[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (125)

Search Parameters:
Keywords = LMWH

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 2996 KiB  
Systematic Review
Incidence and Predictors of Early and Late Radial Artery Occlusion after Percutaneous Coronary Intervention and Coronary Angiography: A Systematic Review and Meta-Analysis
by Aisha Khalid, Hans Mautong, Kayode Ahmed, Zaina Aloul, Jose Montero-Cabezas and Silvana Marasco
J. Clin. Med. 2024, 13(19), 5882; https://doi.org/10.3390/jcm13195882 - 2 Oct 2024
Viewed by 566
Abstract
Introduction: Trans-radial access for coronary angiography and percutaneous coronary intervention (PCI) has gained popularity due to its advantages over the traditional transfemoral approach. However, radial artery occlusion (RAO) remains a common complication following trans-radial procedures. This study aimed to investigate the incidence of [...] Read more.
Introduction: Trans-radial access for coronary angiography and percutaneous coronary intervention (PCI) has gained popularity due to its advantages over the traditional transfemoral approach. However, radial artery occlusion (RAO) remains a common complication following trans-radial procedures. This study aimed to investigate the incidence of early and late RAO along with their risk factors. Methods: Six databases, Medline (Ovid), National Library of Medicine (MeSH), Cochrane Database of Systematic Reviews (Wiley), Embase, Scopus, and Global Index Medicus, were searched. The systematic review and meta-analysis followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Data were extracted and analyzed. Using a random-effect model, the primary endpoint was the overall incidence of RAO after invasive coronary procedures. Subgroup analysis and meta-regression were also performed to identify possible predictors of RAO. Results: A total of 41 studies with 30,020 patients were included. The overall incidence of RAO was 13% (95% CI = 0.09–0.16). The incidence of early RAO (within 24 h) was 14% (95% CI = 0.10–0.18) in 26 studies, while the incidence of late RAO (after 24 h) was 10% (95% CI = 0.04–0.16) in 22 studies. The average incidence rates of early RAO in studies with catheter sizes of <6 Fr, 6 Fr, and >6 Fr were 9.8%, 9.4%, and 8.8%. The overall effect size of female gender as a predictor was 0.22 with a 95% CI of 0.00–0.44. Age was a potential predictor of early RAO (B = 0.000357; 95% CI = −0.015–0.0027, p: 0.006). Conclusions: This meta-analysis provides essential information on the incidence of early (14%) and late (10%) RAO following angiographic procedures. Additionally, our findings suggest that female sex and age are possible predictors of RAO. A larger catheter, especially (6 Fr) and hemostatic compression time <90 min post-procedure, substantially reduced the incidence of RAO. The use of oral anticoagulation and the appropriate dosage of low-molecular-weight heparin (LMWH) does reduce RAO, but a comparison between them showed no statistical significance. Full article
(This article belongs to the Special Issue Advances in Coronary Artery Disease)
Show Figures

Figure 1

Figure 1
<p>PRISMA—selected studies for the meta-analysis [<a href="#B6-jcm-13-05882" class="html-bibr">6</a>].</p>
Full article ">Figure 2
<p>(<b>A</b>) Early and late RAO incidence. (<b>B</b>) Low-ose heparin vs. high-dose heparin. Heterogeneity assessment: I<sup>2</sup> = 61.37%, <span class="html-italic">τ</span><sup>2</sup> = 0.85, H<sup>2</sup> = 2.59, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Subgroup analysis (PCI vs. CA). Heterogeneity assessment: I<sup>2</sup> = 61.37%, <span class="html-italic">τ</span><sup>2</sup> = 0.85, H<sup>2</sup> = 2.59, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Forest plot of the association between different catheter sizes and the incidence of RAO.</p>
Full article ">Figure 5
<p>(<b>A</b>) (<b>panel superior</b>): Forest plot of the incidence of early and late RAO for interventional procedures using oral anticoagulation. (<b>B</b>) (<b>panel inferior</b>): Forest plot of the incidence of early and late RAO for interventional procedures using oral anticoagulation versus LMWH (low vs. high).</p>
Full article ">Figure 6
<p>Forest plot of studies assessing the impact of hemostatic compression times on the incidence of early versus late RAO, summary log odds ratio with 95% confidence interval (CI), and weight (%) of each study.</p>
Full article ">Figure 7
<p>Female gender and RAO incidence across the studies.</p>
Full article ">
18 pages, 1838 KiB  
Article
Influence of Anticoagulants and Heparin Contaminants on the Suitability of MMP-9 as a Blood-Derived Biomarker
by Daniela Küper, Josefin Klos, Friederike Kühl, Rozan Attili, Korbinian Brand, Karin Weissenborn, Ralf Lichtinghagen and René Huber
Int. J. Mol. Sci. 2024, 25(18), 10106; https://doi.org/10.3390/ijms251810106 - 20 Sep 2024
Viewed by 541
Abstract
In contrast to other common anticoagulants such as citrate and low-molecular-weight heparin (LMWH), high-molecular-weight heparin (HMWH) induces the expression of matrix metalloproteinase (MMP)-9, which is also measured as a biomarker for stroke in blood samples. Mechanistically, HMWH-stimulated T cells produce cytokines that induce [...] Read more.
In contrast to other common anticoagulants such as citrate and low-molecular-weight heparin (LMWH), high-molecular-weight heparin (HMWH) induces the expression of matrix metalloproteinase (MMP)-9, which is also measured as a biomarker for stroke in blood samples. Mechanistically, HMWH-stimulated T cells produce cytokines that induce monocytic MMP-9 expression. Here, the influence of further anticoagulants (Fondaparinux, Hirudin, and Alteplase) and the heparin-contaminating glycosaminoglycans (GAG) hyaluronic acid (HA), dermatan sulfate (DS), chondroitin sulfate (CS), and over-sulfated CS (OSCS) on MMP-9 was analyzed to assess its suitability as a biomarker under various conditions. Therefore, starved Jurkat T cells were stimulated with anticoagulants/contaminants. Subsequently, starved monocytic THP-1 cells were incubated with the conditioned Jurkat supernatant, and MMP-9 mRNA levels were monitored (quantitative (q)PCR). Jurkat-derived mediators secreted in response to anticoagulants/contaminants were also assessed (proteome profiler array). The supernatants of HMWH-, Hirudin-, CS-, and OSCS-treated Jurkat cells comprised combinations of activating mediators and led to a significant (in the case of OSCS, dramatic) MMP-9 induction in THP-1. HA induced MMP-9 only in high concentrations, while LMWH, Fondaparinux, Alteplase, and DS had no effect. This indicates that depending on molecular weight and charge (but independent of anticoagulant activity), anticoagulants/contaminants provoke the expression of T-cell-derived cytokines/chemokines that induce monocytic MMP-9 expression, thus potentially impairing the diagnostic validity of MMP-9. Full article
(This article belongs to the Special Issue Glycosaminoglycans, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Supernatant of high-molecular-weight heparin (HMWH)-, but not low-molecular-weight heparin (LMWH)- or Fondaparinux-treated Jurkat cells, induces MMP-9 mRNA in THP-1 cells. Jurkat cells were starved for 24 h and then incubated for 24 h with injection- or diagnostic-grade HMWH (±50 IU per well; equates to 400 µg), diagnostic-grade LMWH (Enoxaparin; ±400 µg), or injection-grade Fondaparinux (±400 µg). Subsequently, the supernatant was harvested and transferred to starved THP-1 cells. Following a 24 h incubation phase, MMP-9 mRNA expression in the THP-1 cells was determined via quantitative (q)PCR. The MMP-9 expression value in starved THP-1 cells at 0 h (i.e., before the transfer of Jurkat supernatant) was set as 1 (n ≥ 4, mean ± SD; Mann–Whitney U-test, * <span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">Figure 2
<p>Supernatant of Hirudin-serum- and Alteplase-treated Jurkat cells induces MMP-9 mRNA in THP-1 cells. Jurkat cells were starved (24 h) and then incubated for 24 h with Hirudin serum (±220 µL per well; derived from a Hirudin blood collection tube) or Alteplase (±50,000 IU). Afterwards, the supernatant was transferred to starved THP-1 cells, and following a 24 h incubation phase, MMP-9 mRNA expression was determined (qPCR). The MMP-9 mRNA level in starved THP-1 cells at 0 h was set as 1 (n ≥ 5, mean ± SD; Mann–Whitney U-test, ** <span class="html-italic">p</span> ≤ 0.01).</p>
Full article ">Figure 3
<p>Effect of the supernatant of hyaluronic acid (HA)-, dermatan sulfate (DS)-, chondroitin sulfate (CS)-, and over-sulfated CS (OSCS)-treated Jurkat cells on MMP-9 mRNA in THP-1 cells. Jurkat cells were starved (24 h) and then incubated for 24 h ±100 or 200 µg HA, 400 µg DS, 400 µg CS, or 400 µg OSCS per well. Afterwards, the supernatant was transferred to starved THP-1 cells, and following a 24 h incubation phase, MMP-9 mRNA expression was determined (qPCR). The MMP-9 mRNA level in starved THP-1 cells at 0 h was set as 1 (n ≥ 3, mean ± SD; Mann–Whitney U-test, * <span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">Figure 4
<p>Induction of MMP-9 expression by T-cell-derived mediators. A total of 0.5 × 10<sup>6</sup> THP-1 cells/well were starved for 24 h. Subsequently, cells were stimulated for 24 h with combinations of soluble intercellular adhesion molecule (sICAM)-1, interleukin (IL)-16, IL-22, IL-5 (5 ng/mL each), interferon (IFN)-γ (10 ng/mL), and kallikrein 3 (1 µM). MMP-9 mRNA expression was determined using qPCR. The MMP-9 mRNA level in starved THP-1 cells at 0 h was set as 1 (n = 3, mean ± SD; Mann–Whitney U-test, * <span class="html-italic">p</span> ˂ 0.05).</p>
Full article ">Figure 5
<p>Extended model of monocytic MMP-9 expression during anticoagulation with specific agents. Our results suggest that in the presence of anticoagulants and heparin contaminants with high charge and molecular weight (MW), T cells secrete sICAM-1 in combination with different interleukins (e.g., IL-5, -16, or -22). In the presence of OSCS, T cells secrete additional mediators (IFN-γ, kallikrein 3) that intensify monocytic activation. * Injection-grade HMWH-induced sICAM-1 and IL-16 activate an additional IL-8-dependent positive autocrine feedback loop [<a href="#B20-ijms-25-10106" class="html-bibr">20</a>]. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 6
<p>Experimental Design. Jurkat T cells were cultivated in starvation medium for 24 h and subsequently stimulated with anticoagulants or contaminants. After 24 h, the conditioned Jurkat medium was transferred to starved THP-1 monocytic cells. In parallel, cytokines secreted in response to the respective stimuli were detected in aliquots of the Jurkat supernatant using the Proteome Profiler Human (XL) Cytokine Array. The MMP-9 mRNA expression in THP-1 cells was assessed via qPCR following a 24 h incubation phase in the T-cell-derived medium. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
9 pages, 811 KiB  
Article
Evaluating the Effectiveness of Enoxaparin in Treating Pediatric Arterial Thrombosis in Saudi Arabia
by Meshary Al-Meshary, Abdulrahman Alotaibi, Nouf S. Alsagri, Asmaa AlZhrani, Husam I. Ardah and Mohammed A. Alnuhait
Children 2024, 11(9), 1139; https://doi.org/10.3390/children11091139 - 19 Sep 2024
Viewed by 524
Abstract
Background: Thrombosis is the abnormal formation of blood clots within blood vessels; it results from an imbalance between fibrinolytic, pro-coagulant, and anticoagulant systems. Pediatric arterial thrombosis, especially related to catheter usage, is an emerging issue with limited evidence. This study evaluates the efficacy [...] Read more.
Background: Thrombosis is the abnormal formation of blood clots within blood vessels; it results from an imbalance between fibrinolytic, pro-coagulant, and anticoagulant systems. Pediatric arterial thrombosis, especially related to catheter usage, is an emerging issue with limited evidence. This study evaluates the efficacy of enoxaparin in treating arterial thrombosis in pediatric patients at a single center. Methods: A retrospective single-center study included children under 14 years old diagnosed with catheter-related arterial thrombosis (CAT) and treated with low-molecular-weight heparin (LMWH) at King Abdulaziz Medical City between 2016 and 2021. Patients without follow-up at our institution or those using other anticoagulants were excluded. Data collected included age, sex, weight, catheter type, location and degree of thrombosis, ultrasonographic results, treatment duration, hemoglobin and platelet levels, and missed refills. Radiologic confirmation of CAT was required for inclusion. Results: This study included 111 children treated with enoxaparin for non-cerebral arterial thrombosis. The median age at diagnosis was 3 months, with 58% being male patients. Most cases (87%) involved cardiac catheterization, and all were confirmed using ultrasonography. Complete thrombus resolution was achieved in 90% of patients, partial resolution in 8.1%, and 1.8% had no resolution. The median duration of enoxaparin therapy was 20 days. Multivariate analysis indicated that higher age and lower body weight were associated with a higher risk of non-resolution. Indwelling catheters also posed a greater risk of non-resolution compared to cardiac catheters. Conclusions: Enoxaparin proved effective in treating catheter-related arterial thrombosis in children, with high resolution rates and few side effects. This study helps inform treatment strategies in pediatric thrombosis management and highlights the need for further research to refine treatment durations and address patient risk factors. Full article
Show Figures

