Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Thromb Haemost ; 16(5): 987-990, 2018 05.
Article in English | MEDLINE | ID: mdl-29518284
2.
J Thromb Haemost ; 13 Suppl 1: S121-32, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26149013

ABSTRACT

Throughout evolution, organisms have developed means to contain wounds by simultaneously limiting bleeding and eliminating pathogens and damaged host cells via the recruitment of innate defense mechanisms. Disease emerges when there is unchecked activation of innate immune and/or coagulation responses. A key component of innate immunity is the complement system. Concurrent excess activation of coagulation and complement - two major blood-borne proteolytic pathways - is evident in numerous diseases, including atherosclerosis, diabetes, venous thromboembolic disease, thrombotic microangiopathies, arthritis, cancer, and infectious diseases. Delineating the cross-talk between these two cascades will uncover novel therapeutic insights.


Subject(s)
Biological Evolution , Blood Coagulation Factors/metabolism , Blood Coagulation , Complement Activation , Complement System Proteins/metabolism , Animals , Cell Communication , Humans , Immunity, Innate , Signal Transduction
3.
J Thromb Haemost ; 13(4): 610-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25556624

ABSTRACT

BACKGROUND: The plasmin(ogen) and complement systems are simultaneously activated at sites of tissue injury, participating in hemostasis, wound healing, inflammation and immune surveillance. In particular, the C3 proteolytic fragment, iC3b, and its degradation product C3dg, which is generated by cleavage by factor I (FI) and the cofactor complement receptor CR1, are important in bridging innate and adaptive immunity. Via a thioester (TE) bond, iC3b and C3dg covalently tag pathogens, modulating phagocytosis and adaptive immune responses. OBJECTIVE: To examine plasmin-mediated proteolysis of iC3b, and to evaluate the functional consequences, comparing the effects with products generated by FI/CR1 cleavage of iC3b. METHODS: Dose-dependent and time-dependent plasmin-mediated cleavage of iC3b were characterized by analytical gel electrophoresis. The properties of the resultant TE bond-containing fragments on phagocytosis and induction of pro-inflammatory cytokines were measured in cell culture systems. RESULTS: At low concentrations, plasmin effectively cleaves iC3b, but at numerous previously undescribed sites, giving rise to novel C3c-like and C3dg-like moieties, the latter of which retain the TE bond. When attached to zymosan or erythrocytes and exposed to THP-1 macrophages, the C3dg-like proteins behave almost identically to the bona fide C3dg, yielding less phagocytosis as compared with the opsonin iC3b, and more macrophage secretion of the pro-inflammatory cytokine, IL-12. CONCLUSION: Plasmin cleavage of iC3b provides a complement regulatory pathway that is as efficient as FI/CR1 but does not require a cellular cofactor.


Subject(s)
Complement Activation , Complement C3 Convertase, Alternative Pathway , Complement C3b/metabolism , Fibrinolysin/metabolism , Fibrinolysis , Immunity, Innate , Macrophages/enzymology , Phagocytosis , Animals , Cell Line , Complement Activation/drug effects , Complement C3 Convertase, Alternative Pathway/drug effects , Complement C3b/immunology , Fibrinolysin/immunology , Fibrinolysin/pharmacology , Fibrinolysis/drug effects , Humans , Immunity, Innate/drug effects , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , Macrophages/immunology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Phagocytosis/drug effects , Proteolysis , Rabbits , Signal Transduction , Time Factors
4.
Oncogene ; 34(29): 3770-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25241898

