Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.969
Filter
1.
Clin Lab ; 70(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38747921

ABSTRACT

BACKGROUND: Pseudothrombocytopenia (PTCP) can be caused by anticoagulants or pre-analytical issues. The authors present a case of PTCP attributed to pre-analytical issues in a 68-year-old male patient. METHODS: The platelet count results were obtained using both the impedance and fluorescence channels of Sysmex XN-10. The blood film was scanned using both Cellavision DM96 and a microscope. RESULTS: The flag for PLT-Clumps and the scattergram from the PLT-F channel indicated the presence of platelet aggregation. Fibrin could be observed at the feathered end of the blood film. A diagnosis of PTCP resulting from pre-analytical issues was made. CONCLUSIONS: The presence of fibrin in a blood film is a critical indicator for diagnosing PTCP due to pre-analytical issues.


Subject(s)
Fibrin , Thrombocytopenia , Humans , Male , Aged , Thrombocytopenia/blood , Thrombocytopenia/diagnosis , Fibrin/metabolism , Fibrin/analysis , Platelet Count/methods , Anticoagulants , Platelet Aggregation , Blood Platelets
2.
Cell Biochem Funct ; 42(4): e4038, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38736214

ABSTRACT

The generation of insulin-producing cells (IPCs) is an attractive approach for replacing damaged ß cells in diabetic patients. In the present work, we introduced a hybrid platform of decellularized amniotic membrane (dAM) and fibrin encapsulation for differentiating adipose tissue-derived stem cells (ASCs) into IPCs. ASCs were isolated from healthy donors and characterized. Human AM was decellularized, and its morphology, DNA, collagen, glycosaminoglycan (GAG) contents, and biocompatibility were evaluated. ASCs were subjected to four IPC differentiation methods, and the most efficient method was selected for the experiment. ASCs were seeded onto dAM, alone or encapsulated in fibrin gel with various thrombin concentrations, and differentiated into IPCs according to a method applying serum-free media containing 2-mercaptoethanol, nicotinamide, and exendin-4. PDX-1, GLUT-2 and insulin expression were evaluated in differentiated cells using real-time PCR. Structural integrity and collagen and GAG contents of AM were preserved after decellularization, while DNA content was minimized. Cultivating ASCs on dAM augmented their attachment, proliferation, and viability and enhanced the expression of PDX-1, GLUT-2, and insulin in differentiated cells. Encapsulating ASCs in fibrin gel containing 2 mg/ml fibrinogen and 10 units/ml thrombin increased their differentiation into IPCs. dAM and fibrin gel synergistically enhanced the differentiation of ASCs into IPCs, which could be considered an appropriate strategy for replacing damaged ß cells.


Subject(s)
Adipose Tissue , Cell Differentiation , Fibrin , Insulin , Stem Cells , Humans , Cell Differentiation/drug effects , Fibrin/chemistry , Fibrin/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Stem Cells/metabolism , Stem Cells/cytology , Insulin/metabolism , Cells, Cultured , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/cytology , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/metabolism , Decellularized Extracellular Matrix/pharmacology , Amnion/cytology , Amnion/metabolism , Amnion/chemistry
3.
Sci Rep ; 14(1): 9225, 2024 04 22.
Article in English | MEDLINE | ID: mdl-38649717

ABSTRACT

Thrombin generation (TG) and fibrin clot formation represent the central process of blood coagulation. Up to 95% of thrombin is considered to be generated after the clot is formed. However, this was not investigated in depth. In this study, we conducted a quantitative analysis of the Thrombin at Clot Time (TCT) parameter in 5758 simultaneously recorded TG and clot formation assays using frozen plasma samples from commercial sources under various conditions of activation. These samples were supplemented with clotting factor concentrates, procoagulant lipid vesicles and a fluorogenic substrate and triggered with tissue factor (TF). We found that TCT is often close to a 10% of thrombin peak height (TPH) yet it can be larger or smaller depending on whether the sample has low or high TPH value. In general, the samples with high TPH are associated with elevated TCT. TCT appeared more sensitive to some procoagulant phenotypes than other commonly used parameters such as clotting time, TPH or Thrombin Production Rate (TPR). In a minority of cases, TCT were not predicted from TG parameters. For example, elevated TCT (above 15% of TPH) was associated with either very low or very high TPR values. We conclude that clotting and TG assays may provide complementary information about the plasma sample, and that the TCT parameter may serve as an additional marker for the procoagulant potential in plasma sample.


