Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
ACS Appl Bio Mater ; 4(6): 5240-5250, 2021 06 21.
Article in English | MEDLINE | ID: mdl-35007006

ABSTRACT

Wound healing materials to prevent blood loss are crucial during emergency medical treatment because uncontrolled bleeding can lead to patient death. Herein, bioabsorbable fibrous architectures of thrombin-loaded poly(ethylene oxide)-PEO/thrombin-are conceptualized and accomplished via electrospinning for faster wound clotting. Membranes with average fiber diameters ranging from 188 to 264 nm are achieved, where the active thrombin is entrapped within the nanofibers. The results of in vitro and in vivo wound healing activity tests revealed that when the nanofibers with thrombin-loaded capacity are in contact with the wound, the presence of water in the skin or blood catalyzes the degradation of the membranes, thus releasing thrombin. Thrombin then accelerates the wound clotting process. In contrast to other hemostatic materials, PEO/thrombin nanofibers do not require mechanical removal after application, and the viscoelastic nature of such biomaterials enables their conformation to a variety of wound topographies. Remarkably, PEO/thrombin membranes are promising functional materials and their use is a powerful strategy for hemostatic treatment, ranging from simple first aid and sealing to a wound to small surgical procedures.


Subject(s)
Chitosan , Hemostatics , Nanofibers , Ethylene Oxide , Hemostatics/pharmacology , Humans , Polyethylene Glycols , Thrombin
2.
Mem Inst Oswaldo Cruz ; 114: e190088, 2019.
Article in English | MEDLINE | ID: mdl-31188952

ABSTRACT

BACKGROUND: Despite treatment with effective antimalarial drugs, the mortality rate is still high in severe cases of the disease, highlighting the need to find adjunct therapies that can inhibit the adhesion of Plasmodium falciparum-infected erythrocytes (Pf-iEs). OBJECTIVES: In this context, we evaluated a new heparan sulfate (HS) from Nodipecten nodosus for antimalarial activity and inhibition of P. falciparum cytoadhesion and rosetting. METHODS: Parasite inhibition was measured by SYBR green using a cytometer. HS was assessed in rosetting and cytoadhesion assays under static and flow conditions using Chinese hamster ovary (CHO) and human lymphatic endothelial cell (HLEC) cells expressing intercellular adhesion molecule-1 (ICAM1) and chondroitin sulfate A (CSA), respectively. FINDINGS: This HS inhibited merozoite invasion similar to heparin. Moreover, mollusk HS decreased cytoadherence of P. falciparum to CSA and ICAM-1 on the surface of endothelial cells under static and flow conditions. In addition, this glycan efficiently disrupted rosettes. CONCLUSIONS: These findings support a potential use for mollusk HS as adjunct therapy for severe malaria.


Subject(s)
Heparitin Sulfate/pharmacology , Merozoites/drug effects , Mollusca/chemistry , Plasmodium falciparum/drug effects , Animals , Cell Adhesion/drug effects , Erythrocytes/drug effects , Protozoan Proteins/drug effects , Reproducibility of Results , Time Factors
3.
Cytotherapy ; 21(4): 444-459, 2019 04.
Article in English | MEDLINE | ID: mdl-30904331

ABSTRACT

BACKGROUND: Endothelial progenitor cells (EPCs) are circulating progenitor cells that can play an essential role in vascular remodelling. In this work, we compared the role of two EPCs cultivated with different mediums in the resolution of the arterial thrombus induced by FeCl3 lesion and in vessel re-endothelization in the mouse carotid artery. METHODS: Mice mononuclear cells were differentiated into EPCs using Dulbecco's Modified Eagle's Medium (DMEM) and vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF) and IGF (Insulin Growth Factor) called EPCs--M1) or with EGM2(endothelial growth medium) (media supplemented with growth factors from Lonza called (EPCs-M2) for 30days and characterized using flow cytometry. The animals received three EPC injections post-lesion, and we analyzed thrombosis time, vessel re-endothelization, metalloproteinases activities, eNOS (endothelial Nitric oxide synthase) presence and SDF-1(Stromal Derived Factor- 1) levels in circulation. RESULTS: EPC-M1 presented a more immature progenitor profile than EPC-M2 cells. The injection of EPC-M1 prolonged the thrombosis time, and the treatment with the different EPCs increased eNOS expression and MMP2 (Metalloproteinase 2) activity and decreased SDF-1 in plasma. Only EPC-M1 treatment increased both MMP2 and MMP9 and reduced thrombus after 7days. Also, both EPCs decreased platelet aggregation in vitro. CONCLUSIONS: EPCs-M1 were more efficient in all of the analyzed assays. EPCsM2 may be a more mature EPC, proliferating less and promoting a less significant matrix remodelling. EPCs can promote vascular remodelling by inhibiting thrombosis and stimulating vascular wall remodelling and the treatment with a more immature progenitor may be more efficient in this process.


