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
    Online Resource
    Online Resource
    American Society of Hematology ; 1998
    In:  Blood Vol. 92, No. 2 ( 1998-07-15), p. 488-495
    In: Blood, American Society of Hematology, Vol. 92, No. 2 ( 1998-07-15), p. 488-495
    Abstract: We have performed a systematic in vivo evaluation of gene expression for the glycoprotein (GP) Ibα subunit of the murine platelet adhesion receptor, GP Ib-IX-V. This study is warranted by in vitro observations of human GP Ibα expression in cells of nonhematopoietic lineage and reports of regulation of the GP Ibα gene by cytokines. However, an in vivo role for a GP Ib-IX-V receptor has not been established beyond that described for normal megakaryocyte/platelet physiology and hemostasis. Our Northern analysis of mouse organs showed high levels of GP Ibα mRNA in bone marrow with a similar expression pattern recapitulated in mice containing a luciferase transgene under the control of the murine GP Ibα promoter. Consistently high levels of luciferase activity were observed in the two hematopoietic organs of mice, bone marrow (1,400 relative light units/μg of protein [RLUs]) and spleen (500 RLUs). Reproducible, but low-levels of luciferase activity were observed in heart, aorta, and lung (30 to 60 RLUs). Among circulating blood cells, the luciferase activity was exclusively localized in platelets. No increase in GP Ibα mRNA or luciferase activity was observed after treatment of mice with lipopolysaccharides (LPS) or tumor necrosis factor-α (TNF-α). We conclude the murine GP Ibα promoter supports a high level of gene expression in megakaryocytes and can express heterologous proteins allowing an in vivo manipulation of platelet-specific proteins in the unique environment of a blood platelet.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 1998
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2005
    In:  Blood Vol. 106, No. 11 ( 2005-11-16), p. 737-737
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 737-737
    Abstract: Insights into the regulation of platelet adhesion have historically been aided by congenital bleeding disorders. Two clinically similar disorders, distinct at the molecular level, are platelet-type von Willebrand disease (Pt-vWD) and type 2B von Willebrand disease (vWD). In each case, the receptor (Pt-vWD) or ligand (type 2B vWD) has undergone a structural change supporting the binding of soluble von Willebrand factor (vWF) to platelets. The net result is an increased affinity between circulating vWF and the platelet glycoprotein (GP) Ib-IX receptor leading to thrombocytopenia and the risk of a bleeding event. In the case of Pt-vWD several missense mutations within the glycoprotein (GP) Ibα subunit of the GP Ib-IX complex have been described as the cause of Pt-vWD. The first described genetic mutation produces a Gly233 to Val233 (G233V) substitution within the GP Ibα amino-terminus. Crystallographic studies of isolated recombinant domains of GP Ibα have documented structural perturbations within the vWF binding site as a consequence of the G233V substitution. The hypothesis exists that these structural changes lead to an increased affinity between GP Ib-IX and vWF. We report the generation of mice expressing a human GP Ibα subunit with a G233V substitution. The animals were generated by traditional transgenic techniques using the human GP Ibα promoter sequence. Human Pt-vWD is autosomally dominant where both wild-type GP Ibα and Pt-vWD variant alleles are present. Mice expressing the G233V transgene were bred to mice deficient in mouse GPIbα producing a number of different genotypes for characterization. Mouse platelets were characterized from animals with (i) wild-type mouse GP Ibα alleles (mGp1b+/+); (ii) devoid of murine GP Ibα but expressing a wild-type human GPIbα allele (mGp1b−/−;hGp1b+); (iii) a single mouse GPIbα wild-type allele and the human G233V variant (mG1b+/−;hGp1bG233V); and (iv) devoid of murine GP Ibα and expressing the human G233V variant (mGp1b−/−;hGp1bG233V). In vivo, the effect of the Pt-vWD G233V human variant produces