Figure 1

Figure 1
<p>The cumulative resolution rate over time for all patients. Of 110 cases, 108 achieved resolutions (complete resolution = 100, partial resolution = 8). In total, 62/108 (57.4%) achieved resolution after 20 days of therapy, 87/108 (80.5%) achieved resolution after 40 days of therapy, 103/108 (95%) achieved resolution after 70 days of therapy, and 108/108 (100%) achieved resolution after 86 days of therapy.</p>
Full article ">Figure 2
<p>Cumulative resolution rate over time according to the type of catheter. One case was considered as an outlier (time to partial resolution = 303 days) and, hence, was excluded from this analysis. The type of catheterization was unknown for 3 cases, which were also excluded. Of 107 cases, 97 patients achieved complete resolution (86 in the cardiac catheter group and 11 in the indwelling catheter group), 8 patients achieved partial resolution (5 in the cardiac catheter group and 3 in the indwelling group), and 2 patients in the cardiac catheter group failed to resolve.</p>
Full article ">
5 pages, 2333 KiB  
Interesting Images
The Aortic Prosthesis and Aortic Valve Bioprosthesis Trombosis as a Late Complication in Patients after the Bentall Procedure Followed by a Valve-in-Valve Transcatheter Aortic Valve Implantation
by Paweł Muszyński, Oliwia Grunwald, Maciej Południewski, Paweł Kralisz, Szymon Kocańda, Tomasz Hirnle, Sławomir Dobrzycki and Marcin Kożuch
Diagnostics 2024, 14(18), 2070; https://doi.org/10.3390/diagnostics14182070 - 19 Sep 2024
Viewed by 487
Abstract
Background: Valve-in-Valve (ViV) transcatheter aortic valve implantation (TAVI) has emerged as a viable therapeutic option for structural valve degeneration following surgical aortic valve replacement (SAVR) or prior TAVI. However, the understanding of long-term complications and their management remains limited. Case presentation: We present [...] Read more.
Background: Valve-in-Valve (ViV) transcatheter aortic valve implantation (TAVI) has emerged as a viable therapeutic option for structural valve degeneration following surgical aortic valve replacement (SAVR) or prior TAVI. However, the understanding of long-term complications and their management remains limited. Case presentation: We present the case of a 69-year-old male with a history of ViV-TAVI, who presented with symptoms of non-ST elevation myocardial infarction (NSTEMI) and transient ischemic attack (TIA). Computed tomography (CT) revealed thrombosis of the ascending aortic graft and aortic valve prosthesis. Transthoracic echocardiography (TTE) further confirmed new valve dysfunction, indicated by an increase in the aortic valve mean gradient. Treatment with low-molecular-weight heparin (LMWH) resulted in partial thrombus resolution. The multidisciplinary Heart Team opted against coronary angiography and recommended the long-term administration of vitamin K antagonists (VKAs). Follow-up CT showed the complete resolution of the thrombus. Conclusions: Thrombosis of the aortic graft and aortic valve following ViV-TAVI may be attributed to alterations in blood flow or mechanical manipulations during the TAVI procedure, yet it can be effectively managed with VKA therapy. CT is a valuable tool in coronary assessment in patients with NSTEMI and aortic valve and/or aortic graft thrombosis. Full article
(This article belongs to the Special Issue Cardiovascular Imaging)
Show Figures