Subject(s)
Endothelial Progenitor Cells/transplantation , Thrombosis/therapy , Animals , Arteries/pathology , Cell Differentiation , Cells, Cultured , Chemokine CXCL12/metabolism , Embolization, Therapeutic , Endothelial Progenitor Cells/metabolism , Gelatinases/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Platelet Aggregation , Thrombosis/enzymology , Thrombosis/pathology , Vascular Endothelial Growth Factor A/metabolism
4.
Thromb Res ; 175: 21-31, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30685522

ABSTRACT

The practice of physical exercise is highly indicated to prevent cardiovascular diseases and is directly related to the improvement of endothelial function and the regulation of arterial blood pressure. The objective of this study was to analyze the effect of physical exercise in vascular remodeling after FeCl3 chemically induced arterial injury on atherosclerotic mice. To analyze the effect of exercises on thrombus formation, LDL receptor-deficient mice were fed for 6 weeks with a high-fat diet and performed or not physical exercises for 2 weeks before the arterial injury. To verify endothelium recovery the animals were exercised or not 2 weeks before the injury, and 3 weeks after it, when the vessels were analyzed. In this work, we observed that physical exercises done only before arterial injury reduced thrombosis time, protected the endothelial layer, promoted the recruitment of CD34 positive progenitor cells, increased the level of eNOS and gelatinases activities and decreased the number of inflammatory cells in the vessel, but do not avoid the growth of neointima. Otherwise exercises done before and continued after injury, increased gelatinase activities, reduced lipid deposition in the aortic arch and prevented neointima formation. Thus, we could conclude that physical exercises are done before and continued after endothelial injury stimulate endothelial recovery by promoting endothelial cell growth, matrix remodeling and decreasing inflammation in the vessel wall.


Subject(s)
Atherosclerosis/therapy , Exercise/physiology , Neointima/therapy , Thrombosis/therapy , Vascular Remodeling/physiology , Animals , Atherosclerosis/pathology , Humans , Male , Mice
5.
Cell Biol Int ; 43(2): 136-146, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30362212

ABSTRACT

Hyperbaric oxygen is a clinical treatment that contributes to wound healing by increasing fibroblasts proliferation, collagen synthesis, and production of growth factors, inducing angiogenesis and inhibiting antimicrobial activity. It also has been shown that hyperbaric oxygen treatment (HBO), through the activation of nitric oxide synthase promotes an increase in the nitric oxide levels that may improve endothelial progenitor cells (EPC) mobilization from bone marrow to the peripheral blood and stimulates the vessel healing process. However, cellular mechanisms involved in cell proliferation and activation of EPC after HBO treatment remain unknown. Therefore, the present work aimed to analyze the effect of HBO on the proliferation of pre-treated bone marrow-derived EPC with TNF-alpha. Also, we investigated the expression of ICAM and eNOS by immunochemistry, the production of reactive species of oxygen and performed an in vitro wound healing. Although 1h of HBO treatment did not alter the rate of in vitro wound closure or cell proliferation, it increased eNOS expression and decreased ICAM expression and reactive oxygen species production in cells pre-treated with TNF-alpha. These results indicate that HBO can decrease the inflammatory response in endothelial cells mediated by TNF-alpha, and thus, promote vascular recovery after injury.


Subject(s)
Cell Proliferation/drug effects , Endothelial Progenitor Cells/metabolism , Oxygen/pharmacology , Animals , Bone Marrow Cells/cytology , Cell Movement/drug effects , Cells, Cultured , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/drug effects , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology
6.
Mem. Inst. Oswaldo Cruz ; 114: e190088, 2019. graf
Article in English | LILACS | ID: biblio-1012679

ABSTRACT

BACKGROUND Despite treatment with effective antimalarial drugs, the mortality rate is still high in severe cases of the disease, highlighting the need to find adjunct therapies that can inhibit the adhesion of Plasmodium falciparum-infected erythrocytes (Pf-iEs). OBJECTIVES In this context, we evaluated a new heparan sulfate (HS) from Nodipecten nodosus for antimalarial activity and inhibition of P. falciparum cytoadhesion and rosetting. METHODS Parasite inhibition was measured by SYBR green using a cytometer. HS was assessed in rosetting and cytoadhesion assays under static and flow conditions using Chinese hamster ovary (CHO) and human lymphatic endothelial cell (HLEC) cells expressing intercellular adhesion molecule-1 (ICAM1) and chondroitin sulfate A (CSA), respectively. FINDINGS This HS inhibited merozoite invasion similar to heparin. Moreover, mollusk HS decreased cytoadherence of P. falciparum to CSA and ICAM-1 on the surface of endothelial cells under static and flow conditions. In addition, this glycan efficiently disrupted rosettes. CONCLUSIONS These findings support a potential use for mollusk HS as adjunct therapy for severe malaria.


