Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    American Society for Clinical Investigation ; 2001
    In:  Journal of Clinical Investigation Vol. 108, No. 4 ( 2001-8-15), p. 537-546
    In: Journal of Clinical Investigation, American Society for Clinical Investigation, Vol. 108, No. 4 ( 2001-8-15), p. 537-546
    Type of Medium: Online Resource
    ISSN: 0021-9738
    Language: English
    Publisher: American Society for Clinical Investigation
    Publication Date: 2001
    detail.hit.zdb_id: 2018375-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2003
    In:  Nature Medicine Vol. 9, No. 3 ( 2003-3), p. 331-337
    In: Nature Medicine, Springer Science and Business Media LLC, Vol. 9, No. 3 ( 2003-3), p. 331-337
    Type of Medium: Online Resource
    ISSN: 1078-8956 , 1546-170X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2003
    detail.hit.zdb_id: 1484517-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Society of Hematology ; 2005
    In:  Blood Vol. 106, No. 11 ( 2005-11-16), p. 529-529
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 529-529
    Abstract: The transmembrane thrombin receptor, thrombomodulin (TM) suppresses excessive activation of blood coagulation. Complete deficiency of TM results in developmental growth arrest and embryonic lethality in mice. These embryos are resorbed by 8.5 day of development, and do not show “head turning” characteristic of the next developmental stage. The mechanism underlying this phenomenon has remained enigmatic. Growth arrest of TM null embryos is mediated by lack of TM expression in fetal-origin cells of the placenta, but is not associated with increased fibrin clots. TM null embryos cannot be rescued by anticoagulation therapy of the mother with heparin or by absence of maternal and fetal fibrinogen. Lethality of TM null embryos is, however, prevented by genetic reduction of embryonic tissue factor expression, suggesting the involvement of coagulation activation in its pathophysiology. We investigated into the role of platelets in this developmental defect. Using appropriate genetic crosses we generated TM null embryos in NF-E2 −/− mice and examined their fate during gestation. Deficiency of NF-E2 has been previously shown to be associated with severe reduction in platelet counts. We show that TM null embryos are rescued from developmental growth arrest in NF-E2 null mothers. We next wanted to determine whether rescue is specific to the absence of maternal platelets or to the absence of the transcription factor NF-E2. We injected pregnant mice from TM +/− intercrosses with platelet depleting antibodies, during midgestation. Antibody induced depletion of maternal platelets also restored the development of TM null embryos. These results demonstrate that in the absence of TM expression in fetal origin cells of the placenta, maternal platelets disrupt placental function resulting in developmental growth arrest of the embryo. To determine if thrombin-mediated platelet activation is required for the deleterious effect of maternal platelets on TM null embryos, we examined the fate of TM null embryos in Protease Activated Receptor 4 (Par4) null mice. Par4 signaling is essential for the activation of mouse platelets by thrombin. Our results show that the absence of maternal Par4 does not rescue TM null embryos. However, anticoagulation therapy of the mother with enoxaparin, in combination with the genetic absence of maternal Par4, results in “turned” emobryos past the developmental block of untreated embryos. Enoxaparin treatment alone is not sufficient to rescue the embryos. These observations implicate maternal platelets, thrombin and the Par4 receptor in mediating the developmental growth arrest of TM null embryos. Toxic effects of platelet mediated excessive thrombin generation, or chemokine secretion, induced by activation of the platelet Par4 receptor are candidate mechanisms. We have substantiated our observations on the role of maternal platelets in pregnancy outcome in another mouse model of fetal loss that we have generated. In this model, combining maternal thrombophilia due to Factor V Leiden polymorphism with embryonic TM Pro mutation results in fetal loss. Depleting platelets in maternal peripheral blood by antibody treatment of pregnant mice rescues the embryos from midgestational lethality. Enoxaparin treatment, on the other hand, is ineffective. Our results highlight an important role of maternal platelets in determining pregnancy outcome. Understanding this role may open avenues for prevention and treatment of pregnancy disorders in prothrombotic mothers through attenuation of platelet function.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 103, No. 5 ( 2004-03-01), p. 1728-1734
    Abstract: Elevated plasma levels of fibrinogen are associated with the presence of cardiovascular disease, but it is controversial whether elevated fibrinogen causally imparts an increased risk, and as such is a true modifier of cardiovascular disease, or is merely associated with disease. By investigating a transgenic mouse model of hyperfibrinogenemia, we show that elevated plasma fibrinogen concentration (1) elicits augmented fibrin deposition in specific organs, (2) interacts with an independent modifier of hemostatic activity to regulate fibrin turnover/deposition, (3) exacerbates neointimal hyperplasia in an experimental model of stasis-induced vascular remodeling, yet (4) may suppress thrombin generation in response to a procoagulant challenge. These findings provide direct experimental evidence that hyperfibrinogenemia is more than a by-product of cardiovascular disease and may function independently or interactively to modulate the severity and/or progression of vascular disease.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of the American Medical Directors Association, Elsevier BV, Vol. 18, No. 12 ( 2017-12), p. 1097.e11-1097.e24
