Contents
Download PDF
pdf Download XML
202 Views
16 Downloads
Share this article
Research Article | Volume 15 Issue 3 (March, 2025) | Pages 565 - 574
Coagulation Cascade and Protein-Protein Interaction in Venous Thromboembolism: A Narrative Review
 ,
 ,
 ,
 ,
 ,
1
Research Scientist-Clinical Proteomics and Genomics research, Scientific officer, Research and Development Department, Kalinga Institute of Medical Sciences, School of Medicine, KIIT DU, Bhubaneswar, Odisha
2
HOD & Associate Professor, CTVS Department, Kalinga Institute of Medical Sciences, School of Medicine, KIIT DU, Bhubaneswar, Odisha
3
Associate Professor, Department of Radiology, Kalinga Institute of medical Sciences, School of Medicine, KIIT DU, Bhubaneswar, Odisha
4
Associate Professor, Department of Nephrology, Kalinga Institute of medical Sciences, School of Medicine, KIIT DU, Bhubaneswar, Odisha
5
Scientist & Program Officer, Division of Non-Communicable Diseases, Indian Council of Medical Research, Head Quarter, New Delhi, India
6
Scientist & Program Officer, Division of Descriptive Research, Indian Council of Medical Research, Head Quarter, New Delhi, India.
Under a Creative Commons license
Open Access
Received
Feb. 11, 2025
Revised
Feb. 22, 2025
Accepted
March 3, 2025
Published
March 20, 2025
Abstract

Purpose of the Review:Venous Thromboembolism is a complication in which blood clots and blocks a vein. Most of the time either it is underdiagnosed or misdiagnosed and becomes serious because of late diagnosis. It encompasses two interrelated conditions such as deep vein thrombosis and pulmonary embolism. Deep vein thrombosis is a significant concern in hospitalized patients and contributes to long hospital stays, morbidity and mortality. The most appropriate inflammatory biomarker for diagnosis and therapeutics in venous thromboembolism remains unclear and is under exploratory condition. The role of molecular markers in the pathophysiology of venous thrombo embolism expands the knowledge of mechanistic action and therapeutic targets. Detection of biomarkers associated with VTE and recurrence of VTE has been explored but limited to few markers like D-dimer, C reactive protein, protein C & S etc. Recent findings: Present study reviewed a total of 44 differentially expressed proteins majorly lipoproteins, glycoproteins, inflammatory proteins and few cytosolic proteins, which were considered as potential biomarker for VTE manifestation. Protein-protein interaction confirmed by a Search tool (STRING) and revealed that CRP, IL6, IL8, IL17, selectin family, JAK-STAT group and tissue factor proteins including VWF proteins are significantly interconnected in disease manifestation, thrombus formation and endothelial cell activation, platelet aggregation, and coagulation.  Summary: The current review and protein protein interaction analysis bring together shreds of evidence of differential expression of protein biomarkers associated with venous-thromboembolism by their function and protein-protein interaction involved in coagulation. 

 

Keywords
INTRODUCTION

Venous thromboembolism is a multifactorial disease and is associated with multiple genetic and acquired factors. Globally the VTE incidence is 0.75 -2.69 per 1000 population and in the elderly population, it is 2.7 per 1000.  Venous thrombosis includes deep vein thrombosis (DVT) and Pulmonary embolism (PE), where DVT is the most common and invariably associated with morbidities and mortalities, whereas, PE is a life-threatening condition with a survival rate of less than 60% [1]. The occurrence of recurrent VTE is about one-third in VTE patients.  The diagnosis of VTE is a challenging point in the case of severely ill patients and it is more difficult in PE cases. The pathophysiology of VTE is still in the exploratory stage.  Thrombosis formation is mostly initiated by coagulation due to activation, adhesion and aggregation of platelets and deposition of fibrins.

 

Platelet deposition forms an obstruction at the site of injury. In thrombosis formation and development of VTE, primary hypercoagulability due to deficiencies in anticoagulant proteins is attributed to 5- 10% whereas, acquired factors such as surgery, malignancy, and immobilization are more associated with VTE[2]. Early diagnosis and treatment of different types of thrombosis are of paramount value. Thereby, the predictive marker for low and high risk for VTE may improve the diagnosis efficiency and early therapeutic action. The role of pro-inflammatory biomarkers and acute phase reactant proteins could give an insight into the pathophysiology and mechanism of coagulation at different sites.

 

Coagulation cascade and disease manifestation:

Blood coagulation is a series of protease conversions, to form thrombin by converting soluble fibrinogen to insoluble fibrin monomer followed by cross linked fibrin polymer and fibrin clot. Protease activator receptors play a major role in the cascade through extrinsic, intrinsic and Common pathway with functional components of tissue factors (TF). TF is released into the micro particle circulation [3]. Under pathological condition, elevated level of TF trigger the thrombosis formation and leads to a sequence of events including DVT, PE and other thrombosis. Figure 1 shows a schematic flow chart of thrombin formation, in which, extrinsic pathways are regulated by tissue factors VII, VIIa, X, Xa, III and II and the intrinsic pathway is by Tissue factors XII, XIIa, XI, XIa, IX, VIIIa and VWF. In common pathway, factor XIII (F13) and XIIIa are involved in Fibrin clot formation [4].    