Figure 1

Figure 1
<p>A 69-year-old male complaining of recurring chest pain, episodes of syncope and visual distortion was admitted to the hospital. His past medical history included the replacement of the ascending aorta with surgical aortic valve replacement (SAVR) in 2011 due to aortic aneurysm and aortic stenosis (Bentall procedure). He underwent transcatheter aortic valve implantation (TAVI) in 2021 due to the degeneration of aortic valve bioprosthesis, using a 26 mm balloon-expandable Sapien 3 Ultra (Edwards Lifesciences, Irvine, CA, USA). The treatment course after TAVI involved longitudinal single antiplatelet therapy (SAPT)—aspirin. In addition, he reported reduced exercise tolerance since the TAVI procedure. The coronarography in 2011 and angio-CT in 2021 excluded the significant coronary artery disease before operations. The angio-CT, including the aorta and head, at the current hospitalization found the thrombosis of the ascending aorta graft causing 80% stenosis (<b>A</b>–<b>C</b>; black arrow). Furthermore, due to the resolution of the neurological dysfunction, the patient was diagnosed with a transient ischemic attack (TIA). The transthoracic echocardiography revealed an increase in gradient through aortic valve bioprosthesis—a mean gradient of 24 mm Hg. The low-molecular-weight heparin (LMWH) in therapeutic dose was initiated. Despite the increase in cardiac enzymes, the chest pain and the diagnosis of non-ST elevation myocardial infarction (NSTEMI), the coronarography was not performed due to the risk involving the dislocation of the thrombi. After three days, the control CT displayed a significant reduction in the thrombus. The patient consulted with the Heart Team and was transferred to a higher reference clinical hospital with the availability of the cardiac surgery department.</p>
Full article ">Figure 2
<p>After the consultation with the cardiac radiologist, the third ECG-gated cardiac CT was performed on the eighth day after the initial CT to access both the aorta and coronary arteries. To increase the quality of the image, an additional beta-blocker and nitroglycerin were administered before examination. The CT revealed aortic valve prosthesis thrombosis (<b>A</b>; red arrow) and a reduction in aortic thrombosis (<b>B</b>; orange arrow). The previous thrombus site was localized to originate within the ascending aorta graft, which was covered by soft tissue that resembled an ulcer. The probable dissection of the neointima along the prosthesis was the suspected cause of the initial aortic thrombosis (<b>C</b>,<b>D</b>; black arrow).</p>
Full article ">Figure 3
<p>The CT discluded significant coronary artery stenosis (Cx—circumflex artery, RCA—right coronary artery, LAD—left anterior descending artery)<b>.</b> The myocardial infarction was treated conservatively with anticoagulation due to the presence of aortic graft and valve thrombosis, and it led to the resolution of the chest pain. The clinically insignificant lesions in the coronary arteries on CT were suggestive of the fact that NSTEMI could be caused by the occlusion of the ostium of the arteries by the aortic valve thrombus.</p>
Full article ">Figure 4
<p>The transthoracic echocardiography showed a reduction in the gradients through the aortic valve prosthesis with parameters similar to that after the TAVI (on left: post-TAVI; on right: post-LMWH treatment). According to the 2021 ESC/EACTS Guidelines, in patients after TAVI, lifelong SAPT is recommended in the lack of indications for oral anticoagulants (OACs) [<a href="#B1-diagnostics-14-02070" class="html-bibr">1</a>]. However, despite similar recommendations for aspirin, the 2020 ACC/AHA Guidelines suggest that in patients with a low bleeding risk, it is worth acknowledging the antithrombotic prophylaxis with DAPT or VKA [<a href="#B2-diagnostics-14-02070" class="html-bibr">2</a>]. The direct-acting oral anticoagulants (DOACs) were not found to be superior to the administering antiplatelet or VKA, and in patients without indications for OAC, were linked to a higher incidence of all-cause mortality [<a href="#B3-diagnostics-14-02070" class="html-bibr">3</a>,<a href="#B4-diagnostics-14-02070" class="html-bibr">4</a>,<a href="#B5-diagnostics-14-02070" class="html-bibr">5</a>]. The 2021 ESC/EACTS and 2020 ACC/AHA Guidelines for managing valvular heart disease indicate that anticoagulation using VKAs or UFH is a first-line therapy for bioprosthetic valve thrombosis. Such an approach is highly effective in the normalization of valve function in 85% of the patients [<a href="#B6-diagnostics-14-02070" class="html-bibr">6</a>].</p>
Full article ">Figure 5
<p>The transesophageal echocardiography failed to assess the aortic valve due to the artefacts. However, it confirmed the suspicion of the visible ruptured neointima within the aorta on the CT (<a href="#app1-diagnostics-14-02070" class="html-app">Video S1</a>—red circle). The patient again consulted with the Heart Team, and the decision about a conservative approach and administration of oral anticoagulation with vitamin K antagonists (VKAs) was sustained. At the 3-month follow-up, the patient was free from angina and syncope, and the control chest CT showed no sign of thrombus presence in the aortic region. The anatomical changes, such as the dimensions and shape of the aortic root, which occurred during TAVI, may have caused the dynamic switch in blood flow through the bioprosthetic valve, thus causing aortic remodelling. This contributes to the fact that the blood stagnates in the prosthetic sinuses, which complements Virchow’s triad and is attributed to thromboembolic events [<a href="#B7-diagnostics-14-02070" class="html-bibr">7</a>]. Furthermore, the ongoing structural bioprosthesis degeneration leading to fibrosis and calcification is often started by leaflet thickening and valve thrombosis [<a href="#B8-diagnostics-14-02070" class="html-bibr">8</a>]. The atherosclerotic plaque in the ascending aorta, or dissection of the neointima along the prosthesis, could also contribute to the thrombotic event. Aortic dissection was reported to be a rare TAVI procedure complication, occurring almost entirely as an acute condition. Regardless, there have been no previous reports of dissection of the neointima in the aortic prosthesis after ViV TAVI [<a href="#B9-diagnostics-14-02070" class="html-bibr">9</a>]. In our case, we suspect that a change in flow through the aorta or mechanical damage during the TAVI could have led to the dissection of the neointima and aortic thrombosis. Additionally, myocardial infarction in patients after TAVI can be the result of co-existing leaflet thrombosis and can cause difficulty in performing PCI procedures due to impaired coronary access [<a href="#B10-diagnostics-14-02070" class="html-bibr">10</a>]. In our case, the patient’s symptoms were most likely associated with the clinical presentation of ViV thrombosis and severe aortic graft thrombosis, in spite of the administration of SAPT—aspirin. The initial treatment with LMWH converted into VKA oral anticoagulants was sufficient to promote thrombus resolution and prevent recurrence at 3-month follow-up.</p>
Full article ">
14 pages, 4377 KiB  
Article
Structural Analysis and Anticoagulant Activity of Fucosylated Glycosaminoglycan from Sea Cucumber Phyllophorus proteus
by Jingwen Liu, Lihua Geng, Jing Wang, Yang Yue, Ning Wu and Quanbin Zhang
Foods 2024, 13(18), 2889; https://doi.org/10.3390/foods13182889 - 12 Sep 2024
Viewed by 625
Abstract
Phyllophorus proteus is a low-value sea cucumber from Indonesia and other tropical peripheral waters. In this study, a fucosylated glycosaminoglycan (FG) was extracted from P. proteus. It consists of GlcA, GalNAc, and Fuc, with a molecular weight of 67.1 kDa. The degraded [...] Read more.
Phyllophorus proteus is a low-value sea cucumber from Indonesia and other tropical peripheral waters. In this study, a fucosylated glycosaminoglycan (FG) was extracted from P. proteus. It consists of GlcA, GalNAc, and Fuc, with a molecular weight of 67.1 kDa. The degraded FG (dFG) was prepared by β-elimination. Structural analysis revealed that the main chain of dFG was composed of GalNAc and GlcA, linked alternately by β1,3 and β1,4 glycosidic bonds. The sulfate group was located at the 4 and 6 positions of GalNAc. Fuc was attached to the 3 position of GlcA by an α1,3 glycosidic bond, and the side chain of Fuc exhibited various sulfate substitutions. FG significantly prolonged the coagulation time of APTT, PT, TT, and FIB, surpassing the effect of LMWH, thereby demonstrating its ability to exert anticoagulant effects in both the endogenous and exogenous coagulation pathways. Conversely, dFG had no significant effect on the clotting time of PT, suggesting its lack of impact on the intrinsic coagulation pathway. This study elucidates the structural properties and potent anticoagulant activities of fucosylated glycosaminoglycan from P. proteus. Full article
(This article belongs to the Special Issue Marine Food: Development, Quality and Functionality)
Show Figures