ABSTRACT

Medulloblastoma (MB) is a highly malignant brain tumor that occurs primarily in children. Although surgery, radiation and high-dose chemotherapy have led to increased survival, many MB patients still die from their disease, and patients who survive suffer severe long-term side effects as a consequence of treatment. Thus, more effective and less toxic therapies for MB are critically important. Development of such therapies depends in part on identification of genes that are necessary for growth and survival of tumor cells. Survivin is an inhibitor of apoptosis protein that regulates cell cycle progression and resistance to apoptosis, is frequently expressed in human MB and when expressed at high levels predicts poor clinical outcome. Therefore, we hypothesized that Survivin may have a critical role in growth and survival of MB cells and that targeting it may enhance MB therapy. Here we show that Survivin is overexpressed in tumors from patched (Ptch) mutant mice, a model of Sonic hedgehog (SHH)-driven MB. Genetic deletion of survivin in Ptch mutant tumor cells significantly inhibits proliferation and causes cell cycle arrest. Treatment with small-molecule antagonists of Survivin impairs proliferation and survival of both murine and human MB cells. Finally, Survivin antagonists impede growth of MB cells in vivo. These studies highlight the importance of Survivin in SHH-driven MB, and suggest that it may represent a novel therapeutic target in patients with this disease.


Subject(s)
Cerebellar Neoplasms/metabolism , Hedgehog Proteins/metabolism , Inhibitor of Apoptosis Proteins/deficiency , Medulloblastoma/metabolism , Repressor Proteins/deficiency , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Biphenyl Compounds/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , Chemoradiotherapy , Child , Hedgehog Proteins/antagonists & inhibitors , Humans , Imidazoles/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Ki-67 Antigen/metabolism , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mice, SCID , Microscopy, Confocal , Naphthoquinones/pharmacology , Pyridines/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Survivin , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Leukemia ; 29(2): 433-40, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24903482

ABSTRACT

The inhibitor of apoptosis protein Survivin regulates hematopoiesis, although its mechanisms of regulation of hematopoietic stem cells (HSCs) remain largely unknown. While investigating conditional Survivin deletion in mice, we found that Survivin was highly expressed in phenotypically defined HSCs, and Survivin deletion in mice resulted in significantly reduced total marrow HSCs and hematopoietic progenitor cells. Transcriptional analysis of Survivin(-/-) HSCs revealed altered expression of multiple genes not previously linked to Survivin activity. In particular, Survivin deletion significantly reduced expression of the Evi-1 transcription factor indispensable for HSC function, and the downstream Evi-1 target genes Gata2, Pbx1 and Sall2. The loss of HSCs following Survivin deletion and impaired long-term HSC repopulating function could be partially rescued by ectopic Evi-1 expression in Survivin -/- HSCs. These data demonstrate that Survivin partially regulates HSC function by modulating the Evi-1 transcription factor and its downstream targets and identify new genetic pathways in HSCs regulated by Survivin.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/cytology , Inhibitor of Apoptosis Proteins/genetics , Proto-Oncogenes/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Alleles , Animals , Bone Marrow Cells/cytology , Cell Cycle , Cell Proliferation , DNA-Binding Proteins/metabolism , Female , GATA2 Transcription Factor/metabolism , Gene Deletion , Hematopoiesis , Homeodomain Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , MDS1 and EVI1 Complex Locus Protein , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Phenotype , Pre-B-Cell Leukemia Transcription Factor 1 , Retroviridae/genetics , Survivin , Transcription Factors/metabolism , Transcription, Genetic
6.
Am J Transplant ; 14(5): 1046-60, 2014 May.
Article in English | MEDLINE | ID: mdl-24731002

ABSTRACT

Ischemia/reperfusion (I/R) injury is a major determinant of graft survival in kidney transplantation. Survivin, an inhibitor of apoptosis that participates in the control of mitosis and cell cycle progression, has been implicated in renal protection and repair after I/R injury; however, no study has been performed in the transplant setting. We investigated the role of survivin in modulating posttransplant I/R injury in syngeneic and allogeneic kidney grafts, and studied whether protection from I/R injury impacted on the recipient immune system, on chronic allograft nephropathy and rejection. We used genetically engineered mice with survivin haploinsufficiency and WT mice in which survivin over-expression was induced by gene-delivery. Survivin haploinsufficiency in syngeneic grafts was associated with exuberant I/R tissue injury, which triggered inflammation eventually resulting in graft loss. Conversely, survivin over-expression in the grafts minimized I/R injury and dysfunction in syngeneic grafts and in a clinically relevant fully MHC-mismatched allogeneic combination. In the latter, survivin over-expression translated into limited anti-donor adaptive immune response and less long-term allograft injury with protection from renal parenchymal damage. Our data support survivin over-expression in the graft as a novel target for protocols aimed at limiting tissue damage at the time of transplant ultimately modulating the recipient immune system.