Subject(s)
Plasmodium falciparum , Malaria, Falciparum , Receptors, Cytoadhesin , Heparitin Sulfate , Mollusca
7.
FASEB J ; 32(8): 4470-4481, 2018 08.
Article in English | MEDLINE | ID: mdl-29558201

ABSTRACT

Cerebral malaria (CM) is a multifactorial syndrome involving an exacerbated proinflammatory status, endothelial cell activation, coagulopathy, hypoxia, and accumulation of leukocytes and parasites in the brain microvasculature. Despite significant improvements in malaria control, 15% of mortality is still observed in CM cases, and 25% of survivors develop neurologic sequelae for life-even after appropriate antimalarial therapy. A treatment that ameliorates CM clinical signs, resulting in complete healing, is urgently needed. Previously, we showed a hyperbaric oxygen (HBO)-protective effect against experimental CM. Here, we provide molecular evidence that HBO targets brain endothelial cells by decreasing their activation and inhibits parasite and leukocyte accumulation, thus improving cerebral microcirculatory blood flow. HBO treatment increased the expression of aryl hydrocarbon receptor over hypoxia-inducible factor 1-α (HIF-1α), an oxygen-sensitive cytosolic receptor, along with decreased indoleamine 2,3-dioxygenase 1 expression and kynurenine levels. Moreover, ablation of HIF-1α expression in endothelial cells in mice conferred protection against CM and improved survival. We propose that HBO should be pursued as an adjunctive therapy in CM patients to prolong survival and diminish deleterious proinflammatory reaction. Furthermore, our data support the use of HBO in therapeutic strategies to improve outcomes of non-CM disorders affecting the brain.-Bastos, M. F., Kayano, A. C. A. V., Silva-Filho, J. L., Dos-Santos, J. C. K., Judice, C., Blanco, Y. C., Shryock, N., Sercundes, M. K., Ortolan, L. S., Francelin, C., Leite, J. A., Oliveira, R., Elias, R. M., Câmara, N. O. S., Lopes, S. C. P., Albrecht, L., Farias, A. S., Vicente, C. P., Werneck, C. C., Giorgio, S., Verinaud, L., Epiphanio, S., Marinho, C. R. F., Lalwani, P., Amino, R., Aliberti, J., Costa, F. T. M. Inhibition of hypoxia-associated response and kynurenine production in response to hyperbaric oxygen as mechanisms involved in protection against experimental cerebral malaria.


Subject(s)
Brain/metabolism , Hypoxia/metabolism , Kynurenine/metabolism , Malaria, Cerebral/metabolism , Oxygen/metabolism , Animals , Cerebrovascular Circulation/physiology , Endothelial Cells/metabolism , Female , Hyperbaric Oxygenation/methods , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Microcirculation/physiology
8.
Sci Rep ; 7(1): 6438, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743887

ABSTRACT

Cancer patients are at an increased risk of developing thromboembolic complications. Several mechanisms have been proposed to explain cancer-associated thrombosis including the release of tumor-derived extracellular vesicles and the activation of host vascular cells. It was proposed that neutrophil extracellular traps (NETs) contribute to the prothrombotic phenotype in cancer. In this study, we evaluated the possible cooperation between tumor-derived exosomes and NETs in cancer-associated thrombosis. Female BALB/c mice were orthotopically injected with 4T1 breast cancer cells. The tumor-bearing animals exhibited increased levels of plasma DNA and myeloperoxidase in addition to significantly increased numbers of circulating neutrophils. Mice were subjected to either Rose Bengal/laser-induced venous thrombosis or ferric chloride-induced arterial thrombosis models. The tumor-bearing mice exhibited accelerated thrombus formation in both models compared to tumor-free animals. Treatment with recombinant human DNase 1 reversed the prothrombotic phenotype of tumor-bearing mice in both models. Remarkably, 4T1-derived exosomes induced NET formation in neutrophils from mice treated with granulocyte colony-stimulating factor (G-CSF). In addition, tumor-derived exosomes interacted with NETs under static conditions. Accordingly, the intravenous administration of 4T1-derived exosomes into G-CSF-treated mice significantly accelerated venous thrombosis in vivo. Taken together, our observations suggest that tumor-derived exosomes and neutrophils may act cooperatively in the establishment of cancer-associated thrombosis.