a mouse thrombocytopenia with increased bleeding as determined in tail bleeding time assays. The most severe phenotype was observed in mice with the mGp1b−/−;hGp1bG233V genotype. One of the diagnostic characteristics of human Pt-vWD is the agglutination of platelets with subthreshold levels of the agonist, ristocetin. Platelet-rich plasma from mice expressing the G233V variant (mGp1b-/-;hGp1bG233V) aggregate with ristocetin (0.25 mg/ml) unlike their counterparts expressing a wild-type human GP Ibα sequence (mGp1b−/−;hGp1b+) that require at least 0.8 mg/ml ristocetin. To observe the ristocetin effect required supplementing mouse platelet-rich plasma with purified human vWF illustrating the species-specificity of ristocetin for human vWF. The characterization of this model documents an animal with hyperfunctional platelets owing to an increased vWF/GP Ib-IX interaction. The generation of a Pt-vWD murine model provides an in vivo setting to examine the role of platelets with increased adhesive potential in diseases where the platelet is gaining recognition as important mediators of inflammatory responses, tumor metastases and angiogenesis.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 104, No. 8 ( 2004-10-15), p. 2339-2344
    Abstract: Here we report the characterization of a mouse model of the Bernard-Soulier syndrome generated by a targeted disruption of the gene encoding the glycoprotein (GP) Ibβ subunit of the GP Ib-IX complex. Similar to a Bernard-Soulier model generated by disruption of the mouse GP Ibα subunit, GP IbβNull mice display macrothrombocytopenia and a severe bleeding phenotype. When examined by transmission electron microscopy, the large platelets produced by a GP IbβNull genotype revealed α-granules with increased size as compared with the α-granules from control mouse platelets. Data are presented linking the overexpression of a septin protein, SEPT5, to the presence of larger α-granules in the GP IbβNull platelet. The SEPT5 gene resides approximately 250 nucleotides 5′ to the GP Ibβ gene and has been associated with modulating exocytosis from neurons and platelets as part of a presynaptic protein complex. Fusion mRNA transcripts present in megakaryocytes can contain both the SEPT5 and GP Ibβ coding sequences as a result in an imperfect polyadenylation signal within the 3′ end of both the human and mouse SEPT5 genes. We observed a 2- to 3-fold increase in SEPT5 protein levels in platelets from GP IbβNull mice. These results implicate SEPT5 levels in the maintenance of normal α-granule size and may explain the variant granules associated with human GP Ibβ mutations and the Bernard-Soulier syndrome.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 114, No. 27 ( 2009-12-24), p. 5541-5546
    Abstract: Platelet-type von Willebrand disease (PT-VWD) is a bleeding disorder of the platelet glycoprotein Ib-IX/von Willebrand factor (VWF) axis caused by mutations in the glycoprotein Ib-IX receptor that lead to an increased affinity with VWF. In this report, platelets from a mouse expressing a mutation associated with PT-VWD have been visualized using state-of-the art image collection and processing. Confocal analysis revealed that VWF bound to the surface of single platelets and bridging micro-aggregates of platelets. Surface-bound VWF appears as a large, linear structure on the surface of 50% of the PT-VWD platelets. In vivo thrombus formation after chemical injury to the carotid artery revealed a severe impairment to occlusion as a consequence of the PT-VWD mutation. In vitro stimulation of PT-VWD platelets with adenosine diphosphate or thrombin demonstrates a significant block in their ability to bind fibrinogen. The impairment of in vivo thrombus formation and in vitro fibrinogen binding are more significant than might be expected from the observed platelet binding to VWF polymers over a small portion of the plasma membrane. Visualization of the receptor/ligand interaction and characterization of a severe antithrombotic phenotype provide a new understanding on the molecular basis of bleeding associated with the PT-VWD phenotype.