Figure 1

Figure 1
<p>Molecular weight determination of the four polysaccharide fractions by HPGPC.</p>
Full article ">Figure 2
<p>Elution curve (<b>A</b>), molecular weight (<b>B</b>), and monosaccharide composition (<b>C</b>) analysis of FG purified by Sephadex G-100 column chromatography. ((<b>C</b>): 1 PMP; 2 GlcN; 3 GlcA; 4 GalN; 5 Glc; 6 GalNAc; 7 Gal; 8 Fuc; 9 Rib).</p>
Full article ">Figure 3
<p>Infrared spectrum of FG (1.1 M elution fraction).</p>
Full article ">Figure 4
<p><sup>1</sup>H-NMR (<b>A</b>) and <sup>13</sup>C-NMR (<b>B</b>) spectra of FG.</p>
Full article ">Figure 5
<p>Molecular weight measurement by HPLC of dFG-P6 elution fractions.</p>
Full article ">Figure 6
<p>1D and 2D-NMR spectra of dFG. <sup>1</sup>H NMR (<b>A</b>); <sup>13</sup>C NMR (<b>B</b>); <sup>1</sup>H-<sup>1</sup>H COSY (<b>C</b>); <sup>1</sup>H-<sup>1</sup>H TOCSY (<b>D</b>); <sup>1</sup>H-<sup>13</sup>C HMBC (<b>E</b>); <sup>1</sup>H-<sup>13</sup>C HSQC (<b>F</b>).</p>
Full article ">Figure 6 Cont.
<p>1D and 2D-NMR spectra of dFG. <sup>1</sup>H NMR (<b>A</b>); <sup>13</sup>C NMR (<b>B</b>); <sup>1</sup>H-<sup>1</sup>H COSY (<b>C</b>); <sup>1</sup>H-<sup>1</sup>H TOCSY (<b>D</b>); <sup>1</sup>H-<sup>13</sup>C HMBC (<b>E</b>); <sup>1</sup>H-<sup>13</sup>C HSQC (<b>F</b>).</p>
Full article ">Figure 6 Cont.
<p>1D and 2D-NMR spectra of dFG. <sup>1</sup>H NMR (<b>A</b>); <sup>13</sup>C NMR (<b>B</b>); <sup>1</sup>H-<sup>1</sup>H COSY (<b>C</b>); <sup>1</sup>H-<sup>1</sup>H TOCSY (<b>D</b>); <sup>1</sup>H-<sup>13</sup>C HMBC (<b>E</b>); <sup>1</sup>H-<sup>13</sup>C HSQC (<b>F</b>).</p>
Full article ">Figure 7
<p>The structure of dFG.</p>
Full article ">Figure 8
<p>Plasma recalcitrance experiments of FG (<b>A</b>) and dFG (<b>B</b>).</p>
Full article ">Figure 9
<p>In vitro anticoagulant assay of FG (blue), dFG (orange) and LMWF (grey). (<b>A</b>) APTT, (<b>B</b>) PT, (<b>C</b>) TT, (<b>D</b>) FIB.</p>
Full article ">
34 pages, 981 KiB  
Review
Evaluating Thromboprophylaxis Strategies for High-Risk Pregnancy: A Current Perspective
by Lucia Stančiaková, Kristína Brisudová, Ingrid Škorňová, Tomáš Bolek, Matej Samoš, Kamil Biringer, Ján Staško and Juraj Sokol
Pharmaceuticals 2024, 17(6), 773; https://doi.org/10.3390/ph17060773 - 13 Jun 2024
Cited by 2 | Viewed by 742
Abstract
Venous thromboembolism (VTE) represents one of the leading causes of death during pregnancy. The greatest risk for it is the presence of medical or family history of VTE, stillbirth, cesarean section and selected thrombophilia. Appropriate thromboprophylaxis has the potential to decrease the risk [...] Read more.
Venous thromboembolism (VTE) represents one of the leading causes of death during pregnancy. The greatest risk for it is the presence of medical or family history of VTE, stillbirth, cesarean section and selected thrombophilia. Appropriate thromboprophylaxis has the potential to decrease the risk of VTE in at-risk pregnant patients by 60–70%. Based on this, the authors reviewed the PubMed, Web of Science and Scopus databases to identify the possibilities of thromboprophylaxis in pregnant patients with a high risk of VTE. Moreover, they summarized its management in specific situations, such as cesarean delivery or neuraxial blockade. Currently, low-molecular-weight heparins (LMWH) are the preferred drugs for anticoagulant thromboprophylaxis in the course of pregnancy and postpartum due to easy administration and a lower rate of adverse events. Full article
(This article belongs to the Special Issue Pharmacotherapy of Thromboembolism)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Pathogenesis of VTE in pregnancy. Adapted from [<a href="#B3-pharmaceuticals-17-00773" class="html-bibr">3</a>].</p>
Full article ">Figure 2
<p>Pregnancy and its impact on the markers of hemostasis [<a href="#B8-pharmaceuticals-17-00773" class="html-bibr">8</a>]. Legend: F1+2; prothrombin fragment 1+2, FII; coagulation factor II, FV; coagulation factor V, FVII; coagulation factor VII, FVIII; coagulation factor VIII, FIX; coagulation factor IX, FX; coagulation factor X, FXI; coagulation factor XI, FXII; coagulation factor XII, FXIII; coagulation factor XIII, PAI-1; plasminogen activator inhibitor-1, PAI-2; plasminogen activator inhibitor 2, PC; protein C, PS; protein S, TAFI; thrombin-activatable fibrinolysis inhibitor, TAT; thrombin–antithrombin complex, tPA; tissue plasminogen activator, vWF; von Willebrand factor (adapted from [<a href="#B8-pharmaceuticals-17-00773" class="html-bibr">8</a>]).</p>
Full article ">Figure 3
<p>Outline of the history of advances in the management of thromboembolic complications [<a href="#B1-pharmaceuticals-17-00773" class="html-bibr">1</a>,<a href="#B14-pharmaceuticals-17-00773" class="html-bibr">14</a>].</p>
Full article ">
13 pages, 4307 KiB  
Article
HEMA-Lysine-Based Cryogels for Highly Selective Heparin Neutralization
by Tommaso Mecca, Fabiola Spitaleri, Rita La Spina, Sabrina Gioria, Valentina Giglio and Francesca Cunsolo
Int. J. Mol. Sci. 2024, 25(12), 6503; https://doi.org/10.3390/ijms25126503 - 13 Jun 2024
Viewed by 694
Abstract
Unfractionated heparin (UFH) and its low-molecular-weight fragments (LMWH) are widely used as anticoagulants for surgical procedures and extracorporeal blood purification therapies such as cardiovascular surgery and dialysis. The anticoagulant effect of heparin is essential for the optimal execution of extracorporeal blood circulation. However, [...] Read more.
Unfractionated heparin (UFH) and its low-molecular-weight fragments (LMWH) are widely used as anticoagulants for surgical procedures and extracorporeal blood purification therapies such as cardiovascular surgery and dialysis. The anticoagulant effect of heparin is essential for the optimal execution of extracorporeal blood circulation. However, at the end of these procedures, to avoid the risk of bleeding, it is necessary to neutralize it. Currently, the only antidote for heparin neutralization is protamine sulphate, a highly basic protein which constitutes a further source of serious side events and is ineffective in neutralizing LMWH. Furthermore, dialysis patients, due to the routine administration of heparin, often experience serious adverse effects, among which HIT (heparin-induced thrombocytopenia) is one of the most severe. For this reason, the finding of new heparin antagonists or alternative methods for heparin removal from blood is of great interest. Here, we describe the synthesis and characterization of a set of biocompatible macroporous cryogels based on poly(2-hydroxyethyl methacrylate) (pHEMA) and L-lysine with strong filtering capability and remarkable neutralization performance with regard to UFH and LMWH. These properties could enable the design and creation of a filtering device to rapidly reverse heparin, protecting patients from the harmful consequences of the anticoagulant. Full article
(This article belongs to the Special Issue Research on Synthesis and Application of Polymer Materials)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Overlayed IR-spectra of the samples; (<b>b</b>) overlayed thermograms of the samples.</p>
Full article ">Figure 2
<p>SEM images of the three samples recorded at 500× magnification.</p>
Full article ">Figure 3
<p>Histograms of swelling degree and porosity of the samples.</p>
Full article ">Figure 4
<p>Neutralization activity (mg Hep/g polymer) of each polymer in comparison with the previously described material (Old-pHEMA-lys). Each histogram represents the average of three independent experiments conducted with materials from three different cryopolymerization reactions. Adapted from a figure reported in the patent.</p>
Full article ">Figure 5
<p>Image of a monolithic sample in dry and wet forms. Sliced sample areas and relative neutralization activity with regard to UFH of Old-pHEMA-lys and pHEMA-lys50.</p>
Full article ">Scheme 1
<p>Synthesis of the HEMA-lys monomer.</p>
Full article ">Scheme 2
<p>General synthesis of pHEMA-lys.</p>
Full article ">
19 pages, 4281 KiB  
Article
NMR Studies of the Interactions between Sialyllactoses and the Polysialytransferase Domain for Polysialylation Inhibition
by Bo Lu, Si-Ming Liao, Shi-Jie Liang, Jian-Xiu Li, Xue-Hui Liu, Ri-Bo Huang and Guo-Ping Zhou
Curr. Issues Mol. Biol. 2024, 46(6), 5682-5700; https://doi.org/10.3390/cimb46060340 - 7 Jun 2024
Viewed by 1052
Abstract
It is known that sialyllactose (SL) in mammalians is a major source of sialic acid (Sia), which can further form cytidine monophosphate sialic acid (CMP-Sia), and the final product is polysialic acid (polySia) using polysialyltransferases (polySTs) on the neural cell adhesion molecule (NCAM). [...] Read more.
It is known that sialyllactose (SL) in mammalians is a major source of sialic acid (Sia), which can further form cytidine monophosphate sialic acid (CMP-Sia), and the final product is polysialic acid (polySia) using polysialyltransferases (polySTs) on the neural cell adhesion molecule (NCAM). This process is called NCAM polysialylation. The overexpression of polysialylation is strongly related to cancer cell migration, invasion, and metastasis. In order to inhibit the overexpression of polysialylation, in this study, SL was selected as an inhibitor to test whether polysialylation could be inhibited. Our results suggest that the interactions between the polysialyltransferase domain (PSTD) in polyST and CMP-Siaand the PSTD and polySia could be inhibited when the 3′-sialyllactose (3′-SL) or 6′-sialyllactose (6′-SL) concentration is about 0.5 mM or 6′-SL and 3 mM, respectively. The results also show that SLs (particularly for 3′-SL) are the ideal inhibitors compared with another two inhibitors, low-molecular-weight heparin (LMWH) and cytidine monophosphate (CMP), because 3’-SL can not only be used to inhibit NCAM polysialylation, but is also one of the best supplements for infant formula and the gut health system. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