Currently, the diagnosis of DVT, Pulmonary Embolism & VTE is confirmed by Doppler, CT angiography, pulmonary angiography, or pathological examination of thrombus extract during surgery or autopsy. Despite extensive research, not a single biomarker reliably predicts VTE occurrence or recurrence because the pathophysiology of VTE is multifactorial with a complex interaction between genetic, environmental and acquired risk factors [6]. 

 

Potential Protein biomarkers differentially expressed in VTE manifestation: A total of 44 potential biomarkers were reviewed based on their expression level in VTE. A meta-analysis has reported 5 inflammatory proteins in 1385 cases of VTE, which revealed that monocytes, CRP and IL6 are significantly elevated in acute conditions. The authors concluded that hs-CRP and CRP were found to be significant markers for VTE recurrence [8]. A meta-analysis of blood markers detected in VTE cancer patients revealed 174 biomarker measurements and concluded with 9 biomarkers, which were significantly associated with VTE [9]. Based on other available literature, the differentially expressed proteins identified through different detection methods from plasma/serum are listed in the table 1. 

 

Based on the functionality and differential expression in VTE, a total of 44 proteins were searched from the literature of 40 published articles. The identified proteins are classified into different sections such as lipoprotein, glycoprotein, Coagulation factors, inflammatory proteins and other cytosolic proteins. Lipoproteins regulate the transportation of dietary lipids, from the liver to peripheral tissue. It plays a major role in cardiovascular diseases especially lipoprotein a (Lpa). An elevated level of Lpa is strongly associated with atherosclerotic cardiovascular disease (ASCVD) [32]. Pro-inflammatory phospholipids are involved in disease manifestation due to the presence of lysine binding sites and is similar to plasminogen, which inhibits fibrinolysis. This inhibition strongly plays a significant role in pulmonary embolism [33]. Other lipoproteins are also differentially expressed in pulmonary embolism such as haptoglobin, serum amyloid A1 (SAA1), retinol-binding protein, and inter alpha trypsin inhibitor heavy chain H4 [10,11]. Biological processes such as protein folding, secretion, adhesion, stability in circulation and signal transduction are associated with glycoprotein. The majority of proteins glycosylated during the synthesis of carbohydrates and a change in glycosylation pattern is a signature of disease. Platelet glycoproteins are actively participate in thrombosis formation [34]. The present study reviewed many published articles, which revealed that glycoproteins such as osteopontin, fibronectin precursor, alpha 1 microglobulin, clusterin, leucine-rich alpha 2 glycoprotein are elevated in pulmonary embolism and lipopolysaccharide-binding proteins are upregulated in VTE [10,11,12-15]. Coagulation factors such as FII, FV, FVII, FVIII, FIX, FX, FXI and Von Willebrand are commonly involved in blood clot formation. Any disproportional change in the expression level leads to thrombosis formation majorly in the deep vein and pulmonary artery and vein [35]. Under VTE manifestation, differentially expressed coagulation factors are FVIII, FIX, and FXI. Factor VIII is an acute phase response protein and the expression level increases in inflammation conditions. In cases of provoked VTE, the level of Factor VIII increases five fold and is identified as a risk factor for recurrent VTE [36]. Soria et al. reported that protein C and FVIII were jointly influenced in thrombosis cases [37]. Factor IX is a vitamin K-dependent glycoprotein that plays a role in hemostasis. It is activated by tissue factors and leads to form a fibrin clot. B Furie et al suggested that Factor IX and Factor VIII have similar effects of high levels of clotting factors on thrombotic risk [38]. Factor X is one of the coagulation factors and occupies a central position in the coagulation system and is a key driver in thrombin formation. In extrinsic and intrinsic pathways factor X is converted to FXa [39]. Factor XI is a 160 KDa disulfide-linked dimeric protein, activated by FXIa and contributes to hemostasis. It is a target site for the therapeutic action of VTE. 53 clinical trials based on FXI inhibitors have been registered. Among them, abelacimab is in phase II trials in comparison to apixaban or dalteparin towards prevention, and treatment of tumor-associated VTE and the completed trials have shown that FXI / FXIa inhibitors are safe and effective in therapeutic application prospects. Arterial thrombosis and inflammation are interlinked; DVT and arterial thrombosis share the same risk factors with having elevated C - reactive protein (CRP). CRP promotes a prothrombotic effect and induces tissue factors, which leads to an extrinsic coagulation cascade. Several studies have reported the elevation of inflammatory markers like CRP, interleukins, ILBeta, IL6, IL8, IL10, IL17, Il15 Receptor, interferon (IFN gamma) and ST2 and monocyte chemotactic protein 1 in acute VTE  patients [17,18,25-28]. Other inflammatory proteins such as D-dimer, selectin family (E-selectin, L-selectin and P-selectin) and micro particles, are reported with a significant increase in expression in VTE manifestation and disease progression [24,28,30,31]. The decreased levels of Calprotectin were reported in VTE patients [11] As D-dimer is a degradation product of cross-linked fibrin, formed when thrombin converts fibrinogen into fibrin, so D-Dimer is considered a significant diagnostic marker for VTE. Several studies reported that D-dimer is a negative predictor and non-specific to DVT [40,41]. Thus it remains as controversial. After completion of anticoagulation therapy, the levels were significantly reduced to half from 500 ng/ml among 247 patients. Few articles reported that VTE recurrence occurred due to discontinuation of oral anticoagulation therapy [43].The association between CRP and VTE has been reported in different prospective studies as observed in the oncologic and obese population. In the acute stage of VTE, CRP activates immune cells (neutrophils, monocytes and macrophages) through the classical complement pathway. As CRP is a pentameric non-glycosylated protein consisting of five identical subunits synthesized in hepatocytes [43,44] and a family member of of Pentraxin along with serum amyloid P component protein (SAP) and Female protein (FP). SAP binds with amyloid fibrils, DNA, chromatin, fibronectin, C4-binding protein and glycosaminoglycan. In response to inflammation, the production of CRP is stimulated by different types of cytokines (IL 6, IL 1, and TNF). Thus, CRP is known to be an acute phase reactant. CRP rises in wide varieties of infections and inflammations.  It initiates precipitation, agglutination and capsular swelling. It has been shown that altered CRP reacts with the FC receptor of monocytes, and lymphocytes and activate the platelet. Therefore, CRP is considered a diagnostic biomarker in monitoring chronic inflammatory conditions promising biomarker for risk stratification in case of short-term mortality, bleeding cases and recurrence due to anticoagulant withdrawal [45]. Selectins are carbohydrate-binding adhesion molecules that actively participate in immune regulation. E selectin is recognized as a cytokine-inducible glycoprotein, which is mostly involved in the new adhesion of molecules.  A strong association of E-Selectin with recurrent VTE has been observed in which a single nucleotide polymorphism “Ser128Arg” was detected in  E-Selectin. An in vitro analysis shows that this polymorphism alters ligand affinity, regulates the interaction of leukocyte-endothelial cell interactions and enhances tissue factor-mediated coagulation, atherosclerosis, myocardial infarction and endotoxin-trigger [46].

