Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
Type of Medium
Language
  • 1
    In: Fertility and Sterility, Elsevier BV, Vol. 110, No. 2 ( 2018-07), p. 185-324.e5
    Type of Medium: Online Resource
    ISSN: 0015-0282
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2018
    detail.hit.zdb_id: 1500469-7
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 2527-2527
    Abstract: Abstract 2527 May-Hegglin Anomaly was originally defined as patients with macrothrombocytopenia. It is now recognized that the phenotypes of May-Hegglin Anomaly and the related syndromes described by Epstein, Fechtner and Sebastian overlap comprising macrothrombocytopenia, neutrophil inclusions, deafness, cataract formation and nephritis. These disorders are defined by mutations in the non-muscle myosin heavy chain IIA gene, MYH9 and have been termed MYH9-Related Disorders. Myh9 is required during murine embryogenesis as Myh9-null embryos die of patterning defects. Megakaryocyte-specific deletion of Myh9 has been performed by the Gachet group and Shivdasani and colleagues examined megakaryocyte function in Myh9-deficient embryonic stem cells induced to differentiate in vitro. The phenotypes of these mice include increased bleeding times, absence of clot retraction and defective Integrin β3 phosphorylation coupling to impaired activation of downstream Rho-Rock signalling and lamellipodia formation. To date, no knock-in MYH9 alleles corresponding to MYH9-related Disorder mutations have been reported. MYH9-related Disorders frequently are misdiagnosed as Chronic Immune Thrombocytopenia, so having enhanced knowledge of the molecular etiology of mutant MYH9 alleles is a priority. Our group has been interested in utilizing random mutagenesis to identify novel alleles of human hematopoietic disorders. We performed a dominant screen in which 129 male mice were mutagenized with ENU and back-crossing was performed on the C57BL/6 background. We identified a heritable line (7238) with macrothrombocytopenia that mapped to a 4.25 Mb region on mouse Chromosome 15. Due to our inability to further refine the interval through back-crossing, we performed high throughput sequencing by comparing SNPs between 129, C57BL/6, 7238/+ and 7238/7238 mice. We identified 18 novel variations, including 1 that corresponded to a coding change - Myh9 Q1443L. Subsequent Sanger sequencing and back-crossing eliminated the remaining 17 variations, confirming our assignment of the 7238 phenotype to a mutation of Myh9. The Myh9 Q1443L allele is within proximity of the MYH9 D1424Y/N/H mutations found within clinical specimens. Myh9 Q1443L/Q1443L mice are viable allowing us to examine the effect of mutation of one or both alleles of Myh9. Myh9Q1443L/Q1443L mice had increased bleeding times, whereas Myh9Q1443L/+ mice were identical to wild type mice. Platelet aggregation experiments were performed after ADP or thrombin stimulation. Decreased aggregation was observed in a dosage-dependent manner, with Myh9Q1443L/+ mice displaying intermediate levels when compared with Myh9Q1443L/Q1443L co-hort. Adherence and aggregation of Myh9Q1443L/Q1443L platelets to a collagen matrix was decreased at shear rates of 350/s and 1800/s. Staining of Myh9Q1443L/+ and Myh9Q1443L/Q1443L neutrophils also revealed neutrophil inclusions. In addition to the hematopoietic phenotypes, we have observed increased cataract formation with a frequency of 36% in wild type animals, 60% in Myh9Q1443L/+ and 92% in Myh9Q1443L/Q1443L mice. Sporadic hematuria and proteinuria is found in Myh9Q1443L mice. We are currently performing albumen loading and lipopolysaccharide activation experiments to determine whether 7238 mice have altered response to nephric stress. We are crossing 7238 mice to the DBA genetic background, which is more sensitive to subtle kidney phenotypes. We performed auditory brain response measurements on aged wild type, Myh9Q1443L/+ and Myh9Q1443L/Q1443L mice and observed no differences in frequency responses ranging from 2 to 24kHz. Myh9Q1443L/+ mice displayed hypertrophy of the preputial gland whereas Myh9Q1443L/Q1443L males showed atrophy of this gland and Myh9Q1443L/Q1443L male mice have reduced fertility. No differences in fertility have been described to date in MYH9 related disorders. In conclusion, Myh9Q1443L/Q1443L mice display macrothrombocytopenia, neutrophil inclusions and cataract formation described in MYH9-related disorders and represent the first animal model of May-Hegglin Anomaly. 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: 2010
    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 ...
