Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 442
Filter
1.
Blood ; 137(21): 2881-2889, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33735914

ABSTRACT

Plasminogen is an abundant plasma protein that exists in various zymogenic forms. Plasmin, the proteolytically active form of plasminogen, is known for its essential role in fibrinolysis. To date, therapeutic targeting of the fibrinolytic system has been for 2 purposes: to promote plasmin generation for thromboembolic conditions or to stop plasmin to reduce bleeding. However, plasmin and plasminogen serve other important functions, some of which are unrelated to fibrin removal. Indeed, for >40 years, the antifibrinolytic agent tranexamic acid has been administered for its serendipitously discovered skin-whitening properties. Plasmin also plays an important role in the removal of misfolded/aggregated proteins and can trigger other enzymatic cascades, including complement. In addition, plasminogen, via binding to one of its dozen cell surface receptors, can modulate cell behavior and further influence immune and inflammatory processes. Plasminogen administration itself has been reported to improve thrombolysis and to accelerate wound repair. Although many of these more recent findings have been derived from in vitro or animal studies, the use of antifibrinolytic agents to reduce bleeding in humans has revealed additional clinically relevant consequences, particularly in relation to reducing infection risk that is independent of its hemostatic effects. The finding that many viruses harness the host plasminogen to aid infectivity has suggested that antifibrinolytic agents may have antiviral benefits. Here, we review the broadening role of the plasminogen-activating system in physiology and pathophysiology and how manipulation of this system may be harnessed for benefits unrelated to its conventional application in thrombosis and hemostasis.


Subject(s)
Plasminogen/physiology , Animals , Antifibrinolytic Agents/therapeutic use , Brain/enzymology , Conjunctivitis/physiopathology , Enzyme Activation , Fibrin/metabolism , Fibrinolysin/physiology , Fibrinolysis/physiology , Fibrinolytic Agents/therapeutic use , Humans , Immunity/physiology , Infections/physiopathology , Inflammation , Mice , Plasminogen/chemistry , Plasminogen/deficiency , Plasminogen/pharmacology , Plasminogen/therapeutic use , Radiodermatitis/drug therapy , Receptors, Cell Surface/physiology , Skin Diseases, Genetic/physiopathology , Thrombosis/diagnosis , Thrombosis/drug therapy , Tranexamic Acid/pharmacology , Tranexamic Acid/therapeutic use , Wound Healing/drug effects , Wound Healing/physiology , Wounds and Injuries/drug therapy
2.
JCI Insight ; 5(15)2020 08 06.
Article in English | MEDLINE | ID: mdl-32759492

ABSTRACT

Stem cell transplantation has emerged as a promising strategy in regenerative medicine. However, the poor survival and persistence of the transplanted cells, including mesenchymal stem cells (MSCs), in the hostile ischemic microenvironments represents a major therapeutic barrier. Here we report that plasminogen (Plg) stimulated MSC functions and promoted MSC survival during tissue repair after ischemia. Genetic Plg ablation abolished MSC survival, migration, and proliferation in mouse ischemic limbs, and abrogated MSC-mediated blood reperfusion, neovascularization, and tissue repair after ischemia, suggesting a critical role for Plg in MSC-mediated tissue repair. Furthermore, multiplex cytokine array analysis identified that Plg cleaved and activated cysteine-rich protein 61 (Cyr61), an ECM-associated growth factor, to stimulate MSC survival and migration. Overexpression with truncated Cyr61 in MSCs rescued blood reperfusion after hind limb ischemia in Plg-deficient mice. Finally, Plg-mediated Cyr61 cleavage promoted endothelial cell migration and neovascularization in vitro and in vivo. Our study reveals that Plg promotes MSC survival, persistence, and paracrine effects and improves postischemic neovascularization and tissue repair through Cyr61 cleavage and activation. Thus, targeting Plg/Cyr61 may offer exciting therapeutic opportunities for strengthening MSC therapy in ischemic diseases.