 

In thrombus formation, the interaction of P selectin and PSGL-1 plays a major role.  P selectin influences fibrin deposition in the thrombus. Rectenwald et al. reported that P-selectin concentration is high in patients with acute DVT and has the clinical applicability to assess the risk of DVT [46]. It is a transmembrane glycoprotein (M. W~140kDa) present in storage granules of endothelial cells (weibal-palade bodies) and stimulated by thrombin and other mediators, intersected with L and E selectin. CD 62 is known to be associated with platelet association [47,48]. L-selectin (Leu-8-TQ1) mostly circulates in lymphocytes, Neutrophils and monocytes. CD36 increases the number of circulating micro particles [49]. Tissue factor activity is significantly correlated with micro particles. In the mice model authors reported that micro particles amplify, the coagulation by binding factor VIIa and are considered an independent risk factor during acute thrombogenesis [50].

 

Protein-protein interaction construction:

A literature review on differentially expressed proteins as involved in VTE manifestation revealed that maximum number of proteins are lipoproteins, glycoproteins, inflammatory proteins and a few are cytosolic proteins. A STRING search tool was used to construct a protein-protein network between differentially expressed proteins in VTE manifestation,  as mentioned in table 1. As VTE is a multifactorial disease, the network speculates the connection and constructed 31 nodes and 116 edges with PPI enrichment p-value <1.0e-16. (Figure 2) Based on their pleiotropic effects, antagonistic master cytokines, CRP, IL6, CXCL8(IL8), IL17 A, SPP, selectins, VWF and SERPINE 1 are centrally located in the network with direct connection with tyrosine-protein kinase JAK 2, PDGEFRB, Tissue factors (TFPI, F12, F8, F9), S100AB, SAA1 and HLA-G.  These clusters are indirectly connected to other clusters leucine-rich glycoprotein (LRG1), fibrinogen protein (FGG), extracellular chaperon (CLU), 35 KDa inter-alpha-trypsin inhibitor heavy chain H4 (ITIH4), haptoglobin (HP), Calcium 2+ dependent actin filament serving protein (SCIN) and retinol transporter (STRA6). Majorly all the proteins are over-expressed except a few such as S100A8, which are downregulated under pulmonary embolism and deep vein thrombosis condition but this is closely connected to IL6, serum amyloid protein (SAA1) and plasminogen activator inhibitor (SERPINE). Five proteins such as Bleomycin hydralase (BLMH), Extracellular matrix protein -Tenascin N (TNN), Follistain-related protein 3 (FSTL 3), deoxyribonuclease gamma (DNASE 1 L3) and Calcium/calmodulin-dependent protein kinase type 1 (CAMK1) are independently activated and not connected the identified network. These proteins can be a potential target point for disease management [51].