Subject(s)
Exosomes/pathology , Mammary Neoplasms, Experimental/pathology , Neutrophils/pathology , Thrombosis/etiology , Animals , Cell Line, Tumor , Disease Models, Animal , Extracellular Traps , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Mammary Neoplasms, Experimental/complications , Mice, Inbred BALB C , Thrombosis/drug therapy , Venous Thrombosis/drug therapy , Venous Thrombosis/etiology
9.
Thromb Res ; 138: 7-15, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26826502

ABSTRACT

INTRODUCTION: MAGP1 is a glycoprotein present in the elastic fibers and is a part of the microfibrils components. MAGP1 interacts with von Willebrand factor and the active form of TGF-ß and BMP. In mice lacking MAGP1, thrombus formation is delayed, increasing the occlusion time of carotid artery despite presenting normal blood coagulation in vitro. MAGP1-containing microfibrils may play a role in hemostasis and thrombosis. In this work, we evaluated the function of MAGP1 and its relation to TGF-ß in the arterial thrombosis process. METHODS AND RESULTS: We analyzed thrombus formation time in wild type and MAGP1-deficient mice comparing Rose Bengal and Ferric Chloride induced arterial lesion. The potential participation of TGF-ß in this process was accessed when we treated both wild type and MAGP1-deficient mice with losartan (an antihypertensive drug that decreases TGF-ß activity) or captopril (an angiotensin converting enzyme inhibitor that was used as a control antihypertensive drug). Besides, we evaluated thrombus embolization and the gelatinolytic activity in the arterial walls in vitro and ex vivo. Losartan and captopril were able to recover the thrombus formation time without changing blood pressure, activated partial thromboplastin time (aPTT), PT (prothrombin time), platelet aggregation and adhesion, but decreased gelatinase activity. CONCLUSIONS: Our results suggest that both treatments are effective in the prevention of the sub-endothelial ECM degradation, allowing the recovery of normal thrombus formation.


Subject(s)
Antihypertensive Agents/therapeutic use , Captopril/therapeutic use , Contractile Proteins/genetics , Extracellular Matrix Proteins/genetics , Losartan/therapeutic use , Thrombosis/drug therapy , Thrombosis/genetics , Animals , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Arteries/physiopathology , Contractile Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Gelatinases/metabolism , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Platelet Aggregation/drug effects , Platelet Function Tests , RNA Splicing Factors , Thrombosis/metabolism , Thrombosis/physiopathology , Transforming Growth Factor beta/metabolism
10.
Cytotherapy ; 17(10): 1447-64, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26349001

ABSTRACT

BACKGROUND AIMS: Dermatan sulfate (DS), an anticoagulant and antithrombotic glycosaminoglycan, also has anti-inflammatory activity. In this study, we investigated the effect of DS treatment in the presence or absence of bone marrow mononuclear cells (MNCs) or endothelial progenitor cells (EPCs) in the vascular response to carotid artery lesion in C57BL6 mice. METHODS: Thrombus formation, the expression of adhesion molecules and factors involved in vascular remodeling, inflammation or vascular tone were analyzed by histologic examination, Western blotting and enzyme-linked immunoassay 1 and 3 days after vascular injury. RESULTS: DS injections prevented thrombus formation and decreased P-selectin expression after 3 days of the injury. DS treatment also increased plasma SDF-1 levels but failed to rescue endothelial nitric oxide synthase (eNOS) expression, which is responsible for vascular tone. Treatment with MNCs alone failed to prevent thrombus formation 1 day after injury and increased intercellular adhesion molecule-1 expression, likely because of the inflammatory nature of these cells. Treatment with EPCs with DS was the most efficient among all therapies studied. Dual administration of EPCs and DS promoted an increase in the expression of adhesion molecules and, at the same time, induced a higher expression of eNOS at the injury site. Furthermore, it stimulated an elevated number of EPCs to migrate and adhere to the vascular wall. DISCUSSION: Simultaneous treatment with EPCs and DS increased the expression of adhesion molecules, prevented thrombosis, rescued the expression of eNOS and increased migration of EPCs to the site of injury, thereby affecting thrombus remodeling and inflammation and can be involved in vessel hemostasis.


Subject(s)
Carotid Artery Injuries/therapy , Dermatan Sulfate/therapeutic use , Endothelial Progenitor Cells/transplantation , Fibrinolytic Agents/therapeutic use , Thrombosis/prevention & control , Vascular Remodeling/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Bone Marrow Cells/cytology , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Artery Injuries/drug therapy , Carotid Artery Injuries/surgery , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Cells, Cultured , Chemokine CXCL12/biosynthesis , Combined Modality Therapy , Intercellular Adhesion Molecule-1/biosynthesis , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/biosynthesis , P-Selectin/biosynthesis
11.
Cell Biol Int ; 39(10): 1138-50, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25965197