Subject(s)
Graft Rejection/prevention & control , Graft Survival/physiology , Inflammation/prevention & control , Inhibitor of Apoptosis Proteins/physiology , Kidney Transplantation/adverse effects , Reperfusion Injury/prevention & control , Repressor Proteins/physiology , Animals , Apoptosis , Cell Proliferation , Female , Gene Transfer Techniques , Graft Rejection/etiology , Graft Rejection/pathology , Inflammation/etiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Survivin , Tissue Donors , Transplantation, Homologous
7.
Oncogene ; 33(17): 2169-78, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-23728349

ABSTRACT

Drug resistance in acute lymphoblastic leukemia (ALL) remains a major problem warranting new treatment strategies. Wnt/catenin signaling is critical for the self-renewal of normal hematopoietic progenitor cells. Deregulated Wnt signaling is evident in chronic and acute myeloid leukemia; however, little is known about ALL. Differential interaction of catenin with either the Kat3 coactivator CREBBP (CREB-binding protein (CBP)) or the highly homologous EP300 (p300) is critical to determine divergent cellular responses and provides a rationale for the regulation of both proliferation and differentiation by the Wnt signaling pathway. Usage of the coactivator CBP by catenin leads to transcriptional activation of cassettes of genes that are involved in maintenance of progenitor cell self-renewal. However, the use of the coactivator p300 leads to activation of genes involved in the initiation of differentiation. ICG-001 is a novel small-molecule modulator of Wnt/catenin signaling, which specifically binds to the N-terminus of CBP and not p300, within amino acids 1-110, thereby disrupting the interaction between CBP and catenin. Here, we report that selective disruption of the CBP/ß- and γ-catenin interactions using ICG-001 leads to differentiation of pre-B ALL cells and loss of self-renewal capacity. Survivin, an inhibitor-of-apoptosis protein, was also downregulated in primary ALL after treatment with ICG-001. Using chromatin immunoprecipitation assay, we demonstrate occupancy of the survivin promoter by CBP that is decreased by ICG-001 in primary ALL. CBP mutations have been recently identified in a significant percentage of ALL patients, however, almost all of the identified mutations reported occur C-terminal to the binding site for ICG-001. Importantly, ICG-001, regardless of CBP mutational status and chromosomal aberration, leads to eradication of drug-resistant primary leukemia in combination with conventional therapy in vitro and significantly prolongs the survival of NOD/SCID mice engrafted with primary ALL. Therefore, specifically inhibiting CBP/catenin transcription represents a novel approach to overcome relapse in ALL.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Peptide Fragments/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Pyrimidinones/pharmacology , Sialoglycoproteins/metabolism , beta Catenin/metabolism , Animals , Asparaginase/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dexamethasone/pharmacology , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Sialoglycoproteins/antagonists & inhibitors , Sialoglycoproteins/genetics , Survivin , Vincristine/pharmacology , Wnt Signaling Pathway , Xenograft Model Antitumor Assays
8.
Cell Death Differ ; 20(6): 834-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23519077

ABSTRACT

The chromosomal passenger complex (CPC) acts as a key regulator of mitosis, preventing asymmetric segregation of chromosomal material into daughter cells. The CPC is composed of three non-enzymatic components termed Survivin, the inner centromere protein (INCENP) and Borealin, and an enzymatic component, Aurora B kinase. Survivin is necessary for the appropriate separation of sister chromatids during mitosis and is involved in liver regeneration, but its role in regenerative processes is incompletely elucidated. Whether Survivin, which is classified as an inhibitor of apoptosis protein (IAP) based on domain composition, also has a role in apoptosis is controversial. The present study examined the in vivo effects of Survivin ablation in the liver and during liver regeneration after 70% hepatectomy in a hepatocyte-specific knockout mouse model. The absence of Survivin caused a reduction in the number of hepatocytes in the liver, together with an increase in cell volume, macronucleation and polyploidy, but no changes in apoptosis. During liver regeneration, mitosis of hepatocytes was associated with mislocalization of the members of the CPC, which were no longer detectable at the centromere despite an unchanged protein amount. Furthermore, the loss of survivin in regenerating hepatocytes was associated with reduced levels of phosphorylated Histone H3 at serine 28 and abolished phosphorylation of CENP-A and Hec1 at serine 55, which is a consequence of decreased Aurora B kinase activity. These data indicate that Survivin expression determines hepatocyte number during liver development and liver regeneration. Lack of Survivin causes mislocalization of the CPC members in combination with reduced Aurora B activity, leading to impaired phosphorylation of its centromeric target proteins and inappropriate cytokinesis.