 

Gene ontology and pathway enrichment analysis:

The network analysis extended the views of pathway involvement through a KEGG pathway enrichment analysis which revealed that the majority of genes participated in the protein translation, which is similar to the pathways and part of the complement and coagulation cascade, malaria pathogenesis, signaling pathway in diabetic complications, other signalling pathway (IL17 & P13K-AKT) cell adhesion molecules, cellular senescence,  tyrosine kinase inhibitor, rheumatoid arthritis and ECM receptor interaction. Figure 3 and 4 explains the involvement of biological processes as derived from the gene ontology based on the protein network.  In Figure 4, it has shown that the interacted proteins are responsible for acute inflammation.

Figure 1: Schematic diagram of coagulation cascade revealing the activators and inhibitors in fibrin clot formation

Figure 2: A protein-protein interaction network consisting of 31 nodes and 116 edges was constructed by using the STRING online tool with PPI enrichment p-value <1.0e-16. Further analysis of k-means clustering reports three clusters; Red colour for cluster 1 corresponds to Acute inflammatory response, the Green colour for cluster 2 corresponds to the acute phase and the blue colour for cluster 3 as calcium-dependent (Ca2+) actin filament-serving protein (SCIN). The dotted lines are for edges. STRING (Search Tool for the Retrieval of Interacting Genes/Proteins)[52]

 

Figure 3: KEGG pathway enrichment analysis in PPI (Protein protein Interaction) by using by using STRING Protein- protein interaction tool. The purple colour shows lower p-value while orange colour shows higher p-value (p-value < 1.0e-16)

Abbreviations: KEGG: Kyoto Encyclopedia of Gene and Genome 

 

Figure 4: Gene Ontology enrichment analysis in PPI by using STRING Protein- protein interaction tool. The purple colour shows lower p-value while orange colour shows higher p-value (Protein protein interaction enrichment p-value < 1.0e-16)

 

Tables & figure legends

SL no

Protein

Purpose

Detection  Method

Type of Fluid

Expression

Reference

Lipoprotein & Glycoprotein

1

Haptoglobin

PE

MALDI

Serum

Up-regulated

Zhang et al[10]

2

Serum amyloid A-1 (SAA1)

PE

Mass spectrometry

Plasma

Up-regulated

Han et al[11]

3

Retinol binding protein 4

PE

2DE &MALDI-TOF-MS

Serum

Up-regulated

Zhang et.al[10]

4

Inter alpha trypsin inhibitor heavy chain H4 (ITIH4)

PE

ELISA

Serum

Up-regulated

Zhang et.al[10]

5

Tenascin- C (TNC)

PE

Antibody-based assay

Plasma

Up-regulated

Han et al[11]

6

Osteopontin

PE

Murine model

Blood

Up-regulated

Kolmel et.al.[12]

7

Fibronectin precursor

PE

LC-MS/MS

Plasma

Up-regulated

Watts et.al[13]

8

LBP(lipo polysaccharide binding protein)

VTE

Mass spectrometry

Plasma

Up-regulated

Liu.et al[14]

9

Alpha-1-Microglobulin/Bikunin precursor

PE

2DE& MALDI-TOF-MS

Serum

Up-regulated

Zhang  et.al[10]

10

Clusterin

PE

2DE&MALDI-TOF-MS

serum

Up-regulated

Zhang et.al[10]

11

Leucine- rich α-2-glycoprotein

PE

LC/MS/MS

Plasma

Up-regulated

Granholm et.al[15]

Coagulation factor

12

Plasminogen activator inhibitor-1(PAI-1)

VTE

ELISA

Plasma

Up-regulated

FRISCHMUTH.et.al[16]

13

Von willebrand Factor

VTE

ELISA

Serum

Up-regulated

Tavares et.al Memon et.al[17,18]

14

Fibrinogen alpha

PE

LC-MS/MS

Plasma

Up-regulated

Watts et.al[19]

15

Fibrinogen beta

PE

LC-MS/MS

Plasma

Up-regulated

Watts et.al[19]

16

Fibrinogen gamma

PE

LC-MS/MS

Plasma

Up-regulated

Watts et.al[19]

17

Factor VIII

VTE

Immunoassay

Plasma

Up-regulated

Vormittag R et.al[20]

18

Factor IX

VTE

ELISA

plasma

Up-regulated

vanHYLCKAMA VLIEG et.al[21]

19

Factor XI

VTE

ElISA

plasma

Up-regulated

MEIJERS Joost C.M et al[22]

Inflammatory protein

20

D-dimer

VTE

ELISA

Serum

Up-regulated

Tavares et.al[17]

21

Crp

VTE

ELISA

Serum

Up-regulated

Tavares et.al[17]