Subject(s)
Cysteine-Rich Protein 61/metabolism , Disease Models, Animal , Ischemia/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Plasminogen/physiology , Animals , Cysteine-Rich Protein 61/genetics , Female , Hindlimb/metabolism , Hindlimb/pathology , Ischemia/etiology , Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Vet Res ; 50(1): 106, 2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31806006

ABSTRACT

The binding and activation of host plasminogen (PLG) by worm surface enolases has been verified to participate in parasite invasion, but the role of this processes during Trichinella spiralis infection has not been clarified. Therefore, the expression and immunolocalization of a T. spiralis enolase (TsENO) and its binding activity with PLG were evaluated in this study. Based on the three-dimensional (3D) molecular model of TsENO, the protein interaction between TsENO and human PLG was analysed by the ZDOCK server. The interacting residues were identified after analysis of the protein-protein interface by bioinformatics techniques. The key interacting residues were confirmed by a series of experiments. The qPCR analysis results demonstrated that Ts-eno was transcribed throughout the whole life cycle of T. spiralis. The immunofluorescence assay (IFA) results confirmed that TsENO was distributed on the T. spiralis surface. The binding assays showed that recombinant TsENO (rTsENO) and native TsENO were able to bind PLG. Four lysine residues (90, 289, 291 and 300) of TsENO were considered to be active residues for PLG interaction. The quadruple mutant (Lys90Ala + Lys289Ala + Lys291Ala + Lys300Ala) TsENO, in which the key lysine residues were substituted with alanine (Ala) residues, exhibited a reduction in PLG binding of nearly 50% (45.37%). These results revealed that TsENO has strong binding activity with human PLG. The four lysine residues (90, 289, 291 and 300) of TsENO play an important role in PLG binding and could accelerate PLG activation and invasion of the host's intestinal wall by T. spiralis.


Subject(s)
Helminth Proteins/genetics , Phosphopyruvate Hydratase/genetics , Plasminogen/physiology , Trichinella spiralis/physiology , Trichinellosis/immunology , Animals , Female , Helminth Proteins/metabolism , Humans , Mice , Mice, Inbred BALB C , Phosphopyruvate Hydratase/metabolism , Trichinella spiralis/genetics , Trichinellosis/parasitology
4.
Invest Ophthalmol Vis Sci ; 60(5): 1571-1580, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30995314

ABSTRACT

Purpose: To understand the role and further dissect pathways downstream of tissue plasminogen activator (tPA) and the fibrinolytic pathway in modulating outflow facility. Methods: Outflow facility of tissue plasminogen activator (Plat) knockout (KO) mice was determined and compared to that of wild-type (WT) littermates. Gene expression of urokinase plasminogen activator (Plau), plasminogen activator inhibitor (Pai-1), plasminogen (Plg), and matrix metalloproteinases (Mmp-2, -9, and -13) was measured in angle tissues. Expression of the same genes and outflow facility were measured in KO and WT mice treated with triamcinolone acetonide (TA). Amiloride was used to inhibit urokinase plasminogen activator (uPA) in Plat KO mice, and outflow facility was measured. Results: Plat deletion resulted in outflow facility reduction and decreased Mmp-9 expression in angle tissues. Plasminogen expression was undetectable in both KO and WT mice. TA led to further reduction in outflow facility and decreases in expression of Plau and Mmp-13 in plat KO mice. Amiloride inhibition of uPA activity prevented the TA-induced outflow facility reduction in Plat KO mice. Conclusions: tPA deficiency reduced outflow facility in mice and was associated with reduced MMP expression. The mechanism of action of tPA is unlikely to involve plasminogen activation. tPA is not the only mediator of TA-induced outflow facility change, as TA caused reduction in outflow facility of Plat KO mice. uPA did not substitute for tPA in outflow facility regulation but abrogated the effect of TA in the absence of tPA, suggesting a complex role of components of the fibrinolytic system in outflow regulation.


Subject(s)
Fibrinolysis/physiology , Plasminogen Activator Inhibitor 1/physiology , Plasminogen/physiology , Tissue Plasminogen Activator/physiology , Trabecular Meshwork/metabolism , Urokinase-Type Plasminogen Activator/physiology , Amiloride/pharmacology , Animals , Diuretics/pharmacology , Gene Expression Regulation/physiology , Glucocorticoids/pharmacology , Injections, Intraocular , Intraocular Pressure/physiology , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Trabecular Meshwork/drug effects , Triamcinolone Acetonide/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
5.
Semin Thromb Hemost ; 43(2): 115-128, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28215042

