1
|
Gendron N, Planquette B, Roche A, Chocron R, Helley D, Philippe A, Morange PE, Gaussem P, Sanchez O, Smadja DM. Circulating CD34 + Cells: A New Biomarker of Residual Pulmonary Vascular Obstruction after Pulmonary Embolism. Stem Cell Rev Rep 2025:10.1007/s12015-025-10865-0. [PMID: 40085375 DOI: 10.1007/s12015-025-10865-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Pulmonary embolism (PE) is a life-threatening condition with long-term complications, including residual pulmonary vascular obstruction (RPVO). RPVO is associated with an increased risk of venous thromboembolism recurrence, chronic symptoms, and reduced quality of life. We hypothesize that an endothelial activation and vascular injury play a central role in the pathophysiology of RPVO. This prospective monocentric study investigates the potential of circulating biomarkers, including CD34⁺ cells, circulating endothelial cells (CECs), and platelet-derived growth factor BB (PDGF-BB), as indicators of vascular sequelae and predictors of RPVO. We included 56 patients with a first episode of PE. Biomarker levels were measured at PE diagnosis and six months later, coinciding with RPVO assessment using ventilation-perfusion lung scans. This defined groups of patients with (RPVO ≥ 10%) and without (RPVO < 10%) perfusion defects. Associations between biomarker levels, presence of perfusion defects, and clinical parameters were analyzed. At PE diagnosis, CEC and PDGF-BB levels were significantly elevated in patients compared to healthy controls, while CD34⁺ levels showed no difference. At the six-month follow-up, patients with perfusion defects exhibited significantly lower CD34⁺ cell levels compared to those without (median 1440 cells/mL vs. 2960 cells/mL). No significant differences in CEC or PDGF-BB levels were observed at follow-up. In conclusion, low CD34⁺ cell levels at RPVO assessment suggest a decreased regenerative potential contributing to thrombus persistence. CD34⁺ cells may serve as biomarkers for perfusion defects and warrant further study for their potential role in guiding clinical management of PE complications.
Collapse
Affiliation(s)
- Nicolas Gendron
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
- F-CRIN INNOVTE, Saint-Étienne, France
| | - Benjamin Planquette
- Respiratory Medicine Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
| | - Anne Roche
- INSERM UMR-S 999 « Pulmonary Hypertension: Pathophysiology and Novel Therapies », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Faculté de Médecine, HPPIT, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Richard Chocron
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
- Emergency Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
| | - Dominique Helley
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
| | - Aurélien Philippe
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
| | - Pierre-Emmanuel Morange
- F-CRIN INNOVTE, Saint-Étienne, France
- Laboratory of Hematology, La Timone Hospital, Marseille, France
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
| | - Pascale Gaussem
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
| | - Olivier Sanchez
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
- F-CRIN INNOVTE, Saint-Étienne, France
- Respiratory Medicine Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
| | - David M Smadja
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France.
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France.
- F-CRIN INNOVTE, Saint-Étienne, France.
| |
Collapse
|
2
|
Kell DB, Pretorius E. The Proteome Content of Blood Clots Observed Under Different Conditions: Successful Role in Predicting Clot Amyloid(ogenicity). Molecules 2025; 30:668. [PMID: 39942772 PMCID: PMC11820299 DOI: 10.3390/molecules30030668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
A recent analysis compared the proteome of (i) blood clots seen in two diseases-sepsis and long COVID-when blood was known to have clotted into an amyloid microclot form (as judged by staining with the fluorogenic amyloid stain thioflavin T) with (ii) that of those non-amyloid clots considered to have formed normally. Such fibrinaloid microclots are also relatively resistant to fibrinolysis. The proteins that the amyloid microclots contained differed markedly both from the soluble proteome of typical plasma and that of normal clots, and also between the diseases studied (an acute syndrome in the form of sepsis in an ITU and a chronic disease represented by Long COVID). Many proteins in the amyloid microclots were low in concentration in plasma and were effectively accumulated into the fibres, whereas many other abundant plasma proteins were excluded. The proteins found in the microclots associated with the diseases also tended to be themselves amyloidogenic. We here ask effectively the inverse question. This is: can the clot proteome tell us whether the clots associated with a particular disease contained proteins that are observed uniquely (or are highly over-represented) in known amyloid clots relative to normal clots, and thus were in fact amyloid in nature? The answer is in the affirmative in a variety of major coagulopathies, viz., venous thromboembolism, pulmonary embolism, deep vein thrombosis, various cardiac issues, and ischaemic stroke. Galectin-3-binding protein and thrombospondin-1 seem to be especially widely associated with amyloid-type clots, and the latter has indeed been shown to be incorporated into growing fibrin fibres. These may consequently provide useful biomarkers with a mechanistic basis.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Søltofts Plads 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch Private Bag X1, Matieland 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch Private Bag X1, Matieland 7602, South Africa
| |
Collapse
|
3
|
Smit ER, Kreft IC, Camilleri E, Burggraaf-van Delft JLI, van Rein N, van Vlijmen BJ, Hulshof AM, van Bussel BC, van Rosmalen F, van der Zwaan C, van de Berg T, Henskens Y, ten Cate H, Coutinho JM, Kruip MJ, Eikenboom JJ, Hoogendijk AJ, Cannegieter SC, van den Biggelaar M. Exploration of the plasma proteomic profile of patients at risk of thromboembolic events. Res Pract Thromb Haemost 2025; 9:102713. [PMID: 40224277 PMCID: PMC11986537 DOI: 10.1016/j.rpth.2025.102713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/15/2024] [Indexed: 04/15/2025] Open
Abstract
Background The elevated health burden of thromboembolic events necessitates development of blood-based risk monitoring tools. Objectives We explored the potential of mass spectrometry-based plasma proteomics to provide insights into underlying plasma protein signatures associated with treatment and occurrence of thromboembolic events. Methods Utilizing a high-throughput, data-independent acquisition, discovery-based proteomics workflow, we analyzed 434 plasma proteomes from different groups of individuals with elevated risk of thromboembolic events, including individuals I) on vitamin K antagonists (VKAs; n = 130), II) with a prior venous thromboembolism (n = 10), III) with acute cerebral venous sinus thrombosis (n = 10, and IV) with SARS-CoV-2 infection (n = 67). Plasma protein levels measured with mass spectrometry were correlated with international normalized ratio and conventional clinical laboratory measurements. Plasma profile differences between different groups were assessed using principal component analysis, moderated t-test, and clustering analysis. Results Plasma protein levels were in agreement with conventional clinical laboratory parameters, including albumin and fibrinogen. Levels of vitamin K-dependent proteins inversely correlated with international normalized ratio. In the individual studies, we found decreased levels of vitamin K-dependent coagulation proteins in patients on VKAs, alterations in inflammatory signatures among CVST patients and a distinctive signature indicative of SARS-CoV-2 infection. However, no protein signature associated with a thromboembolic event could be identified neither in individual nor combined studies. Conclusion Although VKA treatment-specific and disease-specific signatures were captured, our study highlights that the challenges of discovering biomarkers in patients at risk of thromboembolic events lie in the heterogeneity of individual plasma profiles in relation to treatment and etiology.
Collapse
Affiliation(s)
- Eva R. Smit
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Iris C. Kreft
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Eleonora Camilleri
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Nienke van Rein
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bart J.M. van Vlijmen
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne-Marije Hulshof
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Central Diagnostic Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Bas C.T. van Bussel
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Intensive Care Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Frank van Rosmalen
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Intensive Care Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Carmen van der Zwaan
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Tom van de Berg
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Yvonne Henskens
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Central Diagnostic Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Hugo ten Cate
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jonathan M. Coutinho
- Department of Neurology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Marieke J.H.A. Kruip
- Department of Hematology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen J.C. Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Arie J. Hoogendijk
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Suzanne C. Cannegieter
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
4
|
Kim BK, Yang WJ, Seong YS, Choi YJ, Park HJ, Byun MK, Chang YS, Cho JH, Kim CY. Comparative Assessment of Acute Pulmonary Effects Induced by Heat-Not-Burn Tobacco Aerosol Inhalation in a Murine Model. Int J Mol Sci 2025; 26:1135. [PMID: 39940903 PMCID: PMC11817633 DOI: 10.3390/ijms26031135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Tobacco smoking remains a major global health concern, causing preventable deaths and economic strain. Although new tobacco products such as heat-not-burn (HnB) are safer alternatives to traditional cigarettes, research on their associated risks remains limited. This study aimed to investigate the effects of HnB smoke exposure on the lungs compared to those of traditional cigarettes and the combined use of HnB and cigarettes using experiments with a mouse model. We quantitatively analyzed changes in the levels of 92 blood plasma proteins using the proximity extension assay method and observed significant changes in their levels in mice exposed to different smoke conditions; specifically, the levels of certain proteins, including Ccl20, Cxcl1, and Pdgfb, increased in the HnB smoke-exposed group, suggesting activation of nicotine pathways. Comparative analysis with traditional cigarette smoke-exposed mice further highlighted similarities and differences in their protein expression profiles. This study contributes to an improved understanding of the biological mechanisms underlying the harmful effects of alternative nicotine delivery systems and identifies potential biomarkers associated with the harmful effects of HnB smoke exposure. However, the precise impact of nicotine on the immune system may be influenced by various factors, necessitating further research.
Collapse
Affiliation(s)
- Beong Ki Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Dongguk University College of Medicine, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea;
| | - Won Jin Yang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Ye Seul Seong
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul 06273, Republic of Korea;
| | - Yong Jun Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Hye Jung Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Min Kwang Byun
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Yoon Soo Chang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Jae Hwa Cho
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
| | - Chi Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (W.J.Y.); (Y.J.C.); (H.J.P.); (M.K.B.); (Y.S.C.); (J.H.C.)
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University College of Medicine, Korea University Ansan Hospital, Ansan 15355, Republic of Korea
| |
Collapse
|
5
|
Vostatek R, Ay C. Biological Aging and Venous Thromboembolism: A Review of Telomeres and Beyond. Biomedicines 2024; 13:15. [PMID: 39857599 PMCID: PMC11759860 DOI: 10.3390/biomedicines13010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Although venous thromboembolism (VTE) is the third most common cardiovascular disease, and the risk of VTE increases sharply with advancing age, approximately 40% of VTE cases are currently classified as unprovoked, highlighting the importance of risk factor research. While chronological aging is associated with the risk of VTE, the association with biological aging remains unclear. Biological aging is highly complex, influenced by several dysregulated cellular and biochemical mechanisms. In the last decade, advancements in omics methodologies provided insights into the molecular complexity of biological aging. Techniques such as high-throughput genomics, epigenomics, transcriptomics, proteomics, and metabolomics analyses identified and quantified numerous epigenetic markers, transcripts, proteins, and metabolites. These methods have also revealed the molecular alterations organisms undergo as they age. Despite the progress, there is still a lack of consensus regarding the methods for assessing and validating these biomarkers, and their application lacks standardization. This review gives an overview of biomarkers of biological aging, including telomere length, and their potential role for VTE. Furthermore, we critically examine the advantages and disadvantages of the proposed methods and discuss possible future directions for investigating biological aging in VTE.
