GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 407-407
    Abstract: Multiple sclerosis (MS) is common and often devastating chronic inflammatory disease leading to demyelination in the central nervous system and relapsing neurological deficits, including paralysis and vision loss. MS lesions are characterized by blood-brain barrier disruption leading to perivascular deposition of fibrin that correlates with areas of microglia activation and myelin damage. Consistent with multiple studies showing that hemostatic factors can serve as important modulators of the inflammatory response in vivo, we recently identified fibrin(ogen) as a novel regulator of microglial activation/differentiation and showed that fibrinogen plays a causative role in the development of inflammatory demyelination in experimental autoimmune encephalomyelitis (EAE), an established animal model for MS. A working hypothesis that has emerged is that Mac1-mediated microglial cell engagement of fibrin-rich matrices deposited within MS plaques drives activation events leading to neuronal destruction. To better define contribution of the thrombin/fibrin axis in demyelinating neuroinflammatory disease, we challenged conditional “floxed” fII knockout mice carrying low levels (∼10% of normal) of circulation prothrombin (fII) with EAE initiated by immunization with myelin oligodendrocyte peptide. Wild-type mice challenged with EAE typically began to develop overt neurological symptoms within two weeks of immunization. Clinical disease progressed from simple loss of tail tone to ataxia and, ultimately, fore- and hind-limb paralysis. When cohorts were clinically scored in a fashion blinded to animal genotype, prothrombin-deficient mice were found to exhibit significantly reduced disease severity than the control mice tracked in parallel. The timing of disease onset was similar in fII-deficient mice as in controls, consistent with the hypothesis that fII-deficiency diminishes microglial activity. In contrast to control mice that uniformly developed clinically apparent disease, the two-thirds of fII-deficient mice develop either no disease or very mild disease typically limited to the simple loss of tail tone. More detailed studies of fII-deficient mice, including comparative studies of CNS histopathology, are now underway to more fully define the benefits and liabilities of diminished prothrombin on CNS disease. Complementary studies are also underway with gene-targeted mice expressing a mutant form of prothrombin (fII with “specificity switch” favoring protein C over procoagulant substrates). Both prothrombin and fibrinogen are powerful determinants of inflammatory CNS demyelinating disease and a more detailed understanding of the contribution of these factors to disease progression may reveal novel therapeutic strategies for the attenuation of this and other neuroinflammatory diseases.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 113, No. 3 ( 2009-01-15), p. 696-704
    Abstract: Mice carrying a conditional prothrombin knockout allele (fIIlox) were established to develop an experimental setting for exploring the importance of thrombin in the maintenance of vascular integrity, the inflammatory response, and disease processes in adult animals. In the absence of Cre-mediated recombination, homozygous fIIlox/lox mice or compound heterozygous mice carrying one fIIlox allele and one constitutive-null allele were viable. Young adults exhibited neither spontaneous bleeding events nor diminished reproductive success. However, the induction of Cre recombinase in fIIlox mice using the poly I:C-inducible Mx1-Cre system resulted in the rapid and near-complete recombination of the fIIlox allele within the liver, the loss of circulating prothrombin, and profound derangements in coagulation function. Consistent with the notion that thrombin regulates coagulation and inflammatory pathways, an additional early consequence of reducing prothrombin was impaired antimicrobial function in mice challenged with Staphylococcus aureus peritonitis. However, life expectancy in unchallenged adults genetically depleted of prothrombin was very short (∼5-7 days). The loss of viability was associated with the development of severe hemorrhagic events within multiple tissues, particularly in the heart and brain. Unlike the constitutive loss of either clotting or platelet function alone, the conditional loss of prothrombin is uniformly not compatible with maintenance of hemostasis or long-term survival.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 267-267