ABSTRACT

Platelets are crucial to the hemostatic response. Their role in coagulation is well documented and they have been considered for some time to promote resistance of thrombi to fibrinolysis. Platelets confer resistance to lysis by promoting clot retraction of the immediate fibrin network and through release of plasminogen activator inhibitor-1 from their α-granules. However, recent developments in the field indicate that the role of platelets in fibrinolysis is much more diverse. Indeed, novel studies suggest that platelets form different subpopulations upon activation that play varied roles in regulating hemostasis. Likewise the developments in our understanding of thrombus formation, architecture, and changes in fibrin deposition and composition suggest that these different subpopulations of platelets may populate distinct areas within thrombi and potentially dictate the local hemostatic balance in these areas. This review will discuss the diverse roles of platelets in fibrinolysis and highlight the recent developments in the field and the contribution of both the intracellular pool of modulators as well as the membrane surface in regulating these processes.


Subject(s)
Blood Platelets/metabolism , Fibrinolysis/physiology , Plasminogen/physiology , Humans
6.
Arch Oral Biol ; 74: 136-145, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27743595

ABSTRACT

Plasmin is a proteolytic enzyme that is crucial in fibrinolysis. In oral tissues, the plasminogen system plays an essential role in physiological and pathological processes, which in addition to fibrinolysis include degradation of extracellular matrix, inflammation, immune response, angiogenesis, tissue remodeling, cell migration, and wound healing. Oral tissues reveal a change in the plasminogen system during pathological processes such as periodontitis, peri-implantitis, or pulpitis, as well as in response to mechanical load. The plasminogen system is also a key element in tissue regeneration. The number of studies investigating the plasminogen system in dentistry have grown continuously in recent years, highlighting its increasing relevance in dental medicine. In this review, we present the diverse functions of the plasminogen system in physiology and its importance for dental specialists in pathology and regeneration. We thus provide an overview of the current knowledge on the role of the plasminogen system in the different fields of dentistry, including endodontics, orthodontics, periodontics, and oral surgery.


Subject(s)
Plasminogen/physiology , Regeneration/physiology , Extracellular Matrix/metabolism , Fibrinolysin/physiology , Fibrinolysis/physiology , Humans , Inflammation/metabolism , Neovascularization, Pathologic/metabolism , Orthodontics , Peri-Implantitis/metabolism , Periodontics , Periodontitis/metabolism , Plasminogen/immunology , Plasminogen Activators , Pulpitis/metabolism , Specialties, Dental , Surgery, Oral , Wound Healing
8.
Thromb Haemost ; 115(5): 1001-9, 2016 05 02.
Article in English | MEDLINE | ID: mdl-26791370

ABSTRACT

Wound healing is a complicated biological process that consist of partially overlapping inflammatory, proliferation and tissue remodelling phases. A successful wound healing depends on a proper activation and subsequent termination of the inflammatory phase. The failure to terminate the inflammation halts the completion of wound healing and is a known reason for formation of chronic wounds. Previous studies have shown that wound closure is delayed in plasminogen-deficient mice, and a role for plasminogen in dissection of extracellular matrix was suggested. However, our finding that plasminogen is transported to the wound by inflammatory cells early during the healing process, where it potentiates inflammation, indicates that plasminogen may also have other roles in the wound healing process. Here we report that plasminogen-deficient mice have extensive fibrin and neutrophil depositions in the wounded area long after re-epithelialisation, indicating inefficient debridement and chronic inflammation. Delayed formation of granulation tissue suggests that fibroblast function is impaired in the absence of plasminogen. Therefore, in addition to its role in the activation of inflammation, plasminogen is also crucial for subsequent steps, including resolution of inflammation and activation of the proliferation phase. Importantly, supplementation of plasminogen-deficient mice with human plasminogen leads to a restored healing process that is comparable to that in wild-type mice. Besides of being an activator of the inflammatory phase during wound healing, plasminogen is also required for the subsequent termination of inflammation. Based on these results, we propose that plasminogen may be an important future therapeutic agent for wound treatment.