Collapse
Affiliation(s)
| | - Cihan Ay
- Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
6
|
Brækkan SK, Onsaker AL, Nøst TH, Tang W, Hindberg KD, Morelli VM, Guan W, Jonasson C, Folsom AR, Hveem K, Hansen JB. The Plasma Proteome and Risk of Future Venous Thromboembolism-Results from the HUNT Study. Thromb Haemost 2024. [PMID: 39586830 DOI: 10.1055/a-2484-0836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
BACKGROUND This study aimed to identify novel plasma proteins associated with first-lifetime venous thromboembolism (VTE) and molecular pathways involved in VTE pathogenesis. METHODS A case-cohort comprising incident VTE cases (n = 294) and a randomly sampled age- and sex-weighted subcohort (n = 1,066) was derived from the Trøndelag Health Study (HUNT3, n = 50,800). Blood samples were collected and stored at cohort inclusion (2006-2008), and participants were followed up to 5 years. Proteome-wide analyses was performed using the 7k SomaScan® proteomics platform, and weighted Cox-regression models adjusted for age, sex, and sample batch were conducted, with the Bonferroni method applied to account for multiple testing. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were applied on the top-ranked 200 proteins associated with VTE. RESULTS Out of 7,288 human proteins, 7 proteins were significantly associated with higher VTE risk with p-value <6.9 × 10-6 (hazard ratios per 1 standard deviation increase in protein levels ranging from 1.39 to 1.86). Except for coagulation factor VIII and tumor necrosis factor soluble receptor II, these proteins were novel associations and included collagen alpha-3(VI):BPTI/Kunitz inhibitor, histo-blood group ABO system transferase, peroxidasin, human epididymis protein 4, and regulator of G protein signaling 3. KEGG analyses of the top-ranked 200 proteins revealed significant pathway enrichment of nine proteins in the complement (mainly lectin pathway) and coagulation (mainly intrinsic pathway) cascades. CONCLUSION Our proteome-wide analysis led to discovery of five novel protein candidates associated with 5-year risk of future VTE. KEGG analyses supported an interplay between the complement and coagulation pathways in the pathogenesis of VTE.
Collapse
Affiliation(s)
- Sigrid K Brækkan
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
- Thrombosis Research group (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Asbjørn L Onsaker
- Thrombosis Research group (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Therese H Nøst
- HUNT Center for Molecular and Clinical Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Community Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, Minnesota, United States
| | - Kristian D Hindberg
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Vania M Morelli
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
- Thrombosis Research group (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, Minnesota, United States
| | - Christian Jonasson
- HUNT Research Center, Norwegian University of Science and Technology, Levanger, Norway
| | - Aaron R Folsom
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, Minnesota, United States
| | - Kristian Hveem
- HUNT Center for Molecular and Clinical Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Center, Norwegian University of Science and Technology, Levanger, Norway
| | - John-Bjarne Hansen
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
- Thrombosis Research group (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
7
|
Camilleri E, Kruijt M, den Exter PL, Cannegieter SC, van Rein N, Cobbaert CM, van Vlijmen BJM, Ruhaak LR. Quantitative protein mass spectrometry for multiplex measurement of coagulation and fibrinolytic proteins towards clinical application: What, why and how? Thromb Res 2024; 241:109090. [PMID: 39032389 DOI: 10.1016/j.thromres.2024.109090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024]
Abstract
Plasma proteins involved in coagulation and fibrinolysis are essential to hemostasis. Consequently, their circulating levels and functionality are critical in bleeding and thrombosis development. Well-established laboratory tests to assess these are available; however, said tests do not allow high multiplicity, require large volumes of plasma and are often costly. A novel technology to quantify plasma proteins is quantitative protein mass spectrometry (QPMS). Aided by stable isotope-labeled internal standards a large number of proteins can be quantified in one single analytical run requiring <30 μL of plasma. This provides an opportunity to improve insight in the etiology and prognosis of bleeding and thrombotic disorders, in which the balance between different proteins plays a crucial role. This manuscript aims to give an overview of the QPMS potential applications in thrombosis and hemostasis research (quantifying the 38 proteins assigned to coagulation and fibrinolysis by the KEGG database), but also to explore the potential and hurdles if designed for clinical practice. Advantages and limitations of QPMS are described and strategies for improved analysis are proposed, using as an example the test requirements for antithrombin. Application of this technology in the future could represent a step towards individualized patient care.
Collapse
Affiliation(s)
- Eleonora Camilleri
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mirjam Kruijt
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Paul L den Exter
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Suzanne C Cannegieter
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Nienke van Rein
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Pharmacy, Leiden University Medical Center, Leiden, the Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Bart J M van Vlijmen
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
8
|
Vacik Díaz R, Munsch G, Iglesias MJ, Pallares Robles A, Ibrahim-Kosta M, Nourse J, Khan E, Castoldi E, Saut N, Boland A, Germain M, Deleuze JF, Odeberg J, Morange PE, Danckwardt S, Tregouët DA, Goumidi L. Plasma levels of complement components C5 and C9 are associated with thrombin generation. J Thromb Haemost 2024; 22:2531-2542. [PMID: 38838952 DOI: 10.1016/j.jtha.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/30/2024] [Accepted: 04/24/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND The thrombin generation assay (TGA) evaluates the potential of plasma to generate thrombin over time, providing a global picture of an individual's hemostatic balance. OBJECTIVES This study aimed to identify novel biological determinants of thrombin generation using a multiomics approach. METHODS Associations between TGA parameters and plasma levels of 377 antibodies targeting 236 candidate proteins for cardiovascular risk were tested using multiple linear regression analysis in 770 individuals with venous thrombosis from the Marseille Thrombosis Association (MARTHA) study. Proteins associated with at least 3 TGA parameters were selected for validation in an independent population of 536 healthy individuals (Etablissement Français du Sang Alpes-Méditerranée [EFS-AM]). Proteins with strongest associations in both groups underwent additional genetic analyses and in vitro experiments. RESULTS Eighteen proteins were associated (P < 1.33 × 10⁻4) with at least 3 TGA parameters in MARTHA, among which 13 demonstrated a similar pattern of associations in EFS-AM. Complement proteins C5 and C9 had the strongest associations in both groups. Ex vivo supplementation of platelet-poor plasma with purified C9 protein had a significant dose-dependent effect on TGA parameters. No effect was observed with purified C5. Several single nucleotide polymorphisms associated with C5 and C9 plasma levels were identified, with the strongest association for the C5 missense variant rs17611, which was associated with a decrease in C5 levels, endogenous thrombin potential, and peak in MARTHA. No association of this variant with TGA parameters was observed in EFS-AM. CONCLUSION This study identified complement proteins C5 and C9 as potential determinants of thrombin generation. Further studies are warranted to establish causality and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Rocío Vacik Díaz
- Cardiovascular and Nutrition Research Center Centre de recherche en CardioVasculaire et Nutrition (C2VN), Aix-Marseille University, Institut national de la santé et de la recherche médicale 1263, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement 1260, Marseille, France; Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg, Mainz, Germany. https://twitter.com/RocioVacik
| | - Gaëlle Munsch
- Institut national de la santé et de la recherche médicale Unité Mixte de Recherche_S 1219, Bordeaux Population Health Center, University of Bordeaux, Bordeaux, France
| | - Maria Jesus Iglesias
- Science for Life Laboratory, Kungliga Tekniska högskolan-Royal Institute of Technology, Stockholm, Sweden
| | - Alejandro Pallares Robles
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg, Mainz, Germany
| | - Manal Ibrahim-Kosta
- Cardiovascular and Nutrition Research Center Centre de recherche en CardioVasculaire et Nutrition (C2VN), Aix-Marseille University, Institut national de la santé et de la recherche médicale 1263, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement 1260, Marseille, France; Department of Hematology, Centre Hospitalier Universitaire Timone, Marseille, France
| | - Jamie Nourse
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg, Mainz, Germany
| | - Essak Khan
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg, Mainz, Germany
| | - Elisabetta Castoldi
- Department of Biochemistry, Cell Biochemistry of Thrombosis and Haemostasis, Maastricht University, Maastricht, the Netherlands
| | - Noémie Saut
- Department of Hematology, Centre Hospitalier Universitaire Timone, Marseille, France
| | - Anne Boland
- Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, Evry, France
| | - Marine Germain
- Institut national de la santé et de la recherche médicale Unité Mixte de Recherche_S 1219, Bordeaux Population Health Center, University of Bordeaux, Bordeaux, France
| | - Jean-François Deleuze
- Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, Evry, France
| | - Jacob Odeberg
- Science for Life Laboratory, Kungliga Tekniska högskolan-Royal Institute of Technology, Stockholm, Sweden
| | - Pierre-Emmanuel Morange
- Cardiovascular and Nutrition Research Center Centre de recherche en CardioVasculaire et Nutrition (C2VN), Aix-Marseille University, Institut national de la santé et de la recherche médicale 1263, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement 1260, Marseille, France; Department of Hematology, Centre Hospitalier Universitaire Timone, Marseille, France
| | - Sven Danckwardt
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg, Mainz, Germany
| | - David-Alexandre Tregouët
- Institut national de la santé et de la recherche médicale Unité Mixte de Recherche_S 1219, Bordeaux Population Health Center, University of Bordeaux, Bordeaux, France
| | - Louisa Goumidi
- Cardiovascular and Nutrition Research Center Centre de recherche en CardioVasculaire et Nutrition (C2VN), Aix-Marseille University, Institut national de la santé et de la recherche médicale 1263, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement 1260, Marseille, France.
| |
Collapse
|
9
|
Turizo MJF, Patell R, Zwicker JI. Identifying novel biomarkers using proteomics to predict cancer-associated thrombosis. BLEEDING, THROMBOSIS AND VASCULAR BIOLOGY 2024; 3:120. [PMID: 38828226 PMCID: PMC11143428 DOI: 10.4081/btvb.2024.120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/08/2024] [Indexed: 06/05/2024]
Abstract
Comprehensive protein analyses of plasma are made possible by high-throughput proteomic screens, which may help find new therapeutic targets and diagnostic biomarkers. Patients with cancer are frequently affected by venous thromboembolism (VTE). The limited predictive accuracy of current VTE risk assessment tools highlights the need for new, more targeted biomarkers. Although coagulation biomarkers for the diagnosis, prognosis, and treatment of VTE have been investigated, none of them have the necessary clinical validation or diagnostic accuracy. Proteomics holds the potential to uncover new biomarkers and thrombotic pathways that impact the risk of thrombosis. This review explores the fundamental methods used in proteomics and focuses on particular biomarkers found in VTE and cancer-associated thrombosis.