Subject(s)
Inhibitor of Apoptosis Proteins/deficiency , Liver Regeneration/physiology , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/deficiency , Animals , Apoptosis/physiology , Aurora Kinase B , Aurora Kinases , Cell Growth Processes/physiology , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, Transgenic , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Survivin
9.
Neuroscience ; 231: 413-9, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23123921

ABSTRACT

In adult mammals, newborn neural precursor cells (NPCs) derived from either the subventricular zone (SVZ) or the subgranular zone (SGZ) migrate into the olfactory bulb and the dentate gyrus (DG), respectively, where some of them mature into excitatory and inhibitory neurons. There is increasing evidence that this neurogenesis process is important for some types of learning and synaptic plasticity and vice versa. Survivin, a member of the inhibitor-of-apoptosis protein (IAP) family, has been suggested to have a central role in the regulation of neurogenesis. The protein is abundantly expressed in nervous tissue during embryonic development while being restricted postnatally to proliferating and migrating NPCs in SVZ and SGZ. Here we examined adult Survivin(Camcre) mice with a conditional deletion of the survivin gene in embryonic neurogenic regions. Although the deletion of survivin had no effect on basic excitability in DG and CA1-region, there was a marked impairment of long-term potentiation (LTP) in these areas. Our data support a function of survivin in hippocampal synaptic plasticity and learning and underline the importance of adult brain neurogenesis for proper operation of the hippocampal tri-synaptic circuit and the physiological functions that depend on it.


Subject(s)
CA1 Region, Hippocampal/physiology , Dentate Gyrus/physiology , Inhibitor of Apoptosis Proteins/metabolism , Long-Term Potentiation/physiology , Neural Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Dentate Gyrus/metabolism , Electroencephalography , Excitatory Postsynaptic Potentials/physiology , Inhibitor of Apoptosis Proteins/genetics , Mice , Mice, Transgenic , Neurogenesis , Neurons/physiology , Repressor Proteins/genetics , Survivin
10.
Eur Respir J ; 32(4): 862-70, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18508817

ABSTRACT

Ischaemia-reperfusion injury of the lung is a major cause of morbidity and mortality, particularly following lung transplantation, the mainstay treatment for patients with end-stage pulmonary disease. Effective measures to prevent this complication are lacking. Thrombomodulin (TM) is an endothelial cell receptor and cofactor for thrombin-mediated generation of the anticoagulant and anti-inflammatory activated protein C (APC). The N-terminal lectin-like domain (LLD) of TM has no direct effects on coagulation, but has distinct anti-inflammatory properties, interfering with leukocyte adhesion, complement activation and cytokine generation, all of which are hallmarks of ischaemia-reperfusion injury. Using a murine model of lung ischaemia-reperfusion injury (90 min ischaemia, 4 h reperfusion), the present study shows that mice lacking the LLD of TM respond with increased extravasation of neutrophils and macrophages into the lung parenchyma and bronchoalveolar fluid (BALF), with augmented BALF levels of cytokines interleukin (IL)-1beta and granulocyte-monocytic colony-stimulating factor (GM-CSF). Pre-treatment of wild-type mice with recombinant LLD, as compared with controls, significantly suppresses protein leakage and accumulation of leukocytes in the BALF. These novel findings support further evaluation of recombinant lectin-like domain of thrombomodulin to protect the lung against tissue-damaging pro-inflammatory responses following ischaemia-reperfusion.