ABSTRACT

Endothelial progenitor cells (EPCs) can be isolated from bone marrow and characterized by the expression of cellular markers such as CD34, CD133, VEGFR2, CD31, and VE-Cadherin, by the uptake of acetylated low-density lipoprotein and by in vitro tube formation in tridimensional matrices. These cells are able to differentiate into mature endothelial cells and participate in the re-endothelization of damaged vessels. In this work, we tested different cultured media that can promote the proliferation and differentiation of mononuclear cells (MNCs) into early EPCs, with defined concentrations of growth factors and serum in order to establish a composition that may ensure us the reproducibility of our cultures. MNCs from mice bone marrow were cultivated using selective culture media containing DMEM or M199 supplemented with 10% FBS, VEGF, bFGF, and IGF, for 3, 7, and 14 days. Differentiation into early EPCs was analyzed using immunohistochemistry, FACS and western blotting and by functional parameters as uptake of ac-LDL, and formation of vessel-like structures. The cells cultivated with medium DMEM-M1 (DMEM plus VEGF, bFGF and IGF) expressed CD34, CD133, CD31, VEGFR2, and VE-Cadherin at all culture time-points with increased expression of these markers after 7 days. Only EPCs cultured for 30 days were able to form vessel-like structure. The uptake of ac-LDL was observed after 3, 7, 14, and 30 days, confirming the differentiation of mononuclear cells into early EPCs. DMEM-M1 was able to sustain MNCs proliferation and differentiation, increasing the expression of the characteristic EPC markers, allowing the expansion of early EPCs in culture in a similar way to that observed in commercial available media.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation , Endothelial Progenitor Cells/cytology , Animals , Bone Marrow Cells/drug effects , Cell Culture Techniques , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Mice, Inbred C57BL
12.
Thromb Res ; 133(5): 945-51, 2014 May.
Article in English | MEDLINE | ID: mdl-24642009

ABSTRACT

The Bauhinia bauhinioides Kallikrein Inhibitor (BbKI) is a Kunitz-type serine peptidase inhibitor of plant origin that has been shown to impair the viability of some tumor cells and to feature a potent inhibitory activity against human and rat plasma kallikrein (Kiapp 2.4 nmol/L and 5.2 nmol/L, respectively). This inhibitory activity is possibly responsible for an effect on hemostasis by prolonging activated partial thromboplastin time (aPTT). Because the association between cancer and thrombosis is well established, we evaluated the possible antithrombotic activity of this protein in venous and arterial thrombosis models. Vein thrombosis was studied in the vena cava ligature model in Wistar rats, and arterial thrombosis in the photochemical induced endothelium lesion model in the carotid artery of C57 black 6 mice. BbKI at a concentration of 2.0 mg/kg reduced the venous thrombus weight by 65% in treated rats in comparison to rats in the control group. The inhibitor prolonged the time for total artery occlusion in the carotid artery model mice indicating that this potent plasma kallikrein inhibitor prevented thrombosis.


Subject(s)
Fibrinolytic Agents/pharmacology , Plant Proteins/pharmacology , Thrombosis/drug therapy , Animals , Bauhinia , Blood Coagulation/drug effects , Disease Models, Animal , Humans , Male , Mice , Random Allocation , Rats , Rats, Wistar , Thrombin/antagonists & inhibitors , Thrombin/pharmacology , Thrombosis/blood
13.
Antimicrob Agents Chemother ; 58(4): 1862-71, 2014.
Article in English | MEDLINE | ID: mdl-24395239

ABSTRACT

Sequestration of Plasmodium falciparum-infected erythrocytes (Pf-iEs) in the microvasculature of vital organs plays a key role in the pathogenesis of life-threatening malaria complications, such as cerebral malaria and malaria in pregnancy. This phenomenon is marked by the cytoadhesion of Pf-iEs to host receptors on the surfaces of endothelial cells, on noninfected erythrocytes, and in the placental trophoblast; therefore, these sites are potential targets for antiadhesion therapies. In this context, glycosaminoglycans (GAGs), including heparin, have shown the ability to inhibit Pf-iE cytoadherence and growth. Nevertheless, the use of heparin was discontinued due to serious side effects, such as bleeding. Other GAG-based therapies were hampered due to the potential risk of contamination with prions and viruses, as some GAGs are isolated from mammals. In this context, we investigated the effects and mechanism of action of fucosylated chondroitin sulfate (FucCS), a unique and highly sulfated GAG isolated from the sea cucumber, with respect to P. falciparum cytoadhesion and development. FucCS was effective in inhibiting the cytoadherence of Pf-iEs to human lung endothelial cells and placenta cryosections under static and flow conditions. Removal of the sulfated fucose branches of the FucCS structure virtually abolished the inhibitory effects of FucCS. Importantly, FucCS rapidly disrupted rosettes at high levels, and it was also able to block parasite development by interfering with merozoite invasion. Collectively, these findings highlight the potential of FucCS as a candidate for adjunct therapy against severe malaria.