Subject(s)
Blood Coagulation , Fibrin , Thrombin , Thrombin/metabolism , Humans , Fibrin/metabolism , Blood Coagulation Tests/methods , Thromboplastin/metabolism , Thromboplastin/analysis
4.
J Neuroinflammation ; 21(1): 94, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622640

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) causes significant blood-brain barrier (BBB) breakdown, resulting in the extravasation of blood proteins into the brain. The impact of blood proteins, especially fibrinogen, on inflammation and neurodegeneration post-TBI is not fully understood, highlighting a critical gap in our comprehension of TBI pathology and its connection to innate immune activation. METHODS: We combined vascular casting with 3D imaging of solvent-cleared organs (uDISCO) to study the spatial distribution of the blood coagulation protein fibrinogen in large, intact brain volumes and assessed the temporal regulation of the fibrin(ogen) deposition by immunohistochemistry in a murine model of TBI. Fibrin(ogen) deposition and innate immune cell markers were co-localized by immunohistochemistry in mouse and human brains after TBI. We assessed the role of fibrinogen in TBI using unbiased transcriptomics, flow cytometry and immunohistochemistry for innate immune and neuronal markers in Fggγ390-396A knock-in mice, which express a mutant fibrinogen that retains normal clotting function, but lacks the γ390-396 binding motif to CD11b/CD18 integrin receptor. RESULTS: We show that cerebral fibrinogen deposits were associated with activated innate immune cells in both human and murine TBI. Genetic elimination of fibrin-CD11b interaction reduced peripheral monocyte recruitment and the activation of inflammatory and reactive oxygen species (ROS) gene pathways in microglia and macrophages after TBI. Blockade of the fibrin-CD11b interaction was also protective from oxidative stress damage and cortical loss after TBI. CONCLUSIONS: These data suggest that fibrinogen is a regulator of innate immune activation and neurodegeneration in TBI. Abrogating post-injury neuroinflammation by selective blockade of fibrin's inflammatory functions may have implications for long-term neurologic recovery following brain trauma.


Subject(s)
Brain Injuries, Traumatic , Fibrin , Humans , Mice , Animals , Fibrin/genetics , Fibrin/metabolism , Brain Injuries, Traumatic/pathology , Fibrinogen/metabolism , Immunity, Innate , Oxidative Stress , Mice, Inbred C57BL
5.
In Vivo ; 38(3): 1477-1482, 2024.
Article in English | MEDLINE | ID: mdl-38688594

ABSTRACT

BACKGROUND/AIM: Fibrin-associated diffuse large B-cell lymphoma (FA-DLBCL) is frequently associated with the Epstein-Barr virus (EBV) and manifests as non-mass-forming microscopic lesions within fibrin-rich lesions. Herein, we describe the cytological features of FA-DLBCL. CASE REPORT: A 72-year-old man presented with a large retroperitoneal cystic mass that was treated by cyst aspiration and laparoscopic excision. Individually dispersed large, atypical cells in a necrotic background contained scant cytoplasm and hyperchromatic nuclei with irregular nuclear contours, frequent karyorrhectic debris, and mitotic figures. A fibrinous exudate with necrotic material attached to the inner surface of the cystic mass contained large, atypical cells that were individually scattered or arranged in small clusters. These were positive for cluster of differentiation 20 and Epstein-Barr virus-encoded RNA in situ hybridization. CONCLUSION: We cytologically characterized FA-DLBCL as large, atypical cells that were individually scattered or arranged in small clusters in a necrotic background. To the best of our knowledge, we revealed the cytological features of FA-DLBCL.


Subject(s)
Cysts , Fibrin , Lymphoma, Large B-Cell, Diffuse , Humans , Male , Aged , Lymphoma, Large B-Cell, Diffuse/pathology , Fibrin/metabolism , Cysts/pathology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/pathology , Retroperitoneal Neoplasms/pathology , Tomography, X-Ray Computed
6.
Sci Transl Med ; 16(742): eadi4490, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38598613