Collapse
Affiliation(s)
- Maria J Fernandez Turizo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Rushad Patell
- Division of Medical Oncology and Hematology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Jeffrey I Zwicker
- Department of Medicine, Hematology Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Weil Cornell Medical College, New York, NY, United States
| |
Collapse
|
10
|
Mathews R, Hinds MT, Nguyen KP. Venous thromboembolism: diagnostic advances and unaddressed challenges in management. Curr Opin Hematol 2024; 31:122-129. [PMID: 38359323 PMCID: PMC10977858 DOI: 10.1097/moh.0000000000000809] [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] [Indexed: 02/17/2024]
Abstract
PURPOSE OF REVIEW This review summarizes recent advances in developing targeted diagnostics for venous thromboembolism (VTE) and unaddressed knowledge gaps in patient management. Without addressing these critical data needs, the morbidity in VTE patients will persist. RECENT FINDINGS Recent studies investigating plasma protein profiles in VTE patients have identified key diagnostic targets to address the currently unmet need for low-cost, confirmatory, point-of-care VTE diagnostics. These studies and a growing body of evidence from animal model studies have revealed the importance of inflammatory and vascular pathology in driving VTE, which are currently unaddressed targets for VTE therapy. To enhance the translation of preclinical animal studies, clinical quantification of thrombus burden and comparative component analyses between modeled VTE and clinical VTE are necessary. SUMMARY Lead candidates from protein profiling of VTE patients' plasma offer a promising outlook in developing low cost, confirmatory, point-of-care testing for VTE. Additionally, addressing the critical knowledge gap of quantitatively measuring clinical thrombi will allow for an array of benefits in VTE management and informing the translatability of experimental therapeutics.
Collapse
Affiliation(s)
- Rick Mathews
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Monica T Hinds
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Khanh P Nguyen
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
- Research & Development Service, VA Portland Health Care System, Portland, Oregon, USA
- Division of Vascular Surgery, Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
11
|
Gong M, Zhang Y, Chen N, Ma LL, Feng XM, Yan YX. Proteomics in Cardiovascular disease. Clin Chim Acta 2024; 557:117877. [PMID: 38537675 DOI: 10.1016/j.cca.2024.117877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 04/13/2024]
Abstract
This study focuses on recent advances in proteomics and provides an up-to-date use of this technology in identifying cardiovascular disease (CVD) biomarkers. A total of eight electronic databases (PubMed, EMBASE, Web of Science, Cochrane Library, Wanfang, Vip, Sinomed, and CNKI) were searched and five were used for integrative analysis of sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic ratio (DOR) and 1 secondary indicator area under the curve (AUC). This systematic review and integrative analysis summarized potential biomarkers previously identified by proteomics. The integrative analysis suggested that proteomics technology had high clinical value in CVD diagnosis. The findings provided new possible directions for the prevention or diagnosis of CVD.
Collapse
Affiliation(s)
- Miao Gong
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yu Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Ning Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Lin-Lin Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Xu-Man Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yu-Xiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| |
Collapse
|
12
|
Li J, Zhu J, Gray O, Sobreira DR, Wu D, Huang RT, Miao B, Sakabe NJ, Krause MD, Kaikkonen MU, Romanoski CE, Nobrega MA, Fang Y. Mechanosensitive super-enhancers regulate genes linked to atherosclerosis in endothelial cells. J Cell Biol 2024; 223:e202211125. [PMID: 38231044 PMCID: PMC10794123 DOI: 10.1083/jcb.202211125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 10/05/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024] Open
Abstract
Vascular homeostasis and pathophysiology are tightly regulated by mechanical forces generated by hemodynamics. Vascular disorders such as atherosclerotic diseases largely occur at curvatures and bifurcations where disturbed blood flow activates endothelial cells while unidirectional flow at the straight part of vessels promotes endothelial health. Integrated analysis of the endothelial transcriptome, the 3D epigenome, and human genetics systematically identified the SNP-enriched cistrome in vascular endothelium subjected to well-defined atherosclerosis-prone disturbed flow or atherosclerosis-protective unidirectional flow. Our results characterized the endothelial typical- and super-enhancers and underscored the critical regulatory role of flow-sensitive endothelial super-enhancers. CRISPR interference and activation validated the function of a previously unrecognized unidirectional flow-induced super-enhancer that upregulates antioxidant genes NQO1, CYB5B, and WWP2, and a disturbed flow-induced super-enhancer in endothelium which drives prothrombotic genes EDN1 and HIVEP in vascular endothelium. Our results employing multiomics identify the cis-regulatory architecture of the flow-sensitive endothelial epigenome related to atherosclerosis and highlight the regulatory role of super-enhancers in mechanotransduction mechanisms.
Collapse
Affiliation(s)
- Jin Li
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Jiayu Zhu
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Olivia Gray
- Department of Human Genetics, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Débora R. Sobreira
- Department of Human Genetics, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - David Wu
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Ru-Ting Huang
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Bernadette Miao
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Noboru J. Sakabe
- Department of Human Genetics, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Matthew D. Krause
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Minna U. Kaikkonen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Casey E. Romanoski
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Marcelo A. Nobrega
- Department of Human Genetics, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Yun Fang
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
- Committee on Molecular Medicine, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
13
|
Gade IL, Riddersholm SJ, Stilling-Vinther T, Brøndum RF, Bennike TB, Honoré B. A clinical proteomics study of exhaled breath condensate and biomarkers for pulmonary embolism. J Breath Res 2023; 18:016007. [PMID: 37939397 DOI: 10.1088/1752-7163/ad0aaa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/07/2023] [Indexed: 11/10/2023]
Abstract
Pulmonary embolism (PE) can be a diagnostic challenge. Current diagnostic markers for PE are unspecific and new diagnostic tools are needed. The air we exhale is a possible new source for biomarkers which can be tapped into by analysing the exhaled breath condensate (EBC). We analysed the EBC from patients with PE and controls to investigate if the EBC is a useful source for new diagnostic biomarkers of PE. We collected and analysed EBC samples from patients with suspected PE and controls matched on age and sex. Patients in whom PE was ruled out after diagnostic work-up were included in the control group to increase the sensitivity and generalizability of the identified markers. EBC samples were collected using an RTube™. The protein composition of the EBCs were analysed using data dependent label-free quantitative nano liquid chromatography-tandem mass spectrometry. EBC samples from 28 patients with confirmed PE, and 49 controls were analysed. A total of 928 EBC proteins were identified in the 77 EBC samples. As expected, a low protein concentration was determined which resulted in many proteins with unmeasurable levels in several samples. The levels of HSPA5, PEBP1 and SFTPA2 were higher and levels of POF1B, EPPK1, PSMA4, ALDOA, and CFL1 were lower in PE compared with controls. In conclusion, the human EBC contained a variety of endogenous proteins and may be a source for new diagnostic markers of PE and other diseases.
Collapse
Affiliation(s)
- Inger Lise Gade
- Department of Hematology and Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
| | | | | | - Rasmus Froberg Brøndum
- Center for Clinical Data Science, Aalborg University and Aalborg University Hospital, 9260 Gistrup, Denmark
| | - Tue Bjerg Bennike
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Bent Honoré
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
14
|
Iglesias MJ, Sanchez-Rivera L, Ibrahim-Kosta M, Naudin C, Munsch G, Goumidi L, Farm M, Smith PM, Thibord F, Kral-Pointner JB, Hong MG, Suchon P, Germain M, Schrottmaier W, Dusart P, Boland A, Kotol D, Edfors F, Koprulu M, Pietzner M, Langenberg C, Damrauer SM, Johnson AD, Klarin DM, Smith NL, Smadja DM, Holmström M, Magnusson M, Silveira A, Uhlén M, Renné T, Martinez-Perez A, Emmerich J, Deleuze JF, Antovic J, Soria Fernandez JM, Assinger A, Schwenk JM, Souto Andres JC, Morange PE, Butler LM, Trégouët DA, Odeberg J. Elevated plasma complement factor H related 5 protein is associated with venous thromboembolism. Nat Commun 2023; 14:3280. [PMID: 37286573 PMCID: PMC10247781 DOI: 10.1038/s41467-023-38383-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 04/28/2023] [Indexed: 06/09/2023] Open
Abstract
Venous thromboembolism (VTE) is a common, multi-causal disease with potentially serious short- and long-term complications. In clinical practice, there is a need for improved plasma biomarker-based tools for VTE diagnosis and risk prediction. Here we show, using proteomics profiling to screen plasma from patients with suspected acute VTE, and several case-control studies for VTE, how Complement Factor H Related 5 protein (CFHR5), a regulator of the alternative pathway of complement activation, is a VTE-associated plasma biomarker. In plasma, higher CFHR5 levels are associated with increased thrombin generation potential and recombinant CFHR5 enhanced platelet activation in vitro. GWAS analysis of ~52,000 participants identifies six loci associated with CFHR5 plasma levels, but Mendelian randomization do not demonstrate causality between CFHR5 and VTE. Our results indicate an important role for the regulation of the alternative pathway of complement activation in VTE and that CFHR5 represents a potential diagnostic and/or risk predictive plasma biomarker.