    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 ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2002
    In:  Blood Vol. 100, No. 6 ( 2002-09-15), p. 2102-2107
    In: Blood, American Society of Hematology, Vol. 100, No. 6 ( 2002-09-15), p. 2102-2107
    Abstract: An absent platelet glycoprotein (GP) Ib-IX receptor results in the Bernard-Soulier syndrome and is characterized by severe bleeding and the laboratory presentation of macrothrombocytopenia. Although the macrothrombocytopenic phenotype is directly linked to an absent GP Ib-IX complex, the disrupted molecular mechanisms that produce the macrothrombocytopenia are unknown. We have utilized a mouse model of the Bernard-Soulier syndrome to engineer platelets expressing an α-subunit of GP Ib (GP Ibα) in which most of the extracytoplasmic sequence has been replaced by an isolated domain of the α-subunit of the human interleukin-4 receptor (IL-4Rα). The IL-4Rα/GP Ibα fusion is membrane expressed in Chinese hamster ovary (CHO) cells, and its expression is facilitated by the presence of human GP IX and the β-subunit of GP Ib. Transgenic animals expressing a chimeric receptor were generated and bred into the murine Bernard-Soulier syndrome–producing animals devoid of mouse GP Ibα but expressing the IL-4Rα/GP Ibα fusion sequence. The characterization of these mice revealed a 2-fold increase in circulating platelet count and a 50% reduction in platelet size when compared with platelets from the mouse model of the Bernard-Soulier syndrome. Immunoprecipitation confirmed that the IL-4Rα/GP Ibα subunit interacts with filamin-1 and 14-3-3ζ, known binding proteins to the GP Ibα cytoplasmic tail. Mice expressing the chimeric receptor retain a severe bleeding phenotype, confirming a critical role for the GP Ibα extracytoplasmic domain in hemostasis. These results provide in vivo insights into the structural elements of the GP Ibα subunit that contribute to normal megakaryocyte maturation and thrombopoiesis.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2002
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Society of Hematology ; 2004
    In:  Blood Vol. 104, No. 11 ( 2004-11-16), p. 3529-3529
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 3529-3529
    Abstract: Septins form a cytosolic protein family gaining recognition as important effectors in molecular mechanisms involving membrane rearrangements, such as cytokinesis and exocytosis. So far, twelve different members of the family (named SEPT1 to SEPT12) have been characterized in mammals, and their roles are not limited to those mentioned above, but embrace a large number of physiological and pathophysiological events. In particular, reports have associated septins with cancer, apoptosis and neurodegenerative diseases. In this study, we further characterize the platelet septin, SEPT5 (formerly CDCrel-1) as a regulator of platelet physiology. SEPT5 is highly expressed in megakaryocytes, heart and brain and is directly linked to secretion mechanisms in both platelets and neurons. In neurons, SEPT5 negatively modulates neurotransmitter release and has recently been implicated in mechanisms leading to development of schizophrenia in rat models. In platelets, we and others have shown that SEPT5 is preferentially located around α-granules and associates with syntaxin 4 and other members of the septin protein family (SEPT4 and SEPT8) to form a cytoplasmic macromolecular complex. Work from other laboratories has linked SEPT5 to different members of the SNARE family such as syntaxin 1A, the sec6/8 complex, and SNAP25. We have found the overexpression of mouse SEPT5 is associated with fewer α-granules but those present are larger in size suggesting levels of Sept5 maintain normal α-granule size. In a murine model of SEPT5 deficiency, platelets display an increased sensitivity to platelet agonists such as collagen and the thromboxane analogue U46619. To date, SEPT5 is the only mammalian septin whose relevance has been tested in a mouse knockout model. Our current studies use a lumi-aggregometer to document the active secretion of ATP in SEPT5 deficient platelets. Platelets lacking SEPT5 are more responsive to submaximal concentrations of acid insoluble fibrillar type I collagen ( 〈 2.5 mg/mL). We observe a 2- to 3-fold increase in ATP secretion in deficient platelets as compared to their wild-type littermates. The increased dense granule release in the absence of SEPT5 also coincides with increased platelet aggregation. A maximum release of ATP was determined in the presence of the calcium ionophore (A23187, 40μM). Higher concentrations of collagen (5 μg/ml) produced a maximum level of ATP release in SEPT5 deficient mice unlike their wild-type littermates. Our current hypothesis places SEPT5 as a scaffolding protein that serves to anchor other proteins into higher-ordered molecular complexes. SEPT5 biology in platelets needs to be further studied to fully understand its role in granule secretion and formation. While important for platelet biology, these studies may also have implications for mechanisms associated with neurotransmitter release. As such, the platelet becomes an excellent model for analyzing SEPT5 function with implications for hemostasis and beyond.