ABSTRACT

Uncontrolled bleeding after trauma represents a substantial clinical problem. The current standard of care to treat bleeding after trauma is transfusion of blood products including platelets; however, donated platelets have a short shelf life, are in limited supply, and carry immunogenicity and contamination risks. Consequently, there is a critical need to develop hemostatic platelet alternatives. To this end, we developed synthetic platelet-like particles (PLPs), formulated by functionalizing highly deformable microgel particles composed of ultralow cross-linked poly (N-isopropylacrylamide) with fibrin-binding ligands. The fibrin-binding ligand was designed to target to wound sites, and the cross-linking of fibrin polymers was designed to enhance clot formation. The ultralow cross-linking of the microgels allows the particles to undergo large shape changes that mimic platelet shape change after activation; when coupled to fibrin-binding ligands, this shape change facilitates clot retraction, which in turn can enhance clot stability and contribute to healing. Given these features, we hypothesized that synthetic PLPs could enhance clotting in trauma models and promote healing after clotting. We first assessed PLP activity in vitro and found that PLPs selectively bound fibrin and enhanced clot formation. In murine and porcine models of traumatic injury, PLPs reduced bleeding and facilitated healing of injured tissue in both prophylactic and immediate treatment settings. We determined through biodistribution experiments that PLPs were renally cleared, possibly enabled by ultrasoft particle properties. The performance of synthetic PLPs in the preclinical studies shown here supports future translational investigation of these hemostatic therapeutics in a trauma setting.


Subject(s)
Hemostatics , Rodentia , Animals , Mice , Swine , Rodentia/metabolism , Tissue Distribution , Blood Platelets/metabolism , Hemorrhage , Fibrin/chemistry , Fibrin/metabolism
7.
Ann Biomed Eng ; 52(6): 1665-1677, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38459196

ABSTRACT

Prosthetic heart valve (PHV) replacement has increased the survival rate and quality of life for heart valve-diseased patients. However, PHV thrombosis remains a critical problem associated with these procedures. To better understand the PHV flow-related thrombosis problem, appropriate experimental models need to be developed. In this study, we present an in vitro fibrin clot model that mimics clot accumulation in PHVs under relevant hydrodynamic conditions while allowing real-time imaging. We created 3D-printed mechanical aortic valve models that were inserted into a transparent glass aorta model and connected to a system that simulates human aortic flow pulse and pressures. Thrombin was gradually injected into a circulating fibrinogen solution to induce fibrin clot formation, and clot accumulation was quantified via image analysis. The results of valves positioned in a normal versus a tilted configuration showed that clot accumulation correlated with the local flow features and was mainly present in areas of low shear and high residence time, where recirculating flows are dominant, as supported by computational fluid dynamic simulations. Overall, our work suggests that the developed method may provide data on flow-related clot accumulation in PHVs and may contribute to exploring new approaches and valve designs to reduce valve thrombosis.


Subject(s)
Fibrin , Heart Valve Prosthesis , Thrombin , Thrombosis , Humans , Fibrin/metabolism , Models, Cardiovascular , Perfusion , Aortic Valve/surgery
8.
Biomed Pharmacother ; 174: 116449, 2024 May.
Article in English | MEDLINE | ID: mdl-38518607

ABSTRACT

Traumatic nerve injuries are nowadays a significant clinical challenge and new substitutes with adequate biological and mechanical properties are in need. In this context, fibrin-agarose hydrogels (FA) have shown the possibility to generate tubular scaffolds with promising results for nerve repair. However, to be clinically viable, these scaffolds need to possess enhanced mechanical properties. In this line, genipin (GP) crosslinking has demonstrated to improve biomechanical properties with good biological properties compared to other crosslinkers. In this study, we evaluated the impact of different GP concentrations (0.05, 0.1 and 0.2% (m/v)) and reaction times (6, 12, 24, 72 h) on bioartificial nerve substitutes (BNS) consisting of nanostructured FA scaffolds. First, crosslinked BNS were studied histologically, ultrastructurally and biomechanically and then, its biocompatibility and immunomodulatory effects were ex vivo assessed with a macrophage cell line. Results showed that GP was able to improve the biomechanical resistance of BNS, which were dependent on both the GP treatment time and concentration without altering the structure. Moreover, biocompatibility analyses on macrophages confirmed high cell viability and a minimal reduction of their metabolic activity by WST-1. In addition, GP-crosslinked BNS effectively directed macrophage polarization from a pro-inflammatory (M1) towards a pro-regenerative (M2) phenotype, which was in line with the cytokines release profile. In conclusion, this study considers time and dose-dependent effects of GP in FA substitutes which exhibited increased biomechanical properties while reducing immunogenicity and promoting pro-regenerative macrophage shift. These tubular substitutes could be useful for nerve application or even other tissue engineering applications such as urethra.