Collapse
Affiliation(s)
- Maria Jesus Iglesias
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden
- Division of Internal Medicine, University Hospital of North Norway (UNN), PB100, 9038, Tromsø, Norway
- Translational Vascular Research, Department of Clinical Medicine, UiT The Arctic University of Norway, 9019, Tromsø, Norway
| | - Laura Sanchez-Rivera
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden
| | - Manal Ibrahim-Kosta
- Aix-Marseille Univ, INSERM, INRAE, C2VN, Laboratory of Haematology, CRB Assistance Publique-Hôpitaux de Marseille, HemoVasc (CRB AP-HM HemoVasc), Marseille, France
| | - Clément Naudin
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden
- Translational Vascular Research, Department of Clinical Medicine, UiT The Arctic University of Norway, 9019, Tromsø, Norway
| | - Gaëlle Munsch
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, ELEANOR, Bordeaux, France
| | - Louisa Goumidi
- Aix-Marseille Univ, INSERM, INRAE, C2VN, Laboratory of Haematology, CRB Assistance Publique-Hôpitaux de Marseille, HemoVasc (CRB AP-HM HemoVasc), Marseille, France
| | - Maria Farm
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Philip M Smith
- Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Theme of Emergency and Reparative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Florian Thibord
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, USA
- The Framingham Heart Study, Boston University, Framingham, MA, USA
| | - Julia Barbara Kral-Pointner
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Mun-Gwan Hong
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden
| | - Pierre Suchon
- Aix-Marseille Univ, INSERM, INRAE, C2VN, Laboratory of Haematology, CRB Assistance Publique-Hôpitaux de Marseille, HemoVasc (CRB AP-HM HemoVasc), Marseille, France
| | - Marine Germain
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, ELEANOR, Bordeaux, France
- Laboratory of Excellence GENMED (Medical Genomics), Bordeaux, France
| | - Waltraud Schrottmaier
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Philip Dusart
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden
- Translational Vascular Research, Department of Clinical Medicine, UiT The Arctic University of Norway, 9019, Tromsø, Norway
| | - Anne Boland
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
- Laboratory of Excellence GENMED (Medical Genomics), Evry, France
| | - David Kotol
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden
| | - Mine Koprulu
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Scott M Damrauer
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Surgery and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew D Johnson
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, USA
- The Framingham Heart Study, Boston University, Framingham, MA, USA
| | - Derek M Klarin
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Vascular Surgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Nicholas L Smith
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA, USA
| | - David M Smadja
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation), European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, 20 rue Leblanc, Paris, 75015, France
- Innovative Therapies in Haemostasis, INSERM, Université de Paris, 4 avenue de l'Observatoire, Paris, 75270, France
| | - Margareta Holmström
- Coagulation Unit, Department of Haematology, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Maria Magnusson
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Coagulation Unit, Department of Haematology, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, 171 77, Stockholm, Sweden
| | - Angela Silveira
- Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden
| | - Thomas Renné
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Centre Hamburg-Eppendorf, D-20246, Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, D-, 55131, Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, D02 YN77, Ireland
| | - Angel Martinez-Perez
- Genomics of Complex Diseases Group, Research Institute Hospital de la Santa Creu i Sant Pau. IIB Sant Pau, Barcelona, Spain
| | - Joseph Emmerich
- Department of vascular medicine, Paris Saint-Joseph Hospital Group, INSERM 1153-CRESS, University of Paris Cité, 185 rue Raymond Losserand, Paris, 75674, France
| | - Jean-Francois Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
- Laboratory of Excellence GENMED (Medical Genomics), Evry, France
- Centre D'Etude du Polymorphisme Humain, Fondation Jean Dausset, Paris, France
| | - Jovan Antovic
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Jose Manuel Soria Fernandez
- Genomics of Complex Diseases Group, Research Institute Hospital de la Santa Creu i Sant Pau. IIB Sant Pau, Barcelona, Spain
| | - Alice Assinger
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Jochen M Schwenk
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden
| | - Joan Carles Souto Andres
- Unitat d'Hemostàsia i Trombosi. Hospital de la Santa Creu i Sant Pau and IIB-Sant Pau, Barcelona, Spain
| | - Pierre-Emmanuel Morange
- Aix-Marseille Univ, INSERM, INRAE, C2VN, Laboratory of Haematology, CRB Assistance Publique-Hôpitaux de Marseille, HemoVasc (CRB AP-HM HemoVasc), Marseille, France
| | - Lynn Marie Butler
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden
- Translational Vascular Research, Department of Clinical Medicine, UiT The Arctic University of Norway, 9019, Tromsø, Norway
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - David-Alexandre Trégouët
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, ELEANOR, Bordeaux, France.
- Laboratory of Excellence GENMED (Medical Genomics), Bordeaux, France.
| | - Jacob Odeberg
- Science for Life Laboratory, Department of Protein Science, CBH, KTH Royal Institute of Technology, SE-171 21, Stockholm, Sweden.
- Division of Internal Medicine, University Hospital of North Norway (UNN), PB100, 9038, Tromsø, Norway.
- Translational Vascular Research, Department of Clinical Medicine, UiT The Arctic University of Norway, 9019, Tromsø, Norway.
- Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden.
- Coagulation Unit, Department of Haematology, Karolinska University Hospital, SE-171 76, Stockholm, Sweden.
| |
Collapse
|
15
|
Letunica N, McCafferty C, Swaney E, Cai T, Monagle P, Ignjatovic V, Attard C. Proteomic Applications and Considerations: From Research to Patient Care. Methods Mol Biol 2023; 2628:181-192. [PMID: 36781786 DOI: 10.1007/978-1-0716-2978-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Despite technological advancements in the field of proteomics, the rate at which serum and plasma biomarkers identified using proteomic approaches are translated into clinical use remains extremely low. In this chapter, we describe recent technological advancements and analytical strategies in proteomic methods. We also describe the progress of proteomic blood-based biomarkers to date and discuss what the future of proteomics might entail with the use of multi-omic approaches and implementing machine learning on large proteomic datasets. Lastly, we provide several key considerations for biomarker studies, ranging from sample type to the use of reference samples, in order to achieve progress from bench to bedside, ultimately improving patient diagnosis, disease, and/or therapeutic monitoring and care.
Collapse
Affiliation(s)
- Natasha Letunica
- Haematology Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Conor McCafferty
- Haematology Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Ella Swaney
- Haematology Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Tengyi Cai
- Haematology Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul Monagle
- Haematology Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia.,Department of Clinical Haematology, Royal Children's Hospital, Melbourne, VIC, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Vera Ignjatovic
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia.,Institute for Clinical and Translational Research, Johns Hopkins All Children's Hospital, St. Petersburg, USA.,Department of Pediatrics, Johns Hopkins University, Baltimore, USA
| | - Chantal Attard
- Haematology Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia. .,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia. .,The Royal Children's Hospital, Parkville, VIC, Australia.
| |
Collapse
|
16
|
Yuan S, Titova OE, Zhang K, Gou W, Schillemans T, Natarajan P, Chen J, Li X, Åkesson A, Bruzelius M, Klarin D, Damrauer SM, Larsson SC. Plasma protein and venous thromboembolism: prospective cohort and mendelian randomisation analyses. Br J Haematol 2023; 201:783-792. [PMID: 36734038 DOI: 10.1111/bjh.18679] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 02/04/2023]
Abstract
We conducted cohort and Mendelian randomisation (MR) analyses to examine the associations of circulating proteins with risk of venous thromboembolism (VTE) to provide evidence basis for disease prevention and drug development. Cohort analysis was performed in 11 803 participants without baseline VTE. Cox regression was used to estimate the associations between 257 proteins and VTE risk. A machine-learning model was constructed to compare the importance of identified proteins and traditional risk factors. Genetic association data on VTE were obtained from a genome-wide meta-analysis (26 066 cases and 624 053 controls) and FinnGen (14 454 cases and 294 700 controls). The cohort analysis, including 353 incident VTE cases diagnosed during a 6.6-year follow-up, identified 21 proteins associated with VTE risk after false discovery rate correction. The machine-learning model indicated that body mass index and von Willebrand factor (vWF) made the same as well as most of the contributions to the overall model prediction. MR analysis found that genetically predicted levels of vWF, SERPINE1 (plasminogen activator inhibitor 1, known as PAI-1), EPHB4 (ephrin type-B receptor 4), TYRO3 (tyrosine-protein kinase receptor TYRO3), TNFRSF11A (tumour necrosis factor receptor superfamily member 11A), and BOC (brother of CDO) were causally associated with VTE risk.
Collapse
Affiliation(s)
- Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Olga E Titova
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Ke Zhang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Wanglong Gou
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Tessa Schillemans
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Pradeep Natarajan
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jie Chen
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Agneta Åkesson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Maria Bruzelius
- Department of Medicine, Solna, Karolinska Institute, Stockholm, Sweden
- Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Derek Klarin
- VA Palo Alto Healthcare System, Palo Alto, California, USA
- Department of Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Scott M Damrauer
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Wu T, Xu Y, Zhang L, Liang Z, Zhou X, Evans SM, Chen J. Filamin C is Essential for mammalian myocardial integrity. PLoS Genet 2023; 19:e1010630. [PMID: 36706168 PMCID: PMC9907827 DOI: 10.1371/journal.pgen.1010630] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/08/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
FLNC, encoding filamin C, is one of the most mutated genes in dilated and hypertrophic cardiomyopathy. However, the precise role of filamin C in mammalian heart remains unclear. In this study, we demonstrated Flnc global (FlncgKO) and cardiomyocyte-specific knockout (FlnccKO) mice died in utero from severely ruptured ventricular myocardium, indicating filamin C is required to maintain the structural integrity of myocardium in the mammalian heart. Contrary to the common belief that filamin C acts as an integrin inactivator, we observed attenuated activation of β1 integrin specifically in the myocardium of FlncgKO mice. Although deleting β1 integrin from cardiomyocytes did not recapitulate the heart rupture phenotype in Flnc knockout mice, deleting both β1 integrin and filamin C from cardiomyocytes resulted in much more severe heart ruptures than deleting filamin C alone. Our results demonstrated that filamin C works in concert with β1 integrin to maintain the structural integrity of myocardium during mammalian heart development.
Collapse
Affiliation(s)
- Tongbin Wu
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Yujun Xu
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Lunfeng Zhang
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Zhengyu Liang
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Xiaohai Zhou
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Sylvia M. Evans
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
18
|
Namba S, Konuma T, Wu KH, Zhou W, Okada Y. A practical guideline of genomics-driven drug discovery in the era of global biobank meta-analysis. CELL GENOMICS 2022; 2:100190. [PMID: 36778001 PMCID: PMC9903693 DOI: 10.1016/j.xgen.2022.100190] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 07/27/2022] [Accepted: 09/08/2022] [Indexed: 11/06/2022]
Abstract
Genomics-driven drug discovery is indispensable for accelerating the development of novel therapeutic targets. However, the drug discovery framework based on evidence from genome-wide association studies (GWASs) has not been established, especially for cross-population GWAS meta-analysis. Here, we introduce a practical guideline for genomics-driven drug discovery for cross-population meta-analysis, as lessons from the Global Biobank Meta-analysis Initiative (GBMI). Our drug discovery framework encompassed three methodologies and was applied to the 13 common diseases targeted by GBMI (N mean = 1,329,242). Individual methodologies complementarily prioritized drugs and drug targets, which were systematically validated by referring previously known drug-disease relationships. Integration of the three methodologies provided a comprehensive catalog of candidate drugs for repositioning, nominating promising drug candidates targeting the genes involved in the coagulation process for venous thromboembolism and the interleukin-4 and interleukin-13 signaling pathway for gout. Our study highlighted key factors for successful genomics-driven drug discovery using cross-population meta-analyses.
Collapse
Affiliation(s)
- Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| | - Takahiro Konuma
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki 569-1125, Japan
| | - Kuan-Han Wu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Wei Zhou
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Global Biobank Meta-analysis Initiative
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki 569-1125, Japan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita 565-0871, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita 565-0871, Japan
| |
Collapse
|
19
|
Smith P, Rosell A, Farm M, Bruzelius M, Aguilera Gatica K, Mackman N, Odeberg J, Thålin C. Markers of neutrophil activation and neutrophil extracellular traps in diagnosing patients with acute venous thromboembolism: A feasibility study based on two VTE cohorts. PLoS One 2022; 17:e0270865. [PMID: 35901107 PMCID: PMC9333265 DOI: 10.1371/journal.pone.0270865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 06/19/2022] [Indexed: 11/21/2022] Open
Abstract
Background Venous 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. Objectives To assess the levels of specific markers of neutrophil activation and NETs and compare their diagnostic accuracy to D-dimer. Methods We 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. Results Higher 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. Conclusions Our 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.
