeutic index and are expected to enter clinical trials for cancer-associated thrombosis.
- Published
- 2023
39. Data from Epidermal Growth Factor Receptor and PTEN Modulate Tissue Factor Expression in Glioblastoma through JunD/Activator Protein-1 Transcriptional Activity
- Author
-
Daniel J. Brat, Nigel Mackman, Erwin G. Van Meir, Jeffrey J. Olson, Donald L. Durden, Gang Chen, Carol Tucker-Burden, Vladimir E. Belozerov, and Yuan Rong
- Abstract
Hypoxia and necrosis are fundamental features of glioblastoma (GBM) and their emergence is critical for the rapid biological progression of this fatal tumor; yet, underlying mechanisms are poorly understood. We have suggested that vaso-occlusion following intravascular thrombosis could initiate or propagate hypoxia and necrosis in GBM. Tissue factor (TF), the main cellular initiator of coagulation, is overexpressed in GBMs and likely favors a thrombotic microenvironment. Epidermal growth factor receptor (EGFR) amplification and PTEN loss are two common genetic alterations seen in GBM but not in lower-grade astrocytomas that could be responsible for TF up-regulation. The most frequent EGFR mutation in GBM involves deletion of exons 2 to 7, resulting in the expression of a constitutively active receptor, EGFRvIII. Here, we show that overexpression of EGFR or EGFRvIII in human glioma cells causes increased basal TF expression and that stimulation of EGFR by its ligand, EGF, leads to a marked dose-dependent up-regulation of TF. In all cases, increased TF expression led to accelerated plasma coagulation in vitro. EGFR-mediated TF expression depended most strongly on activator protein-1 (AP-1) transcriptional activity and was associated with c-Jun NH2-terminal kinase (JNK) and JunD activation. Restoration of PTEN expression in PTEN-deficient GBM cells diminished EGFR-induced TF expression by inhibiting JunD/AP-1 transcriptional activity. PTEN mediated this effect by antagonizing phosphatidylinositol 3-kinase activity, which in turn attenuated both Akt and JNK activities. These mechanisms are likely at work in vivo, as EGFR expression was highly correlated with TF expression in human high-grade astrocytoma specimens. [Cancer Res 2009;69(6):2540–9]
- Published
- 2023
40. Supplementary Figures 1-3, Tables 1-3, Methods from Tissue Factor–Activated Coagulation Cascade in the Tumor Microenvironment Is Critical for Tumor Progression and an Effective Target for Therapy
- Author
-
Cheng Liu, Kim D. Janda, Nigel Mackman, Subhash C. Sinha, Longwu Ye, Dong Xue, Katerina Capkova, Pengfei Jiang, and Yuan Liu
- Abstract
PDF file - 910K
- Published
- 2023
41. Supplementary Figures and Tables from Epidermal Growth Factor Receptor and PTEN Modulate Tissue Factor Expression in Glioblastoma through JunD/Activator Protein-1 Transcriptional Activity
- Author
-
Daniel J. Brat, Nigel Mackman, Erwin G. Van Meir, Jeffrey J. Olson, Donald L. Durden, Gang Chen, Carol Tucker-Burden, Vladimir E. Belozerov, and Yuan Rong
- Abstract
Supplementary Figures and Tables from Epidermal Growth Factor Receptor and PTEN Modulate Tissue Factor Expression in Glioblastoma through JunD/Activator Protein-1 Transcriptional Activity
- Published
- 2023
42. Data from Tissue Factor–Activated Coagulation Cascade in the Tumor Microenvironment Is Critical for Tumor Progression and an Effective Target for Therapy
- Author
-
Cheng Liu, Kim D. Janda, Nigel Mackman, Subhash C. Sinha, Longwu Ye, Dong Xue, Katerina Capkova, Pengfei Jiang, and Yuan Liu
- Abstract
Tissue factor (TF), a rate-limiting enzyme cofactor in activating coagulation, is highly expressed in a wide spectrum of human tumor and tumor stromal cells. Using TF-deficient cancer cells and a conditional TF-knockout mouse model, we show that TF expressed by cancer cells, but not by the host stromal cells, plays a critical role in tumor growth. In the tumor microenvironment, serum coagulation factors are readily extravasated and therefore lead to continuous TF-mediated activation of coagulation proteases. To target this highly specific cascade of serine proteases, we used both a TF:VIIa inhibitor and doxorubicin-based prodrugs that are selectively activated by TF:FVIIa, FXa, and thrombin. Treatment with the TF:FVIIa inhibitor led to growth retardation in breast tumor models. In contrast, treatment with the prodrug eliminated primary tumor cells and lung metastases without apparent toxicity. Our findings offer preclinical proof of principle that targeting the coagulation cascade that is activated in the tumor microenvironment can be a highly effective approach for cancer therapy. Cancer Res; 71(20); 6492–502. ©2011 AACR.