    Abstract: Sickle cell anemia (SCA) is caused by a point mutation in the beta-globin gene. SCA has potentially devastating consequences including chronic hemolytic anemia, episodic vascular occlusion, inflammation and oxidative stress, and cumulative multi-organ damage resulting in early mortality. In fact, with reduction of childhood mortality due to early diagnosis and infection prophylaxis, end-organ damage is the major cause of death in SCA. SCA patients show hyper coagulative state in the absence of vascular occlusion. Recently, our group has shown that reduction of circulating major clotting factor, thrombin, in SCA mice significantly improves survival, reduces inflammation, results in reduced multi-organ damage and prolongs survival (Arumugam 2015). However, the mechanism(s) by which thrombin contributes to the pathophysiology of SCA remains undefined. While fibrinogen functions primarily to occlude blood vessels after cleavage by thrombin into fibrin, and thereby prevents excessive bleeding, it also plays an important role in the pathologic inflammatory disease processes through mitogenic, chemotactic, and immune-regulatory activities by interacting with neutrophils/ macrophages via the Mac-1 receptor. To test the hypothesis that leukocyte engagement of fibrinogen via Mac-1 and secondary inflammation are drivers of SCA-associated organ pathologies, hematopoietic stem cells (HSC) from the well-characterized humanized murine model of SCA, Berkeley sickle mice (HbS or SS) were transplanted into the Fibγ390-396A mice, the later expressing a mutant fibrinogen γ-chain which does not interact with Mac-1 (named as F1M SS). Fibγ390-396 mutation has no effect on clotting functions, supports platelet adhesion, and does not cause spontaneous hemorrhagic events in mice during development or adulthood (Flick 2004). As a control, normal fibrinogen expressing mice were also transplanted with HSC from sickle mice (named as F1WT SS). One year after HSC transplant, the F1M SS and F1WT SS mice were analyzed for blood parameters, reactive oxygen species (ROS), inflammatory cytokines, and organ functions and pathologies. We found that genetically imposed inhibition of fibrinogen binding to Mac-1 resulted in mild reduction (not statistically significant) of neutrophil, monocyte, and platelet counts. There was no significant difference in RBC parameters between F1WT SS and F1M SS mice. However, we found an impressive reduction in WBC ROS and decreased circulating inflammatory cytokines, IL-6 (79.1 ± 19.5 vs. 303.5 ± 73.9, P 〈 0.001), KC (132.7 ± 19.4 vs. 200.7 ± 33.6, P=0.04) and TNF-α (25.9 ± 2.8 vs. 37.7 ± 3.9, P=0.02) in F1M SS mice compared to F1WT SS mice. We also found significant improvement in SCA-associated glomerular pathologies such as reduced glomerulosclerosis, inflammatory cell infiltration, ischemic lesions, mesangial thickening, mesangial hyper cellularity and glomerular enlargement; associated with reduced albuminuria (123.3 ± 19.5 vs. 322.1 ± 91.4, P=0.03) in F1M SS mice compared to F1WT SS mice. However, tubular pathologies and urine osmolality were not improved in F1M SS mice. RNA-Seq analyses of glomerular preparation revealed improved glomerular protective responses and diminished loss of podocyte/mesangial cell signatures in F1M SS mice compared to F1WT SS mice. Interestingly, unlike the pan-organ improvement with thrombin reduction (Arumugam, 2015), fibrinogen binding to Mac-1 had minimal to no effect on cardiac, lung, and liver functions and pathologies. Taken together, our data support that fibrinogen significantly contributes to the WBC-driven inflammation and ROS production, and represents a key driver of SCA-associated glomerulopathy, and may provide a novel therapeutic target against irreversible kidney damage in SCA. Disclosures Mullins: Shire: Honoraria, Membership on an entity's Board of Directors or advisory committees. Malik:CSL Behring: Patents & Royalties.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 1753-1753
    Abstract: Interplay between the hemostatic and innate immune systems appears to be an important determinant of tumor metastasis. Studies of mice with selected hemostatic and/or immunological deficits have been particularly revealing, and indicate that platelets and fibrinogen support metastatic potential in part by impeding the clearance of newly formed micrometastases by natural killer (NK) cells. A key step in the formation of stable platelet/fibrin thrombi is the fXIII-mediated cross-linking of fibrin matrices. In order to examine the role of fXIII in tumor dissemination in detail, we studied tumor growth and metastasis in gene-targeted mice lacking the catalytic fXIII-A subunit (fXIII−/−). Comparative analyses of experimental lung metastases in immunocompetent fXIII−/− and wild-type mice showed that elimination of fXIII diminished the metastatic potential of both Lewis lung carcinoma (LLC) and B16-BL6 melanoma cells 5- to 10-fold. Loss of fXIII activity also significantly diminished tumor cell metastatic potential in spontaneous metastasis assays in which lung and liver lesions were quantified ∼2 weeks after resection of primary subcutaneous tumors. These differences were not the result of any genotype-dependent difference in tumor growth, as tumors transplanted into the dorsal subcutis of fXIII−/− and control mice grew at similar rates. In order to determine if fXIII was coupled to