Subject(s)
Antimalarials/pharmacology , Chondroitin Sulfates/pharmacology , Merozoites/drug effects , Plasmodium falciparum/drug effects , Animals , Antimalarials/adverse effects , Cells, Cultured , Chondroitin Sulfates/adverse effects , Erythrocytes/drug effects , Erythrocytes/parasitology , Hep G2 Cells , Humans , Sea Cucumbers/chemistry
14.
Int J Med Sci ; 10(12): 1746-54, 2013.
Article in English | MEDLINE | ID: mdl-24155660

ABSTRACT

INTRODUCTION: Mature circulating endothelial cells (CEC) and circulating endothelial progenitor cells (EPC) have been described in several conditions associated with endothelial injury. Their role in deep vein thrombosis (DVT) has not been previously evaluated. PATIENTS AND METHODS: In this pilot study we evaluated the time course of CEC and EPC release after vena cava experimental DVT in mice, using the FeCl3 model. We also evaluated their presence in patients with DVT at different phases of the disease (acute and chronic phase). CEC and EPC were evaluated by Flow Cytometry. RESULTS: In mice, both CEC and EPC were increased 24 hours after DVT induction, peaking 48 hours thereafter. After 72 hours, CEC counts decreased sharply, whereas EPC counts decreased less substantially. In DVT patients we observed a significant increase in CEC counts immediately after DVT compared to healthy individuals. Patients with chronic disease also presented a significant elevation of these cell count. In a subgroup of patients for whom serial samples were available, CEC counts decreased significantly after 9-15 months of the acute event. CONCLUSIONS: Our results suggest the participation of these cells in the reparative processes that follows DVT, both at immediate and late time-points. The different kinetics of CEC and EPC release in experimental DVT suggests a heterogeneous role for these cells in the reparative events after DVT.


Subject(s)
Cell Count , Endothelial Cells/pathology , Stem Cells/pathology , Venous Thrombosis/pathology , Animals , Disease Models, Animal , Female , Flow Cytometry , Humans , Iron Compounds/toxicity , Male , Mice , Venous Thrombosis/blood , Venous Thrombosis/chemically induced
15.
Thromb Res ; 130(4): 646-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22178578

ABSTRACT

INTRODUCTION: The thrombin mutant W215A/E217A (WE thrombin) has greatly reduced procoagulant activity, but it activates protein C in the presence of thrombomodulin and inhibits binding of platelet glycoprotein Ib to von Willebrand factor and collagen under flow conditions. Both thrombomodulin-dependent protein C activation and inhibition of platelet adhesion could contribute to the antithrombotic activity of WE thrombin. MATERIALS AND METHODS: To assess the role of thrombomodulin, we administered WE thrombin to thrombomodulin-deficient (TM(Pro/Pro)) mice and measured the time to occlusive thrombus formation in the carotid artery after photochemical injury of the endothelium. RESULTS AND CONCLUSIONS: Doses of WE thrombin ≥10µg/kg prolonged the thrombosis time of wild-type mice (>1.6-fold), while doses ≥100µg/kg only slightly prolonged the thrombosis time of TM(Pro/Pro) mice. We conclude that thrombomodulin plays a predominate role in mediating the antithrombotic effect of WE thrombin in the arterial circulation of mice after endothelial injury. Thrombomodulin-independent effects may occur only when high doses of WE thrombin are administered.


Subject(s)
Carotid Arteries/drug effects , Carotid Artery Thrombosis/drug therapy , Carotid Artery Thrombosis/pathology , Fibrinolytic Agents/therapeutic use , Thrombin/therapeutic use , Thrombomodulin/metabolism , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Thrombosis/metabolism , Fibrinolytic Agents/metabolism , Gene Deletion , Mice , Mice, Inbred C57BL , Point Mutation , Protein Engineering , Thrombin/genetics , Thrombomodulin/genetics
16.
Thromb Haemost ; 106(4): 712-23, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21800005

ABSTRACT

Shedding of microvesicles (MVs) by cancer cells is implicated in a variety of biological effects, including the establishment of cancer-associated hypercoagulable states. However, the mechanisms underlying malignant transformation and the acquisition of procoagulant properties by tumour-derived MVs are poorly understood. Here we investigated the procoagulant and prothrombotic properties of MVs produced by a melanocyte-derived cell line (melan-a) as compared to its tumourigenic melanoma counterpart Tm1. Tumour cells exhibit a two-fold higher rate of MVs production as compared to melan-a. Melanoma MVs display greater procoagulant activity and elevated levels of the clotting initiator protein tissue factor (TF). On the other hand, tumour- and melanocyte-derived MVs expose similar levels of the procoagulant lipid phosphatidylserine, displaying identical abilities to support thrombin generation by the prothrombinase complex. By using an arterial thrombosis model, we observed that melanoma- but not melanocyte-derived MVs strongly accelerate thrombus formation in a TF-dependent manner, and accumulate at the site of vascular injury. Analysis of plasma obtained from melanoma-bearing mice showed the presence of MVs with a similar procoagulant pattern as compared to Tm1 MVs produced in vitro. Remarkably, flow-cytometric analysis demonstrated that 60% of ex vivo MVs are TF-positive and carry the melanoma-associated antigen, demonstrating its tumour origin. Altogether our data suggest that malignant transformation in melanocytes increases the production of procoagulant MVs, which may contribute for a variety of coagulation-related protumoural responses.