22

E-selectin

VTE

ELISA

Serum

Up-regulated

Watany M et al[23]

23

L-selectin

VTE

ELISA

Serum

Up-regulated

Watany Met.al[23]

24

P-selectin

DVT

Proximity extension assays

Plasma

Up-regulated

Memon et.al[18]

25

Micro particles

VTE

ELISA/flow cytometry

Serum

Up-regulated

Zhou L et.al[24]

26

Calprotectin (S100A8),

PE

Antibody based assay

Plasma

Down-regulated

Han et al[11]

27

Interleukin-15Rα

PE

Proximity extension assays

plasma

Up-regulated

Ten cate et.al[25]

28

IL-6

VTE

ELISA

Plasma

Up-regulated

Roumen-Klappe et.al[26]

29

IL-8

VTE

ELISA

Plasma

Up-regulated

Roumen-klappe et.al[26]

30

IL-17

VTE

Mice model

plasma

Up-regulated

P.Ding et.al[27]

31

Interferon-gamma

PE

Proximity extension assays

Plasma

Up-regulated

Ten cate et.al[25]

32

ST2

DVT

Proximity extension assays

plasma

Up-regulated

Memon et.al[18]

33

Monocyte chemotactic protein-1(MCP-1)

VTE

ELISA

Plasma

Up-regulated

M.F. Matos et.al[28]

34

CFHR5

VTE

Mass spectrometry

plasma

Up-regulated

Razzaq et.al[29]

35

Histidine-rich glycoprotein (HRG)

PE

Antibody based assay

Plasma

Down-regulated

Han et al[11]

Cytosolic protein

36

Bleomycin Hydrolase

DVT

Proximity extension assays

Plasma

Up-regulated

Memon et.al[18]

37

LDHB (Lactate dehydrogenase B)

VTE

Mass spectrometry

Plasma

Up-regulated

Liu.et.al[14]

38

Human immunodeficiency virus type 1 enhancer binding protein1(HIVEP1)

VTE

IC-MS

Plasma

Up-regulated

Bruzelius et.al[30]

39

Platelet derived growth factor β(PDGFB)

VTE

IC-MS

Plasma

Up-regulated

Bruzelius et.al[30]

40

GAS6

VTE

ELISA

Plasma

Up-regulated

A.Schnegg-kaufmam et.al[31]

41

Transferrin receptor protein-1

DVT

Proximity extension assays

Plasma

Up-regulated

Memon et.al[18]

42

TF pathway inhibitor

DVT

Proximity extension assays

Plasma

Up-regulated

Memon et.al[18]

43

Glial cell line derived neutrophic factor(GDNF)

PE

Proximity extension assays

Plasma

Up-regulated

Ten cate et.al[25]

44

Gelsolin (GSN),

PE

Antibody based assay

Plasma

Down-regulated

Han et al[11]

 

Table 1: A list of differentially expressed proteins in venous thromboembolic disorder

CONCLUSION

At present, radiological diagnosis of VTE is one of the reliable ways to diagnose VTE, but it is difficult in severely ill patients because of the uncomfortable invasion and complex procedures. Though VTE is a multifactorial disease, pro-inflammatory biomarkers are more desirable for the early diagnosis of thrombosis and critically ill patients. As a therapeutic aspect, D-dimer and Factor VIII have been proven for the prediction of severity in VTE and treatment action on the duration of anticoagulant use. Lipoproteins, glycoproteins, and inflammatory proteins have the potential for diagnosis and therapeutic action. The interaction of proteins confirmed that CRP, IL6, IL8, IL17, selectin family, JAK-STAT group and tissue factor proteins including VWF proteins are significantly interconnected in disease manifestation. The mechanism of P-selectin, FVIII and IL-10 in coagulation gives a new insight into the pathophysiology and inhibition of anti-inflammatory cytokines in VTE, which may help in dealing with inflammation due to DVT /PE and will give some hope in immunotherapy implementation. Genetic factors are also essential for the pathogenesis of DVT. Further investigations, including full genome sequencing and genome-wide association studies (GWAS), lipidomics may help to elucidate the mechanisms underlying the risk of thrombosis and therapeutic strategies for precision medicine to reduce morbidity and mortality.

Author Contribution: Study design, conceptualization, literature search, Interpretation, Interaction analysis, KEGG pathway analysis and Original draft, done by BP.  Literature search and data collection done by BP & NR. Critical review and final approval done by CKRM, BP, HT and SA.