Collapse
Affiliation(s)
- Philip Smith
- Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden
- Theme of Emergency and Reparative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Axel Rosell
- Division of Internal Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Maria Farm
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Bruzelius
- Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden
- Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Katherina Aguilera Gatica
- Division of Internal Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jacob Odeberg
- Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden
- Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, University Hospital of North Norway (UNN), Tromsø, Norway
| | - Charlotte Thålin
- Division of Internal Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
20
|
Edfors F, Iglesias MJ, Butler LM, Odeberg J. Proteomics in thrombosis research. Res Pract Thromb Haemost 2022; 6:e12706. [PMID: 35494505 PMCID: PMC9039028 DOI: 10.1002/rth2.12706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/24/2022] Open
Abstract
A State of the Art lecture titled “Proteomics in Thrombosis Research” was presented at the ISTH Congress in 2021. In clinical practice, there is a need for improved plasma biomarker‐based tools for diagnosis and risk prediction of venous thromboembolism (VTE). Analysis of blood, to identify plasma proteins with potential utility for such tools, could enable an individualized approach to treatment and prevention. Technological advances to study the plasma proteome on a large scale allows broad screening for the identification of novel plasma biomarkers, both by targeted and nontargeted proteomics methods. However, assay limitations need to be considered when interpreting results, with orthogonal validation required before conclusions are drawn. Here, we review and provide perspectives on the application of affinity‐ and mass spectrometry‐based methods for the identification and analysis of plasma protein biomarkers, with potential application in the field of VTE. We also provide a future perspective on discovery strategies and emerging technologies for targeted proteomics in thrombosis research. Finally, we summarize relevant new data on this topic, presented during the 2021 ISTH Congress.
Collapse
Affiliation(s)
- Fredrik Edfors
- Science for Life Laboratory Department of Protein Science CBH KTH Royal Institute of Technology Stockholm Sweden
- Karolinska University Laboratory Karolinska University Hospital Stockholm Sweden
| | - Maria Jesus Iglesias
- Science for Life Laboratory Department of Protein Science CBH KTH Royal Institute of Technology Stockholm Sweden
| | - Lynn M. Butler
- Science for Life Laboratory Department of Protein Science CBH KTH Royal Institute of Technology Stockholm Sweden
- Clinical Chemistry and Blood Coagulation Research Department of Molecular Medicine and Surgery Karolinska Institute Stockholm Sweden
- Clinical Chemistry Karolinska University Laboratory Karolinska University Hospital Stockholm Sweden
- Department of Clinical Medicine The Arctic University of Norway Tromsø Norway
| | - Jacob Odeberg
- Science for Life Laboratory Department of Protein Science CBH KTH Royal Institute of Technology Stockholm Sweden
- Department of Clinical Medicine The Arctic University of Norway Tromsø Norway
- Division of Internal Medicine University Hospital of North Norway Tromsø Norway
- Coagulation Unit Department of Hematology Karolinska University Hospital Stockholm Sweden
- Department of Medicine Solna Karolinska Institute Stockholm Sweden
| |
Collapse
|
21
|
SAKÇİ D, SAYHAN MB, SALT Ö, DİBİRDİK İ. A Novel Diagnostic Marker in Pulmonary Thromboembolism? Platelet-Derived Growth Factor –β. DICLE MEDICAL JOURNAL 2022. [DOI: 10.5798/dicletip.1085918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
22
|
Détriché G, Gendron N, Philippe A, Gruest M, Billoir P, Rossi E, Guerin CL, Lokajczyk A, Brabant S, Prié D, Mirault T, Smadja DM. Gonadotropins as novel active partners in vascular diseases: Insight from angiogenic properties and thrombotic potential of endothelial colony-forming cells. J Thromb Haemost 2022; 20:230-237. [PMID: 34623025 DOI: 10.1111/jth.15549] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/06/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND The impact of estrogen and testosterone on atherosclerotic cardiovascular disease is well known, but the role of the gonadotropins follicle-stimulating hormone (FSH), luteinizing hormone (LH), and prolactin (PRL) to some extent remain less studied. OBJECTIVES To explore the angiogenic potential of gonadotropins on endothelial colony-forming cells (ECFCs). METHODS We examined the effects of various doses of gonadotropins on ECFCs obtained from cord blood by assessing colony number, proliferation, migration, and sprouting ability. Moreover, we studied thrombin generation in ECFCs exposed to gonadotropins by performing a thrombin generation assay. Finally, we determined the levels of circulating gonadotropins in 30 men, to exclude the effect of estrogen, with lower extremity arterial disease (LEAD), in comparison with age- and sex-matched controls. RESULTS Exposure to FSH, LH, or PRL resulted in an increase in ECFC migration but showed no effect on proliferation or ECFC commitment from cord blood mononuclear cells. Using a three-dimensional fibrin gel assay, we showed that ECFC sprouting was significantly enhanced by gonadotropins. Exposure to FSH also increased the thrombin generation of ECFCs exposed to FSH. Finally, FSH and LH levels in men with LEAD were higher than those in controls. CONCLUSION Gonadotropins increase ECFC-related angiogenesis and may be involved in thrombin generation in cardiovascular disease. Gonadotropins may act as biomarkers; moreover, we hypothesize that gonadotropin-blocking strategies may be a novel interesting therapeutic approach in atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Grégoire Détriché
- INSERM, Innovative Therapies in Haemostasis, Université de Paris, Paris, France
- Biosurgical Research Lab (Carpentier Foundation), Vascular Medicine Department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
| | - Nicolas Gendron
- INSERM, Innovative Therapies in Haemostasis, Université de Paris, Paris, France
- Biosurgical Research Lab (Carpentier Foundation), Hematology Department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
| | - Aurélien Philippe
- INSERM, Innovative Therapies in Haemostasis, Université de Paris, Paris, France
- Biosurgical Research Lab (Carpentier Foundation), Hematology Department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
| | - Maxime Gruest
- INSERM, Innovative Therapies in Haemostasis, Université de Paris, Paris, France
- Biosurgical Research Lab (Carpentier Foundation), Hematology Department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
| | - Paul Billoir
- Vascular Hemostasis Unit, UNIROUEN, INSERM U1096, Rouen University Hospital, Normandie Univ, Rouen, France
| | - Elisa Rossi
- INSERM, Innovative Therapies in Haemostasis, Université de Paris, Paris, France
| | - Coralie L Guerin
- INSERM, Innovative Therapies in Haemostasis, Université de Paris, Paris, France
- Cytometry Platform, Institut Curie, Paris, France
- Department of Infection and Immunity, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Anna Lokajczyk
- INSERM, Innovative Therapies in Haemostasis, Université de Paris, Paris, France
| | - Séverine Brabant
- AP-HP, Department of Functional Explorations, Necker Enfants Malades Hospital, Paris-Centre University, Paris Cedex, France
| | - Dominique Prié
- AP-HP, Department of Functional Explorations, Necker Enfants Malades Hospital, Paris-Centre University, Paris Cedex, France
| | - Tristan Mirault
- INSERM, Innovative Therapies in Haemostasis, Université de Paris, Paris, France
- Biosurgical Research Lab (Carpentier Foundation), Vascular Medicine Department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
| | - David M Smadja
- INSERM, Innovative Therapies in Haemostasis, Université de Paris, Paris, France
- Biosurgical Research Lab (Carpentier Foundation), Hematology Department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
| |
Collapse
|
23
|
Letunica N, Van Den Helm S, McCafferty C, Swaney E, Cai T, Attard C, Karlaftis V, Monagle P, Ignjatovic V. Proteomics in Thrombosis and Haemostasis. Thromb Haemost 2021; 122:1076-1084. [PMID: 34753192 DOI: 10.1055/a-1690-8897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
No Abstract.
Collapse
Affiliation(s)
- Natasha Letunica
- Haematology, Murdoch Childrens Research Institute, Parkville, Australia
| | | | - Conor McCafferty
- Haematology, Murdoch Childrens Research Institute, Parkville, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Ella Swaney
- Haematology, Murdoch Childrens Research Institute, Parkville, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Tengyi Cai
- Haematology, Murdoch Childrens Research Institute, Parkville, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Chantal Attard
- Haematology, Murdoch Childrens Research Institute, Parkville, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Vasiliki Karlaftis
- Haematology, Murdoch Childrens Research Institute, Parkville, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Paul Monagle
- Haematology, Murdoch Childrens Research Institute, Parkville, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia.,Department of Clinical Haematology, The Royal Children's Hospital Melbourne, Parkville, Australia.,Kids Cancer Centre, Sydney Children's Hospital Randwick, Randwick, Australia
| | - Vera Ignjatovic
- Haematology, Murdoch Childrens Research Institute, Parkville, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
24
|
Deutsch EW, Omenn GS, Sun Z, Maes M, Pernemalm M, Palaniappan KK, Letunica N, Vandenbrouck Y, Brun V, Tao SC, Yu X, Geyer PE, Ignjatovic V, Moritz RL, Schwenk JM. Advances and Utility of the Human Plasma Proteome. J Proteome Res 2021; 20:5241-5263. [PMID: 34672606 PMCID: PMC9469506 DOI: 10.1021/acs.jproteome.1c00657] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The study of proteins circulating in blood offers tremendous opportunities to diagnose, stratify, or possibly prevent diseases. With recent technological advances and the urgent need to understand the effects of COVID-19, the proteomic analysis of blood-derived serum and plasma has become even more important for studying human biology and pathophysiology. Here we provide views and perspectives about technological developments and possible clinical applications that use mass-spectrometry(MS)- or affinity-based methods. We discuss examples where plasma proteomics contributed valuable insights into SARS-CoV-2 infections, aging, and hemostasis and the opportunities offered by combining proteomics with genetic data. As a contribution to the Human Proteome Organization (HUPO) Human Plasma Proteome Project (HPPP), we present the Human Plasma PeptideAtlas build 2021-07 that comprises 4395 canonical and 1482 additional nonredundant human proteins detected in 240 MS-based experiments. In addition, we report the new Human Extracellular Vesicle PeptideAtlas 2021-06, which comprises five studies and 2757 canonical proteins detected in extracellular vesicles circulating in blood, of which 74% (2047) are in common with the plasma PeptideAtlas. Our overview summarizes the recent advances, impactful applications, and ongoing challenges for translating plasma proteomics into utility for precision medicine.