Subject(s)
Plasminogen/physiology , Skin Physiological Phenomena , Wound Healing/physiology , Animals , Burns/pathology , Burns/physiopathology , Fibrinogen/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Neutrophils/pathology , Plasminogen/deficiency , Plasminogen/genetics , Skin/injuries , Skin/pathology , Skin/physiopathology
9.
Zhonghua Zhong Liu Za Zhi ; 36(5): 329-34, 2014 May.
Article in Chinese | MEDLINE | ID: mdl-25030586

ABSTRACT

OBJECTIVE: To explore the combined anti-tumor effect of radiation therapy and gene-targeted suppression of tumor neovasculature in lung adenocarcinoma in vivo, and to explore the feasibility of micro-PET/CT in dynamic evaluation of treatment effectiveness. METHODS: Thirty 5-6-week old male BALB/c nude mice were used in this study. The mouse models of xenotransplanted human lung adenocarcinoma were divided into 5 groups at random, six mice in each group: the control group, radiation treatment alone group and three groups of recombinant baculovirus plus radiation treatment (intratumoral injection, tail vein injection, and intramuscular injection). The tumor volume was measured every 2 days. Growth delay time (GD) and growth inhibition rate was calculated. FDG metabolism was evaluated by micro-PET-CT before and after treatment. The expressions of VEGF, CD31 and Ki-67 were detected by immunohistochemistry (IHC). RESULTS: The tumor growth delay was >12 days, and the tumor inhibition rate was >45% in the recombinant baculovirus combined with radiotherapy groups, significantly higher than that of the radiotherapy alone group (P < 0.05). Immunohistochemical analysis showed that the expressions of VEGF, CD31 and Ki-67 were significantly lower than that in other groups (P < 0.05). The micro-PET-CT assessment showed that the FDG-metabolism in the recombinant baculovirus combined with radiotherapy groups was significantly reduced (P < 0.05), and the SUVmax (FDG metabolism) of transplanted tumors after treatment was also markedly decreased in comparison with that of the control group. The tumor volume after treatment was significantly correlated with SUVmax in the recombinant baculovirus intratumoral injection + radiotherapy group(r = 0.976), recombinant baculovirus intravenous injection + radiotherapy group (r = 0.954), recombinant baculovirus intramuscular injection + radiotherapy group (r = 0.929), and radiotherapy alone group (r = 0.871, P < 0.05). CONCLUSIONS: The recombinant baculovirus containing Egr1 promoter and K5 gene combined with radiotherapy enhances the suppressing effect on the growth of lung adenocarcinoma in the tumor-bearing nude mice. The inducibility of Egr1 promoter by radiation allows the targeting and controllability of treatment. Micro-PET-CT results have a good correlation with the treatment effectiveness. Therefore, it can be used in real-time evaluation of tumor metabolic function in vivo.


Subject(s)
Adenocarcinoma/radiotherapy , Early Growth Response Protein 1/genetics , Genetic Therapy , Lung Neoplasms/radiotherapy , Molecular Targeted Therapy , Peptide Fragments/genetics , Plasminogen/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Baculoviridae/genetics , Cell Line, Tumor , Combined Modality Therapy , Early Growth Response Protein 1/physiology , Fluorodeoxyglucose F18 , Genetic Vectors , Humans , Ki-67 Antigen/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Peptide Fragments/physiology , Plasminogen/physiology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Positron-Emission Tomography , Promoter Regions, Genetic , Random Allocation , Recombinant Proteins/genetics , Tomography, X-Ray Computed , Tumor Burden , Vascular Endothelial Growth Factor A/metabolism
10.
Semin Thromb Hemost ; 40(5): 585-91, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25000957

ABSTRACT

Great advances have been made in our understanding of the fibrinolytic system from the initial discovery of proteolysis of fibrin by plasmin to the multifaceted and complex role of the plasminogen-plasmin (P-P) system. We now know that the P-P system is composed of several serine proteases and their inhibitors (serpins). This system is involved in many physiological functions, including embryogenesis, cell migration, and wound healing. They also play an important role in the pathogenesis of many diseases, including atherosclerosis, obesity, cancer, and even autoimmune disorders, and neuronal degeneration. Knowledge of their role in cancer enables their use as a prognostic factor. Therapeutic use of various forms of proteases derived from this system has been employed as thrombolytic agents. In addition, small molecules designed to inhibit many of the components of the P-P system are now available for clinical trial, aimed at treatment of these various disorders. The history of such remarkable development of our knowledge on fibrinolysis is reviewed in this article.