    Type of Medium: Online Resource
    ISSN: 1525-8610
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Annals of Surgery, Ovid Technologies (Wolters Kluwer Health), Vol. 267, No. 6 ( 2018-06), p. 1093-1099
    Abstract: To characterize the economic hardship for uninsured patients admitted for trauma using catastrophic health expenditure (CHE) risk. Background: Medical debts are the greatest cause of bankruptcies in the United States. Injuries are often unpredictable, expensive to treat, and disproportionally affect uninsured patients. Current measures of economic hardship are insufficient and exclude those at greatest risk. Methods: We performed a retrospective review, using data from the 2007–2011 Nationwide Inpatient Samples of all uninsured nonelderly adults (18—64 yrs) admitted with primary diagnoses of trauma. We used US Census data to estimate annual postsubsistence income and inhospital charges for trauma-related admission. Our primary outcome measure was catastrophic health expenditure risk, defined as any charges ≥40% of annual postsubsistence income. Results: Our sample represented 579,683 admissions for uninsured nonelderly adults over the 5-year study period. Median estimated annual income was $40,867 (interquartile range: $21,286–$71.733). Median inpatient charges were $27,420 (interquartile range: $15,196–$49,694). Overall, 70.8% (95% posterior confidence interval: 70.7%–71.1%) of patients were at risk for CHE. The risk of CHE was similar across most demographic subgroups. The greatest risk, however, was concentrated among patients from low-income communities (77.5% among patients in the lowest community income quartile) and among patients with severe injuries (81.8% among those with ISS ≥ 16). Conclusions: Over 7 in 10 uninsured patients admitted for trauma are at risk of catastrophic health expenditures. This analysis is the first application of CHE to a US trauma population and will be an important measure to evaluate the effectiveness of health care and coverage strategies to improve financial risk protection.
    Type of Medium: Online Resource
    ISSN: 0003-4932 , 1528-1140
    RVK:
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2018
    detail.hit.zdb_id: 2641023-0
    detail.hit.zdb_id: 2002200-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Rockefeller University Press ; 2011
    In:  Journal of Experimental Medicine Vol. 208, No. 5 ( 2011-05-09), p. 923-935
    In: Journal of Experimental Medicine, Rockefeller University Press, Vol. 208, No. 5 ( 2011-05-09), p. 923-935
    Abstract: Mice and rats lacking the guanosine nucleotide-binding protein Gimap5 exhibit peripheral T cell lymphopenia, and Gimap5 can bind to Bcl-2. We show that Gimap5-deficient mice showed progressive multilineage failure of bone marrow and hematopoiesis. Compared with wild-type counterparts, Gimap5-deficient mice contained more hematopoietic stem cells (HSCs) but fewer lineage-committed hematopoietic progenitors. The reduction of progenitors and differentiated cells in Gimap5-deficient mice resulted in a loss of HSC quiescence. Gimap5-deficient HSCs and progenitors underwent more apoptosis and exhibited defective long-term repopulation capacity. Absence of Gimap5 disrupted interaction between Mcl-1—which is essential for HSC survival—and HSC70, enhanced Mcl-1 degradation, and compromised mitochondrial integrity in progenitor cells. Thus, Gimap5 is an important stabilizer of mouse hematopoietic progenitor cell survival.
    Type of Medium: Online Resource
    ISSN: 1540-9538 , 0022-1007
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2011
    detail.hit.zdb_id: 1477240-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    Rockefeller University Press ; 2007
    In:  The Journal of Experimental Medicine Vol. 204, No. 5 ( 2007-05-14), p. 1049-1056
    In: The Journal of Experimental Medicine, Rockefeller University Press, Vol. 204, No. 5 ( 2007-05-14), p. 1049-1056
    Abstract: We describe a mouse model of fetal loss in factor V Leiden (FvL) mothers in which fetal loss is triggered when the maternal prothrombotic state coincides with fetal gene defects that reduce activation of the protein C anticoagulant pathway within the placenta. Fetal loss is caused by disruption of placental morphogenesis at the stage of labyrinth layer formation and occurs in the absence of overt placental thrombosis, infarction, or perfusion defects. Platelet depletion or elimination of protease-activated receptor 4 (Par4) from the mother allows normal placentation and prevents fetal loss. These findings establish a cause–effect relationship for the observed epidemiologic association between maternal FvL status and fetal loss and identify fetal gene defects as risk modifiers of pregnancy failure in prothrombotic mothers. Pregnancy failure is mediated by Par4-dependent activation of maternal platelets at the fetomaternal interface and likely involves a pathogenic pathway independent of occlusive thrombosis. Our results further demonstrate that the interaction of two given thrombosis risk factors produces markedly disparate consequences on disease manifestation (i.e., thrombosis or pregnancy loss), depending on the vascular bed in which this interaction occurs.