Subject(s)
Cross-Linking Reagents , Iridoids , Macrophages , Tissue Scaffolds , Iridoids/pharmacology , Animals , Macrophages/drug effects , Macrophages/metabolism , Tissue Scaffolds/chemistry , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Mice , Hydrogels/chemistry , Hydrogels/pharmacology , Biomechanical Phenomena , Cell Survival/drug effects , Fibrin/metabolism , Sepharose/chemistry , Sepharose/pharmacology , Tissue Engineering/methods , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , RAW 264.7 Cells
9.
Biophys J ; 123(5): 610-621, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38356261

ABSTRACT

We modify a three-dimensional multiscale model of fibrinolysis to study the effect of plasmin-mediated degradation of fibrin on tissue plasminogen activator (tPA) diffusion and fibrinolysis. We propose that tPA is released from a fibrin fiber by simple kinetic unbinding, as well as by "forced unbinding," which occurs when plasmin degrades fibrin to which tPA is bound. We show that, if tPA is bound to a small-enough piece of fibrin that it can diffuse into the clot, then plasmin can increase the effective diffusion of tPA. If tPA is bound to larger fibrin degradation products (FDPs) that can only diffuse along the clot, then plasmin can decrease the effective diffusion of tPA. We find that lysis rates are fastest when tPA is bound to fibrin that can diffuse into the clot, and slowest when tPA is bound to FDPs that can only diffuse along the clot. Laboratory experiments confirm that FDPs can diffuse into a clot, and they support the model hypothesis that forced unbinding of tPA results in a mix of FDPs, such that tPA bound to FDPs can diffuse both into and along the clot. Regardless of how tPA is released from a fiber, a tPA mutant with a smaller dissociation constant results in slower lysis (because tPA binds strongly to fibrin), and a tPA mutant with a larger dissociation constant results in faster lysis.


Subject(s)
Fibrinolysin , Fibrinolysis , Fibrinolysin/metabolism , Fibrinolysin/pharmacology , Tissue Plasminogen Activator/metabolism , Tissue Plasminogen Activator/pharmacology , Fibrin/metabolism , Kinetics , Plasminogen/metabolism
10.
Biol Pharm Bull ; 47(1): 303-310, 2024.
Article in English | MEDLINE | ID: mdl-38281774

ABSTRACT

Methotrexate (MTX) is an indispensable drug used for the treatment of many autoimmune and cancerous diseases. However, its clinical use is associated with serious side effects, such as lung fibrosis. The main objective of this study is to test the hypothesis that hydroxytyrosol (HT) can mitigate MTX-induced lung fibrosis in rats while synergizing MTX anticancer effects. Pulmonary fibrosis was induced in the rats using MTX (14 mg/kg/week, per os (p.o.)). The rats were treated with or without HT (10, 20, and 40 mg/kg/d p.o.) or dexamethasone (DEX; 0.5 mg/kg/d, intraperitoneally (i.p.)) for two weeks concomitantly with MTX. Transforming growth factor beta 1 (TGF-ß1), interleukin-4 (IL-4), thromboxane A2 (TXA2), vascular endothelial growth factor (VEGF), 8-hydroxy-2-deoxy-guanosine (8-OHdG), tissue factor (TF) and fibrin were assessed using enzyme-linked immunosorbent assay (ELISA), immunofluorescence, and RT-PCR. Pulmonary fibrosis was manifested by an excessive extracellular matrix (ECM) deposition and a marked increase in TGF-ß1 and IL-4 in lung tissues. Furthermore, cotreatment with HT or dexamethasone (DEX) significantly attenuated MTX-induced ECM deposition, TGF-ß1, and IL-4 expression. Similarly, HT or DEX notably reduced hydroxyproline contents, TXA2, fibrin, and TF expression in lung tissues. Moreover, using HT or DEX downregulated the gene expression of TF. A significant decrease in lung contents of VEGF, IL-8, and 8-OHdG was also observed in HT + MTX- or DEX + MTX -treated animals in a dose-dependent manner. Collectively, the results of our study suggest that HT might represent a potential protective agent against MTX-induced pulmonary fibrosis.