Subject(s)
Fibrinolysin/history , Fibrinolysis/physiology , Plasminogen/history , Animals , Fibrinolysin/physiology , History, 18th Century , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Neoplasms/blood , Neoplasms/history , Plasminogen/physiology , Plasminogen Activator Inhibitor 1/history , Thrombolytic Therapy/history
11.
Fiziol Zh (1994) ; 60(1): 25-33, 2014.
Article in Ukrainian | MEDLINE | ID: mdl-24809171

ABSTRACT

The effects of several forms of plasminogen on the state of actin cytoskeleton of human platelets were studied. A ratio between various actin pools, which were detected by immunoblotting, was taken as indicator of platelet cytoskeleton reorganization. It was revealed that intact platelets contain globular (G) actin and membrane cortex (MC) actin in amounts that are 56 and 40% of filamentous (F) actin level, respectively. In both thrombin- and collagen-activated platelets, actin is almost entirely presented in F-form. Incubation of resting platelets with Lys-plasminogen causes elevation of MC-actin level up to 79% in respect to F-form content. In addition, Lys-plasminogen inhibits reorganization of actin cytoskeleton typical for activated platelets. In contrast to Lys-form, Glu-plasminogen affects neither platelet aggregation nor redistribution of actin pools. Thus, these data indicate that cytoskeletal structures of platelets are involved in realization of anti-aggregating effects of Lys-plasminogen.


Subject(s)
Actins/metabolism , Blood Platelets/drug effects , Cytoskeleton/drug effects , Peptide Fragments/pharmacology , Plasminogen/pharmacology , Platelet Aggregation/drug effects , Blood Platelets/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Humans , Peptide Fragments/physiology , Plasminogen/physiology , Platelet Aggregation/physiology
12.
PLoS One ; 9(3): e92326, 2014.
Article in English | MEDLINE | ID: mdl-24647588

ABSTRACT

BACKGROUND: Both tumor cells and their supporting endothelial cells should be considered for targeted cell killing when designing cancer treatments. Here we investigated the feasibility of combining radioiodide and antiangiogenic therapies after baculovirus-mediated transfer of genes encoding the sodium iodide symporter (NIS) and plasminogen kringle 5 (K5). METHODS: A recombinant baculovirus containing the NIS gene under control of the human telomerase reverse transcriptase (hTERT) promoter and the K5 gene driven by the early growth response 1 (Egr1) promoter was developed. Dual-luciferase reporter assay was performed to confirm the activation of hTERT transcription. NIS and K5 gene expression were identified by Western blot and Real-Time PCR. Functional NIS activity in baculovirus-infected Hela cells was confirmed by the uptake of 125I and cytotoxicity of 131I. The apoptotic effect of 131I-induced K5 on baculovirus-infected human umbilical vein endothelial cells (HUVECs) was analyzed by a flow cytometry-based assay. In vivo, NIS reporter gene imaging and therapeutic experiments with 131I were performed. Finally, the microvessel density (MVD) in tumors after treatment was determined by CD31 immunostaining. RESULTS: The activation of hTERT transcription was specifically up-regulated in tumor cells. NIS gene expression markedly increased in baculovirus-infected HeLa cells, but not in MRC5 cells. The Hela cells showed a significant increase of 125I uptake, which was inhibited by NaClO4, and a notably decreased cell survival rate by 131I treatment. Expression of the K5 gene induced by 131I was elevated in a dose- and time-dependent manner and resulted in the apoptosis of HUVECs. Furthermore, 131I SPECT imaging clearly showed cervical tumor xenografts infected with recombinant baculovirus. Following therapy, tumor growth was significantly retarded. CD31 immunostaining confirmed a significant decrease of MVD. CONCLUSION: The recombinant baculovirus supports a promising strategy of NIS-based raidoiodide therapy combined with K5-based antiangiogenic therapy by targeting both the tumor and its supporting vessels.


Subject(s)
Baculoviridae/genetics , Genetic Vectors/genetics , Iodine Radioisotopes/therapeutic use , Peptide Fragments/genetics , Plasminogen/genetics , Symporters/genetics , Cell Line, Tumor , Female , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Iodine Radioisotopes/metabolism , Peptide Fragments/physiology , Plasminogen/physiology , Promoter Regions, Genetic , Radiography , Symporters/physiology , Telomerase/genetics , Uterine Cervical Neoplasms/diagnostic imaging , Uterine Cervical Neoplasms/metabolism , Xenograft Model Antitumor Assays
13.
J Lipid Res ; 55(4): 625-34, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24478033