    Type of Medium: Online Resource
    ISSN: 1540-9538 , 0022-1007
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2007
    detail.hit.zdb_id: 1477240-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 422-422
    Abstract: Introduction: Tissue factor pathway inhibitor (TFPI) and thrombomodulin (TM) are the two anticoagulant proteins directly bound to endothelium. TFPI inhibits the initiation of coagulation by inhibiting TF-FVIIa and early forms of prothrombinase. TM slows the propagation of coagulation by promoting the activation of protein C (APC), which inactivates FVa and FVIIIa. An in vitro model system of purified coagulation proteins has shown that TFPI and thrombomodulin act synergistically to quench tissue factor mediated thrombin generation via neutralization of prothrombinase activity. However, it is unclear how these two proteins cooperate within different vascular beds in vivo, particularly in the brain that has large amounts of tissue factor procoagulant activity. We have previously shown that mice with decreased thrombomodulin function (TMpro) do not have cerebral fibrin deposition even following LPS injection, while partial TFPI deficiency does induce intravascular fibrin deposition in the brain of the TMpro mice, suggesting that these two anticoagulant proteins have distinct functions within the brain vasculature. TFPI null mice die in utero with brain fibrin deposition. A hyperactivatable murine protein C (hMPC) can be produced by altering two acidic amino acids near the thrombin cleavage site. hMPC is activated 30-fold more efficiently by thrombin than wild type protein C and its activation does not require TM. We have used murine model systems to investigate how hMPC expression alters the (patho)physiology of mice with TFPI deficiency. Methods: Transgenic mice producing hMPC under control of the transthyretin promoter were produced. hMPC was bred into TFPI+/- mice, which were then characterized to define the anticoagulant activity of the transgene. Offspring from these initial matings were interbred to determine if hMPC expression would rescue the embryonic lethality of TFPI null mice. Results: hMPC expression elevated plasma protein C 2-fold and plasma APC 3-fold producing a potent anticoagulant effect. In thrombin generation assays, plasma from TFPI+/-/hMPC+ mice had peak thrombin generation of 44.9±7.5 nM vs. 63.0±3.5 nM in TFPI+/-/hMPC- mice (p 〈 0.001). In a TF-induced pulmonary embolism model 5 of 14 TFPI+/-/hMPC+ mice survived over 5 minutes, while only 1 of 10 TFPI+/-/hMPC- did (p=0.02). When TFPI+/-/hMPC+ mice were mated with TFPI+/-/hMPC- mice, TFPI-/-/hMPC+ pups were born at approximately 30% of the expected frequency regardless of whether the transgene was expressed by the mother or the father. About 35% of the surviving TFPI-/-/hMPC+ mice developed a domed shaped head and succumbed to severe hydrocephalus by 8 weeks of age. Those surviving beyond 8 weeks did not develop severe hydrocephalus and were fertile. One TFPI-/-/hMPC+ mouse with a domed shaped head survived long enough to be examined by MRI, which documented severe hydrocephalus. In addition, two lesions (1mm and 0.5 mm diameter) were present in remaining brain tissue. These lesions contained iron suggesting they were areas of blood clot or hemorrhage. MRI exam of 12 week old TFPI-/-/hMPC+ mice identified subclinical hydrocephalus in 1 of 4 mice. Hydrocephalus did not occur in TFPI+/+/hMPC+ or TFPI+/-/hMPC+ mice. Histological examination of brain tissue from TFPI-/-/hMPC+ mice confirmed hydrocephalus with little remaining normal brain tissue. Mice with hydrocephalus had hemorrhage in the ventricles and brain parenchyma that was associated with areas of fibrin(ogen) deposition. There was also congestion of pial vessels and hemorrhage within the subarachnoid space. These findings were not observed in TFPI+/-/hMPC+ mice. Conclusions: Expression of the hMPC transgene produces a potent anticoagulant effect that partially rescues TFPI null embryonic lethality. Surviving TFPI-/-/hMPC+ pups are susceptible to death from severe hydrocephalus associated with hemorrhage and vascular abnormalities in the brain as they mature. These findings demonstrate that TFPI produces physiologically important anticoagulant activity within murine brain vasculature that is not fully compensated by over-expression of activated protein C. Disclosures Mast: Novo Nordisk: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 125, No. 26 ( 2015-06-25), p. 4078-4084
    Abstract: Modulation of thrombin-dependent platelet activation by TFPI is required for successful embryonic development. TFPI dampens intravascular thrombin generation even in the absence of thrombin-mediated platelet activation.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages