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
    Springer Science and Business Media LLC ; 2012
    In:  Investigational New Drugs Vol. 30, No. 1 ( 2012-02), p. 98-104
    In: Investigational New Drugs, Springer Science and Business Media LLC, Vol. 30, No. 1 ( 2012-02), p. 98-104
    Type of Medium: Online Resource
    ISSN: 0167-6997 , 1573-0646
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2012
    detail.hit.zdb_id: 2009846-7
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Biochemical Journal, Portland Press Ltd., Vol. 427, No. 2 ( 2010-04-15), p. 305-312
    Abstract: EPO (erythropoietin), the major hormone regulating erythropoiesis, functions via activation of its cell-surface receptor (EPO-R) present on erythroid progenitor cells. One of the most striking properties of EPO-R is its low expression on the cell surface, as opposed to its high intracellular levels. The low cell-surface expression of EPO-R may thus limit the efficacy of EPO that is routinely used to treat primary and secondary anaemia. In a recent study [Nahari, Barzilay, Hirschberg and Neumann (2008) Biochem. J. 410, 409–416] we have shown that insertion of an NPVY sequence into the intracellular domain of EPO-R increases its cell-surface expression. In the present study we demonstrate that this NPVY EPO-R insert has a selective effect on EPO-mediated downstream signalling in Ba/F3 cells expressing this receptor (NPVY-EPO-R). This is monitored by increased phosphorylation of the NPVY-EPO-R (on Tyr479), Akt, JAK2 (Janus kinase 2) and ERK1/2 (extracellular-signal-regulated kinase 1/2), but not STAT5 (signal transducer and activator of transcription 5), as compared with cells expressing wild-type EPO-R. This enhanced signalling is reflected in augmented proliferation at low EPO levels (0.05 units/ml) and protection against etoposide-induced apoptosis. Increased cell-surface levels of NPVY-EPO-R are most probably not sufficient to mediate these effects as the A234E-EPO-R mutant that is expressed at high cell-surface levels does not confer an augmented response to EPO. Taken together, we demonstrate that insertion of an NPVY sequence into the cytosolic domain of the EPO-R confers not only improved maturation, but also selectively affects EPO-mediated signalling resulting in an improved responsiveness to EPO reflected in cell proliferation and protection against apoptosis.
    Type of Medium: Online Resource
    ISSN: 0264-6021 , 1470-8728
    RVK:
    Language: English
    Publisher: Portland Press Ltd.
    Publication Date: 2010
    detail.hit.zdb_id: 1473095-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal of Endocrinology, Bioscientifica, Vol. 205, No. 1 ( 2010-01-8), p. 87-95
    Abstract: Erythropoietin (EPO) regulates proliferation and differentiation of erythroid precursor cells into erythrocytes. The last decade has revealed non-renal sites of EPO production and extrahematopoietic expression of the EPO receptor, thus suggesting that EPO has pleiotropic functions. Here, we addressed the interplay between EPO/glucose metabolism/body weight by employing a panel of relevant experimental murine models. The models focused on situations of increased EPO levels, including EPO-injected C57BL/6 and BALB/c mice, as well as transgenic mice (tg6) constitutively overexpressing human EPO, thus exposed to constantly high EPO serum levels. As experimental models for diabetes and obesity, we employed protein Tyr phosphatase 1B (PTP1B) knockout mice associated with resistance to diabetes (PTP1B −/− ), and ob/ob mice susceptible to diabetes and obesity. The data presented herein demonstrate EPO-mediated decrease in blood glucose levels in all mice models tested. Moreover, in the ob/ob mice, we observed EPO-mediated attenuation of body weight gain and reduction of hemoglobin A1c. Taken together, our data bear significant clinical implications of EPO treatment in the management of a wide range of metabolic diseases, thus adding an important novel therapeutic potential to this pleiotropic hormone.