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Proceedings of the National Academy of Sciences ; 2000
    In:  Proceedings of the National Academy of Sciences Vol. 97, No. 6 ( 2000-03-14), p. 2803-2808
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 97, No. 6 ( 2000-03-14), p. 2803-2808
    Abstract: The human Bernard-Soulier syndrome is an autosomal recessive disorder of platelet dysfunction presenting with mild thrombocytopenia, circulating “giant” platelets and a bleeding phenotype. The bleeding in patients with the Bernard-Soulier syndrome is disproportionately more severe than suggested by the reduced platelet count and is explained by a defect in primary hemostasis owing to the absence of the platelet glycoprotein (GP) Ib-IX-V membrane receptor. However, the molecular basis for the giant platelet phenotype and thrombocytopenia have remained unresolved but assumed to be linked to an absent receptor complex. We have disrupted the gene encoding the α-subunit of mouse GP Ib-IX-V (GP Ibα) and describe a murine model recapitulating the hallmark characteristics of the human Bernard-Soulier syndrome. The results demonstrate a direct link between expression of a GP Ib-IX-V complex and normal megakaryocytopoiesis and platelet morphogenesis. Moreover, using transgenic technology the murine Bernard-Soulier phenotype was rescued by expression of a human GP Ibα subunit on the surface of circulating mouse platelets. Thus, an in vivo model is defined for analysis of the human GP Ib-IX-V receptor and its role in the processes performed exclusively by megakaryocytes and platelets.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2000
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 102, No. 5 ( 2003-09-01), p. 1701-1707
    Abstract: Platelet interaction with exposed adhesive ligands at sites of vascular injury is required to initiate a normal hemostatic response and may become a pathogenic factor in arterial diseases leading to thrombosis. We report a targeted disruption in a key receptor for collagen-induced platelet activation, glycoprotein (GP) VI. The breeding of mice with heterozygous GP VI alleles produced the expected frequency of wild-type, heterozygous, and homozygous genotypes, indicating that these animals had no reproductive problems and normal viability. GP VInull platelets failed to aggregate in response to type I fibrillar collagen or convulxin, a snake venom protein and known platelet agonist of GP VI. Nevertheless, tail bleeding time measurements revealed no severe bleeding tendency as a consequence of GP VI deficiency. Ex vivo platelet thrombus formation on type I collagen fibrils was abolished using blood from either GP VInull or FcR-γnull animals. Reflection interference contrast microscopy revealed that the lack of thrombus formation by GP VInull platelets could be linked to a defective platelet activation following normal initial tethering to the surface, visualized as lack of spreading and less stable adhesion. These results illustrate the role of GP VI in postadhesion events leading to the development of platelet thrombi on collagen fibrils.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2003
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 1463-1463
    Abstract: Successful tumor formation and metastatic spread depend on the ability of tumor cells to interact with their host microenvironment. Studies from our groups and others indicate that platelets contribute to tumor host interactions through adhesive and stimulatory functions that may promote tumor growth and dissemination. Here we show that platelet receptor GPIbα plays a critical role in the ability of platelets to support primary tumor development and hematogenous metastasis. We previously developed knockout mice lacking platelet GP Ibα (GPIbαNull, a model of the human Bernard-Soulier syndrome) and mice missing GPIbα, but expressing a fusion protein of cytoplasmic GP Ibα and the