Subject(s)
Cell-Derived Microparticles/metabolism , Melanocytes/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Thromboplastin/metabolism , Animals , Blood Coagulation , Cell Line, Tumor , Cell Transformation, Neoplastic , Cell-Derived Microparticles/pathology , Coagulants/metabolism , Humans , Melanocytes/pathology , Melanocytes/transplantation , Melanoma/pathology , Melanoma/physiopathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Plasma/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/physiopathology , Thrombophilia , Thrombosis , Tumor Microenvironment
17.
BMC Biochem ; 12: 29, 2011 May 29.
Article in English | MEDLINE | ID: mdl-21619699

ABSTRACT

BACKGROUND: Previously, we have reported the presence of highly sulfated dermatans in solitary ascidians from the orders Phlebobranchia (Phallusia nigra) and Stolidobranchia (Halocynthia pyriformis and Styela plicata). Despite the identical disaccharide backbone, consisting of [→4IdoA(2S)ß-1→3GalNAcß-1→], those polymers differ in the position of sulfation on the N-Acetyl galactosamine, which can occur at carbon 4 or 6. We have shown that position rather than degree of sulfation is important for heparin cofactor II activity. As a consequence, 2,4- and 2,6-sulfated dermatans have high and low heparin cofactor II activities, respectively. In the present study we extended the disaccharide analysis of ascidian dermatan sulfates to additional species of the orders Stolidobranchia (Herdmania pallida, Halocynthia roretzi) and Phlebobranchia (Ciona intestinalis), aiming to investigate how sulfation evolved within Tunicata. In addition, we analysed how heparin cofactor II activity responds to dermatan sulfates containing different proportions of 2,6- or 2,4-disulfated units. RESULTS: Disaccharide analyses indicated a high content of disulfated disaccharide units in the dermatan sulfates from both orders. However, the degree of sulfation decreased from Stolidobranchia to Phlebobranchia. While 76% of the disaccharide units in dermatan sulfates from stolidobranch ascidians are disulfated, 53% of disulfated disaccharides are found in dermatan sulfates from phlebobranch ascidians. Besides this notable difference in the sulfation degree, dermatan sulfates from phlebobranch ascidians contain mainly 2,6-sulfated disaccharides whereas dermatan sulfate from the stolidobranch ascidians contain mostly 2,4-sulfated disaccharides, suggesting that the biosynthesis of dermatan sulfates might be differently regulated during tunicates evolution. Changes in the position of sulfation on N-acetylgalactosamine in the disaccharide [→4IdoA(2-Sulfate)ß-1→3GalNAcß-1→] modulate heparin cofactor II activity of dermatan sulfate polymers. Thus, high and low heparin cofactor II stimulating activity is observed in 2,4-sulfated dermatan sulfates and 2,6-sulfated dermatan sulfates, respectively, confirming the clear correlation between the anticoagulant activities of dermatan sulfates and the presence of 2,4-sulfated units. CONCLUSIONS: Our results indicate that in ascidian dermatan sulfates the position of sulfation on the GalNAc in the disaccharide [→4IdoA(2S)ß-1→3GalNAcß-1→] is directly related to the taxon and that the 6-O sulfation is a novelty apparently restricted to the Phlebobranchia. We also show that the increased content of [→4IdoA(2S)ß-1→3GalNAc(4S)ß-1→] disaccharide units in dermatan sulfates from Stolidobranchia accounts for the increased heparin cofactor II stimulating activity.


Subject(s)
Dermatan Sulfate/chemistry , Dermatan Sulfate/metabolism , Heparin Cofactor II/metabolism , Phylogeny , Urochordata/metabolism , Animals , Antithrombins/chemistry , Antithrombins/metabolism , Carbohydrate Sequence , Chondroitin ABC Lyase/metabolism , Electrophoresis, Agar Gel , Electrophoresis, Polyacrylamide Gel , Evolution, Molecular , Hexuronic Acids/metabolism , Humans , Molecular Sequence Data , Partial Thromboplastin Time , Species Specificity , Urochordata/genetics
18.
Cytotherapy ; 13(6): 695-704, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21250866