    Type of Medium: Online Resource
    ISSN: 0022-0795 , 1479-6805
    Language: Unknown
    Publisher: Bioscientifica
    Publication Date: 2010
    detail.hit.zdb_id: 1474892-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: ChemInform, Wiley, Vol. 39, No. 25 ( 2008-06-17)
    Type of Medium: Online Resource
    ISSN: 0931-7597 , 1522-2667
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2008
    detail.hit.zdb_id: 2110203-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: ChemInform, Wiley, Vol. 40, No. 6 ( 2009-02-10)
    Type of Medium: Online Resource
    ISSN: 0931-7597 , 1522-2667
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2009
    detail.hit.zdb_id: 2110203-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 3694-3694
    Abstract: Background: Murine models of myelodysplastic syndromes (MDS) exhibit lower bone mass (B-Mass), and several reports suggest increased incidence of osteoporosis and fractures in MDS patients. Whether MDS is associated with lower B-Mass is unknown. Aims: 1) to develop a simple method to estimate trabecular B-Mass from bone marrow biopsies (BMB); 2) to compare the trabecular B-Mass of MDS patients at diagnosis and non-MDS controls; 3) to develop an artificial intelligence (AI) algorithm for automated trabecular B-Mass assessment. Methods: In this study we included male patients age ≥ 65 years old with "lower-risk" (LR) MDS, diagnosed at Tel Aviv Sourasky Medical Center, between 2011 and 2019 and age-matched controls undergoing bone marrow (BM) biopsy for either unexplained anemia or as a part of staging work-up for newly diagnosed lymphoma. BM slides (H & E stain) were digitally scanned. The total relevant area (TRA, Figure 1A) was identified and bone trabeculae were manually contoured using Adobe Photoshop software. Bone area (BA, figure 1B) was calculated as the sum of all contoured pixels of bone, and B-Mass = BA/TRA(%). The manually annotated data were used to develop a preliminary AI algorithm to automatically detect the BA as well as BM fat for use in the current and future research in the field. The Sørensen-Dice similarity coefficient (DSC) was used to assess agreement between manual and AI annotations. Results: There were 43 MDS and 36 control patients with mean ages of 80 and 78 years (p & gt;0.05), respectively. Comorbidities in both groups were similar. Notably, trabecular B-Mass of MDS was significantly lower than that of controls: 11.6% [95%CI 9.9-13.3] vs 18.3% [16.6-20.3] , respectively (p & lt;0.0001). This represented a 37% relative reduction for MDS patients compared to controls. Next, the manually annotated data used to develop an AI algorithm automatically assessed trabecular B-Mass. The devised algorithm, tested on 17 patient samples, yielded a DSC (Mean ± SEM) of 0.76 ± 0.015 and 0.81 ± 0.02 for bone and fat respectively (Figure 1C and 1D). Conclusions: We have developed a simple technique to estimate trabecular B-Mass based on available BM sections. Importantly, we found that trabecular B-Mass of LR-MDS patients is compromised already at diagnosis. In addition, automated (AI) assessment of B-Mass using available H & E-stained BM slides is feasible and clinically relevant for patients with MDS and other hematological malignancies. Future work will develop the AI technique and expand it to identify all components of the BM, including the cellular compartment. This will enhance our understanding of the osteohematological niche in such patients, and aid in diagnosis as well as treatment planning. This study was supported by a grant from the Dotan Hemato-oncology Fund, the Cancer Biology Research Center, Tel Aviv University to DN, MM and HSO. Figure 1: Bone marrow biopsy (BMB) slide of a representative patient showing (A) the total relevant area, and (B) the bone area - TRA and BA respectively. Demonstrating the artificial intelligence (AI) technique for the same patient, we compare (C) manual annotation to (D) AI-based annotation of both bone (red) and fat (green). Figure 1 Figure 1. Disclosures Mittelman: Janssen · Roche · Novartis · Takeda · Medison / Amgen · Neopharm / Celgene / BMS · Abbvie · Gilead: Research Funding; Novartis · Takeda · Fibrogen · Celgene / BMS · Onconova · Geron: Other: Clini; Onconova · Novartis · Takeda · Silence: Membership on an entity's Board of Directors or advisory committees; MDS HUB: Consultancy; Celgene / BMS · Novartis: Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    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: Blood, American Society of Hematology, Vol. 136, No. Supplement 1 ( 2020-11-5), p. 13-13