Figure 1
<p>CD spectra of the PSTD alone (black), and in the presence of polySia/CMP-Sia (red), or in the presence of polySia/CMP-Sia/3′-SL (green) (<b>a</b>); CD spectra of the PSTD peptide in the absence of any ligand black and in the presence of polySia/CMP-Sia (red), or in the presence of polySia/CMP-Sia/6′-SL (green) (<b>b</b>).</p>
Full article ">Figure 2
<p>The overlaid <sup>1</sup>H-<sup>15</sup>N HSQC spectra of the PSTD in the absence and presence of 0.5 mM 3′-SL (<b>a</b>), and 1 mM 3′-SL (<b>b</b>), respectively. The obvious changes in chemical shift are residues located in the N-terminus and C-terminal, comparing (<b>a</b>) with (<b>b</b>).</p>
Full article ">Figure 3
<p>CSPs of the PSTD for the PSTD-(CMP-1 mM Sia), the PSTD-(0.5 mM 3′-SL), the PSTD-(1 mM 3′-SL), and the PSTD-(3 mM 3′-SL) interactions, respectively (<b>a</b>); the CSPs of the PSTD for the PSTD-(0.1 mM polySia or PSA), the PSTD-(0.5 mM 3′-SL) and the PSTD-(2 mM 3′-SL) interactions, respectively (<b>b</b>).</p>
Full article ">Figure 3 Cont.
<p>CSPs of the PSTD for the PSTD-(CMP-1 mM Sia), the PSTD-(0.5 mM 3′-SL), the PSTD-(1 mM 3′-SL), and the PSTD-(3 mM 3′-SL) interactions, respectively (<b>a</b>); the CSPs of the PSTD for the PSTD-(0.1 mM polySia or PSA), the PSTD-(0.5 mM 3′-SL) and the PSTD-(2 mM 3′-SL) interactions, respectively (<b>b</b>).</p>
Full article ">Figure 4
<p>The binding ranges of the three ligands, CMP-Sia, 3′-SL and 6′-SL on the PSTD based on the results shown in <a href="#cimb-46-00340-f002" class="html-fig">Figure 2</a>, <a href="#cimb-46-00340-f003" class="html-fig">Figure 3</a>, <a href="#cimb-46-00340-f005" class="html-fig">Figure 5</a> and <a href="#cimb-46-00340-f006" class="html-fig">Figure 6</a>, respectively.</p>
Full article ">Figure 5
<p>The overlaid <sup>1</sup>H-<sup>15</sup>N HSQC spectra of the PSTD in the absence and presence of 1.0 mM 6′-SL (<b>a</b>), and 2.0 mM 6′-SL (<b>b</b>), respectively. The obvious changes in chemical shift are residues K246, R252, T253 in the N-terminus, comparing (<b>a</b>) and (<b>b</b>).</p>
Full article ">Figure 6
<p>The chemical shift perturbations (CSPs) of the PSTD in the presence of 0.5, 1.0, 2.0, and 3.0 mM 6′-SL (<b>a</b>); the CSPs of the PSTD when interacting with 1 mM CMP-Sia, and 1.0 mM 6-SL, respectively (<b>b</b>); and the CSPs of the PSTD when interacting with 0.1 mM PSA, 2.0 mM and 3 mM 6′-SL, respectively (<b>c</b>).</p>
Full article ">Figure 7
<p>Comparison of the CSPs in the PSTD when PSTD interacted with different ligands. The CSPs of the PSTD for the PSTD-0.5 mM 3′-SL, the PSTD-1 mM 6′-SL, the PSTD-1 mM (CMP-Sia), and the PSTD-80 μM LMWH interactions (<b>a</b>); CSPs of the PSTD for the PSTD-0.5 mM 3′-SL, the PSTD-3 mM 6′-SL, the PSTD-80 μM LMWH, and the PSTD-0.1 mM polySia interactions (<b>b</b>).</p>
Full article ">Figure 7 Cont.
<p>Comparison of the CSPs in the PSTD when PSTD interacted with different ligands. The CSPs of the PSTD for the PSTD-0.5 mM 3′-SL, the PSTD-1 mM 6′-SL, the PSTD-1 mM (CMP-Sia), and the PSTD-80 μM LMWH interactions (<b>a</b>); CSPs of the PSTD for the PSTD-0.5 mM 3′-SL, the PSTD-3 mM 6′-SL, the PSTD-80 μM LMWH, and the PSTD-0.1 mM polySia interactions (<b>b</b>).</p>
Full article ">Figure 8
<p>Comparison of the inhibitions of the SL CMP for the interaction between the PSTD and CMP-Sia, and the interaction between the PSTD and polySia. The CSPs of the PSTD for the PSTD-(0.5 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(1 mM CMP-Sia) interactions (<b>a</b>); CSPs of the PSTD for the PSTD-(0.5 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(0.1 mM polySia) interactions (<b>b</b>); CSPs of the PSTD for the PSTD-(1 mM 6′-SL), the PSTD-1 mM CMP, and the PSTD-(1 mM CMP-Sia) interactions (<b>c</b>); CSPs of the PSTD for the PSTD-(3 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(0.1 mM polySia) interactions (<b>d</b>).</p>
Full article ">Figure 8 Cont.
<p>Comparison of the inhibitions of the SL CMP for the interaction between the PSTD and CMP-Sia, and the interaction between the PSTD and polySia. The CSPs of the PSTD for the PSTD-(0.5 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(1 mM CMP-Sia) interactions (<b>a</b>); CSPs of the PSTD for the PSTD-(0.5 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(0.1 mM polySia) interactions (<b>b</b>); CSPs of the PSTD for the PSTD-(1 mM 6′-SL), the PSTD-1 mM CMP, and the PSTD-(1 mM CMP-Sia) interactions (<b>c</b>); CSPs of the PSTD for the PSTD-(3 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(0.1 mM polySia) interactions (<b>d</b>).</p>
Full article ">
17 pages, 1535 KiB  
Review
Pancreatic Cancer and Venous Thromboembolism
by Teagan Prouse, Mohammad A. Mohammad, Sonali Ghosh, Narender Kumar, Ma. Lorena Duhaylungsod, Rinku Majumder and Samarpan Majumder
Int. J. Mol. Sci. 2024, 25(11), 5661; https://doi.org/10.3390/ijms25115661 - 23 May 2024
Cited by 2 | Viewed by 1413
Abstract
Pancreatic ductal adenocarcinoma (PDAC) accounts for more than 90% of all pancreatic cancers and is the most fatal of all cancers. The treatment response from combination chemotherapies is far from satisfactory and surgery remains the mainstay of curative strategies. These challenges warrant identifying [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) accounts for more than 90% of all pancreatic cancers and is the most fatal of all cancers. The treatment response from combination chemotherapies is far from satisfactory and surgery remains the mainstay of curative strategies. These challenges warrant identifying effective treatments for combating this deadly cancer. PDAC tumor progression is associated with the robust activation of the coagulation system. Notably, cancer-associated thrombosis (CAT) is a significant risk factor in PDAC. CAT is a concept whereby cancer cells promote thromboembolism, primarily venous thromboembolism (VTE). Of all cancer types, PDAC is associated with the highest risk of developing VTE. Hypoxia in a PDAC tumor microenvironment also elevates thrombotic risk. Direct oral anticoagulants (DOACs) or low-molecular-weight heparin (LMWH) are used only as thromboprophylaxis in PDAC. However, a precision medicine approach is recommended to determine the precise dose and duration of thromboprophylaxis in clinical setting. Full article
(This article belongs to the Special Issue Molecular Insights into Platelet Biology and Function)
Show Figures