metastasis by a mechanism linked to NK cell function, we compared the early fate/survival of radiolabeled LLC cells in cohorts of fXIII−/− and wild-type mice pretreated with either an anti-asialo GM1 antibody known to deplete NK cells or control Ig. Here, the residual radiolabel present within lungs, blood and abdominal organs was measured 30 minutes or 26 hours after injection. Neither loss of fXIII nor NK cells had any impact on the initial localization of tumor cells within the lungs. The majority of the inoculum (∼90%) was present in the lungs 30 minutes after intravenous inoculation, with only scant amounts present in the other organs evaluated, regardless of mouse genotype or antibody treatment. Twenty-six hours after injection, fXIII deficiency resulted in a significant diminution in the apparent number of tumor cells remaining in the lungs in mice with intact NK cells. However, in mice immunologically depleted of NK cells, fXIII ceased to be a determinant of early tumor cell survival. These analyses identify fXIII as a significant determinant of metastatic potential and indicate that at least one mechanism whereby fXIII increases metastatic success is by impeding NK cell-mediated clearance of tumor cells. Given that these findings closely parallel previous observations made in fibrinogen-deficient mice, an attractive but still unproven model is that fXIII-mediated stabilization of fibrin/platelet aggregates associated with newly-formed micrometastases increases tumor cell survival in large part by limiting NK cell function. These studies also suggest that therapeutic strategies directed at fXIII might be useful in limiting malignant disease.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 3079-3079
    Abstract: Inflammatory bowel disease has been recognized as an important risk factor in the development of colon cancer for decades. However, the factors that support the inflammatory response in this setting are not fully defined. Given that activation of the hemostatic system is a consistent feature of colitis, and previous studies have shown that hemostatic factors are key regulators of inflammation in other settings, we hypothesized that the platelet/fibrinogen axis plays an important role in inflammatory colitis. In order to explore whether fibrinogen interactions with leukocytes or platelets contribute to colitis, we challenged gene-targeted mice expressing mutant forms of fibrinogen lacking the binding motifs recognized by the leukocyte integrin αMβ2 (Fibγ390–396A mice) or the platelet integrin αIIbβ3 (FibγΔ5 mice) with dextran sodium sulfate (DSS). The severity of colonic injury and inflammation in Fibγ390–396A mice were dramatically diminished relative to control animals based on multiple histopathological criteria, including edema, erosion/ulceration, crypt loss, and inflammatory cell infiltration. DSS-challenged Fibγ390–396A mice also exhibited significantly diminished circulating levels of the inflammatory cytokine IL-6. These studies support the general conclusion that fibrinogen-αMβ2 interactions are an important determinant of inflammatory processes within the colon. Complementary studies of FibγΔ5 mice suggest that fibrinogen interaction with the platelet integrin αIIbβ3 is also a determinant of colitis-associated pathologies. Here, initial studies of FibγΔ5 mice revealed that the loss of platelet-fibrinogen interactions results in a significant, but counterintuitive, diminution in colitis-associated GI bleeding and weight loss. Fibrinogen-supported platelet deposition and activation are likely to contribute to inflammatory disease processes though multiple mechanisms, but a contribution of the many proinflammatory cytokines and chemokines present within the platelet secretome may be a significant factor. In addition to supporting colitis, hemostatic factors contribute to the more complex process of colitis-associated cancer (CAC) progression. In this regard, it is notable that Fibγ390–396A mice developed significantly fewer colonic adenomas relative to control mice when challenged with a combination of azoxymethane, a DNA alkylator, and DSS. Indeed, ~30% of Fibγ390–396A mice had no discernable adenomas, while penetrance was 100% in control mice. Furthermore, the tumors harvested from Fibγ390–396A mice were significantly smaller than those observed in wild-type mice, resulting in a profound diminution in total tumor burden. These results support the conclusion that the platelet/fibrinogen axis is a major determinant of inflammatory colitis and colitis-associated cancer progression, and therapies designed to disrupt inflammatory pathways at the level of hemostatic factors could be useful in the treatment or prevention of colitis or colitis-associated colon cancer.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 267-267