- Published
- 2023
43. Loss of P2Y1 receptor desensitization does not impact hemostasis or thrombosis despite increased platelet reactivity in vitro
- Author
-
David S. Paul, Tasha N. Blatt, Wyatt J. Schug, Emily G. Clark, Tomohiro Kawano, Nigel Mackman, Sebastian Murcia, Kathryn O. Poe, Jean Marie N. Mwiza, T. Kendall Harden, Wolfgang Bergmeier, and Robert A. Nicholas
- Subjects
Hematology - Published
- 2023
44. Tissue factor activity of small and large extracellular vesicles in different diseases
- Author
-
Ana T.A. Sachetto, Sierra J. Archibald, Yohei Hisada, Axel Rosell, Sebastian Havervall, Nick van Es, Rienk Nieuwland, Robert A. Campbell, Elizabeth A. Middleton, Matthew T. Rondina, Charlotte Thålin, Nigel Mackman, Vascular Medicine, ACS - Pulmonary hypertension & thrombosis, Laboratory for Experimental Clinical Chemistry, ACS - Microcirculation, CCA - Cancer biology and immunology, and CCA - Imaging and biomarkers
- Subjects
sepsis ,cancer ,COVID-19 ,Hematology ,extracellular vesicles ,tissue factor ,procoagulant activity - Abstract
Background: Circulating procoagulant extracellular vesicles (EVs) are increased in diseases, such as cancer, sepsis, and COVID-19. EV tissue factor (TF) activity is associated with disseminated intravascular coagulation in sepsis and venous thrombosis in patients with pancreatic cancer and COVID-19. EVs are commonly isolated by centrifugation at ∼20,000 g. Objectives: In this study, we analyzed the TF activity of 2 EV populations enriched for large and small EVs in patients with either sepsis, pancreatic cancer, or COVID-19. Methods: EVs were isolated from plasma by sequential centrifugation at 20,000 g (large EVs, LEVs) and then 100,000 g (small EVs, SEVs). We analyzed EVs from plasma prepared from whole blood samples from healthy individuals with or without lipopolysaccharide (LPS) stimulation as well as EVs from plasma samples from patients with either sepsis, pancreatic cancer, or COVID-19. TF-dependent (EV-TF activity) and TF-independent factor Xa (FXa) generation of the EVs was measured. Results: LPS increased EV-TF activity in LEVs but not SEVs. Similarly, in 2 patients with sepsis who had EV-TF activity above the background of the assay we observed EV-TF activity in LEVs but not SEVs. Patients with pancreatic cancer or COVID-19 had circulating EV-TF activity in both LEVs and SEVs. Conclusion: We recommend that EVs are isolated from plasma from patients by centrifugation at 100,000 g rather than 20,000 g to obtain a more accurate measure of levels of circulating EV-TF activity.
- Published
- 2023
45. Direct Oral Anticoagulants and Coronary Artery Disease
- Author
-
Ravi Choxi, Kunal Kapoor, Nigel Mackman, and Ion S. Jovin
- Subjects
Percutaneous Coronary Intervention ,Atrial Fibrillation ,Administration, Oral ,Anticoagulants ,Humans ,Hemorrhage ,Coronary Artery Disease ,Cardiology and Cardiovascular Medicine ,Platelet Aggregation Inhibitors - Abstract
Coronary artery disease is a leading cause of morbidity and mortality worldwide. Acute coronary syndrome as a first presentation is common and patients with established disease have a high rate of recurrent ischemic events, despite antiplatelet therapy. Over the past several years, direct oral anticoagulants have become available and have been studied in patients with coronary artery disease. These medications directly inhibit either thrombin or factor Xa which contribute to atherothrombosis. This review will summarize the clinical data regarding the use of direct oral anticoagulants in different patient populations with coronary disease and the balance between protection against ischemia and bleeding. Additionally, the review will summarize the available data on the use of direct oral anticoagulants periprocedurally in patients undergoing percutaneous coronary intervention. The future direction of coronary artery disease and the role of direct oral anticoagulants will rely on further studies determining the optimal combination of antiplatelet and oral anticoagulant regimens that derive ischemic benefit without increased rates of bleeding. Additional upstream blockade of the coagulation cascade with factor XIIa and factor XIa inhibitors may also improve treatment in the future.