REFERENCES
  1. Cushman M(2007) :Epidemiology and risk factors for venous thrombosis. Seminars in hematology 44: 62-69. doi: 10.1053/j.seminhematol.2007.02.004.
  2. Schulman S, Lindmarker P, Holmström M, et al (2006): Post-thrombotic syndrome, recurrence, and death 10 years after the first episode of venous thromboembolism treated with warfarin for 6 weeks or 6 months. J Thrombo and Haemost 4: 734–742. doi: 10.1111/j.1538-7836.2006.01795.x.
  3. Owens AP 3rd, Mackman N. Tissue factor and thrombosis: The clot starts here. Thromb Haemost. 2010 ;104(3):432-9. doi: 10.1160/TH09-11-0771.
  4. Mackman N, Tilley RE, Key NS. Role of the extrinsic pathway of blood coagulation in hemostasis and thrombosis. Arterioscler Thromb Vasc Biol. 2007;27(8):1687-1693. doi:10.1161/ATVBAHA.107.141911
  5. Heit JA, Silverstein MD, Mohr DN, Petterson TM, O'Fallon WM, Melton LJ 3rd. Predictors of survival after deep vein thrombosis and pulmonary embolism: a population-based, cohort study. Arch Intern Med. 1999 ;159(5):445-53. doi: 10.1001/archinte.
  6. Freund Y, Cohen-Aubart F, Bloom B. Acute Pulmonary Embolism: A Review. JAMA. 2022 ;328(13):1336-1345. doi: 10.1001/jama.2022.16815.
  7. Gomes MP, Solomon AL. Vascular Disease Patient Information Page: Venous thrombosis at unusual sites. Vasc Med. 2022 ;27(3):312-316. doi: 10.1177/1358863X221087251.
  8. Ding, J., Yue, X., Tian, X. et al.Association between inflammatory biomarkers and venous thromboembolism: a systematic review and meta-analysis. Thrombosis J 21, 82 (2023). https://doi.org/10.1186/s12959-023-00526-y
  9. Roy DC, Wang TF, Lun R.et al. Circulating Blood Biomarkers and Risk of Venous Thromboembolism in Cancer Patients: A Systematic Review and Meta-Analysis. Thromb Haemost. 2024 ;124(12):1117-1133. doi: 10.1055/a-2330-1371.
  10. Zhang YX, Li JF, Yang YH, Huang K, Miao R, Zhai ZG, Wang C. Identification of haptoglobin as a potential diagnostic biomarker of acute pulmonary embolism. Blood Coagul Fibrinolysis. 2018 ;29(3):275-281. doi: 10.1097/MBC.0000000000000715.
  11. Han B, Li C, Li H et al. Discovery of plasma biomarkers with data-independent acquisition mass spectrometry and antibody microarray for diagnosis and risk stratification of pulmonary embolism. J Thromb Haemost. 2021 ;19(7):1738-1751. doi: 10.1111/jth.15324.
  12. Kölmel S, Hobohm L, Käberich A et.al. Potential Involvement of Osteopontin in Inflammatory and Fibrotic Processes in Pulmonary Embolism and Chronic Thromboembolic Pulmonary Hypertension. Thromb Haemost. 2019 ;119(8):1332-1346. doi: 10.1055/s-0039-1692174.
  13. Watts JA, Lee YY, Gellar MA, Fulkerson MB, Hwang SI, Kline JA. Proteomics of microparticles after experimental pulmonary embolism. Thromb Res. 2012 Jul;130(1):122-8. doi: 10.1016/j.thromres.2011.09.016.
  14. Liu Y, Gao L, Fan Y, Ma R, An Y, Chen G, Xie Y. Discovery of protein biomarkers for venous thromboembolism in non-small cell lung cancer patients through data-independent acquisition mass spectrometry. Front Oncol. 2023 9;13:1079719. doi: 10.3389/fonc.2023.1079719.
  15. Granholm F., Bylund D., Shevchenko G., Lind S. B., Henriksson A. E. A Feasibility Study on the Identification of Potential Biomarkers in Pulmonary Embolism Using Proteomic Analysis // Clinical and Applied Thrombosis/Hemostasis. 2022. Vol. 28. p. 107602962210743.
  16. Frischmuth T, Hindberg K, Aukrust P, Ueland T, Braekkan SK, Hansen JB, Morelli VM. Elevated plasma levels of plasminogen activator inhibitor-1 are associated with risk of future incident venous thromboembolism. J Thromb Haemost. 2022 ;20(7):1618-1626. doi: 10.1111/jth.15701.
  17. Tavares IR, Caffaro RA, Portugal MF et al. Biomarkers Profile in Provoked VersusUnprovoked Deep Venous Thrombosis. Clin Appl Thromb Hemost. 2024 :10760296241238211. doi: 10.1177/10760296241238211. 2024;30.
  18. Memon AA, Sundquist K, PirouziFard M,et al. Identification of novel diagnostic biomarkers for deep venous thrombosis. Br J Haematol. 2018 ;181(3):378-385. doi: 10.1111/bjh.15206. Epub 2018