  • 3
    Online Resource
    Online Resource
    American Association for the Advancement of Science (AAAS) ; 2019
    In:  Science Signaling Vol. 12, No. 598 ( 2019-09-10)
    In: Science Signaling, American Association for the Advancement of Science (AAAS), Vol. 12, No. 598 ( 2019-09-10)
    Abstract: Inflammation alters bone marrow hematopoiesis to favor the production of innate immune effector cells at the expense of lymphoid cells and erythrocytes. Furthermore, proinflammatory cytokines inhibit steady-state erythropoiesis, which leads to the development of anemia in diseases with chronic inflammation. Acute anemia or hypoxic stress induces stress erythropoiesis, which generates a wave of new erythrocytes to maintain erythroid homeostasis until steady-state erythropoiesis can resume. Although hypoxia-dependent signaling is a key component of stress erythropoiesis, we found that inflammation also induced stress erythropoiesis in the absence of hypoxia. Using a mouse model of sterile inflammation, we demonstrated that signaling through Toll-like receptors (TLRs) paradoxically increased the phagocytosis of erythrocytes (erythrophagocytosis) by macrophages in the spleen, which enabled expression of the heme-responsive gene encoding the transcription factor SPI-C. Increased amounts of SPI-C coupled with TLR signaling promoted the expression of Gdf15 and Bmp4 , both of which encode ligands that initiate the expansion of stress erythroid progenitors (SEPs) in the spleen. Furthermore, despite their inhibition of steady-state erythropoiesis in the bone marrow, the proinflammatory cytokines TNF-α and IL-1β promoted the expansion and differentiation of SEPs in the spleen. These data suggest that inflammatory signals induce stress erythropoiesis to maintain erythroid homeostasis when inflammation inhibits steady-state erythropoiesis.
    Type of Medium: Online Resource
    ISSN: 1945-0877 , 1937-9145
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2019
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 2007
    In:  Blood Vol. 109, No. 10 ( 2007-05-15), p. 4494-4502
    In: Blood, American Society of Hematology, Vol. 109, No. 10 ( 2007-05-15), p. 4494-4502
    Abstract: The erythroid response to acute anemia relies on the rapid expansion in the spleen of a specialized population of erythroid progenitors termed stress BFU-E. This expansion requires BMP4/Madh5-dependent signaling in vivo; however, in vitro, BMP4 alone cannot recapitulate the expansion of stress BFU-E observed in vivo, which suggests that other signals are required. In this report we show that mutation of the Kit receptor results in a severe defect in the expansion of stress BFU-E, indicating a role for the Kit/SCF signaling pathway in stress erythropoiesis. In vitro analysis showed that BMP4 and SCF are necessary for the expansion of stress BFU-E, but only when spleen cells were cultured in BMP4 + SCF at low-oxygen concentrations did we recapitulate the expansion of stress BFU-E observed in vivo. Culturing spleen cells in BMP4, SCF under hypoxic conditions resulted in the preferential expansion of erythroid progenitors characterized by the expression of Kit, CD71, and TER119. This expression pattern is also seen in stress erythroid progenitors isolated from patients with sickle cell anemia and patients with β-thalassemia. Taken together these data demonstrate that SCF and hypoxia synergize with BMP4 to promote the expansion and differentiation of stress BFU-E during the recovery from acute anemia.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Hematology & Oncology, Springer Science and Business Media LLC, Vol. 12, No. 1 ( 2019-12)
    Type of Medium: Online Resource
    ISSN: 1756-8722
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 2429631-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal of Leukocyte Biology, Oxford University Press (OUP), Vol. 111, No. 2 ( 2022-01-28), p. 427-437