Subject(s)
Lectins/chemistry , Lung Injury/pathology , Reperfusion Injury/metabolism , Thrombomodulin/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Anticoagulants/chemistry , Blood Coagulation , Bronchoalveolar Lavage Fluid , Endothelial Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lung Injury/mortality , Mice , Models, Biological , Protein C/chemistry , Protein Structure, Tertiary , Reperfusion Injury/prevention & control
11.
J Thromb Haemost ; 4(8): 1813-24, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16879225

ABSTRACT

BACKGROUND: Thrombomodulin (TM) is predominantly a vascular endothelial cell plasma membrane glycoprotein that, via distinct structural domains, interacts with multiple ligands, thereby modulating coagulation, fibrinolysis, complement activation, inflammation and cell proliferation. We previously reported that by mediating signals that interfere with mitogen-activated protein kinase and nuclear factor kappaB pathways, the amino-terminal C-type lectin-like domain of TM has direct anti-inflammatory properties. METHODS: In the current study, we use murine models of acute inflammatory arthritis and biochemical approaches to assess the mechanism by which the lectin-like domain of TM modifies disease progression. RESULTS: Mice lacking the lectin-like domain of TM (TM(LeD/LeD)mice) develop inflammatory arthritis that is more rapid in onset and more severe than that developed in their wildtype counterparts. In two models of arthritis, treatment of mice with recombinant soluble lectin-like domain of TM significantly suppresses clinical evidence of disease and diminishes monocyte/macrophage infiltration into the synovium, with weaker expression of the pro-inflammatory high mobility group box chromosomal protein 1. While thrombin-TM mediated activation of thrombin activatable fibrinolysis inhibitor inactivates complement factors C3a and C5a, we show that TM has a second independent mechanism to regulate complement: the lectin-like domain of TM directly interferes with complement activation via the classical and lectin pathways. CONCLUSIONS: These data extend previous insights into the mechanisms by which TM modulates innate immunity, and highlight its potential as a therapeutic target for inflammatory diseases.


Subject(s)
Arthritis/metabolism , Carboxypeptidase B2/metabolism , Lectins/chemistry , Thrombomodulin/chemistry , Thrombomodulin/genetics , Animals , Arthritis/prevention & control , Arthritis, Experimental/metabolism , Blood Coagulation , Complement Activation , Gene Transfer Techniques , Humans , Inflammation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Protein Structure, Tertiary , Synovial Membrane/pathology , Thrombin/metabolism , Thrombomodulin/metabolism
12.
Ann Clin Biochem ; 41(Pt 6): 474-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15588437

ABSTRACT

BACKGROUND: Clinical samples were distributed on 10 occasions to six UK laboratories that perform urinary steroid profile analysis. Urine samples were from normal adult men and women, normal children and neonates. Samples from patients with Cushing's syndrome, virilization, adrenarche, obesity and congenital adrenal hyperplasia (21 and 17-hydroxylase defects) were also used for evaluation. METHODS: Samples were analysed by capillary column gas chromatography (all laboratories) after hydrolysis of conjugates and derivative formation (five laboratories) or by variation of 17-oxogenic steroid methodology (one laboratory). RESULTS: For each distribution of samples, the performance of the participants was compared for quantitative analysis, and user comments were summarized. Quantitative results showed variation without necessarily biasing the result. Comments varied considerably in length. The interpretations did not always lead to a clear diagnosis or advise about appropriate further tests. CONCLUSIONS: This pilot urine steroid profiling scheme has clearly identified the requirement for external quality assessment. It is now hoped to offer this scheme worldwide in collaboration with the European Research Network for the Evaluation and Improvement of Screening, Diagnosis and Treatment of Inherited Disorders of Metabolism (ERNDIM).