    Abstract: Background and aims: Immunotherapy with anti-CD20-specific antibodies (e.g. rituximab), has become the standard of care for B cell lymphoproliferative disorders and many autoimmune diseases. Despite previously demonstrated role for B cells in bone metabolism, the effect of anti-CD20-mediated B cell depletion on bone mass in human patients has not been thoroughly studied. For example, in rheumatological patients the effect of rituximab on bone mass yielded conflicting results, while in lymphoma patients it has not yet been described. Here, we describe the effect of treatment with anti-CD20-specific antibodies on bone mass in a cohort of patients with follicular lymphoma and propose a plausible mechanism using murine model. Methods: To assess the effect of rituximab on bone mass in lymphoma patients, we retrospectively studied the bone mass in patients with follicular lymphoma (FL) during the maintenance phase of chemoimunotherapy, i.e. when rituximab is administered as monotherapy. FL patients on no maintenance (historical controls) or patients with marginal zone lymphoma were include as a control group. Cross-sectional X-ray imaging (CT/PET-CT), performed at the completion of the induction phase and 6-12 months thereafter, were used to serially assess bone density. Wild-type female C57BL/6J mice and mouse anti-mouse CD20 antibody (Genentech) were used for the animal experimental system. Murine bone structure was assessed by the microCT method. Immunophenotyping of the bone marrow (BM), spleen and peripheral blood cells was performed. ELISA and "real-time" quantitative PCR were used to measure the levels of key mediators of bone remodeling, e.g. RANKL, OPG and TNFα. Standard osteoclastogenic assay was used to assesses the osteoclastogenic potential of BM cells. Results: Rituximab treatment prevented the decline in bone mass observed in patients who did not receive active maintenance therapy, both in the lumbar spine (-2.6% vs -8%) and femoral head (-0.5% vs -5.1%) (n=12 patients in each group, p & lt;0.05 for the comparisons in the control group, calculated by Wilcoxon matched-pairs signed rank test). Anti-CD20-mediated B cell depletion in mice led to a significant increase in bone mass as reflected by: 7.7% increase in bone mineral density (whole femur) and ~5% increase in cortical as well as trabecular tissue mineral density (n=17-21 mice in each group, p & lt;0.05). Mechanistically, treating mice with anti-CD20 antibodies significantly decreased the osteoclastogenic signals, including RANKL and TNFα, along with a substantial downregulation of RANK (the RANKL receptor). This correlated with nearly a 50% reduction in osteoclastogenic potential of BM cells derived from B-cell-depleted animals (p & lt;0.05). The decline in the RANKL output was observed both at the bone level (≈25% relative reduction in the mRNA levels), as measured in the whole bone and BM cells, as well as in the serum (18% relative reduction) of the anti-CD20-treated mice as compared to diluent-treated controls (p & lt;0.05 for all comparisons). No significant changes in the OPG levels were noted. Conclusions: While many lymphoma patients may suffer from bone loss due to advanced age and glucocorticoid administration, our data suggest additional favorable effect of anti-CD20 treatment in bone preservation. Importantly, our murine studies indicate that B cell depletion has a direct effect on bone remodeling, primarily by reducing the osteoclastogenic signals, thus potentially diminishing bone resorption. This novel unrecognized effect should be taken into consideration when maintenance treatment is considered. MM and DN contributed equally to this work. Figure 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: 2020
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 850-850