Collapse
Affiliation(s)
- Eric W Deutsch
- Institute for Systems Biology, Seattle, Washington 98109, United States
| | - Gilbert S Omenn
- Institute for Systems Biology, Seattle, Washington 98109, United States.,Departments of Computational Medicine & Bioinformatics, Internal Medicine, and Human Genetics and School of Public Health, University of Michigan, Ann Arbor, Michigan 48109-2218, United States
| | - Zhi Sun
- Institute for Systems Biology, Seattle, Washington 98109, United States
| | - Michal Maes
- Institute for Systems Biology, Seattle, Washington 98109, United States
| | - Maria Pernemalm
- Department of Oncology and Pathology/Science for Life Laboratory, Karolinska Institutet, 171 65 Stockholm, Sweden
| | | | - Natasha Letunica
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville 3052, Victoria, Australia
| | - Yves Vandenbrouck
- Université Grenoble Alpes, CEA, Inserm U1292, Grenoble 38000, France
| | - Virginie Brun
- Université Grenoble Alpes, CEA, Inserm U1292, Grenoble 38000, France
| | - Sheng-Ce Tao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, B207 SCSB Building, 800 Dongchuan Road, Shanghai 200240, China
| | - Xiaobo Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Philipp E Geyer
- OmicEra Diagnostics GmbH, Behringstr. 6, 82152 Planegg, Germany
| | - Vera Ignjatovic
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville 3052, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, 50 Flemington Road, Parkville 3052, Victoria, Australia
| | - Robert L Moritz
- Institute for Systems Biology, Seattle, Washington 98109, United States
| | - Jochen M Schwenk
- Affinity Proteomics, Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Tomtebodavägen 23, SE-171 65 Solna, Sweden
| |
Collapse
|
25
|
Su Y, Li Q, Zheng Z, Wei X, Hou P. Identification of genes, pathways and transcription factor-miRNA-target gene networks and experimental verification in venous thromboembolism. Sci Rep 2021; 11:16352. [PMID: 34381164 PMCID: PMC8357955 DOI: 10.1038/s41598-021-95909-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
Venous thromboembolism (VTE) is a complex, multifactorial life-threatening disease that involves vascular endothelial cell (VEC) dysfunction. However, the exact pathogenesis and underlying mechanisms of VTE are not completely clear. The aim of this study was to identify the core genes and pathways in VECs that are involved in the development and progression of unprovoked VTE (uVTE). The microarray dataset GSE118259 was downloaded from the Gene Expression Omnibus database, and 341 up-regulated and 8 down-regulated genes were identified in the VTE patients relative to the healthy controls, including CREB1, HIF1α, CBL, ILK, ESM1 and the ribosomal protein family genes. The protein-protein interaction (PPI) network and the transcription factor (TF)-miRNA-target gene network were constructed with these differentially expressed genes (DEGs), and visualized using Cytoscape software 3.6.1. Eighty-nine miRNAs were predicted as the targeting miRNAs of the DEGs, and 197 TFs were predicted as regulators of these miRNAs. In addition, 237 node genes and 4 modules were identified in the PPI network. The significantly enriched pathways included metabolic, cell adhesion, cell proliferation and cellular response to growth factor stimulus pathways. CREB1 was a differentially expressed TF in the TF-miRNA-target gene network, which regulated six miRNA-target gene pairs. The up-regulation of ESM1, HIF1α and CREB1 was confirmed at the mRNA and protein level in the plasma of uVTE patients. Taken together, ESM1, HIF1α and the CREB1-miRNA-target genes axis play potential mechanistic roles in uVTE development.
Collapse
Affiliation(s)
- Yiming Su
- Department of Vascular Surgery, LiuzhouWorker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545005, Guangxi Province, China
| | - Qiyi Li
- Department of Vascular Surgery, LiuzhouWorker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545005, Guangxi Province, China
| | - Zhiyong Zheng
- Department of Vascular Surgery, LiuzhouWorker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545005, Guangxi Province, China
| | - Xiaomin Wei
- Department of Vascular Surgery, LiuzhouWorker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545005, Guangxi Province, China
| | - Peiyong Hou
- Department of Vascular Surgery, LiuzhouWorker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545005, Guangxi Province, China.
| |
Collapse
|
26
|
An artificial neural network approach integrating plasma proteomics and genetic data identifies PLXNA4 as a new susceptibility locus for pulmonary embolism. Sci Rep 2021; 11:14015. [PMID: 34234248 PMCID: PMC8263618 DOI: 10.1038/s41598-021-93390-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023] Open
Abstract
Venous thromboembolism is the third common cardiovascular disease and is composed of two entities, deep vein thrombosis (DVT) and its potential fatal form, pulmonary embolism (PE). While PE is observed in ~ 40% of patients with documented DVT, there is limited biomarkers that can help identifying patients at high PE risk. To fill this need, we implemented a two hidden-layers artificial neural networks (ANN) on 376 antibodies and 19 biological traits measured in the plasma of 1388 DVT patients, with or without PE, of the MARTHA study. We used the LIME algorithm to obtain a linear approximate of the resulting ANN prediction model. As MARTHA patients were typed for genotyping DNA arrays, a genome wide association study (GWAS) was conducted on the LIME estimate. Detected single nucleotide polymorphisms (SNPs) were tested for association with PE risk in MARTHA. Main findings were replicated in the EOVT study composed of 143 PE patients and 196 DVT only patients. The derived ANN model for PE achieved an accuracy of 0.89 and 0.79 in our training and testing sets, respectively. A GWAS on the LIME approximate identified a strong statistical association peak (rs1424597: p = 5.3 × 10-7) at the PLXNA4 locus. Homozygote carriers for the rs1424597-A allele were then more frequently observed in PE than in DVT patients from the MARTHA (2% vs. 0.4%, p = 0.005) and the EOVT (3% vs. 0%, p = 0.013) studies. In a sample of 112 COVID-19 patients known to have endotheliopathy leading to acute lung injury and an increased risk of PE, decreased PLXNA4 levels were associated (p = 0.025) with worsened respiratory function. Using an original integrated proteomics and genetics strategy, we identified PLXNA4 as a new susceptibility gene for PE whose exact role now needs to be further elucidated.
Collapse
|
27
|
Plasma levels of von Willebrand factor and future risk of incident venous thromboembolism. Blood Adv 2021; 5:224-232. [PMID: 33570640 DOI: 10.1182/bloodadvances.2020003135] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/25/2020] [Indexed: 01/01/2023] Open
Abstract
Several case-control studies have reported elevated plasma von Willebrand factor (VWF) levels in patients with venous thromboembolism (VTE) compared with controls. However, because few studies have investigated the association in a prospective design, it is unclear whether elevated plasma VWF is a risk factor or a consequence of the VTE event. Therefore, we aimed to investigate the prospective association between plasma VWF levels and risk of VTE, as well as to perform subgroup analyses of deep vein thrombosis (DVT) and pulmonary embolism. We established a population-based nested case-control study of 414 VTE cases and 843 age- and sex-matched controls based on the Tromsø study cohort (1994-2007). Blood samples were collected at cohort baseline (1994-1995). Odds ratios (ORs) with 95% confidence intervals (CIs) for VTE were estimated across quartiles of VWF levels. We found that the risk of VTE increased linearly across quartiles of VWF levels (P for trend = .023). Participants with VWF in the highest quartile had an OR of 1.45 (95% CI, 1.03-2.03) for VTE compared with those in the lowest quartile. The association was strongest for unprovoked VTE (OR, 2.74; 95% CI, 1.66-4.54) and unprovoked DVT in particular (OR, 6.73; 95% CI, 3.07-14.76). Further adjustment for body mass index, C-reactive protein, hypertension, estrogen use, and smoking had a modest effect on the risk estimates. To conclude, we found a dose-dependent relationship between plasma VWF levels and future risk of incident VTE, and unprovoked events in particular. Our findings suggest that VWF may represent a promising biomarker for future risk of incident VTE.
Collapse
|
28
|
Iglesias MJ, Schwenk JM, Odeberg J. Affinity Proteomics Assays for Cardiovascular and Atherosclerotic Disease Biomarkers. Methods Mol Biol 2021; 2344:163-179. [PMID: 34115359 DOI: 10.1007/978-1-0716-1562-1_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Systematic exploration of the dynamic human plasma proteome enables the discovery of novel protein biomarkers. Using state-of-the-art technologies holds the promise to facilitate a better diagnosis and risk prediction of diseases. Cardiovascular disease (CVD) pathophysiology is characterized for unbalancing of processes such as vascular inflammation, endothelial dysfunction, or lipid profiles among others. Such processes have a direct impact on the dynamic and complex composition of blood and hence the plasma proteome. Therefore, the study of the plasma proteome comprises an excellent exploratory source of biomarker research particularly for CVD. We describe the protocol for performing the discovery of protein biomarker candidates using the suspension bead array technology. The process does not require depletion steps to remove abundant proteins and consumes only a few microliters of sample from the body fluid of interest. The approach is scalable to measure many analytes as well as large numbers of samples. Moreover, we describe a bead-assisted antibody-labeling process that helps to develop quantitative assays for validation purposes and facilitate the translation of the identified candidates into clinical studies.
Collapse
Affiliation(s)
- Maria Jesus Iglesias
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Solna, Sweden. .,Department of Clinical Medicine, Faculty of Health Science, The Arctic University of Tromsø, Tromsø, Norway.
| | - Jochen M Schwenk
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Solna, Sweden
| | - Jacob Odeberg
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Solna, Sweden.,Department of Clinical Medicine, Faculty of Health Science, The Arctic University of Tromsø, Tromsø, Norway.,Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Predicting the Risk of Recurrent Venous Thromboembolism: Current Challenges and Future Opportunities. J Clin Med 2020; 9:jcm9051582. [PMID: 32456008 PMCID: PMC7290951 DOI: 10.3390/jcm9051582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Acute venous thromboembolism (VTE) is a commonly diagnosed condition and requires treatment with anticoagulation to reduce the risk of embolisation as well as recurrent venous thrombotic events. In many cases, cessation of anticoagulation is associated with an unacceptably high risk of recurrent VTE, precipitating the use of indefinite anticoagulation. In contrast, however, continuing anticoagulation is associated with increased major bleeding events. As a consequence, it is essential to accurately predict the subgroup of patients who have the highest probability of experiencing recurrent VTE, so that treatment can be appropriately tailored to each individual. To this end, the development of clinical prediction models has aided in calculating the risk of recurrent thrombotic events; however, there are several limitations with regards to routine use for all patients with acute VTE. More recently, focus has shifted towards the utility of novel biomarkers in the understanding of disease pathogenesis as well as their application in predicting recurrent VTE. Below, we review the current strategies used to predict the development of recurrent VTE, with emphasis on the application of several promising novel biomarkers in this field.