Subject(s)
Laboratories/standards , Quality Control , Steroids/urine , Adrenal Cortex Diseases/diagnosis , Adrenal Cortex Diseases/urine , Adrenal Hyperplasia, Congenital/diagnosis , Adrenal Hyperplasia, Congenital/urine , Adult , Child , Child, Preschool , Cushing Syndrome/diagnosis , Cushing Syndrome/urine , Female , Humans , Male , Obesity/diagnosis , Obesity/urine , Pilot Projects , Reproducibility of Results , Sensitivity and Specificity , Virilism/diagnosis , Virilism/urine
14.
Arterioscler Thromb Vasc Biol ; 21(9): 1531-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11557684

ABSTRACT

Mutations in the gene encoding thrombomodulin (TM), a thrombin regulator, are suspected risk factors for venous and arterial thrombotic disease. We have previously described the generation of TM(Pro/Pro) mice carrying a TM gene mutation that disrupts the TM-dependent activation of protein C. Here, it is shown that inbred C57BL/6J TM(Pro/Pro) mice exhibit a hypercoagulable state and an increased susceptibility to thrombosis and sepsis. Platelet thrombus growth after FeCl(3)-induced acute endothelial injury was accelerated in mutant mice. Vascular stasis after permanent ligation of the carotid artery precipitated thrombosis in mutant but not in normal mice. Mutant mice showed increased mortality after exposure to high doses of endotoxin and demonstrated altered cytokine production in response to low-dose endotoxin. The severity of the hypercoagulable state and chronic microvascular thrombosis caused by the TM(Pro) mutation is profoundly influenced by mouse strain-specific genetic differences between C57BL/6 and 129SvPas mice. These data demonstrate that in mice, TM is a physiologically relevant regulator of platelet- and coagulation-driven large-vessel thrombosis and modifies the response to endotoxin-induced inflammation. The phenotypic penetrance of the TM(Pro) mutation is determined by as-yet-uncharacterized genetic modifiers of thrombosis other than TM.


Subject(s)
Thrombomodulin/genetics , Thrombomodulin/physiology , Thrombosis/etiology , Animals , Blood Coagulation , Carotid Artery Thrombosis/chemically induced , Carotid Artery Thrombosis/pathology , Chlorides , Cytokines/biosynthesis , Ferric Compounds , Fibrin/metabolism , Genetic Predisposition to Disease , Ligation , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Sepsis/chemically induced , Sepsis/immunology , Survival Analysis , Thrombosis/blood , Thrombosis/pathology
15.
Cardiovasc Res ; 49(3): 507-21, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11166264

ABSTRACT

The basic molecular mechanisms governing how endothelial cells, periendothelial cells and matrix molecules interact with each other and with numerous growth factors and receptors, to form blood vessels have been presented. The many insights gained from this basic knowledge are being extended to further understand pathological angiogenesis associated with disorders such as arterial stenosis, myocardial ischemia, atherosclerosis, allograft transplant stenosis. wound healing and tissue repair. As a result, novel angiogenic and anti-angiogenic molecules are rapid-ly entering the clinic, with the promise of relief from a host of medical disorders.


Subject(s)
Blood Vessels/embryology , Growth Substances/genetics , Signal Transduction/physiology , Animals , Arteries , Cell Differentiation , Cell Division , Endothelium, Vascular/embryology , Gene Expression , Growth Substances/physiology , Morphogenesis/physiology , Oxygen/metabolism , Stress, Mechanical , Veins , Yolk Sac/blood supply
16.
Blood ; 96(1): 145-8, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10891443

ABSTRACT

The coagulation protease factor Xa induces cellular responses implicated in cardiovascular and inflammatory disease. Effector-cell protease receptor 1 (EPR-1) is a functionally characterized receptor of factor Xa, and the EPR-1 complementary DNA (cDNA) was published. Remarkably, the cDNA encoding an inhibitor of apoptosis, survivin, is reportedly identical to that of EPR-1 except for a few nucleotide differences and its orientation opposite to EPR-1. To isolate the EPR-1 cDNA and gene, we surveyed gene databases for expressed sequence tags (ESTs) that could be derived from EPR-1. All ESTs with strong homology to EPR-1/survivin were derived from survivin and could not encode EPR-1. By polymerase chain reaction and Southern blot hybridization, EPR-1 was not detectable in the human or murine genome, but survivin was. Our data suggest that EPR-1 is either highly cell-specific or the published EPR-1 cDNA includes sequences from clones derived from survivin messenger RNA. The means by which factor Xa mediates its cellular effects requires further evaluation.