    Abstract: Background and aims: Erythropoietin (EPO) is a pleiotropic cytokine, which besides its classical role in driving erythropoiesis, displays tissue protective and immunomodulatory activities. EPO also induces bone loss. While hematopoiesis is mediated via the homodimeric EPO receptor (EPOR), tissue protection is conferred via a heteromer composed of EPOR and CD131. Cibinetide (CIB), a non-erythropoietic analogue of EPO, specifically binds to the heteromeric receptor and confers tissue protection. Our published findings that EPO stimulates osteoclast precursors and entrains a decrease in bone density, raise questions regarding the underlying molecular mechanisms. Here, we evaluated the role of the heteromeric complex in bone metabolism using CIB alone and in combination with EPO in vivo and in vitro. Results: CIB injections to 12-week-old female mice (120 µg/kg thrice weekly for 4 weeks) resulted in a significant increase in tissue mineral density in cortical bone by 5.8% (1416.4±39.27 vs 1338.74±16.56 mgHA/cm 3) and in trabecular bone by 5.2% (1056.52±30.94 vs 1004.13±16.91 mg HA/cm 3) (n=10 in each group, p & lt; 0.05 versus saline-injected controls), as measured by microCT (Figure 1A). To evaluate the capacity of CIB to attenuate EPO mediated bone loss, we administered CIB (300 µg/kg) for 5 consecutive days, to 13-week-old female mice that also received 2 injections of 120U EPO on days 1 and 4. Flow cytometry analysis revealed a 1.8-fold reduction in the number of osteoclast progenitors, defined as Lin -CD11b −CD115 +Ly6C hi, in the EPO + CIB injected mice, compared to the mice injected with EPO alone (n=7 in each group, p & lt; 0.05). Hemoglobin levels and TER119 + bone marrow (BM) erythroid progenitors were similar in both groups. In vitro, EPO administration to BM-derived macrophages (BMDM) enhanced osteoclastogenesis, whereas CIB had an opposite, dose-dependent effect. Combining CIB with EPO inhibited osteoclastogenesis in BMDM, suggesting that CIB overrides the pro-osteoclastogenic effect of EPO (Figure 1B). Conclusions: Our findings highlight the increasing complexity of EPOR signaling in bone and pave the way for clinical translation through potential combination therapy of EPO and CIB in anemic and in cancer patients. Adjunctive administration of CIB may prevent or attenuate bone loss while preserving the erythropoietic actions of EPO. This study was supported by a grant from the Dotan Hemato-oncology Fund, the Cancer Biology Research Center, Tel Aviv University to DN and YG. Figure 1 Figure 1. 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: 2021
    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. 122, No. 21 ( 2013-11-15), p. 2174-2174
    Abstract: Absolute erythrocytosis are rare disorders characterized by a significant increase of the red blood cell mass which is reflected by an increased number of circulating erythrocytes, high hematocrit and hemoglobin. Primary familial and congenital polycythemia (PFCP, familial erythrocytosis type 1, OMIM #133100) is an inherited form of this disease, caused by erythropoietin (EPO) hypersensitivity of erythroid precursor cells. A number of mutations in the intracellular region of the EPO receptor (EPOR) have been found in recent years to be associated with manifestation of the disease. The objective of the study was to explore the mechanisms by which these mutations in the cytosolic domain of EPOR may induce the erythrocytosis phenotype. The research strategy was to construct chimeric receptors that contain the extracellular and transmembrane regions of the human epidermal growth factor receptor (EGFR) fused to the cytosolic domain of the human EPOR. The moiety of the EPOR intracellular region was mutated according to 4 novel mutations discovered in the cytosolic domain of the EPOR in PFCP patients. This design enabled EPOR signaling to be triggered by EGF binding to the extracellular domain of the chimera. The working hypothesis was that since the cytosolic, EPOR region of the chimera was generated based on mutations discovered in PFCP patients, any differences in the chimera-related effects would be specifically attributed to the mutations. The experiments were performed in an in vitro system, in which the Wild-Type (WT) EGFR– EPOR and the EGFR– EPOR chimeras bearing the mutated EPOR cytosolic domain were stably transfected into the EPO-dependent human erythroleukemia cell-line UT7. As these cells lack the EGF receptor, the natural EPOR signaling can be avoided when the transfected cells are grown with, or activated by, EGF. Using this system we have analyzed 4 novel EPOR mutations discovered in PFCP patients: 2 deletions which lead to frameshifts, premature stop codons and truncations (Del1387-1390 and Del1378-1412), a nonsense mutation (C1371A) which results in truncation of the receptor, and a missense mutation (G1445A) which results in the substitution of arginine 437 to histidine. A panel of experiments was performed on UT7 cells expressing the WT EGFR-EPOR