  19. Watts JA, Lee YY, Gellar MA, Fulkerson MB, Hwang SI, Kline JA. Proteomics of microparticles after experimental pulmonary embolism. Thromb Res. 2012 Jul;130(1):122-8. doi: 10.1016/j.thromres.2011.09.016.
  20. Vormittag R, Vukovich T, Mannhalter C,et al. Haptoglobin phenotype 2-2 as a potentially new risk factor for spontaneous venous thromboembolism. Haematologica. 2005 ;90(11):1557-61.
  21. van Hylckama Vlieg A, van der Linden IK, Bertina RM, Rosendaal FR. High levels of factor IX increase the risk of venous thrombosis. Blood. 2000 ;95(12):3678-82. 
  22. Meijers JC, Tekelenburg WL, Bouma BN, Bertina RM, Rosendaal FR. High levels of coagulation factor XI as a risk factor for venous thrombosis. N Engl J Med. 2000;342(10):696-701. doi: 10.1056/NEJM200003093421004.
  23. Watany MM, Abdou S, Elkolaly R, Elgharbawy N, Hodeib H. Evaluation of admission levels of P, E and L selectins as predictors for thrombosis in hospitalized COVID-19 patients. Clin Exp Med. 2022 ;22(4):567-575. doi: 10.1007/s10238-021-00787-9.
  24. Zhou L, Qi XL, Xu MX, Mao Y, Liu ML, Song HM. Microparticles: new light shed on the understanding of venous thromboembolism. Acta Pharmacol Sin. 2014 ;35(9):1103-10. doi: 10.1038/aps.2014.73.
  25. Ten Cate V, Prochaska JH, Schulz A,et al. Protein expression profiling suggests relevance of noncanonical pathways in isolated pulmonary embolism. Blood. 2021 ;137(19):2681-2693. doi: 10.1182/blood.2019004571.
  26. Roumen-Klappe EM, den Heijer M, van Uum SH, van der Ven-Jongekrijg J, van der Graaf F, Wollersheim H. Inflammatory response in the acute phase of deep vein thrombosis. J Vasc Surg. 2002 ;35(4):701-6. doi: 10.1067/mva.2002.121746. 
  27. Ding P, Zhang S, Yu M,et al. IL-17A promotes the formation of deep vein thrombosis in a mouse model. Int Immunopharmacol. 2018 ;57:132-138. doi: 10.1016/j.intimp.2018.02.006.
  28. Matos MF, Lourenço DM, Orikaza CM, Bajerl JA, Noguti MA, Morelli VM. The role of IL-6, IL-8 and MCP-1 and their promoter polymorphisms IL-6 -174GC, IL-8 -251AT and MCP-1 -2518AG in the risk of venous thromboembolism: a case-control study. Thromb Res. 2011 ;128(3):216-20. doi: 10.1016/j.thromres.2011.04.016.
  29. Razzaq M, Iglesias MJ, Ibrahim-Kosta M, et al.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(1):14015. doi: 10.1038/s41598-021-93390-7. 
  30. Bruzelius M, Iglesias MJ, Hong MG, et al. PDGFB, a new candidate plasma biomarker for venous thromboembolism: results from the VEREMA affinity proteomics study. Blood. 2016 ;128(23):e59-e66. doi: 10.1182/blood-2016-05-711846.
  31. Schnegg-Kaufmann, A., Calzavarini, S., Limacher, A., Mean, M., Righini, M., Staub, D., Beer, J. H., Frauchiger, B., Osterwalder, J., Kucher, N., Matter, C. M., Husmann, M., Banyai, M., Aschwanden, M., Mazzolai, L., Hugli, O., Nagler, M., Daskalakis, M., Rodondi, N., Aujesky, D., … Angelillo-Scherrer, A. (2019). A high Gas6 level in plasma predicts venous thromboembolism recurrence, major bleeding and mortality in the elderly: a prospective multicenter cohort study. Journal of thrombosis and haemostasis : JTH17(2), 306–318. https://doi.org/10.1111/jth.14365.
  32. Bhatia HS, Wilkinson MJ. Lipoprotein(a): Evidence for Role as a Causal Risk Factor in Cardiovascular Disease and Emerging Therapies. J Clin Med. 2022 ;11(20):6040. doi: 10.3390/jcm11206040.
  33. Gressenberger P, Posch F, Pechtold M.etal. Lipoprotein(a) and Pulmonary Embolism Severity-A Retrospective Data Analysis. Front Cardiovasc Med. 2022 Feb 7;9:808605. doi: 10.3389/fcvm.2022.808605.
  34. Koupenova M, Kehrel BE, Corkrey HA, Freedman JE. Thrombosis and platelets: an update. Eur Heart J. 2017 14;38(11):785-791. doi: 10.1093/eurheartj/ehw550.
  35. Waheed SM, Kudaravalli P, Hotwagner DT. Deep Vein Thrombosis. [Updated 2023 Jan 19]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 -. Available from: https://www.ncbi.nlm.nih.gov/books/NBK507708/.
  36. O'Donnell J, Tuddenham EG, Manning R, Kemball-Cook G, Johnson D, Laffan M. High prevalence of elevated factor VIII levels in patients referred for thrombophilia screening: role of increased synthesis and relationship to the acute phase reaction. Thromb Haemost. 1997 ;77(5):825-8.