Subject(s)
Methotrexate , Phenylethyl Alcohol , Pulmonary Fibrosis , Animals , Rats , Dexamethasone/pharmacology , Fibrin/metabolism , Interleukin-4/metabolism , Lung/pathology , Methotrexate/adverse effects , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/prevention & control , Thromboplastin/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
11.
Sci Rep ; 14(1): 2623, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38297113

ABSTRACT

Blood clots, which are composed of blood cells and a stabilizing mesh of fibrin fibers, are critical in cessation of bleeding following injury. However, their action is transient and after performing their physiological function they must be resolved through a process known as fibrinolysis. Internal fibrinolysis is the degradation of fibrin by the endogenous or innate presence of lytic enzymes in the bloodstream; under healthy conditions, this process regulates hemostasis and prevents bleeding or clotting. Fibrin-bound tissue plasminogen activator (tPA) converts nearby plasminogen into active plasmin, which is bound to the fibrin network, breaking it down into fibrin degradation products and releasing the entrapped blood cells. It is poorly understood how changes in the fibrin structure and lytic protein ratios influence the biochemical regulation and behavior of internal fibrinolysis. We used turbidity kinetic tracking and microscopy paired with mathematical modeling to study fibrin structure and lytic protein ratios that restrict internal fibrinolysis. Analysis of simulations and experiments indicate that fibrinolysis is driven by pore expansion of the fibrin network. We show that this effect is strongly influenced by the ratio of fibrin:tPAwhen compared to absolute tPA concentration. Thus, it is essential to consider relative protein concentrations when studying internal fibrinolysis both experimentally and in the clinic. An improved understanding of effective internal lysis can aid in development of better therapeutics for the treatment of bleeding and thrombosis.


Subject(s)
Fibrinolysis , Thrombosis , Humans , Tissue Plasminogen Activator/metabolism , Blood Coagulation , Fibrin/metabolism
12.
Blood ; 143(6): 548-560, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-37944157

ABSTRACT

ABSTRACT: Nonmuscle cell contractility is an essential feature underlying diverse cellular processes such as motility, morphogenesis, division and genome replication, intracellular transport, and secretion. Blood clot contraction is a well-studied process driven by contracting platelets. Megakaryocytes (MKs), which are the precursors to platelets, can be found in bone marrow and lungs. Although they express many of the same proteins and structures found in platelets, little is known about their ability to engage with extracellular proteins such as fibrin and contract. Here, we have measured the ability of MKs to compress plasma clots. Megakaryocytes derived from human induced pluripotent stem cells (iPSCs) were suspended in human platelet-free blood plasma and stimulated with thrombin. Using real-time macroscale optical tracking, confocal microscopy, and biomechanical measurements, we found that activated iPSC-derived MKs (iMKs) caused macroscopic volumetric clot shrinkage, as well as densification and stiffening of the fibrin network via fibrin-attached plasma membrane protrusions undergoing extension-retraction cycles that cause shortening and bending of fibrin fibers. Contraction induced by iMKs involved 2 kinetic phases with distinct rates and durations. It was suppressed by inhibitors of nonmuscle myosin IIA, actin polymerization, and integrin αIIbß3-fibrin interactions, indicating that the molecular mechanisms of iMK contractility were similar or identical to those in activated platelets. Our findings provide new insights into MK biomechanics and suggest that iMKs can be used as a model system to study platelet contractility. Physiologically, the ability of MKs to contract plasma clots may play a role in the mechanical remodeling of intravascular blood clots and thrombi.


Subject(s)
Induced Pluripotent Stem Cells , Thrombosis , Humans , Megakaryocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Blood Platelets/metabolism , Thrombosis/metabolism , Fibrin/metabolism , Plasma
13.
Oral Maxillofac Surg ; 28(1): 413-424, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37269407