Collapse
|
30
|
Tala JA, Polikoff LA, Pinto MG, Li S, Trakas E, Miksa M, Gertz S, Faustino EVS. Protein biomarkers for incident deep venous thrombosis in critically ill adolescents: An exploratory study. Pediatr Blood Cancer 2020; 67:e28159. [PMID: 31904170 DOI: 10.1002/pbc.28159] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/18/2019] [Accepted: 12/11/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND There are no tests to identify critically ill children at high risk of deep venous thrombosis (DVT). In this exploratory study, we aimed to identify proteins that are associated with incident DVT in critically ill adolescents. PROCEDURE Plasma samples were obtained from critically ill adolescents within 24 hours after initiation of cardiopulmonary support. The adolescents were followed with ultrasound to detect the development of DVT of the lower extremity and clinically for bleeding. Thrombin-antithrombin complex and prothrombin fragment 1+2 were measured using immunosorbent assays, whereas procoagulation and anticoagulation factors were measured using multiplex assays. Plasma samples were also analyzed using SOMAscan, an aptamer-based capture assay. The associations between DVT and the log-transformed level of the proteins were assessed using logistic regression adjusting for the presence of femoral venous catheter and severity of illness. Associations were expressed as odds ratio (OR) for every log-fold increase in level of the protein with 95% confidence interval (CI). RESULTS Plasma from 59 critically ill adolescents, of whom 9 developed incident DVT, was analyzed. The median age of the adolescents was 15.1 years (interquartile range, 14.0-16.7 years). Higher levels of thrombin-antithrombin complex (OR: 31.54; 95% CI: 2.09-475.92) and lower levels of factor XIII (OR: 0.03; 95% CI: 0.002-0.44) were associated with DVT. CD36, MIC-1, and EpoR were marginally associated with DVT. Only factor XIII was associated with clinically relevant bleeding (OR: 0.27; 95% CI: 0.08-0.97). CONCLUSIONS We identified candidate protein biomarkers for incident DVT. We plan to validate our findings in adequately powered studies.
Collapse
Affiliation(s)
- Joana A Tala
- Yale-New Haven Children's Hospital, New Haven, Connecticut
| | - Lee A Polikoff
- The Warren Alpert School of Medicine at Brown University, Providence, Rhode Island
| | | | - Simon Li
- New York Medical College, Valhalla, New York
| | - Erin Trakas
- University of Rochester Golisano Children's Hospital, Rochester, New York
| | - Michael Miksa
- Children's Hospital at Montefiore, New York City, New York
| | - Shira Gertz
- Saint Barnabas Medical Center, Livingston, New Jersey
| | | | | |
Collapse
|
31
|
Smith JD, Narayanan P, Li N. Biomarkers of platelet dysfunction in non-clinical safety studies and humans. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
32
|
Wu FL, Lai DY, Ding HH, Tang YJ, Xu ZW, Ma ML, Guo SJ, Wang JF, Shen N, Zhao XD, Qi H, Li H, Tao SC. Identification of Serum Biomarkers for Systemic Lupus Erythematosus Using a Library of Phage Displayed Random Peptides and Deep Sequencing. Mol Cell Proteomics 2019; 18:1851-1863. [PMID: 31308251 PMCID: PMC6731078 DOI: 10.1074/mcp.ra119.001582] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 07/01/2019] [Indexed: 12/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is one of the most serious autoimmune diseases, characterized by highly diverse clinical manifestations. A biomarker is still needed for accurate diagnostics. SLE serum autoantibodies were discovered and validated using serum samples from independent sample cohorts encompassing 306 participants divided into three groups, i.e. healthy, SLE patients, and other autoimmune-related diseases. To discover biomarkers for SLE, a phage displayed random peptide library (Ph.D. 12) and deep sequencing were applied to screen specific autoantibodies in a total of 100 serum samples from 50 SLE patients and 50 healthy controls. A statistical analysis protocol was set up for the identification of peptides as potential biomarkers. For validation, 10 peptides were analyzed using enzyme-linked immunosorbent assays (ELISA). As a result, four peptides (SLE2018Val001, SLE2018Val002, SLE2018Val006, and SLE2018Val008) were discovered with high diagnostic power to differentiate SLE patients from healthy controls. Among them, two peptides, i.e. SLE2018Val001 and SLE2018Val002, were confirmed between SLE with other autoimmune patients. The procedure we established could be easily adopted for the identification of autoantibodies as biomarkers for many other diseases.
Collapse
Affiliation(s)
- Fan-Lin Wu
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; ¶School of Agriculture, Ludong University, Yantai 264025, China
| | - Dan-Yun Lai
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Hui-Hua Ding
- ‖Shanghai Institute of Rheumatology, Department of rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai 200240, China
| | - Yuan-Jia Tang
- ‖Shanghai Institute of Rheumatology, Department of rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai 200240, China
| | - Zhao-Wei Xu
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Ming-Liang Ma
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Shu-Juan Guo
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jing-Fang Wang
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Nan Shen
- ‖Shanghai Institute of Rheumatology, Department of rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai 200240, China; **State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, 2200 Lane 25 Xietu Road, Shanghai 200240, China
| | - Xiao-Dong Zhao
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Huan Qi
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Hua Li
- §Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Sheng-Ce Tao
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; §Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
33
|
Ten Cate V, Koeck T, Panova-Noeva M, Rapp S, Prochaska JH, Lenz M, Schulz A, Eggebrecht L, Hermanns MI, Heitmeier S, Krahn T, Laux V, Münzel T, Leineweber K, Konstantinides SV, Wild PS. A prospective cohort study to identify and evaluate endotypes of venous thromboembolism: Rationale and design of the Genotyping and Molecular Phenotyping in Venous ThromboEmbolism project (GMP-VTE). Thromb Res 2019; 181:84-91. [PMID: 31374513 DOI: 10.1016/j.thromres.2019.07.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/17/2019] [Accepted: 07/21/2019] [Indexed: 01/02/2023]
Abstract
Several clinical, genetic and acquired risk factors for venous thromboembolism (VTE) have been identified. However, the molecular pathophysiology and mechanisms of disease progression remain poorly understood. This is reflected by uncertainties regarding the primary and secondary prevention of VTE and the optimal duration of antithrombotic therapy. A growing body of literature points to clinically relevant differences between VTE phenotypes (e.g. deep vein thrombosis (DVT) versus pulmonary embolism (PE), unprovoked versus provoked VTE). Extensive links to cardiovascular, inflammatory and immune-related morbidities are testament to the complexity of the disease. The GMP-VTE project is a prospective, multi-center cohort study on individuals with objectively confirmed VTE. Sequential data sampling was performed at the time of the acute event and during serial follow-up investigations. Various data levels (e.g. clinical, genetic, proteomic and platelet data) are available for multi-dimensional data analyses by means of advanced statistical, bioinformatic and machine learning methods. The GMP-VTE project comprises n = 663 individuals with acute VTE (mean age: 60.3 ± 15.9 years; female sex: 42.8%). In detail, 28.4% individuals (n = 188) had acute isolated DVT, whereas 71.6% subjects (n = 475) had PE with or without concomitant DVT. In the study sample, 28.9% (n = 129) of individuals with PE and 30.1% (n = 55) of individuals with isolated DVT had a recurrent VTE event at the time of study enrolment. The systems-oriented approach for the comprehensive dataset of the GMP-VTE project may generate new biological insights into the pathophysiology of VTE and refine our current understanding and management of VTE.
Collapse
Affiliation(s)
- V Ten Cate
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - T Koeck
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - M Panova-Noeva
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, Germany
| | - S Rapp
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - J H Prochaska
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, Germany
| | - M Lenz
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Organismic and Molecular Evolution, Johannes Gutenberg University Mainz, Mainz, Germany
| | - A Schulz
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - L Eggebrecht
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - M I Hermanns
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; School of Chemistry, Biology and Pharmacy, Fresenius University of Applied Sciences, Idstein, Germany
| | | | - T Krahn
- Bayer AG, Wuppertal, Germany
| | - V Laux
- Bayer AG, Wuppertal, Germany
| | - T Münzel
- Center for Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - S V Konstantinides
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Department of Cardiology, Democritus University of Thrace, University General Hospital, Greece
| | - P S Wild
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, Germany.
| | | |
Collapse
|
34
|
Alhabibi AM, Eldewi DM, Wahab MAA, Farouk N, El-Hagrasy HA, Saleh OI. Platelet-derived growth factor-beta as a new marker of deep venous thrombosis. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2019; 24:48. [PMID: 31160915 PMCID: PMC6540930 DOI: 10.4103/jrms.jrms_965_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/16/2019] [Accepted: 02/28/2019] [Indexed: 12/15/2022]
Abstract
Background: Deep venous thrombosis (DVT) is associated with significant morbidity and mortality. Thus, there is a great need to demonstrate a more efficient biomarker that would confirm the diagnosis of DVT. Our work aimed to evaluate the role of platelet-derived growth factor-beta (PDGF-B) as a new marker of DVT and its correlation with other radiological and laboratory tools used for the diagnosis. Materials and Methods: A case–control study enrolled forty patients selected from our university hospital between April 2018 and August 2018, who divided into two groups: Group I (n = 20) consisted of patients diagnosed with acute venous thrombosis and Group II (n = 20) consisted of patients diagnosed with chronic venous thrombosis. Twenty samples were collected from age- and gender-matched apparently healthy controls to be used as a control. Venous duplex ultrasonography, routine laboratory investigations, D-dimer (DD), and protein expression of PDGF-B were performed on all patients. Results: There was a highly significant increase in a protein expression of PDFG-B in all cases of acute and chronic venous thrombosis compared to the control group with P < 0.001; furthermore, it was more specific than DD for the detection of DVT (specificity 95% and 90%, respectively). Conclusion: Our study submits a novel association of PDGF-B plasma levels with DVT, and PDGF-B is considered to be a more specific indicator for DVT than is DD.
Collapse
Affiliation(s)
- Alshaymaa M Alhabibi
- Department of Clinical Pathology, Faculty of Medicine (For Girls), Al-Azhar University, Cairo, Egypt
| | - Dalia Mahmoud Eldewi
- Department of Clinical Pathology, Faculty of Medicine (For Girls), Al-Azhar University, Cairo, Egypt
| | - Maisa A Abdel Wahab
- Department of Vascular Surgery, Faculty of Medicine (For Girls), Al-Azhar University, Cairo, Egypt
| | - Nehal Farouk
- Department of Vascular Surgery, Faculty of Medicine (For Girls), Al-Azhar University, Cairo, Egypt
| | - Hanan A El-Hagrasy
- Department of Clinical Pathology, Faculty of Medicine (For Girls), Al-Azhar University, Cairo, Egypt
| | - Ola I Saleh
- Department of Radio-Diagnosis, Faculty of Medicine (For Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
35
|
Sendama W, Musgrave KM. Decision-Making with D-Dimer in the Diagnosis of Pulmonary Embolism. Am J Med 2018; 131:1438-1443. [PMID: 30125536 DOI: 10.1016/j.amjmed.2018.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 01/13/2023]
Abstract
The measurement of D-dimer can provide useful information to aid in the diagnosis of patients with suspected venous thromboembolism. D-dimer measurement in conjunction with adequate estimation of pretest probability can facilitate the safe discharge of patients with suspected pulmonary embolism and limit unnecessary investigation or anticoagulation. We review clinical decision-making strategies in the diagnosis of pulmonary embolism and propose means to minimize diagnostic error that might arise from knowledge of the D-dimer result prior to clinical assessment.