    Abstract: The prognosis for acute myeloid leukemia (AML) relapse post allogeneic hematopoietic stem cell transplantation (alloSCT) is dismal. Novel effective treatment is urgently needed. Clinical benefit of alloSCT greatly relies on the graft-versus-leukemia (GVL) effect. The mechanisms that mediate immune escape of leukemia (thus causing GVL failure) remain poorly understood. Studies of human GVL have been hindered by the lack of optimal clinically relevant models. Here, using our large, longitudinal clinical tissue bank that include AML cells and G-CSF mobilized donor hematopoietic stem cells (HSCs), we successfully established a novel GVL model in humanized mice. Donor HSCs were injected into immune-deficient NOD-Cg-PrkdcscidIL2rgtm1Wjl/SzJ (NSG) mice to build humanized mice. Immune reconstitution in these mice recapitulated some clinical scenario in the patient who received the corresponding HSCs. Allogeneic but HLA partially matched patient-derived AML cells were successfully engrafted in these humanized mice. Importantly, we observed a significantly reduced (yet incomplete elimination of) leukemia growth in humanized mice compared with that in control NSG mice, demonstrating a functional (but defective) GVL effect. Thus, for the first time, we established a novel humanized mouse model that can be used for studying human GVL responses against human AML cells in vivo. This novel clinically relevant model provides a valuable platform for investigating the mechanisms of human GVL and development of effective leukemia treatments.
    Type of Medium: Online Resource
    ISSN: 0741-5400 , 1938-3673
    RVK:
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2022
    detail.hit.zdb_id: 2026833-6
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 113, No. 4 ( 2009-01-22), p. 911-918
    Abstract: The production of mature cells necessitates that lineage-committed progenitor cells be constantly generated from multipotential progenitors. In addition, the ability to respond rapidly to physiologic stresses requires that the signals that regulate the maintenance of progenitor populations be coordinated with the signals that promote differentiation of progenitors. Here we examine the signals that are necessary for the maintenance of the BMP4-dependent stress erythropoiesis pathway. Our previous work demonstrated that BMP4, stem cell factor, and hypoxia act in concert to promote the expansion of a specialized population of stress erythroid progenitors in the spleen during the recovery from acute anemia. Our analysis shows that acute anemia leads to an almost complete mobilization of BMP4-responsive stress erythroid burst-forming units; therefore, new stress progenitors must be recruited to the spleen to replenish this system. We show that bone marrow cells can home to the spleen and, in response to a signal in the spleen microenvironment, Hedgehog, they develop into BMP4-responsive stress progenitors. Hedgehog induces the expression of BMP4, and together these 2 signals are required for the development of BMP4-responsive stress progenitors. These data demonstrate that the interplay between these 2 signals is crucial for maintenance of this stress response pathway.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 323-323
    Abstract: Acute myeloid leukemia (AML) is a heterogeneous and rapidly progressing blood cell cancer caused by numerous cytogenetic alterations. Although significant improvement in treatment of AML has been made, the unfortunate reality is that currently available treatments are largely ineffective for most AML patients. Thus, there is a critical need for new therapeutic targets and agents for the treatment of AML. The sphingolipid metabolic pathway is an untapped source of new therapeutic targets for the treatment of AML. Sphingosine Kinase 1 (SphK1) plays a central role in the sphingolipid metabolic pathway as the key enzyme regulating the intracellular equilibrium between pro-apoptotic Ceramide (Cer) and pro-mitogenic/pro-survival Sphingosine-1-phosphate (S1P), a.k.a. the “Sphingolipid Rheostat”. As SphK1 activity increases in the cell, pro-mitogenic/pro-survival S1P signaling predominates and AML cells become dependent upon S1P signaling (non-oncogene addiction), while depletion of Cer levels makes them more resistant to chemotherapies. We previously developed a novel SphK1-selective inhibitor (SKI-178) that is potently cytotoxic to multiple AML cell lines including multi-drug resistant lines. Our recent, thorough examination of