  37. Soria JM, Almasy L, Souto JC, et.al. A new locus on chromosome 18 that influences normal variation in activated protein C resistance phenotype and factor VIII activity and its relation to thrombosis susceptibility. Blood. 2003 ;101(1):163-7. doi: 10.1182/blood-2002-06-1792.
  38. Furie B, Furie BC. The molecular basis of blood coagulation. Cell. 1988 ;53(4):505-18. doi: 10.1016/0092-8674(88)90567-3.
  39. Hertzberg M. Biochemistry of factor X. Blood Rev. 1994 ;8(1):56-62. doi: 10.1016/0268-960x(94)90007-8
  40. Wells PS, Anderson DR, Rodger M,et al. Evaluation of D-dimer in the diagnosis of suspected deep-vein thrombosis. N Engl J Med. 2003 ;349(13):1227-35. doi: 10.1056/NEJMoa023153.
  41. Douma RA, le Gal G, Söhne M,et al. Potential of an age adjusted D-dimer cut-off value to improve the exclusion of pulmonary embolism in older patients: a retrospective analysis of three large cohorts. BMJ. 2010 30;340:c1475. doi: 10.1136/bmj.c1475.
  42. Rodger MA, Kahn SR, Wells PS,et al. Identifying unprovoked thromboembolism patients at low risk for recurrence who can discontinue anticoagulant therapy. CMAJ. 2008 ;179(5):417-26. doi: 10.1503/cmaj.080493.
  43. Pulivarthi S, Gurram MK. Effectiveness of d-dimer as a screening test for venous thromboembolism: an update. N Am J Med Sci. 2014 ;6(10):491-9. doi: 10.4103/1947-2714.
  44. Clyne B, Olshaker JS. The C-reactive protein. J Emerg Med. 1999 ;17(6):1019-25. doi: 10.1016/s0736-4679(99)00135-3.
  45. proston NR, Ashworth JJ. Role of C-Reactive Protein at Sites of Inflammation and Infection. Front Immunol. 2018 13;9:754. doi: 10.3389/fimmu.2018.00754.
  46. Miszti-Blasius K, Debreceni IB, Felszeghy S, Dezso B, Kappelmayer J. Lack of P-selectin glycoprotein ligand-1 protects mice from thrombosis after collagen/epinephrine challenge. Thromb Res. 2011 ;127(3):228-34. doi: 10.1016/j.thromres.2010.11.022.
  47. Kline JA, Zeitouni R, Marchick MR, Hernandez-Nino J, Rose GA. Comparison of 8 biomarkers for prediction of right ventricular hypokinesis 6 months after submassive pulmonary embolism. Am Heart J. 2008 ;156(2):308-14. doi: 10.1016/j.ahj.2008.03.026.
  48. Key NS, Chantrathammachart P, Moody PW, Chang JY. Membrane microparticles in VTE and cancer. Thromb Res. 2010 ;125 Suppl 2:S80-3. doi: 10.1016/S0049-3848(10)70020-7. 
  49. Shimada Y, Hasegawa M, Takehara K, Sato S. Elevated serum L-selectin levels and decreased L-selectin expression on CD8(+) lymphocytes in systemic sclerosis. Clin Exp Immunol. 2001 ;124(3):474-9. doi: 10.1046/j.1365-2249.
  50. Wang JG, Manly D, Kirchhofer D, Pawlinski R, Mackman N. Levels of microparticle tissue factor activity correlate with coagulation activation in endotoxemic mice. J Thromb Haemost. 2009 ;7(7):1092-8. doi: 10.1111/j.1538-7836.2009.03448.x.
  51. Turizo MJF, Patell R, Zwicker JI. Identifying novel biomarkers using proteomics to predict cancer-associated thrombosis. Bleeding Thromb Vasc Biol. 2024;3(Suppl 1):120. doi: 10.4081/btvb.2024.120.
  52. Snel B, Lehmann G, Bork P, Huynen MA. STRING: a web-server to retrieve and display the repeatedly occurring neighbourhood of a gene. Nucleic Acids Res. 2000 Sep 15;28(18):3442-4. doi: 10.1093/nar/28.18.3442.
Recommended Articles
Research Article
Detection Of Hcv Rna and Clinico Epidemiological Profile of Seropositive Hepatitis C Patients Attending a Tertiary Center in Central Kerala
...
Published: 28/04/2025
Download PDF
Case Series
Ischemic Stroke: A Case Series Unraveling Benign and Malignant Causes of High Prothrombotic Tendency
...
Published: 28/04/2025
Download PDF
Research Article
Analysis Of Thyroid Lesions Cytology by The Bethesda System and Its Histopathological Correlation.
...
Published: 28/04/2025
Download PDF
Research Article
Retrospective Study of Uterine Corpus Lesions Over a Period of One Year in Tertiary Care Centre
...
Published: 25/04/2025
Download PDF
Chat on WhatsApp
Copyright © EJCM Publisher. All Rights Reserved.