ABSTRACT

OBJECTIVES: This cross-sectional invitro research aimed to compare and contrast the macroscopic and microscopic, mechanical and biochemical features of leukocyte-rich platelet-rich fibrin, advanced platelet-rich fibrin, and injectable platelet-rich fibrin. MATERIALS AND METHODS: In all, 150 samples were taken from males aged 18 to 25 with good systemic health (n = 50 each for i-PRF, A-PRF, and L-PRF). The samples were assessed for clot length, clot width, membrane length and width. Microscopic parameters assessed were the distribution of cells and fibrin structure. Mechanical tests were performed for tensile strength using a universal testing machine and growth factor analysis was performed for platelet derived growth factor (PDGF), vascular endothelial growth factor (VEGF), and transforming growth factor (TGF)- ß on Days 1, 3 and 7 using commercially available ELISA kits. The osteogenic potential was analyzed in a culture of human periodontal ligament cells for 21 days using cell viability assay, alkaline phosphatase formation and alizarin red staining for mineralization. RESULTS: L-PRF demonstrates statistically superior clot length, width, weight, membrane length, width and weight in comparison to A-PRF (p < 0.05). L-PRF demonstrates a denser fibrin structure in comparison to A-PRF and i-PRF (p < 0.05). The cells in L-PRF are most commonly situated in the proximal of the clot where as they are distributed in the proximal and middle aspect for A-PRF(p < 0.05). A-PRF demonstrates the highest tensile strength followed by L-PRF (p < 0.05). When growth factor release was evaluated, A-PRF showed noticeably increased release of all growth factors, namely PDGF-BB, TGF-ß, and VEGF, in comparison to i-PRF and L-PRF (p < 0.05). On days 7 and 14, the cell viability of human periodontal ligament cells in co-culture with A-PRF was statistically substantially greater than that of L-PRF and i-PRF (p < 0.05). Alkaline phosphatase levels were statistically substantially higher in A-PRF, followed by i-PRF and L-PRF on days 14 and 21 (p < 0.05). After 21 days of culture, A-PRF treated cultures had much more Alizarin Red staining than L-PRF and i-PRF cultures did (p < 0.05). CONCLUSION: It was determined that although L-PRF exhibits greater size and weight in comparison to A-PRF and i-PRF, A-PRF has superior mechanical properties, increased growth factor releases of TGF-b, PDGF-BB, and VEGF as well as superior cell viability, alkaline phosphatase production, and mineralization on human periodontal ligament cells. CLINICAL RELEVANCE: Based on these findings, A-PRF can be recommended for improved delivery of growth factors and osteogenesis whereas L-PRF is better-suited for applications relying on the size of membrane.


Subject(s)
Anthraquinones , Platelet-Rich Fibrin , Male , Humans , Platelet-Rich Fibrin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Fibrin/pharmacology , Fibrin/metabolism , Osteogenesis , Becaplermin/metabolism , Alkaline Phosphatase/metabolism , Cross-Sectional Studies , Periodontal Ligament/metabolism , Intercellular Signaling Peptides and Proteins , Leukocytes/metabolism
14.
Cell Prolif ; 57(1): e13528, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37539497

ABSTRACT

Cellular spheroids have been described as an appropriate culture system to restore human follicle dermal papilla cells (hFDPc) intrinsic properties; however, they show a low and variable efficiency to promote complete hair follicle formation in in vivo experiments. In this work, a conscientious analysis revealed a 25% cell viability in the surface of the dermal papilla spheroid (DPS) for all culture conditions, questioning whether it is an appropriate culture system for hFDPc. To overcome this problem, we propose the use of human blood plasma for the generation of fibrin microgels (FM) with encapsulated hFDPc to restore its inductive signature, either in the presence or in the absence of blood platelets. FM showed a morphology and extracellular matrix composition similar to the native dermal papilla, including Versican and Collagen IV and increasing cell viability up to 85%. While both systems induce epidermal invaginations expressing hair-specific keratins K14, K15, K71, and K75 in in vitro skin cultures, the number of generated structures increases from 17% to 49% when DPS and FM were used, respectively. These data show the potential of our experimental setting for in vitro hair follicle neogenesis with wild adult hFDPc using FM, being a crucial step in the pursuit of human hair follicle regeneration therapies.


Subject(s)
Hair Follicle , Microgels , Humans , Fibrin/metabolism , Skin , Epidermis , Cells, Cultured
16.
J ISAKOS ; 9(2): 215-220, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37562572

ABSTRACT

This classic discusses the original publication of Dohan Eherenfest et al. on "Classification of platelet concentrates: from pure platelet-rich plasma (P-PRP) to leucocyte- and platelet-rich fibrin (L-PRF)", in which the authors propose four categories of platelet concentrates depending on their leucocyte and fibrin content (P-PRP, leucocyte- and platelet-rich plasma (L-PRP), pure platelet-rich fibrin (P-PRF), and L-PRF) to group a "jungle" of products in which the term platelet-rich plasma (PRP) was used indistinctly. They were able to identify common factors such as: (1) the use of anticoagulants and immediate centrifugation of the blood after its collection; (2) most preparation techniques allowed platelet concentrate preparation within an hour; (3) the centrifugation aimed to separate the blood in layers that would allow the extraction of specific fractions; and (4) the product was activated with thrombin or calcium chloride. The reviewed manuscript has been listed among the most cited PRP articles in regenerative medicine, with more than 800 citations, driving current scientific research and clinical practise by categorising L-PRP and P-PRP (now, leucocyte-poor PRP). The classification has also opened the door to understanding intrinsic biological mechanisms between platelets, leukocytes, fibrin, and growth factors, which will later be considered for studying the proliferation and differentiation of cells in different tissues affected by PRP. Since the initial classification of platelet concentrates, several other classification systems have been proposed and published in the current literature such as platelet, activation, white blood cell (PAW), Mishra, platelet, leucocyte, red blood cells, and activation (PLRA), dose of platelet, efficiency, purity, and activation (DEPA), method, activation, red blood cells, spin, platelets, image guidance, leukocytes, and light activation (MARSPILL), etc. These classifications have identified important aspects of PRP that affect the biological composition and, ultimately, the indications and outcomes. To date, there is still a lack of standardisation in sample preparation, cohort heterogeneity, and incomplete reporting of sample preparation utilised, leading to a lack of clarity and challenging researchers and clinicians.