    Abstract: Thrombin is central in thrombus formation as both a positive mediator of thrombus formation through the proteolytic activation of PARs, fibrinogen, fXI and other prothrombotic substrates, and a negative modulator of the coagulation cascade through the activation of protein C. Detailed structure-function studies have revealed that thrombin can be redesigned to favor either procoagulant or anticoagulant substrates. The introduction of W215A/E217A substitutions in the murine thrombin active site (fIIWE) results in a pronounced “specificity switch” that reduces catalytic efficiency with fibrinogen by at least 3-orders-magnitude while only modestly reducing activity for protein C activation. To evaluate the effects of fIIWE activity in vivo, we have used a gene-targeting strategy to generate mice carrying the W215A/E217A mutations in the endogenous murine prothrombin gene. The mutant allele was transmitted through the germline and was found to support the expression of normal levels of hepatic fII mRNA and plasma fII in both heterozygous and homozygous neonates. Unlike fII knockout mice, homozygous fIIWE mice were observed at term with the expected Mendelian frequency. Nevertheless, homozygous fIIWE offspring uniformly succumbed to spontaneous bleeding events within days of birth. Heterozygous fIIWE/WT animals generally survived to adulthood, were capable of carrying multiple liters to term, and unchallenged mice displayed a hematological profile similar to wildtype mice. However, consistent with a predicted anticoagulant phenotype, adult fIIWE/WT heterozygotes exhibited significantly delayed thrombus formation following ferric chloride injury of mesenteric arterioles and extended bleeding times following tail tip excision relative to control mice expressing wildtype fII. Given that activated protein C has been shown to be efficacious in the treatment of sepsis, we explored whether the shift in thrombin specificity in heterozygous fIIWE/WT mice would confer the benefit of rendering animals tolerant to acute septic challenges. Kaplan-Meier analyses following intravenous administration of S. aureus revealed that fIIWE/WT mice exhibited a significant survival advantage over littermate wildtype animals challenged in parallel and tracked over a 7-day observation period. Notably, extended thrombus formation and bleeding times as well as resistance to sepsis was not simply a function of half normal wildtype fII expression. When these analyses were performed in animals carrying one wildtype allele and one null mutation allele, results were similar to wiltype. These studies further underscore the interplay between the hemostatic and inflammatory systems in vivo and highlight the possible therapeutic utility of recombinant (pro)thrombin derivatives with selected alterations in substrate specificity.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: The Journal of Neuroscience, Society for Neuroscience, Vol. 37, No. 14 ( 2017-04-05), p. 3776-3788
    Abstract: Multiple sclerosis (MS) is a neuroinflammatory, demyelinating disease of the CNS. Fibrinogen deposition at sites of blood–brain barrier breakdown is a prominent feature of neuroinflammatory disease and contributes to disease severity. Plasminogen, the primary fibrinolytic enzyme, also modifies inflammatory processes. We used a murine model of MS, experimental autoimmune encephalomyelitis (EAE), to evaluate the hypothesis that the loss of plasminogen would exacerbate neuroinflammatory disease. However, contrary to initial expectations, EAE-challenged plasminogen-deficient (Plg − ) mice developed significantly delayed disease onset and reduced disease severity compared with wild-type (Plg + ) mice. Similarly, pharmacologic inhibition of plasmin activation with tranexamic acid also delayed disease onset. The T-cell response to immunization was similar between genotypes, suggesting that the contribution of plasminogen was downstream of the T-cell response. Spinal cords from EAE-challenged Plg − mice demonstrated significantly decreased demyelination and microglial/macrophage accumulation compared with Plg + mice. Although fibrinogen-deficient mice or mice with combined deficiencies of plasminogen and fibrinogen had decreased EAE severity, they did not exhibit the delay in EAE disease onset, as seen in mice with plasminogen deficiency alone. Together, these data suggest that plasminogen and plasmin-mediated fibrinolysis is a key modifier of the onset of neuroinflammatory demyelination. SIGNIFICANCE STATEMENT Multiple sclerosis is a severe, chronic, demyelinating disease. Understanding the pathobiology related to the autoreactive T-cell and microglial/macrophage demyelinating response is critical to effectively target therapeutics. We describe for the first time that deficiency of plasminogen, the key fibrinolytic enzyme, delays disease onset and protects from the development of the paralysis associated with a murine model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Administration of a widely used, pharmacologic inhibitor of plasminogen activation, tranexamic acid, also delays the onset of neuroinflammation associated with EAE.