ABSTRACT

Apo(a), the distinguishing protein component of lipoprotein(a) [Lp(a)], exhibits sequence similarity to plasminogen and can inhibit binding of plasminogen to cell surfaces. Plasmin generated on the surface of vascular cells plays a role in cell migration and proliferation, two of the fibroproliferative inflammatory events that underlie atherosclerosis. The ability of apo(a) to inhibit pericellular plasminogen activation on vascular cells was therefore evaluated. Two isoforms of apo(a), 12K and 17K, were found to significantly decrease tissue-type plasminogen activator-mediated plasminogen activation on human umbilical vein endothelial cells (HUVECs) and THP-1 monocytes and macrophages. Lp(a) purified from human plasma decreased plasminogen activation on THP-1 monocytes and HUVECs but not on THP-1 macrophages. Removal of kringle V or the strong lysine binding site in kringle IV10 completely abolished the inhibitory effect of apo(a). Treatment with carboxypeptidase B to assess the roles of carboxyl-terminal lysines in cellular receptors leads in most cases to decreases in plasminogen activation as well as plasminogen and apo(a) binding; however, inhibition of plasminogen activation by apo(a) was unaffected. Our findings directly demonstrate that apo(a) inhibits pericellular plasminogen activation in all three cell types, although binding of apo(a) to cell-surface receptors containing carboxyl-terminal lysines does not appear to play a major role in the inhibition mechanism.


Subject(s)
Apolipoproteins A/physiology , Plasminogen/physiology , Apolipoproteins A/chemistry , Cells, Cultured , Human Umbilical Vein Endothelial Cells , Humans , Leukocytes, Mononuclear/metabolism , Lysine/physiology , Macrophages/metabolism , Plasminogen Activators/chemistry , Plasminogen Activators/pharmacology , Protein Binding , Protein Interaction Domains and Motifs
15.
J Neurochem ; 126(1): 58-69, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23646899

ABSTRACT

The α2-Antiplasmin (α2AP) protein is known as a principal physiological inhibitor of plasmin, but we previously demonstrated that it acts as a regulatory factor for cellular functions independent of plasmin. α2AP is highly expressed in the hippocampus, suggesting a potential role for α2AP in hippocampal neuronal functions. However, the role for α2AP was unclear. This study is the first to investigate the involvement of α2AP in the dendritic growth of hippocampal neurons. The expression of microtubule-associated protein 2, which contributes to neurite initiation and neuronal growth, was lower in the neurons from α2AP⁻/⁻ mice than in the neurons from α2AP⁺/⁺ mice. Exogenous treatment with α2AP enhanced the microtubule-associated protein 2 expression, dendritic growth and filopodia formation in the neurons. This study also elucidated the mechanism underlying the α2AP-induced dendritic growth. Aprotinin, another plasmin inhibitor, had little effect on the dendritic growth of neurons, and α2AP induced its expression in the neurons from plaminogen⁻/⁻ mice. The activation of p38 MAPK was involved in the α2AP-induced dendritic growth. Therefore, our findings suggest that α2AP induces dendritic growth in hippocampal neurons through p38 MAPK activation, independent of plasmin, providing new insights into the role of α2AP in the CNS.


Subject(s)
Dendrites/physiology , Hippocampus/cytology , Hippocampus/growth & development , Neurons/physiology , alpha-2-Antiplasmin/physiology , Animals , Blotting, Western , Cells, Cultured , Fibrinolysin/physiology , Hippocampus/physiology , Immunohistochemistry , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/physiology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Plasminogen/genetics , Plasminogen/physiology , Real-Time Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/physiology
17.
Semin Thromb Hemost ; 39(4): 373-81, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23504608

ABSTRACT

Lung and pleural injuries are characterized by inflammation, fibrinous transitional matrix deposition, and ultimate scarification. The accumulation of extravascular fibrin is due to concurrently increased local coagulation and decreased fibrinolysis, the latter mainly as a result of increased plasminogen activator inhibitor-1 (PAI-1) expression. Therapeutic targeting of disordered fibrin turnover has long been used for the treatment of pleural disease. Intrapleural fibrinolytic therapy has been found to be variably effective in clinical trials, which likely reflects empiric dosing that does not account for the wide variation in pleural fluid PAI-1 levels in individual patients. The incidence of empyema is increasing, providing a strong rationale to identify more effective, nonsurgical treatment to improve pleural drainage and patient outcomes. Therapeutics designed to resist inhibition by PAI-1 are in development for the treatment of pleural loculation and impaired drainage. The efficacy and safety of these strategies remains to be proven in clinical trial testing. Fibrinolytic therapy administered via the airway has also been proposed for the treatment of acute lung injury, but this approach has not been rigorously validated and is not part of routine clinical management at this time. Challenges to airway delivery of fibrinolysins relate to bioavailability, distribution, and dosing of the interventional agents.