- Published
- 2022
46. Effect of combining aspirin and rivaroxaban on atherosclerosis in mice
- Author
-
Steven P. Grover, Taylor Coughlin, Salma M. Fleifil, Jens J.N. Posma, Henri H.M. Spronk, Stefan Heitmeier, A. Phillip Owens, Nigel Mackman, Biochemie, RS: Carim - B04 Clinical thrombosis and Haemostasis, and Interne Geneeskunde
- Subjects
Male ,Mice, Knockout ,ANTICOAGULANT ,Inflammation ,Aspirin ,COAGULATION ,PROGRESSION ,Atherosclerosis ,DEFICIENT MICE ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,DOUBLE-BLIND ,INITIATION ,Apolipoproteins E ,Receptors, LDL ,Rivaroxaban ,CLOPIDOGREL ,Animals ,CORONARY-ARTERY-DISEASE ,Cardiology and Cardiovascular Medicine ,PLATELET ,Aorta - Abstract
BACKGROUND AND AIMS: The platelet inhibitor aspirin reduces inflammation and atherosclerosis in both apolipoprotein E deficient (apoE-/-) mice and low-density lipoprotein receptor deficient (Ldlr-/-) mice. Similarly, the factor Xa inhibitor rivaroxaban reduces atherosclerosis in both apoE-/- and Ldlr-/- mice. We tested the hypothesis that the combination of aspirin and rivaroxaban reduces atherosclerosis in mice to a greater extent than either agent alone.METHODS: Male Ldlr-/- mice were fed a western-type diet for 12 weeks to induce atherosclerosis. Cohorts of mice received aspirin in the water and/or rivaroxaban in the diet. Atherosclerosis and lesion composition were measured in the aortic sinus and the aorta. Expression of 55 proteins in the aorta and plasma was determined using multiplex ELISA assays.RESULTS: Aspirin alone, rivaroxaban alone, and the combination of both agents significantly reduced atherosclerosis in the Ldlr-/- mice compared with control Ldlr-/- mice fed a western-type diet. However, there were no significant differences in atherosclerosis in the group receiving aspirin and rivaroxaban compared with the groups that received aspirin or rivaroxaban alone. Aspirin, rivaroxaban and the combination reduced macrophage content and apoptosis in the lesions compared with controls but there was no difference between the three treatment groups. We observed statistically significant changes in the expression of a small number of proteins in the aorta and plasma in mice treated with aspirin and/or rivaroxaban.CONCLUSIONS: Contrary to our expectation, the combination of aspirin and rivaroxaban did not further reduce atherosclerosis in Ldlr-/- mice beyond the level observed with each agent alone.
- Published
- 2022
47. C1 inhibitor deficiency enhances contact pathway mediated activation of coagulation and venous thrombosis
- Author
-
Steven Philip Grover, Tomohiro Kawano, Jun Wan, Pansakorn Tanratana, Zsofia Polai, Young Jun Shim, Omri Snir, Sigrid K. Brækkan, Sophia Dhrolia, Rohan Raj Kasthuri, Tem Bendapudi, Keith R McCrae, Alisa S. Wolberg, John-Bjarne Hansen, Henriette Farkas, and Nigel Mackman
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
C1 inhibitor (C1INH) is a multifunctional serine protease inhibitor that functions as a major negative regulator of several biological pathways, including the contact pathway of blood coagulation. In humans, congenital C1INH deficiency results in a rare episodic bradykinin-mediated swelling disorder called hereditary angioedema (HAE). Patients with C1INH deficiency associated HAE (C1INH-HAE) have increased circulating markers of activation of coagulation. Further, we recently reported that patients with C1INH-HAE had a moderate but significant increased risk of venous thromboembolism. To further investigate the impact of C1INH deficiency on activation of coagulation and thrombosis, we conducted studies using patient samples and mouse models. Plasmas from patients with C1INH-HAE supported significantly increased contact pathway-mediated thrombin generation. C1INH-deficient mice, which have been used as a model of C1INH-HAE, had significantly increased baseline circulating levels of prothrombin fragment 1+2 and thrombin-antithrombin complexes. In addition, whole blood from C1INH-deficient mice supported significantly increased contact pathway-mediated thrombin generation. Importantly, C1INH-deficient mice exhibited significantly enhanced venous, but not arterial, thrombus formation. Further, purified human C1INH normalized contact pathway-mediated thrombin generation and venous thrombosis in C1INH-deficient mice. These findings suggest a key role for endogenous C1INH as a negative regulator of contact pathway-mediated coagulation in humans and mice. Further, this work identifies endogenous C1INH as an important negative regulator of venous thrombus formation in mice complementing the phenotype associated with C1INH-HAE.