    Type of Medium: Online Resource
    ISSN: 0270-6474 , 1529-2401
    Language: English
    Publisher: Society for Neuroscience
    Publication Date: 2017
    detail.hit.zdb_id: 1475274-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 7 ( 2010-04-01), p. 2634-2643
    Abstract: A link between colitis and colon cancer is well established, but the mechanisms regulating inflammation in this context are not fully defined. Given substantial evidence that hemostatic system components are powerful modulators of both inflammation and tumor progression, we used gene-targeted mice to directly test the hypothesis that the coagulation factor fibrinogen contributes to colitis-associated colon cancer in mice. This fundamental provisional matrix protein was found to be an important determinant of colon cancer. Fibrinogen deficiency resulted in a dramatic diminution in the number of colonic adenomas formed following azoxymethane/dextran sodium sulfate challenge. More detailed analyses in mice expressing a mutant form of fibrinogen that retains clotting function, but lacks the leukocyte integrin receptor αMβ2 binding motif (Fibγ390-396A), revealed that αMβ2-mediated engagement of fibrin(ogen) is mechanistically coupled to local inflammatory processes (e.g., interleukin-6 elaboration) and epithelial alterations that contribute to adenoma formation. Consistent with these findings, the majority of Fibγ390-396A mice developed no discernable adenomas, whereas penetrance was 100% in controls. Furthermore, the adenomas harvested from Fibγ390-396A mice were significantly smaller than those from control mice and less proliferative based on quantitative analyses of mitotic indices, suggesting an additional role for fibrin(ogen) in the growth of established adenomas. These studies show, for the first time, a unique link between fibrin(ogen) and the development of inflammation-driven malignancy. Given the importance of antecedent inflammation in the progression of numerous cancers, these studies suggest that therapies targeting fibrin(ogen)-αMβ2 interactions may be useful in preventing and/or treating this important subset of malignancies. Cancer Res; 70(7); 2634–43
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 496-496
    Abstract: Abstract 496 Thrombin-mediated proteolysis is the central event not only of hemostasis and thrombosis but also in distinct physiological and pathological contexts such as development, inflammation, cancer biology, and cardiovascular disease. The proteolytic conversion of prothrombin to α-thrombin (fIIa) by the prothrombinase complex occurs through two possible pathways: i) the inactive intermediate termed prethrombin 2, or ii) the proteolytically active two-chain intermediate termed meizothrombin (fIIaMZ). fIIaMZ, unlike fIIa, retains the γ-carboxyglutamic acid-rich gla domain and two kringle domains of prothrombin and has distinct catalytic properties relative to fIIa that could be biologically meaningful in vivo, including a diminished capacity to cleave fibrinogen and a significantly increased capacity to activate protein C in the presence of thrombomodulin. Here, the endogenous prothrombin gene was modified in mice to better explore the properties of fIIaMZin vivo and to test the hypotheses that mice carrying a mutant form of prothrombin with a terminal activation product of fIIaMZ will: 1) develop to term and survive to adulthood despite an altered hemostatic profile; and 2) exhibit significantly dampened inflammatory responses due to enhanced protein C activation. In order to limit cleavage events to the single site yielding fIIaMZ, alterations were introduced into the endogenous prothrombin gene resulting in three amino acid substitutions (R157A, R268A, and K281A) located at the P1 positions of potential fXa or autocatalytic cleavage sites. Unlike prothrombin-null mice, mice homozygous for these mutations (hereafter referred to as fIIMZ mice) were found to be present at weaning age and viable into adulthood. Furthermore, unlike mice with a major deficit in either clotting function (e.g., fibrinogen-null) or platelet function (e.g., Gαq-null), fIIMZ females were capable of successfully carrying a litter to term. However, analysis of over 700 progeny generated from crosses of heterozygous fIIMZ/WT mice revealed that only about half of the number homozygous fIIMZ mice expected, based on Mendelian transmission rates, were observed in weaning-age offspring. Complementary studies suggest that the fraction of fIIMZ offspring that fail are lost primarily in utero, but occasional post-partum failures were observed associated with hemorrhagic events. Successful adult fIIMZ mice were found to have similar prothrombin mRNA levels and equivalent fII protein expression to fIIWT mice. Furthermore, fIIMZ animals exhibited normal complete blood cell counts and predictably normal thrombin times, but prolonged PTs and aPTTs. Thrombin generation assays revealed a prolonged time to peak thrombin production, but