Subject(s)
Fibrinolysin/physiology , Lung Injury/blood , Plasminogen/physiology , Pleura/injuries , Pleura/pathology , Animals , Fibrinolysin/metabolism , Fibrinolysis , Humans , Lung Injury/therapy , Plasminogen/metabolism , Pleura/metabolism , Pleural Effusion/metabolism , Pleural Effusion/pathology , Pleural Effusion/therapy , Thrombolytic Therapy
19.
Circulation ; 127(11): 1209-18, e1-16, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23401155

ABSTRACT

BACKGROUND: Deciphering the molecular and cellular processes that govern macrophage foam cell formation is critical to understanding the basic mechanisms underlying atherosclerosis and other vascular pathologies. METHODS AND RESULTS: Here, we identify a pivotal role of plasminogen (Plg) in regulating foam cell formation. Deficiency of Plg inhibited macrophage cholesterol accumulation on exposure to hyperlipidemic conditions in vitro, ex vivo, and in vivo. Gene expression analysis identified CD36 as a regulated target of Plg, and macrophages from Plg(-/-) mice had decreased CD36 expression and diminished foam cell formation. The Plg-dependent CD36 expression and foam cell formation depended on conversion of Plg to plasmin, binding to the macrophage surface, and the consequent intracellular signaling that leads to production of leukotriene B4. Leukotriene B4 rescued the suppression of CD36 expression and foam cell formation arising from Plg deficiency. CONCLUSIONS: Our findings demonstrate an unanticipated role of Plg in the regulation of gene expression and cholesterol metabolism by macrophages and identify Plg-mediated regulation of leukotriene B4 as an underlying mechanism.


Subject(s)
Cell Differentiation/physiology , Foam Cells/cytology , Foam Cells/metabolism , Gene Expression Regulation/physiology , Macrophages/cytology , Macrophages/metabolism , Plasminogen/physiology , Animals , CD36 Antigens/metabolism , Cholesterol/metabolism , In Vitro Techniques , Leukotriene B4/metabolism , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Plasminogen/deficiency , Plasminogen/genetics , Signal Transduction/physiology
20.
Mol Biol Cell ; 24(3): 210-21, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23243000

ABSTRACT

Lipoprotein(a) (Lp(a)) is associated with cardiovascular disease risk. This may be attributable to the ability of Lp(a) to elicit endothelial dysfunction. We previously reported that apolipoprotein(a) (apo(a); the distinguishing kringle-containing component of Lp(a)) elicits cytoskeletal rearrangements in vascular endothelial cells, resulting in increased cellular permeability. These effects require a strong lysine-binding site (LBS) in apo(a). We now report that apo(a) induces both nuclear ß-catenin-mediated cyclooxygenase-2 (COX-2) expression and prostaglandin E2 secretion, indicating a proinflammatory role for Lp(a). Apo(a) caused the disruption of VE-cadherin/ß-catenin complexes in a Src-dependent manner, decreased ß-catenin phosphorylation, and increased phosphorylation of Akt and glycogen synthase kinase-3ß, ultimately resulting in increased nuclear translocation of ß-catenin; all of these effects are downstream of apo(a) attenuation of phosphatase and tensin homologue deleted on chromosome 10 activity. The ß-catenin-mediated effects of apo(a) on COX-2 expression were absent using a mutant apo(a) lacking the strong LBS. Of interest, the normal and LBS mutant forms of apo(a) bound to human umbilical vein endothelial cells in a similar manner, and the binding of neither was affected by lysine analogues. Taken together, our findings suggest a novel mechanism by which apo(a) can induce proinflammatory and proatherosclerotic effects through modulation of vascular endothelial cell function.


Subject(s)
Apoprotein(a)/physiology , Cell Nucleus/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , beta Catenin/metabolism , Antigens, CD/metabolism , Atherosclerosis/metabolism , Binding Sites , Cadherins/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Enzyme Induction , HEK293 Cells , Humans , Inflammation Mediators/physiology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Plasminogen/physiology , Protein Binding , Protein Isoforms/physiology , Protein Transport , Proteolysis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...