Figure 1

Figure 1
<p>Factors that increase VTE risk in PDAC patients (created using <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">Figure 2
<p>Summary of PDAC and VTE (created using <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">
11 pages, 2712 KiB  
Article
Evaluating Anticoagulant and Antiplatelet Therapies in Rhesus and Cynomolgus Macaques for Predictive Modeling in Humans
by Sydney N. Phu, David J. Leishman, Sierra D. Palmer, Scott H. Oppler, Melanie N. Niewinski, Lucas A. Mutch, Jill S. Faustich, Andrew B. Adams, Robert T. Tranquillo and Melanie L. Graham
Surgeries 2024, 5(2), 423-433; https://doi.org/10.3390/surgeries5020035 - 17 May 2024
Viewed by 679
Abstract
Anticoagulant and antiplatelet therapies are used to prevent life-threatening complications associated with thrombosis. While there are numerous clinical guidelines for antithrombotic medications, there is an incomplete understanding of whether these interventions yield similar effects in preclinical models, potentially impacting their predictive value for [...] Read more.
Anticoagulant and antiplatelet therapies are used to prevent life-threatening complications associated with thrombosis. While there are numerous clinical guidelines for antithrombotic medications, there is an incomplete understanding of whether these interventions yield similar effects in preclinical models, potentially impacting their predictive value for translational studies on the development of medical devices, therapies, and surgical techniques. Due to their close physiologic similarities to humans, we employed nonhuman primates (NHPs) using a reverse translational approach to analyze the response to clinical regimens of unfractionated heparin, low-molecular-weight heparin (LMWH) and aspirin to assess concordance with typical human responses and evaluate the predictive validity of this model. We evaluate activated clotting time (ACT) in nine rhesus and six cynomolgus macaques following the intraoperative administration of intravenous unfractionated heparin (100–300 U/kg) reflecting the clinical dose range. We observed a significant dose-dependent effect of heparin on ACT (low-dose average = 114.1 s; high-dose average = 148.3 s; p = 0.0011). LMWH and aspirin, common clinical antithrombotic prophylactics, were evaluated in three rhesus macaques. NHPs achieved therapeutic Anti-Xa levels (mean = 0.64 U/mL) and ARU (mean = 459) via VerifyNow, adhering to clinical guidance using 1.0 mg/kg enoxaparin and 81 mg aspirin. Clinical dosing strategies for unfractionated heparin, LMWH, and aspirin were safe and effective in NHPs, with no development of thrombosis or bleeding complications intraoperatively, postoperatively, or for prophylaxis. Our findings suggest that coagulation studies, performed as an integrative part of studies on biologics, bioengineered devices, or transplantation in NHPs, can be extrapolated to the clinical situation with high predictive validity. Full article
Show Figures

Figure 1

Figure 1
<p>Overview of different intraoperative heparin dosing strategy groups. NHPs were divided into low-dose (100 U/kg), high-dose (300 U/kg), and re-dose (100 U/kg followed by 50 U/kg) groups. In preparation for anastomosis creation and prior to vascular clamping, intravenous unfractionated heparin was administered, and ACT was measured 60–150 min afterward. In the re-dose group, an additional 50 U/kg was administered 30 min after initial heparin injection.</p>
Full article ">Figure 2
<p>Overview of LMWH and aspirin administration and dose adjustment. (<b>A</b>) Administration and sampling timeline for the dose optimization of LMWH and aspirin. (<b>B</b>) Protocol for adjusting LMWH dosage based on measured Anti-Xa level. (<b>C</b>) Protocol for adjusting aspirin dosage based on measured ARU.</p>
Full article ">Figure 3
<p>Mean activated clotting time (ACT) following intraoperative unfractionated heparin administration in rhesus and cynomolgus macaques. (<b>A</b>) Rhesus (<span class="html-italic">n</span> = 4) and cynomolgus (<span class="html-italic">n</span> = 5) ACT response to low-dose unfractionated heparin. Rhesus macaques had a mean ACT of 117.5 s, and cynomolgus macaques had a mean ACT of 111.4 s. There was no significant difference in ACT between species in the low-dose group (<span class="html-italic">p</span> = 0.6111). (<b>B</b>) ACT response by dosing group for both rhesus and cynomolgus macaques combined. The low-dose group had a mean ACT of 114.1 s. The high-dose group had a mean ACT of 148.3 s. The re-dose group had a mean ACT of 132.7 s. There was a significant difference between low-dose (<span class="html-italic">n</span> = 9) and high-dose (<span class="html-italic">n</span> = 3) groups (<span class="html-italic">p</span> = 0.0190), but no significant difference between low-dose and re-dose (<span class="html-italic">n</span> = 3) groups (<span class="html-italic">p</span> = 0.2881) or between high-dose and re-dose groups (<span class="html-italic">p</span> ≥ 0.9999). Data are presented as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05, ns = not significant.</p>
Full article ">Figure 4
<p>Anti-Xa levels following the subcutaneous administration of low-molecular-weight heparin (LMWH) in rhesus macaques (<span class="html-italic">n</span> = 3). (<b>A</b>) Mean anti-Xa levels with standard deviation at baseline and post low-molecular-weight heparin and aspirin treatment. Animals had a mean Anti-Xa level of 0.29 IU/mL at baseline and mean Anti-Xa level of 0.82 IU/mL with treatment. (<b>B</b>) Individual Anti-Xa levels across treatment days with target levels of 0.5–1.0 IU/mL, as shown by the dotted lines. There was a significant increase in Anti-Xa levels with treatment (<span class="html-italic">p</span> = 0.0238). Data are presented as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05, ns = not significant</p>
Full article ">Figure 5
<p>Aspirin reaction units (ARU) following oral aspirin administration in rhesus macaques. (<b>A</b>) Mean ARU with standard deviation at baseline and after low-molecular-weight heparin and aspirin treatment. Animals had a mean of 636 ARU at baseline and a mean of 459 ARU with treatment. (<b>B</b>) Individual ARU values over the course of treatment with target ARU levels of &lt;550, as shown by the dotted line. There was a significant reduction in ARU levels with treatment (<span class="html-italic">p</span> = 0.0091). Data are presented as mean ± SD. ** <span class="html-italic">p</span> &lt; 0.01, ns = not significant.</p>
Full article ">
16 pages, 2842 KiB  
Article
The Bifunctional Effects of Lactoferrin (LFcinB11) in Inhibiting Neural Cell Adhesive Molecule (NCAM) Polysialylation and the Release of Neutrophil Extracellular Traps (NETs)
by Bo Lu, Si-Ming Liao, Shi-Jie Liang, Li-Xin Peng, Jian-Xiu Li, Xue-Hui Liu, Ri-Bo Huang and Guo-Ping Zhou
Int. J. Mol. Sci. 2024, 25(9), 4641; https://doi.org/10.3390/ijms25094641 - 24 Apr 2024
Cited by 1 | Viewed by 1139
Abstract
The expression of polysialic acid (polySia) on the neuronal cell adhesion molecule (NCAM) is called NCAM-polysialylation, which is strongly related to the migration and invasion of tumor cells and aggressive clinical status. Thus, it is important to select a proper drug to block [...] Read more.
The expression of polysialic acid (polySia) on the neuronal cell adhesion molecule (NCAM) is called NCAM-polysialylation, which is strongly related to the migration and invasion of tumor cells and aggressive clinical status. Thus, it is important to select a proper drug to block tumor cell migration during clinical treatment. In this study, we proposed that lactoferrin (LFcinB11) may be a better candidate for inhibiting NCAM polysialylation when compared with CMP and low-molecular-weight heparin (LMWH), which were determined based on our NMR studies. Furthermore, neutrophil extracellular traps (NETs) represent the most dramatic stage in the cell death process, and the release of NETs is related to the pathogenesis of autoimmune and inflammatory disorders, with proposed involvement in glomerulonephritis, chronic lung disease, sepsis, and vascular disorders. In this study, the molecular mechanisms involved in the inhibition of NET release using LFcinB11 as an inhibitor were also determined. Based on these results, LFcinB11 is proposed as being a bifunctional inhibitor for inhibiting both NCAM polysialylation and the release of NETs. Full article
(This article belongs to the Special Issue Mechanisms of Small Molecule Inhibitors Targeting Cancer)
Show Figures