no significant difference in peak thrombin levels. Consistent with these findings, tail bleeding times in fIIMZ mice were significantly prolonged. None of the fIIMZ mice assayed achieved hemostasis within the 10 minute observation window, whereas fIIWT mice all stopped within 2 minutes of challenge. More sophisticated comparative studies of thrombus formation in mesenteric arterioles following FeCl3 injury using a real-time intravital microscopy approach established that the time to first thrombus formation in fIIMZ mice was almost twice that of wildtype animals. The majority of the fIIMZ mice failed to occlude the injured vessel within a 25 min observation window, whereas all fIIWT mice successfully formed occlusive thrombi with a mean time of 12.5 minutes. In summary, site-directed alterations of the endogenous prothrombin gene in mice leading to a terminal prothrombin activation product of meizothrombin were found to be compatible with development, growth to adulthood and reproductive success, albeit with modified hemostatic function. Based on the catalytic properties of fIIaMZ favoring protein C activation, studies are underway to compare APC generation in control and fIIMZ mice and explore the theory that physiological and pathological inflammatory responses will be dampened in fIIMZ mice. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2018
    In:  Blood Vol. 132, No. Supplement 1 ( 2018-11-29), p. 136-136
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 136-136
    Abstract: Mounting evidence ties both fibrin(ogen) and plasmin(ogen) to inflammatory diseases. Indeed, both fibrin(ogen) and plasmin(ogen) have been linked to critical macrophage functions in multiple disease processes. Migration of macrophages to sites of sterile inflammation is, at least partially, dependent on plasmin(ogen). Mice lacking plasminogen, when challenged with sterile thioglycollate-induced peritonitis, have both diminished overall leukocyte migration and decreased macrophage migration. Additionally, macrophage migration defects have been identified in both mice lacking plasminogen and plasminogen receptors. Plasmin has many targets that may play a role in supporting macrophage migration. In addition to proteolysis of fibrin(ogen), plasmin activates matrix metalloprotease (MMP) 2 and MMP9 and cleaves collagen and laminin. Indeed, mice that lack MMP9 have a migration defect similar to mice that lack plasminogen, suggesting that MMP9 is a biologically relevant proteolytic target in this context. To further examine the targets of plasmin that regulate macrophage migration, we challenged animals that have individual and combined genetic deficiencies in fibrinogen and plasminogen with thioglycollate-induced peritonitis. We have found that mice that lack fibrinogen alone have a significantly increased migration of macrophages to the peritoneal cavity. Mice that lack plasminogen alone demonstrated the expected diminution in macrophage migration to the peritoneal cavity. However, mice that were deficient in both plasminogen and fibrinogen demonstrated macrophage migration that was indistinguishable from wildtype. These data suggest that fibrin(ogen) impedes macrophage migration to the peritoneal cavity. To further confirm this mechanism, we examined macrophage migration in a transwell assay in vitro. Here, a macrophage cell line (RAW 264.7 or BMDM) migration was examined in the absence and presence of fibrin matrices. Macrophages, in the presence of plasminogen, did demonstrate a modest, but statistically significant, increase in migration across the transwell membrane in the absence of fibrinogen. When a fibrin matrix was generated on the transwell membrane, macrophages were essentially unable to cross in the absence of plasminogen. These data further support the concept that macrophages require plasmin(ogen) to cross fibrin matrices. We further sought to determine if the fibrin-αMβ2 interaction was implicated in the macrophage migration phenotype. To do this, we first examined macrophage migration in vivo in mice expressing a form of fibrinogen that cannot interact with αMβ2, Fibγ390-396A mice. Similar to mice lacking fibrinogen, an increase in peritoneal macrophages was observed at 72 hours following a challenge with 4% thioglycollate. To confirm that this was related to the fibrin-αMβ2 interaction, and not due to abnormal factor XIII crosslinking, factor XIII deficient animals were also challenged with thioglycollate induced peritonitis. Mice lacking factor XIII exhibited no difference from wildtype in this model of peritonitis. We further confirmed in the in vitro transwell migration assay that macrophages were able to cross a fibrin barrier, derived from Fibγ390-396A mice, in the absence of plasminogen. Taken together, these data suggest that plasmin allows macrophage migration via liberation from the fibrin-αMβ2 interaction. Disclosures Mullins: Shire: Honoraria, Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...