Subject(s)
Microtubule-Associated Proteins , Platelet Membrane Glycoproteins/genetics , Receptors, Cell Surface/genetics , Animals , DNA, Complementary , Expressed Sequence Tags , Factor Xa/metabolism , Humans , Inhibitor of Apoptosis Proteins , Mice , Neoplasm Proteins , Polymerase Chain Reaction , Proteins/genetics , RNA, Messenger/genetics , Survivin , Transcription, Genetic
17.
Blood ; 95(4): 1435-42, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10666222

ABSTRACT

Survivin is a member of the inhibitor of apoptosis protein (IAP) family that is believed to play a role in oncogenesis. To elucidate further its physiologic role(s), we have characterized the murine survivin gene and complementary DNA (cDNA). The structural organization of the survivin gene, located on chromosome 11E2, is similar to that of its human counterpart, both containing 4 exons. Surprisingly, 3 full-length murine survivin cDNA clones were isolated, predicting the existence of 3 distinct survivin proteins. The longest open reading frame, derived from all 4 exons, predicts a 140-amino acid residue protein, survivin(140), similar to human survivin, which contains a single IAP repeat and a COOH-terminal coiled-coil domain that links its function to the cell cycle. A second cDNA, which retains intron 3, predicts the existence of a 121-amino acid protein, survivin(121) that lacks the coiled-coil domain. Removal of exon 2-derived sequences by alternative pre-messenger RNA (mRNA) splicing results in a third 40-amino acid residue protein, survivin(40), lacking the IAP repeat and coiled-coil structure. Predictably, only recombinant survivin(140) and survivin(121) inhibited caspase-3 activity. All 3 mRNA species were variably expressed during development from 7.5 days postcoitum. Of the adult tissues surveyed, thymus and testis accumulated high levels of survivin(140) mRNA, whereas survivin(121)-specific transcripts were detected in all tissues, while those representing survivin(40) were absent. Human counterparts to the 3 survivin mRNA transcripts were identified in a study of human cells and tissues. The presence of distinct isoforms of survivin that are expressed differentially suggests that survivin plays a complex role in regulating apoptosis. (Blood. 2000;95:1435-1442)


Subject(s)
Alternative Splicing , Apoptosis/physiology , Genetic Variation , Microtubule-Associated Proteins , Proteins/genetics , Proteins/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle/physiology , Chromosome Mapping , DNA, Complementary , Exons , Gene Expression Regulation, Developmental , Humans , Inhibitor of Apoptosis Proteins , Introns , Male , Mice , Molecular Sequence Data , Neoplasm Proteins , Open Reading Frames , RNA, Messenger/genetics , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Survivin , Testis/metabolism , Transcription, Genetic , Transfection
18.
Thromb Haemost ; 84(6): 1023-30, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11154109

ABSTRACT

The development of the embryonic coagulation system, and its contribution to the maintenance of vascular integrity during the formation of embryonic blood vessels, remain poorly understood. We have characterized the temporal expression patterns of 27 hemostasis-related genes during murine development. We show that, although most coagulation and fibrinolysis-related factors are expressed coordinately by 7.5 dpc, several, including FIX, FXII and PAI2, are not detectable until later developmental timepoints. The expression of hemostasis-specific genes prior to the formation of a functional circulatory system supports the view that some coagulation factors have additional non-hemostatic functions during development. In addition, the discordant expression of some factors suggests that the embryonic hemostatic system may be distinct from that of the adult. These analyses will help to elucidate the regulation of hemostasis during embryonic/vascular development, and will provide a framework to facilitate the interpretation of gene inactivation studies.