Subject(s)
Platelet-Rich Fibrin , Platelet-Rich Plasma , Humans , Platelet-Rich Fibrin/metabolism , Platelet-Rich Plasma/metabolism , Leukocytes/metabolism , Blood Platelets/metabolism , Fibrin/metabolism
17.
J Thromb Thrombolysis ; 57(2): 248-259, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37932588

ABSTRACT

BACKGROUND: Residual pulmonary vascular obstruction (RPVO) is common following pulmonary embolism (PE) but its association with fibrin clot properties is poorly understood. We investigated whether prothrombotic state and hypofibrinolysis markers can identify patients with RPVO. METHODS: In 79 normotensive noncancer patients (aged 56 ± 13.3 years) with acute PE, we determined fibrin clot permeability (Ks), clot lysis time (CLT), endogenous thrombin potential (ETP), fibrinolysis proteins, oxidative stress markers, and E-selectin on admission before initiation of anticoagulant therapy, after 5-7 days, and 3 months of anticoagulation. RPVO was diagnosed using computed tomography angiography 3-6 months since PE. RESULTS: Patients with RPVO (n = 23, 29.1%) had at baseline higher simplified Pulmonary Embolism Severity Index (sPESI) (P = 0.004), higher N-terminal brain natriuretic propeptide (P = 0.006) and higher D-dimer (P = 0.044). Patients with versus without RPVO had lower Ks (P < 0.001) and longer CLT (P < 0.05), both at baseline and 5-7 days since admission, but not at 3 months. Patients with RPVO showed 40.6% higher E-selectin (P < 0.001) solely at 3 months. By multivariable logistic regression, baseline Ks (odds ratio [OR] 0.010, 95% confidence interval [CI] 0.001-0.837, P = 0.042, per 10- 9 cm2), baseline D-dimer (OR 1.105, 95% CI 1.000-1.221, P = 0.049, per 100 ng/ml), and E-selectin levels after 3 months (OR 3.874, 95% CI 1.239-12.116, P = 0.020, per 1 ng/ml) were associated with RPVO. CONCLUSIONS: RPVO patients despite anticoagulation characterize with the formation of denser fibrin clots on admission and higher E-selectin at 3 months. Those parameters could be the potential novel RPVO risk factors that warrant further evaluation in an independent cohort.


Subject(s)
Pulmonary Embolism , Thrombosis , Vascular Diseases , Humans , E-Selectin , Pulmonary Embolism/diagnosis , Thrombosis/complications , Risk Factors , Fibrinolysis , Fibrin/metabolism , Fibrin Clot Lysis Time , Anticoagulants , Permeability
18.
Biochemistry ; 63(2): 202-211, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38156948

ABSTRACT

Based on the high structural homology between vascular endothelial (VE)-cadherin and neural (N)-cadherin, we hypothesized that fibrin, which is known to interact with VE-cadherin and promote angiogenesis through this interaction, may also interact with N-cadherin. To test this hypothesis, we prepared fibrin and its plasmin-produced and recombinant fragments covering practically all parts of the fibrin molecule. We also prepared the soluble extracellular portion of N-cadherin (sN-cadherin), which includes all five extracellular N-cadherin domains, and studied its interaction with fibrinogen, fibrin, and the aforementioned fibrin fragments using two independent methods, ELISA and SPR. The experiments confirmed our hypothesis, revealing that fibrin interacts with sN-cadherin with high affinity. Furthermore, the experiments localized the N-cadherin binding site within the fibrin ßN-domains. Notably, the recombinant dimeric (ß15-66)2 fragment, corresponding to these domains and mimicking their dimeric arrangement in fibrin, preserved the N-cadherin-binding properties of fibrin. To localize the fibrin binding site within N-cadherin, we performed ELISA and SPR experiments with (ß15-66)2 and recombinant N-cadherin fragments representing its individual extracellular domains and combinations thereof. The results obtained indicate that the interaction of fibrin with N-cadherin occurs through the third and fifth extracellular domains of the latter. This is in contrast to our previous study, which revealed that fibrin interacts only with the third extracellular domain of VE-cadherin. In conclusion, our study identified N-cadherin as a novel receptor for fibrin and localized complementary binding sites within both fibrin and N-cadherin. The pathophysiological role of this interaction remains to be established.