Figure 1

Figure 1
<p>The figure shows the CD spectra of the PSTD in the absence (red) and presence of the mixture of CMP-Sia and polySia (green) and the mixture of CMP-Sia, polySia, and LFcinB11 (blue). The helical contents of these three CD spectra of the PSTD are 23%, 16.4%, and 14.8%, respectively.</p>
Full article ">Figure 2
<p>The figure shows the overlaid 1H-15N HSQC spectra of the 2 mM PSTD in the absence and presence of 20 μM LFcinB11 (<b>A</b>), 40 μM LFcinB11 (<b>B</b>), 60 μM LFcinB11 (<b>C</b>) and 80 μM LFcinB11 (<b>D</b>), respectively.</p>
Full article ">Figure 3
<p>The figure shows the chemical shift perturbations (CSPs) of the PSTD when the PSTD interacted with 20 μM LFcinB11 (blue), 40 μM LFcinB11 (orange), 60 μM LFcinB11 (gray), and 1 mM CMP--Sia (red), respectively (<b>A</b>); chemical shift perturbations (CSPs) of the PSTD when the PSTD interacted with 20 μM LFcinB11(blue), 40 μM LFcinB11 (black), 60 μM LFcinB11 (gray), 80 μM LFcinB11 (orange), and 0.1 mM polySia (red), respectively (<b>B</b>).</p>
Full article ">Figure 4
<p>The figure shows the overlaid 1H-15N HSQC spectra of 50 μM LFcin11 in the absence (black) and presence of 50 μM polySia (red), respectively.</p>
Full article ">Figure 5
<p>The figure shows the chemical shift perturbations (CSPs) of the PSTD when interacting with 1 mM CMP-Sia, 80 μM CMP, 40 μM, and 60 μM LFcinB11, respectively (<b>A</b>), and the CSPs of the PSTD when interacting with 0.1 mM PSA, 1 mM CMP, 40 μM, and 60 μM LFcinB11, respectively (<b>B</b>).</p>
Full article ">Figure 6
<p>The figure shows the chemical shift perturbations (CSPs) of the PSTD when interacting with 1 mM CMP-Sia, 80 μM heparin LMWH, 40 μM, and 60 μM LFcinB11, respectively (<b>A</b>), and the CSPs of the PSTD when interacting with 0.1 mM PSA, 80 μM heparin LWH, and 40 μM and 60 μM LFcinB11, respectively (<b>B</b>).</p>
Full article ">
10 pages, 379 KiB  
Review
Primary Thromboprophylaxis for the Prevention of Venous Thromboembolism in Cancer Patients with Central Venous Catheters: A Literature Review
by Hikmat Abdel-Razeq and Mohammed J. Al-Jaghbeer
J. Clin. Med. 2024, 13(6), 1660; https://doi.org/10.3390/jcm13061660 - 14 Mar 2024
Cited by 1 | Viewed by 1454
Abstract
Cancer is a known risk factor for venous thromboembolism (VTE). The wider adoption of immunotherapy and anti-angiogenic drugs in recent years have increased this risk further. Central venous catheters (CVCs) are widely used access devices utilized to deliver infusion therapy, mostly in ambulatory [...] Read more.
Cancer is a known risk factor for venous thromboembolism (VTE). The wider adoption of immunotherapy and anti-angiogenic drugs in recent years have increased this risk further. Central venous catheters (CVCs) are widely used access devices utilized to deliver infusion therapy, mostly in ambulatory settings. The endothelial injury associated with the use of these catheters adds to the risk of VTE to already high-risk patients. The introduction of direct oral anticoagulants (DOACs), with its proven efficacy and safety in multiple clinical indications, have renewed the attention to VTE prophylaxis in cancer patients with CVC. Several clinical trials and meta-analyses had shown that both apixaban and rivaroxaban are effective in lowering the risk of VTE, without increasing the risk of bleeding. Several risk assessment models (RAM) have utilized patient-related, tumor-related, and treatment-related factors, in addition to widely available biomarkers, like Hemoglobin (Hb) level, white blood cell (WBC) and platelets counts to stratify patients into two or three VTE risk levels. In this manuscript, we review the published clinical trials and meta-analyses that attempted to study the efficacy and safety of anticoagulants, mostly the DOACs, in cancer patients with CVCs. We will also propose a practical risk-directed approach to enhance VTE prophylaxis rate. Full article
(This article belongs to the Special Issue Thromboembolic Disease and Antithrombotic Therapy)
Show Figures

Figure 1

Figure 1
<p>Efficacy and safety endpoints: Thromboprophylaxis versus placebo or observation. VTE: Venous thromboembolism; CRT: Catheter-related thrombosis; CRNMB: Clinically-relevant no major bleeding.</p>
Full article ">
14 pages, 2963 KiB  
Article
Thrombosis and Bleeding Risk Scores Are Strongly Associated with Mortality in Hospitalized Patients with COVID-19: A Multicenter Cohort Study
by Kunapa Iam-Arunthai, Supat Chamnanchanunt, Pravinwan Thungthong, Poj Intalapaporn, Chajchawan Nakhahes, Tawatchai Suwanban and Ponlapat Rojnuckarin
J. Clin. Med. 2024, 13(5), 1437; https://doi.org/10.3390/jcm13051437 - 1 Mar 2024
Cited by 1 | Viewed by 1077
Abstract
Background: Internationally established guidelines mention pharmacological prophylaxis for all hospitalized COVID-19 patients. However, there are concerns regarding the efficacy and safety of anticoagulants. This study investigated the associations between thrombosis/bleeding risk scores and clinical outcomes. Methods: We conducted a retrospective review of adult [...] Read more.
Background: Internationally established guidelines mention pharmacological prophylaxis for all hospitalized COVID-19 patients. However, there are concerns regarding the efficacy and safety of anticoagulants. This study investigated the associations between thrombosis/bleeding risk scores and clinical outcomes. Methods: We conducted a retrospective review of adult patients admitted to two hospitals between 2021 and 2022. We analyzed clinical data, laboratory results, low molecular weight heparin (LMWH) use, thrombosis, bleeding, and 30-day survival. Results: Of the 160 patients, 69.4% were female, and the median age was 59 years. The rates of thrombotic complications and mortality were 12.5% and 36.3%, respectively. LMWH prophylaxis was administered to 73 of the patients (45.6%). The patients with high Padua prediction scores (PPS) and high IMPROVEVTE scores had a significantly higher risk of venous thromboembolism (VTE) compared to those with low scores (30.8% vs. 9.0%, p = 0.006 and 25.6% vs. 7.7%, p = 0.006). Similarly, elevated IMPROVEVTE and IMPROVEBRS scores were associated with increased mortality (hazard ratios of 7.49 and 6.27, respectively; p < 0.001). Interestingly, LMWH use was not associated with a decreased incidence of VTE when stratified by risk groups. Conclusions: this study suggests that COVID-19 patients with high thrombosis and bleeding risk scores have a higher mortality rate. Full article
Show Figures