Subject(s)
Blood Coagulation Factors/genetics , Embryonic and Fetal Development , Mice, Inbred Strains/embryology , Age Factors , Animals , Cells, Cultured/metabolism , Cells, Cultured/ultrastructure , Gene Expression Regulation , Hemostasis/genetics , Hemostasis/physiology , Mice , Models, Animal , RNA/metabolism , Yolk Sac/cytology , Yolk Sac/embryology , Yolk Sac/metabolism , Yolk Sac/ultrastructure
19.
Thromb Haemost ; 81(5): 815-21, 1999 May.
Article in English | MEDLINE | ID: mdl-10365758

ABSTRACT

The pathophysiologic events leading to organ damage in Plasmodium falciparum malaria infections involve adhesion and sequestration of parasite-infected erythrocytes (PRBC) to the vascular endothelium and syncytiotrophoblast. Several potential receptors to which the PRBCs may bind have recently been identified, one of which is thrombomodulin (TM). TM has been implicated particularly in mediating sequestration of P. falciparum-infected erythrocytes in the placenta and brain, two sites of disease associated with high morbidity. In order to establish that binding of parasite-infected red blood cells to TM is dependent on its containing chondroitin-4-sulfate (CSA), we have mutated the CSA-attachment site of murine TM, and expressed this mutant form (TMsergly) in COS-7 cells. In cytoadhesion assays, we demonstrate that, in contrast to wild-type TM which contains CSA and supports the adhesion of 1466 PRBCs/mm2, TMser-gly does not contain CSA and adhesion of PRBCs to those cells expressing TMser-gly is entirely abrogated (200 PRBCs/mm2). These studies further confirm that the CSA of TM may play a role in the pathophysiology of malaria by providing a binding site for PRBCs.


Subject(s)
Erythrocytes/parasitology , Plasmodium falciparum , Thrombomodulin/metabolism , Animals , Binding Sites , Cell Adhesion , Cells, Cultured , DNA Mutational Analysis , Erythrocytes/metabolism , Erythrocytes/pathology , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Thrombomodulin/genetics
20.
Blood ; 93(10): 3442-50, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10233896

ABSTRACT

Thrombomodulin (TM) is a widely expressed glycoprotein receptor that plays a physiologically important role in maintaining normal hemostatic balance postnatally. Inactivation of the TM gene in mice results in embryonic lethality without thrombosis, suggesting that structures of TM not recognized to be involved in coagulation might be critical for normal fetal development. Therefore, the in vivo role of the cytoplasmic domain of TM was studied by using homologous recombination in ES cells to create mice that lack this region of TM (TMcyt/cyt). Cross-breeding of F1 TMwt/cyt mice (1 wild-type and 1 mutant allele) resulted in more than 300 healthy offspring with a normal Mendelian inheritance pattern of 25.7% TMwt/wt, 46.6% TMwt/cyt, and 27.7% TMcyt/cyt mice, indicating that the tail of TM is not necessary for normal fetal development. Phenotypic analyses showed that the TMcyt/cyt mice responded identically to their wild-type littermates after procoagulant, proinflammatory, and skin wound challenges. Plasma levels of plasminogen, plasminogen activator inhibitor 1 (PAI-1), and alpha2-antiplasmin were unaltered, but plasmin:alpha2-antiplasmin (PAP) levels were significantly lower in TMcyt/cyt mice than in TMwt/wt mice (0.46 +/- 0.2 and 1.99 +/- 0.1 ng/mL, respectively; P <.001). Tissue levels of TM antigen were also unaffected. However, functional levels of plasma TM in the TMcyt/cyt mice, as measured by thrombin-dependent activation of protein C, were significantly increased (P <.001). This supported the hypothesis that suppression in PAP levels may be due to augmented activation of thrombin-activatable fibrinolysis inhibitor (TAFI), with resultant inhibition of plasmin generation. In conclusion, these studies exclude the cytoplasmic domain of TM from playing a role in the early embryonic lethality of TM-null mice and support its function in regulating plasmin generation in plasma.


Subject(s)
Embryonic and Fetal Development/genetics , Thrombomodulin/genetics , Thrombomodulin/metabolism , Animals , Base Sequence , COS Cells , Cytoplasm/metabolism , Embryonic and Fetal Development/physiology , Fetal Death , Genomic Library , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides, Antisense , Phenotype , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...