extracytoplasmic domain of the interleukin-4 receptor (Ibα/IL-4R). Expression of the Ibα/IL-4R ameliorates macrothrombocytopenia, a characteristic of the Bernard-Soulier syndrome. However, both models result in a severe hemorrhagic state owing to the absence of GPIbα. Congenic mouse strains of both GP Ibα models were generated by 10-generation backcrosses with wild-type (WT) C57BL/6J mice to study the tumorgenic and metastatic activity of syngeneic tumor cells. To analyze primary tumor growth, Lewis lung carcinoma cells (D121) were injected intradermally in the dorsal flank of the hind limb (105 cells/injection) and tumor development analyzed in its initial and advanced stages. Seventeen days after injection, tumors in GPIbαNull and Ibα/IL-4R mice were several-fold smaller than those in WT C57BL/6J mice. Histological examination of early and end-stage tumors revealed three major differences: a severe and immediate onset of tumor necrosis, a lack of new blood vessels large enough to connect to the circulation, and a significant reduction in tumor-associated macrophages. While the proliferation rate of tumor cells in non-necrotic areas of the tumors was unaffected by the lack of GPIbα, the percentage of necrosis in these tumors was 6-fold increased in GPIbαNull mice. Each necrotic area showed increased hemorrhage in the absence of GPIbα. The overall density of microvessels in early primary tumors appeared unchanged, but the number of larger vessels associated with tumors was reduced ~2-fold and the density of tumor-associated macrophages was reduced ~6-fold compared to tumors developing in WT mice. To analyze a contribution of platelet GPIbα to hematogenous tumor metastasis, another syngeneic tumor cell model was used and B16F10.1 melanoma cells were injected into the tail vein (105 cells/mouse) of GPIbαNull, Ibα/IL-4R, or WT mice, to determine the ability of tumor cells to colonize the lungs 14 days later. In the absence of functional GPIbα, the number of metastatic foci at the surface of the lungs was significantly reduced (15-fold, p = 0.0002). These results indicate that successful target organ colonization by tumor cells from the blood stream is severely affected by the lack of extracytoplasmic GPIbα in Ibα/IL-4R animals, and not necessarily by the reduced number of platelets in GPIbαNull animals. The results demonstrate a significant involvement of platelet GP Ibα in primary tumor growth by affecting tumor angiogenesis and the recruitment of tumor-associated macrophages while providing evidence GPIbα directly contributes to target organ colonization by circulating tumor cells. These studies identify platelet GP Ibα as a potential new target for anti cancer therapy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    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 ; 1998
    In:  Blood Vol. 92, No. 2 ( 1998-07-15), p. 488-495
    In: Blood, American Society of Hematology, Vol. 92, No. 2 ( 1998-07-15), p. 488-495
    Abstract: We have performed a systematic in vivo evaluation of gene expression for the glycoprotein (GP) Ibα subunit of the murine platelet adhesion receptor, GP Ib-IX-V. This study is warranted by in vitro observations of human GP Ibα expression in cells of nonhematopoietic lineage and reports of regulation of the GP Ibα gene by cytokines. However, an in vivo role for a GP Ib-IX-V receptor has not been established beyond that described for normal megakaryocyte/platelet physiology and hemostasis. Our Northern analysis of mouse organs showed high levels of GP Ibα mRNA in bone marrow with a similar expression pattern recapitulated in mice containing a luciferase transgene under the control of the murine GP Ibα promoter. Consistently high levels of luciferase activity were observed in the two hematopoietic organs of mice, bone marrow (1,400 relative light units/μg of protein [RLUs]) and spleen (500 RLUs). Reproducible, but low-levels of luciferase activity were observed in heart, aorta, and lung (30 to 60 RLUs). Among circulating blood cells, the luciferase activity was exclusively localized in platelets. No increase in GP Ibα mRNA or luciferase activity was observed after treatment of mice with lipopolysaccharides (LPS) or tumor necrosis factor-α (TNF-α). We conclude the murine GP Ibα promoter supports a high level of gene expression in megakaryocytes and can express heterologous proteins allowing an in vivo manipulation of platelet-specific proteins in the unique environment of a blood platelet.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 1998
    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...