the apoptotic mechanism-of-action of SKI-178, including direct target engagement assays employing the Cellular Thermal Shift Assay (CETSA) revealed that SKI-178 also acts as a colchicine binding site directed microtubule disrupting agent (MDA). Numerous studies have demonstrated that agents that promote Cer accumulation, including SphK inhibitors, synergistically induce apoptosis, in combination with MDAs by the simultaneous activation of pro-apoptotic Bcl-2 family proteins and inhibition of anti-apoptotic Bcl-2 family proteins, respectively. SKI-178 uniquely accomplishes these two separate cellular effects as a single agent. We examined the therapeutic efficacy of SKI-178 in three mouse models of AML. Using a retro-viral transduction model of the MLL/AF9 t(9;11)(p22;q23) translocation, we have shown that SphK1 is necessary for the development of MLL/AF9-driven AML and that SKI-178 effectively induces complete remission of AML in this model system. Separately, in a human AML cell line (MOLM-13; MLL/AF9+, FLT3-ITD) xenograft model, SKI-178 significantly extended survival relative to vehicle controls. Lastly, employing a Patient Derived Xenograft model of a human primary AML sample (FLT3-ITD, NPM1+), SKI-178 also significantly extended survival relative to vehicle treated cohorts. In all 3 models, SKI-178 was well tolerated and did not affect normal hematopoiesis. Together, these data demonstrate the therapeutic efficacy of a strategy co-targeting SphK1 inhibition and microtubule dynamics and suggest that SKI-178 should be further developed as a novel therapeutic agent for AML. Citation Format: Taryn E. Dick, Jeremy A. Hengst, Laura M. Geffert, Robert F. Paulson, Hong-Gang Wang, David F. Claxton, Mark Kester, Thomas P. Loughran, Jong K. Yun. SKI-178, a single agent co-targeting sphingosine kinase 1 and microtubule dynamics, as a therapeutic strategy for treatment of acute myeloid leukemia. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 323.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: The Journal of Immunology, The American Association of Immunologists, ( 2023-09-15)
    Abstract: Allogeneic hematopoietic stem cell transplantation (alloSCT) is, in many clinical settings, the only curative treatment for acute myeloid leukemia (AML). The clinical benefit of alloSCT greatly relies on the graft-versus-leukemia (GVL) effect. However, AML relapse remains the top cause of posttransplant death; this highlights the urgent need to enhance GVL. Studies of human GVL have been hindered by the lack of optimal clinically relevant models. In this article, we report, the successful establishment of a novel (to our knowledge) humanized GVL model system by transplanting clinically paired donor PBMCs and patient AML into MHC class I/II knockout NSG mice. We observed significantly reduced leukemia growth in humanized mice compared with mice that received AML alone, demonstrating a functional GVL effect. Using this model system, we studied human GVL responses against human AML cells in vivo and discovered that AML induced T cell depletion, likely because of increased T cell apoptosis. In addition, AML caused T cell exhaustion manifested by upregulation of inhibitory receptors, increased expression of exhaustion-related transcription factors, and decreased T cell function. Importantly, combined blockade of human T cell–inhibitory pathways effectively reduced leukemia burden and reinvigorated CD8 T cell function in this model system. These data, generated in a highly clinically relevant humanized GVL model, not only demonstrate AML-induced inhibition of alloreactive T cells but also identify promising therapeutic strategies targeting T cell depletion and exhaustion for overcoming GVL failure and treating AML relapse after alloSCT.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2023
    detail.hit.zdb_id: 1475085-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Archives of Biochemistry and Biophysics, Elsevier BV, Vol. 460, No. 2 ( 2007-04), p. 306-313
    Type of Medium: Online Resource
    ISSN: 0003-9861
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2007
    detail.hit.zdb_id: 1461378-5
    SSG: 12
    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