Figure 1

Figure 1
<p>Receiver operating characteristic (ROC) curves of the venous thromboembolism (PPS<sub>VTE</sub>, IMPROVE<sub>VTE)</sub> and bleeding (IMPROVE<sub>BRS)</sub> risk score models for mortality prediction. The areas under the curves (AUC) were 0.68 (PPS<sub>VTE</sub>), 0.79 (IMPROVE<sub>VTE</sub>), 0.67 (IMPROVE<sub>BRS</sub>), and 0.82 (IMPROVEDD<sub>VTE</sub>).</p>
Full article ">Figure 2
<p>Kaplan–Meier curve for cumulative survival according to difference venous thromboembolism and bleeding risk scores among 160 patients. (<b>A</b>) Comparison of cumulative survival rates between a Padual prediction score of VTE of &lt;4 and ≥4 subgroup; <span class="html-italic">p</span>-value &lt; 0.001. (<b>B</b>) Comparison of cumulative survival rates between an IMPROVE prediction score of VTE of &lt;2 and ≥2 subgroup; <span class="html-italic">p</span>-value &lt; 0.001. (<b>C</b>) Comparison of cumulative survival rates between an IMPROVEDD prediction score of VTE of &lt;2 and ≥2 subgroup; <span class="html-italic">p</span>-value &lt; 0.001. (<b>D</b>) Comparison of cumulative survival rates between an IMPROVE prediction score of bleeding of &lt;7 and ≥7 subgroup; <span class="html-italic">p</span>-value &lt; 0.001. (<b>E</b>) Comparison of cumulative survival rates between a combination of PPS<sub>VTE</sub> (≥4) + IMPROVE<sub>BRS</sub> (≥7) (high risk) and a combination of PPS<sub>VTE</sub> (&lt;4) or IMPROVE<sub>BRS</sub> (&lt;7) (low risk) prediction score subgroups; <span class="html-italic">p</span>-value &lt; 0.001. (<b>F</b>) Comparison of cumulative survival rates between a combination of IMPROVE<sub>VTE</sub> (≥2) + IMPROVE<sub>BRS</sub> (≥7) (high risk) and a combination of IMPROVE<sub>VTE</sub> (&lt;2) or IMPROVE<sub>BRS</sub> (&lt;7) (low risk) prediction score subgroups; <span class="html-italic">p</span>-value &lt; 0.001. (<b>G</b>) Comparison of cumulative survival rates between a combination of IMPROVEDD<sub>VTE</sub> (≥2) + IMPROVE<sub>BRS</sub> (≥7) (high risk) and a combination of IMPROVEDD<sub>VTE</sub> (&lt;2) or IMPROVE<sub>BRS</sub> (&lt;7) (low risk) prediction score subgroups; <span class="html-italic">p</span>-value &lt; 0.001.</p>
Full article ">Figure 2 Cont.
<p>Kaplan–Meier curve for cumulative survival according to difference venous thromboembolism and bleeding risk scores among 160 patients. (<b>A</b>) Comparison of cumulative survival rates between a Padual prediction score of VTE of &lt;4 and ≥4 subgroup; <span class="html-italic">p</span>-value &lt; 0.001. (<b>B</b>) Comparison of cumulative survival rates between an IMPROVE prediction score of VTE of &lt;2 and ≥2 subgroup; <span class="html-italic">p</span>-value &lt; 0.001. (<b>C</b>) Comparison of cumulative survival rates between an IMPROVEDD prediction score of VTE of &lt;2 and ≥2 subgroup; <span class="html-italic">p</span>-value &lt; 0.001. (<b>D</b>) Comparison of cumulative survival rates between an IMPROVE prediction score of bleeding of &lt;7 and ≥7 subgroup; <span class="html-italic">p</span>-value &lt; 0.001. (<b>E</b>) Comparison of cumulative survival rates between a combination of PPS<sub>VTE</sub> (≥4) + IMPROVE<sub>BRS</sub> (≥7) (high risk) and a combination of PPS<sub>VTE</sub> (&lt;4) or IMPROVE<sub>BRS</sub> (&lt;7) (low risk) prediction score subgroups; <span class="html-italic">p</span>-value &lt; 0.001. (<b>F</b>) Comparison of cumulative survival rates between a combination of IMPROVE<sub>VTE</sub> (≥2) + IMPROVE<sub>BRS</sub> (≥7) (high risk) and a combination of IMPROVE<sub>VTE</sub> (&lt;2) or IMPROVE<sub>BRS</sub> (&lt;7) (low risk) prediction score subgroups; <span class="html-italic">p</span>-value &lt; 0.001. (<b>G</b>) Comparison of cumulative survival rates between a combination of IMPROVEDD<sub>VTE</sub> (≥2) + IMPROVE<sub>BRS</sub> (≥7) (high risk) and a combination of IMPROVEDD<sub>VTE</sub> (&lt;2) or IMPROVE<sub>BRS</sub> (&lt;7) (low risk) prediction score subgroups; <span class="html-italic">p</span>-value &lt; 0.001.</p>
Full article ">Figure 3
<p>Kaplan–Meier curve for cumulative survival rates between patients with and without low molecular weight heparin, according to each low venous thromboembolism and bleeding risk score. (<b>A</b>) Comparison of cumulative survival rates among patients with Padual prediction scores of VTE ≥ 4 subgroup; <span class="html-italic">p</span>-value = 0.054. (<b>B</b>) Comparison of cumulative survival rates among patients with IMPROVE prediction scores of VTE ≥ 2; <span class="html-italic">p</span>-value = 0.035. (<b>C</b>) Comparison of cumulative survival rates among patients with IMPROVEDD prediction scores of VTE ≥ 2; <span class="html-italic">p</span>-value = 0.123. (<b>D</b>) Comparison of cumulative survival rates among patients with IMPROVE prediction scores of bleeding ≥ 7; <span class="html-italic">p</span>-value = 0.002. (<b>E</b>) Comparison of cumulative survival rates among patients with PPS<sub>VTE</sub> (≥4) + IMPROVE<sub>BRS</sub> (≥7) (high risk) prediction score subgroup; <span class="html-italic">p</span>-value = 0.022. (<b>F</b>) Comparison of cumulative survival rates among patients with IMPROVE<sub>VTE</sub> (≥2) + IMPROVE<sub>BRS</sub> (≥7) (high risk) prediction score subgroup; <span class="html-italic">p</span>-value &lt; 0.003. (<b>G</b>) Comparison of cumulative survival rates among patients with IMPROVEDD<sub>VTE</sub> (≥2) + IMPROVE<sub>BRS</sub> (≥7) (high risk) prediction score subgroup; <span class="html-italic">p</span>-value &lt; 0.003.</p>
Full article ">Figure 3 Cont.
<p>Kaplan–Meier curve for cumulative survival rates between patients with and without low molecular weight heparin, according to each low venous thromboembolism and bleeding risk score. (<b>A</b>) Comparison of cumulative survival rates among patients with Padual prediction scores of VTE ≥ 4 subgroup; <span class="html-italic">p</span>-value = 0.054. (<b>B</b>) Comparison of cumulative survival rates among patients with IMPROVE prediction scores of VTE ≥ 2; <span class="html-italic">p</span>-value = 0.035. (<b>C</b>) Comparison of cumulative survival rates among patients with IMPROVEDD prediction scores of VTE ≥ 2; <span class="html-italic">p</span>-value = 0.123. (<b>D</b>) Comparison of cumulative survival rates among patients with IMPROVE prediction scores of bleeding ≥ 7; <span class="html-italic">p</span>-value = 0.002. (<b>E</b>) Comparison of cumulative survival rates among patients with PPS<sub>VTE</sub> (≥4) + IMPROVE<sub>BRS</sub> (≥7) (high risk) prediction score subgroup; <span class="html-italic">p</span>-value = 0.022. (<b>F</b>) Comparison of cumulative survival rates among patients with IMPROVE<sub>VTE</sub> (≥2) + IMPROVE<sub>BRS</sub> (≥7) (high risk) prediction score subgroup; <span class="html-italic">p</span>-value &lt; 0.003. (<b>G</b>) Comparison of cumulative survival rates among patients with IMPROVEDD<sub>VTE</sub> (≥2) + IMPROVE<sub>BRS</sub> (≥7) (high risk) prediction score subgroup; <span class="html-italic">p</span>-value &lt; 0.003.</p>
Full article ">
11 pages, 247 KiB  
Article
Comparative Analysis of Antithrombotic Therapy Outcomes in Mild Traumatic Brain-Injury Patients: A Focus on Bleeding Risk and Hospital-Stay Duration
by Antonio Desai, Dana Shiffer, Mauro Giordano, Alice Giotta Lucifero, Elena Generali, Francesco Reggiani, Marta Calatroni, Gabriele Savioli, Sabino Luzzi and Antonio Voza
Life 2024, 14(3), 308; https://doi.org/10.3390/life14030308 - 27 Feb 2024
Viewed by 959
Abstract
Background: Traumatic brain injury (TBI) in the elderly is a noteworthy pathology due to the exponential increase in population age, and the effects of antiplatelet and anticoagulation on patients’ outcomes are still a matter of dispute. The aim of the present study was [...] Read more.
Background: Traumatic brain injury (TBI) in the elderly is a noteworthy pathology due to the exponential increase in population age, and the effects of antiplatelet and anticoagulation on patients’ outcomes are still a matter of dispute. The aim of the present study was to evaluate the impact of various antithrombotic agents on patients with mild TBI, focusing on the risk of intracranial bleeding (ICH) and length of hospitalization (LOS). Methods: A retrospective analysis was conducted, including patients with a diagnosis of TBI admitted to the Emergency Department between 2021 and 2022. Patients were classified according to the concurrent antithrombotic therapy as aspirin (ASA), antiplatelets, direct oral anticoagulants (DOACs), and low-molecular-weight heparin (LMWH). The primary outcome was the ICH occurrence, while the secondary outcome was the LOS. The statistical analysis was performed via logistic regression models in R and STATA 13.1 software. Fisher’s exact test was used for the statistical significance. Results: 267 patients with mild TBI were included; 148 were not on antithrombotic agents, 43 were on aspirin, 33 on DOACs, 5 on LMWH, 22 on antiplatelets, and 16 on VKA. Out of the total, 9 patients experienced ICH, none of which were on DOACs, LMWH, or VKA, but 4—out of 65—were on antiplatelets, and 5—out of 148—were not on antithrombotic therapies. Patients not on antithrombotic therapy had the shortest LOS at 0.46 days, while those on VKA had the longest LOS at 1.19 days; similar trends were observed for patients on DOAC and LMWH. Conclusions: The results reveal that TBI patients on anticoagulants/antiplatelets had longer hospital stays compared with those on aspirin alone. Notably, VKA was the strongest predictor for an extended LOS. Regarding ICH, patients taking only aspirin were twice as likely to experience bleeding compared with those on anticoagulants/antiplatelets. However, to achieve statistically significant evidence, further research with a larger cohort of patients is needed. Full article
(This article belongs to the Special Issue Trauma and Emergency: Beyond Damage Control Surgery: 2nd Edition)
22 pages, 841 KiB  
Review
Lung Cancer Related Thrombosis (LCART): Focus on Immune Checkpoint Blockade
by Andriani Charpidou, Grigorios Gerotziafas, Sanjay Popat, Antonio Araujo, Arnaud Scherpereel, Hans-Georg Kopp, Paolo Bironzo, Gilbert Massard, David Jiménez, Anna Falanga, Anastasios Kollias and Konstantinos Syrigos
Cancers 2024, 16(2), 450; https://doi.org/10.3390/cancers16020450 - 20 Jan 2024
Cited by 2 | Viewed by 2994
Abstract
Cancer-associated thrombosis (CAT) is a common complication in lung cancer patients. Lung cancer confers an increased risk of thrombosis compared to other solid malignancies across all stages of the disease. Newer treatment agents, including checkpoint immunotherapy and targeted agents, may further increase the [...] Read more.
Cancer-associated thrombosis (CAT) is a common complication in lung cancer patients. Lung cancer confers an increased risk of thrombosis compared to other solid malignancies across all stages of the disease. Newer treatment agents, including checkpoint immunotherapy and targeted agents, may further increase the risk of CAT. Different risk-assessment models, such as the Khorana Risk Score, and newer approaches that incorporate genetic risk factors have been used in lung cancer patients to evaluate the risk of thrombosis. The management of CAT is based on the results of large prospective trials, which show similar benefits to low-molecular-weight heparins (LMWHs) and direct oral anticoagulants (DOACs) in ambulatory patients. The anticoagulation agent and duration of therapy should be personalized according to lung cancer stage and histology, the presence of driver mutations and use of antineoplastic therapy, including recent curative lung surgery, chemotherapy or immunotherapy. Treatment options should be evaluated in the context of the COVID-19 pandemic, which has been shown to impact the thrombotic risk in cancer patients. This review focuses on the epidemiology, pathophysiology, risk factors, novel predictive scores and management of CAT in patients with active lung cancer, with a focus on immune checkpoint inhibitors. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Risk factors and molecular mechanisms for cancer-associated thrombosis (CAT) in lung cancer (LC) patients.</p>
Full article ">
Back to TopTop