- Published
- 2023
48. High plasma levels of C1-inhibitor are associated with lower risk of future venous thromboembolism
- Author
-
Steven P. Grover, Omri Snir, Kristian Hindberg, Tatianna M. Englebert, Sigrid K. Braekkan, Vânia M. Morelli, Søren B. Jensen, Alisa S. Wolberg, Tom Eirik Mollnes, Thor Ueland, Nigel Mackman, and John-Bjarne Hansen
- Subjects
Hematology - Abstract
Background C1-inhibitor (C1INH) is a broad-acting serine protease inhibitor with anticoagulant activity. The impact of C1INH plasma levels within the normal physiological range on risk of venous thromboembolism (VTE) is unknown. We assessed the association of plasma C1INH levels and VTE risk and evaluated the impact of C1INH on thrombin and plasmin generation in ex vivo assays. Methods A nested case-control study with 405 patients with VTE and 829 age- and sex-matched controls was derived from the Tromsø Study. Odds ratios (ORs) with 95% confidence intervals (95% CI) for VTE were estimated across plasma C1INH quartiles. Genetic regulation of C1INH was explored using quantitative trait loci analysis of whole exome sequencing data. The effect of plasma C1INH levels on coagulation was evaluated ex vivo by calibrated automated thrombography. Results Individuals with C1INH levels in the highest quartile had a lower risk of VTE (OR 0.68, 95% CI: 0.49-0.96) compared with those with C1INH in the lowest quartile. In subgroup analysis, the corresponding ORs were 0.60 (95% CI: 0.39-0.89) for deep vein thrombosis and 0.85 (95% CI: 0.52-1.38) for pulmonary embolism, respectively. No significant genetic determinants of plasma C1INH levels were identified. Addition of exogenous C1INH to normal human plasma reduced thrombin generation triggered by an activator of the intrinsic coagulation pathway, but not when triggered by an activator of the extrinsic coagulation pathway. Conclusions High plasma levels of C1INH were associated with lower risk of VTE, and C1INH inhibited thrombin generation initiated by the intrinsic coagulation pathway ex vivo.
- Published
- 2023
49. A novel mouse whole blood thrombin generation assay sensitive to FXI- and FIX-mediated amplification of coagulation
- Author
-
Jun Wan, Pansakorn Tanratana, Mark Roest, Andras Gruber, Rafal Pawlinski, Alisa S. Wolberg, Nigel Mackman, and Steven P. Grover
- Subjects
Hematology - Abstract
Thrombin generation (TG) assays serve as a valuable tool to study the amplifying roles of intrinsic pathway factors in human coagulation and provide functional insights into the increased bleeding observed in individuals deficient in factors (F) XI, IX, or VIII. Mice are used extensively in hemostasis research owing to the availability of coagulation factor–deficient mice. However, phenotypic differences between mouse and human TG have become apparent. In this study, we describe a novel, calibrated mouse whole blood (WB) TG assay used to assess the amplifying roles of intrinsic pathway factors in mouse coagulation. WB- and plasma-TG was triggered with either silica or tissue factor (TF) in samples from wild-type mice and mice deficient for FXII, FXI, or FIX. Expectedly, silica-triggered WB-TG and platelet-poor plasma (PPP)-TG were significantly reduced by deficiencies for FXII, FXI, or FIX. FXII deficiency had no effect on WB-TG or PPP-TG when triggered with TF. However, FXI deficiency resulted in significantly reduced WB-TG triggered by low concentrations of TF but had no effect on TF-triggered PPP-TG. FIX deficiency profoundly reduced WB-TG when triggered by low or high concentrations of TF whereas TG in PPP or platelet-rich plasma was only moderately reduced under these conditions. In conclusion, we have developed a novel mouse WB-TG assay with enhanced sensitivity to FXI- and FIX-dependent amplification of coagulation compared with an established plasma-TG assay. The enhanced sensitivity of WB-TG to FXI and FIX-dependent amplification of coagulation suggests an important role of blood cells in this process.
- Published
- 2022
50. Circulating Tumor Cells and Cancer-Associated Venous Thrombosis: A Missing Link
- Author
-
Nigel Mackman and Yohei Hisada
- Subjects
Cardiology and Cardiovascular Medicine - Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.