Subject(s)
Endothelial Cells , Fibrin , Fibrin/metabolism , Binding Sites , Endothelial Cells/metabolism , Fibrinogen/metabolism , Cadherins/metabolism
19.
Int J Mol Sci ; 24(23)2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38069268

ABSTRACT

The effective and long-term treatment of cartilage defects is an unmet need among patients worldwide. In the past, several synthetic and natural biomaterials have been designed to support functional articular cartilage formation. However, they have mostly failed to enhance the terminal stage of chondrogenic differentiation, leading to scar tissue formation after the operation. Growth factors substantially regulate cartilage regeneration by acting on receptors to trigger intracellular signaling and cell recruitment for tissue regeneration. In this study, we investigated the effect of recombinant insulin-like growth factor 1 (rIGF-1), loaded in fibrin microbeads (FibIGF1), on cartilage regeneration. rIGF-1-loaded fibrin microbeads were injected into full-thickness cartilage defects in the knees of goats. The stability, integration, and quality of tissue repair were evaluated at 1 and 6 months by gross morphology, histology, and collagen type II staining. The in vivo results showed that compared to plain fibrin samples, particularly at 6 months, FibIGF1 improved the functional cartilage formation, confirmed through gross morphology, histology, and collagen type II immunostaining. FibIGF1 could be a promising candidate for cartilage repair in the clinic.


Subject(s)
Cartilage Diseases , Cartilage, Articular , Humans , Animals , Collagen Type II/metabolism , Fibrin/metabolism , Goats , Cartilage, Articular/metabolism , Cartilage Diseases/metabolism , Chondrocytes
20.
RMD Open ; 9(4)2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38097272

ABSTRACT

OBJECTIVES: This study investigates the diagnostic role of synovial tissue analysis in children presenting with arthritis and assesses its prognostic significance to predict clinical outcome in juvenile idiopathic arthritis (JIA). METHODS: Synovial samples of paediatric patients undergoing synovial biopsy between 1995 and 2020 were analysed histologically and immunohistochemically. Relationships between histological/immunohistochemical parameters and clinical variables were assessed. RESULTS: Synovial biopsy was performed for diagnosis in 65 cases allowing to correctly classify 79% of patients.At histological analysis on 42 JIA samples, any difference in the number of synovial lining layers, subsynovial elementary lesions, fibrin deposit, Krenn Synovitis Score, inflammatory infiltrate score and pattern emerged between JIA subsets or on treatment exposure. Synovial tissue analysis predicted outcome: higher number of synovial layers predicted worse disease course (>4 flares during follow-up; 4.5 vs 3.0, p=0.035), even after adjusting for age at diagnosis and observation time (OR 2.2, p=0.007); subjects who had switched>2 biological disease-modifying antirheumatic drugs had higher prevalence of subsynovial elementary lesions (55.6% vs 10.3%, p=0.005) and fibrin deposits in synovial lining (60.0% vs 22.6%, p=0.049), even after adjustment for observation time and age at diagnosis (OR 8.1, p=0.047). At immunohistochemistry on 31 JIA samples, higher CD3 expression was described in polyarticular compared with oligoarticular subset (p=0.040). Patients with severe disease course had higher CD20+ rate (OR 7, p=0.023), regardless of JIA subset and treatment exposure. CONCLUSIONS: Synovial tissue analysis might support the clinicians in the diagnostic approach of paediatric patients presenting with arthritis and guide the clinical management in JIA.


Subject(s)
Arthritis, Juvenile , Synovitis , Humans , Child , Arthritis, Juvenile/diagnosis , Arthritis, Juvenile/drug therapy , Prognosis , Synovial Membrane/metabolism , Synovitis/pathology , Disease Progression , Fibrin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...