Collapse
Affiliation(s)
- Wezi Sendama
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.
| | - Kathryn M Musgrave
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
36
|
Jensen SB, Hindberg K, Solomon T, Smith EN, Lapek JD, Gonzalez DJ, Latysheva N, Frazer KA, Braekkan SK, Hansen JB. Discovery of novel plasma biomarkers for future incident venous thromboembolism by untargeted synchronous precursor selection mass spectrometry proteomics. J Thromb Haemost 2018; 16:1763-1774. [PMID: 29964323 PMCID: PMC6123273 DOI: 10.1111/jth.14220] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Indexed: 01/08/2023]
Abstract
Essentials Discovery of predictive biomarkers of venous thromboembolism (VTE) may aid risk stratification. A case-control study where plasma was sampled before the occurrence of VTE was established. We generated untargeted plasma proteomic profiles of 200 individuals by use of mass spectrometry. Assessment of the biomarker potential of 501 proteins yielded 46 biomarker candidates. ABSTRACT Background Prophylactic anticoagulant treatment may substantially reduce the incidence of venous thromboembolism (VTE) but entails considerable risk of severe bleeding. Identification of individuals at high risk of VTE through the use of predictive biomarkers is desirable in order to achieve a favorable benefit-to-harm ratio. Objective We aimed to identify predictive protein biomarker candidates of VTE. Methods We performed a case-control study of 200 individuals that participated in the Tromsø Study, a population-based cohort, where blood samples were collected before the VTE events occurred. Untargeted tandem mass tag-synchronous precursor selection-mass spectrometry (TMT-SPS-MS3)-based proteomic profiling was used to study the plasma proteomes of each individual. Results Of the 501 proteins detected in a sufficient number of samples to allow multivariate analysis, 46 proteins were associated with VTE case-control status with P-values below the 0.05 significance threshold. The strongest predictive biomarker candidates, assessed by statistical significance, were transthyretin, vitamin K-dependent protein Z and protein/nucleic acid deglycase DJ-1. Conclusions Our untargeted approach of plasma proteome profiling revealed novel predictive biomarker candidates of VTE and confirmed previously reported candidates, thereby providing conceptual support for the validity of the study. A larger nested case-control study will be conducted to validate our findings.
Collapse
Affiliation(s)
- S B Jensen
- K. G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - K Hindberg
- K. G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - T Solomon
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California, USA
| | - E N Smith
- K. G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
- Department of Pediatrics and Rady's Children's Hospital, University of California San Diego, La Jolla, California, USA
| | - J D Lapek
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - D J Gonzalez
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - N Latysheva
- K. G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - K A Frazer
- K. G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
- Department of Pediatrics and Rady's Children's Hospital, University of California San Diego, La Jolla, California, USA
- Institute of Genomic Medicine, University of California San Diego, La Jolla, California, USA
| | - S K Braekkan
- K. G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - J-B Hansen
- K. G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
37
|
Lygirou V, Latosinska A, Makridakis M, Mullen W, Delles C, Schanstra JP, Zoidakis J, Pieske B, Mischak H, Vlahou A. Plasma proteomic analysis reveals altered protein abundances in cardiovascular disease. J Transl Med 2018; 16:104. [PMID: 29665821 PMCID: PMC5905170 DOI: 10.1186/s12967-018-1476-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/06/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) describes the pathological conditions of the heart and blood vessels. Despite the large number of studies on CVD and its etiology, its key modulators remain largely unknown. To this end, we performed a comprehensive proteomic analysis of blood plasma, with the scope to identify disease-associated changes after placing them in the context of existing knowledge, and generate a well characterized dataset for further use in CVD multi-omics integrative analysis. METHODS LC-MS/MS was employed to analyze plasma from 32 subjects (19 cases of various CVD phenotypes and 13 controls) in two steps: discovery (13 cases and 8 controls) and test (6 cases and 5 controls) set analysis. Following label-free quantification, the detected proteins were correlated to existing plasma proteomics datasets (plasma proteome database; PPD) and functionally annotated (Cytoscape, Ingenuity Pathway Analysis). Differential expression was defined based on identification confidence (≥ 2 peptides per protein), statistical significance (Mann-Whitney p value ≤ 0.05) and a minimum of twofold change. RESULTS Peptides detected in at least 50% of samples per group were considered, resulting in a total of 3796 identified proteins (838 proteins based on ≥ 2 peptides). Pathway annotation confirmed the functional relevance of the findings (representation of complement cascade, fibrin clot formation, platelet degranulation, etc.). Correlation of the relative abundance of the proteins identified in the discovery set with their reported concentrations in the PPD was significant, confirming the validity of the quantification method. The discovery set analysis revealed 100 differentially expressed proteins between cases and controls, 39 of which were verified (≥ twofold change) in the test set. These included proteins already studied in the context of CVD (such as apolipoprotein B, alpha-2-macroglobulin), as well as novel findings (such as low density lipoprotein receptor related protein 2 [LRP2], protein SZT2) for which a mechanism of action is suggested. CONCLUSIONS This proteomic study provides a comprehensive dataset to be used for integrative and functional studies in the field. The observed protein changes reflect known CVD-related processes (e.g. lipid uptake, inflammation) but also novel hypotheses for further investigation including a potential pleiotropic role of LPR2 but also links of SZT2 to CVD.
Collapse
Affiliation(s)
- Vasiliki Lygirou
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, 115 27, Athens, Greece
| | | | - Manousos Makridakis
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, 115 27, Athens, Greece
| | - William Mullen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institute of Cardiovascular and Metabolic Disease, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Jerome Zoidakis
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, 115 27, Athens, Greece
| | - Burkert Pieske
- Deutsches Herzzentrum Berlin, Augustenburger Pl. 1, 13353, Berlin, Germany
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, Rotenburger Straße 20, 30659, Hannover, Germany
| | - Antonia Vlahou
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, 115 27, Athens, Greece.
| |
Collapse
|
38
|
Alshanwani AR, Riches-Suman K, O'Regan DJ, Wood IC, Turner NA, Porter KE. MicroRNA-21 drives the switch to a synthetic phenotype in human saphenous vein smooth muscle cells. IUBMB Life 2018; 70:649-657. [DOI: 10.1002/iub.1751] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 03/22/2018] [Indexed: 01/09/2023]
Affiliation(s)
- Aliah R. Alshanwani
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine; University of Leeds; Leeds UK
- Multidisciplinary Cardiovascular Research Centre (MCRC), University of Leeds; Leeds UK
| | - Kirsten Riches-Suman
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine; University of Leeds; Leeds UK
- School of Chemistry and Biosciences; University of Bradford; Bradford UK
| | - David J. O'Regan
- Multidisciplinary Cardiovascular Research Centre (MCRC), University of Leeds; Leeds UK
- Department of Cardiac Surgery; The Yorkshire Heart Centre, Leeds General Infirmary; Leeds UK
| | - Ian C. Wood
- Faculty of Biological Sciences, School of Biomedical Sciences; University of Leeds; Leeds UK
| | - Neil A. Turner
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine; University of Leeds; Leeds UK
- Multidisciplinary Cardiovascular Research Centre (MCRC), University of Leeds; Leeds UK
| | - Karen E. Porter
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine; University of Leeds; Leeds UK
- Multidisciplinary Cardiovascular Research Centre (MCRC), University of Leeds; Leeds UK
| |
Collapse
|
39
|
Trégouët DA, Morange PE. What is currently known about the genetics of venous thromboembolism at the dawn of next generation sequencing technologies. Br J Haematol 2018; 180:335-345. [PMID: 29082522 DOI: 10.1111/bjh.15004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Venous thromboembolism (VTE) has a strong genetic component. This review summarizes what is known at the seventeen genes that are now well established to harbour VTE-associated genetic variants. In addition, it discusses additional candidate genes that deserve further validation before being claimed as VTE associated genes. Finally, several research strategies are briefly described to identify other molecular determinants of the disease.
Collapse
Affiliation(s)
- David-Alexandre Trégouët
- Department of Genomics & Pathophysiology of Cardiovascular Diseases, Sorbonne Universités, UPMC Univ. Paris 06, Institut National pour la Santé et la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR_S) 1166, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Pierre-Emmanuel Morange
- Laboratory of Haematology, La Timone Hospital, Marseille, France
- INSERM UMR_S 1062, Nutrition Obesity and Risk of Thrombosis, Aix-Marseille University, Marseille, France
| |
Collapse
|
40
|
Harahsheh Y, Ho KM. Use of viscoelastic tests to predict clinical thromboembolic events: A systematic review and meta-analysis. Eur J Haematol 2018; 100:113-123. [DOI: 10.1111/ejh.12992] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Yusrah Harahsheh
- Department of Intensive Care Medicine; Royal Perth Hospital; Perth WA Australia
- School of Medicine and Pharmacology; University of Western Australia; Perth WA Australia
| | - Kwok M. Ho
- Department of Intensive Care Medicine; Royal Perth Hospital; Perth WA Australia
- School of Population and Global Health; University of Western Australia; Perth WA Australia
- School of Veterinary and Life Sciences; Murdoch University; Perth WA Australia
| |
Collapse
|
41
|
Zhang Z, Tang L, Hu Y. Progress in the research on venous thromboembolism. Curr Med Sci 2017; 37:811-815. [DOI: 10.1007/s11596-017-1811-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/02/2017] [Indexed: 01/05/2023]
|
42
|
Multiplexed targeted proteomic assay to assess coagulation factor concentrations and thrombosis-associated cancer. Blood Adv 2017; 1:1080-1087. [PMID: 29296750 DOI: 10.1182/bloodadvances.2017007955] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/26/2017] [Indexed: 12/21/2022] Open
Abstract
The plasma levels of pro- and anticoagulant proteins are important markers for venous thrombosis (VT) risk and can be affected by both genetic and acquired factors, including cancer. Generally, these markers are measured using activity- or antibody-based assays. Targeted proteomics with stable-isotope-labeled internal standards has proven adept at the rapid, multiplex, and precise quantification of proteins in complex biological samples such as plasma. We used liquid chromatography coupled to multiple reaction monitoring (MRM) mass spectrometry to evaluate the concentrations of 31 coagulation- and fibrinolysis-related proteins in plasma from 25 healthy controls, 25 patients with VT, and 25 patients with VT who were also diagnosed with cancer. The concentration level of 1 to 3 proteotypic peptides per protein was determined, and all samples were previously characterized using traditional antibody- or activity-based methods. When comparing the conventional and the MRM strategies, the mean Pearson correlation for the 13 proteins (covered by 36 target peptides) shared between the 2 approaches was 0.77, indicating a good correlation. Additionally, MRM offers higher sensitivity (mean regression slope, 0.81), higher multiplicity in a single run, and good ability to leverage all measurements to discriminate groups using unsupervised clustering, which identified vitamin K antagonist users as well as patients with VT and cancer. The data collected using MRM show that the combination of coagulation factor levels yields signature information on VT and cancer, which was not obvious from a single measurement. These results encourage the further validation and investigation of MRM in profiling protein signature of disease.
Collapse
|