ABSTRACT

BACKGROUND AIMS: Previously, we have demonstrated that administration of dermatan sulfate (DS) suppresses neointima formation in the mouse carotid artery by activating heparin co-factor II. A similar suppressive effect was observed by increasing the number of progenitor cells in circulation. In this study, we investigated the combination of DS and bone marrow mononuclear cells (MNC), which includes potential endothelial progenitors, in neointima formation after arterial injury. METHODS: Arterial injury was induced by mechanical dilation of the left common carotid artery. We analyzed the extension of endothelial lesion, thrombus formation, P-selectin expression and CD45(+) cell accumulation 1 and 3 days post-injury, and neointima formation 21 days post-injury. Animals were injected with MNC with or without DS during the first 48 h after injury. RESULTS: The extension of endothelial lesion was similar in all groups 1 day after surgery; however, in injured animals treated with MNC and DS the endothelium recovery seemed to be more efficient 21 days after lesion. Treatment with DS inhibited thrombosis, decreased CD45(+) cell accumulation and P-selectin expression at the site of injury, and reduced the neointimal area by 56%. Treatment with MNC reduced the neointimal area by 54%. The combination of DS and MNC reduced neointima formation by more than 91%. In addition, DS promoted a greater accumulation of MNC at the site of injury. CONCLUSIONS: DS inhibits the initial thrombotic and inflammatory processes after arterial injury and promotes migration of MNC to the site of the lesion, where they may assist in the recovery of the injured endothelium.


Subject(s)
Bone Marrow Cells/cytology , Carotid Arteries/drug effects , Dermatan Sulfate/therapeutic use , Neointima/prevention & control , Neointima/therapy , Animals , Anticoagulants/therapeutic use , Bone Marrow Cells/physiology , Carotid Arteries/metabolism , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , P-Selectin/metabolism , Thrombosis/prevention & control , Thrombosis/therapy
19.
Blood ; 111(8): 4137-44, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18281502

ABSTRACT

Mice lacking the extracellular matrix protein microfibril-associated glycoprotein-1 (MAGP1) display delayed thrombotic occlusion of the carotid artery following injury as well as prolonged bleeding from a tail vein incision. Normal occlusion times were restored when recombinant MAGP1 was infused into deficient animals prior to vessel wounding. Blood coagulation was normal in these animals as assessed by activated partial thromboplastin time and prothrombin time. Platelet number was lower in MAGP1-deficient mice, but the platelets showed normal aggregation properties in response to various agonists. MAGP1 was not found in normal platelets or in the plasma of wild-type mice. In ligand blot assays, MAGP1 bound to fibronectin, fibrinogen, and von Willebrand factor, but von Willebrand factor was the only protein of the 3 that bound to MAGP1 in surface plasmon resonance studies. These findings show that MAGP1, a component of microfibrils and vascular elastic fibers, plays a role in hemostasis and thrombosis.


Subject(s)
Carotid Arteries/pathology , Contractile Proteins/deficiency , Extracellular Matrix Proteins/deficiency , Thrombosis/pathology , Animals , Bleeding Time , Blood Coagulation/drug effects , Blood Platelets/drug effects , Blood Pressure/drug effects , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Cattle , Contractile Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Fibrinogen/metabolism , Fibronectins/metabolism , Humans , Immunohistochemistry , Injections , Mice , Mice, Inbred C57BL , Platelet Function Tests , Protein Binding/drug effects , RNA Splicing Factors , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Surface Plasmon Resonance , von Willebrand Factor/metabolism
20.
Blood ; 111(8): 4118-25, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18281504

ABSTRACT

Heparin cofactor II (HCII)-deficient mice form occlusive thrombi more rapidly than do wild-type mice following injury to the carotid arterial endothelium. Dermatan sulfate (DS) and heparan sulfate (HS) increase the rate of inhibition of thrombin by HCII in vitro, but it is unknown whether vascular glycosaminoglycans play a role in the antithrombotic effect of HCII in vivo. In this study, we found that intravenous injection of either wild-type recombinant HCII or a variant with low affinity for HS (K173H) corrected the abnormally short thrombosis time of HCII-deficient mice, while a variant with low affinity for DS (R189H) had no effect. When HCII was incubated with frozen sections of the mouse carotid artery, it bound specifically to DS in the adventitia. HCII was undetectable in the wall of the uninjured carotid artery, but it became concentrated in the adventitia following endothelial injury. These results support the hypothesis that HCII interacts with DS in the vessel wall after disruption of the endothelium and that this interaction regulates thrombus formation in vivo.


Subject(s)
Carotid Arteries/metabolism , Dermatan Sulfate/metabolism , Fibrinolytic Agents/metabolism , Heparin Cofactor II/metabolism , Animals , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Arteries/radiation effects , Chondroitin Lyases/metabolism , Female , Heparin Cofactor II/deficiency , Heparin Cofactor II/pharmacokinetics , Heparin Cofactor II/pharmacology , Heparitin Sulfate/metabolism , Humans , Light , Male , Mice , Mice, Inbred C57BL , Mutant Proteins/pharmacology , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Substrate Specificity/drug effects , Substrate Specificity/radiation effects , Swine , Thrombin/antagonists & inhibitors , Thrombosis/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...