chimera or EGFR– EPOR chimeras bearing the EPOR cytosolic PFCP-associated mutations. We addressed the degradation rates of the chimeric receptors, the kinetics of EGF induced STAT5 and MAPK/ERK signaling cascades and the levels of glycan-maturation and cell-surface expression of the chimeras. Cell viability analysis revealed that UT7 cells expressing either one of the 4 mutated chimeric receptors proliferated even under very low (0.01 ng/ml) EGF concentrations, as opposed to cells expressing the WT chimera, which did not proliferate under these conditions. UT7 cells expressing the mutated chimeras also displayed enhanced growth, which may indicate that indeed the mutations are the underlying cause of PFCP. Notably, our findings are the first description of a missense EPOR mutation that is associated with over-activity of the EPOR and with development of a polycythemic phenotype. Furthermore, we found that each of the 4 EPOR mutations differentially affected signaling and metabolism of the chimeric receptors. Hence, (i) Del1387-1390, showed higher levels of glycan-mature protein, as well as slower degradation rates, and conferred slower attenuation of EGF-induced pSTAT5. (ii) Del1378-1412 exhibited slower degradation rates and higher levels of glycan-mature protein. (iii) The nonsense mutation, C1371A, displayed higher cell surface levels, slower degradation rates and relayed constitutive activation of STAT5. (iv) The missense mutation, G1445A, mediated a slower attenuation of EGF induced MAPK/ERK signaling. Our study supports the evidence regarding the role of the cytosolic region of EPOR in metabolism of the receptor and sheds new light on the mechanisms underlying erythrocytosis-inducing mutations in the EPOR. HH, MFMcM, MJP, CB, HC and DN are members MPN & MPNr-EuroNet (COST Action BM0902) 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: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 3165-3165
    Abstract: Background: The immune system has been shown to be involved in the pathogenesis of myelodysplastic syndromes (MDS), and is also affected by the disease. Recombinant erythropoietin (rHuEPO), or in general, erythroid stimulating agents (ESAs) have become a standard treatment for anemic patients with MDS. They were found to improve anemia, quality of life, and possibly survival. We have previously demonstrated that EPO has effects on cellular and humoral immunity and specifically, on immune function in patients with multiple myeloma (MM). Here we report our findings demonstrating the effect of ESAs on T cell (CD4+, CD8+ and CD4+CD25+) number and function in patients with MDS. Patients and Methods: We examined three groups: healthy subjects ('Control', 20 participants), MDS patients without ESA treatment ('MDS', 13), and MDS patients treated with an ESA ('MDS+EPO', 17). All diagnosed patients gave informed consent as approved by our IRB. Cell numbers were evaluated with flow cytometry. In a subset of patients, cell activation was assessed in response to phytohemagglutinin (PHA) by examining CD69 expression in both CD4+ and CD8+ cells. The co-stimulatory marker, CD28, and the inhibitory marker CTLA-4 (CD152) were evaluated as well. We also examined World Health Organization (WHO) subgroups, refractory anemia (RA) and RA with ringed sideroblasts (RARS) versus more advanced disease. Results: CD4+ and CD8+ T cell levels are reduced and increased respectively in MDS patients compared to control, and these changes are reversed in MDS+EPO (Table 1, CD4+, p 〈 0.01; CD8+, p=0.05). The CD4+:CD8+ ratio (Table 1) is reduced and nearly equalized in MDS (1.16), but approaches that of the control (2.24) in MDS+EPO (1.94). CD4+CD25+ T cell numbers (including regulatory T cells), were lower in MDS patients and improve in the MDS+EPO group (Table 1). In vitro activation of T cells (CD4+CD69+ and CD8+CD69+) achieves an approximately 15-fold increase in healthy subjects. MDS patients without EPO sustained only a 7.17 fold increase in CD4+ activation versus 13.64 fold for the MDS+EPO group (p 〈 0.01); for CD8+ T cells, 10.20 fold (MDS) versus 18.56 fold (MDS+EPO, p 〈 0.01). The expression of the co-stimulatory marker CD28 was decreased in both CD4+ and CD8+ T cells in MDS, and approached normal in MDS+EPO in CD4+ T cells (Table 1). There was no significant change in inhibitory CTLA-4 (CD152) expression among the groups (not shown). Subgroup analysis demonstrated that ESA has a similar effect on CD4+ and CD8+ cells and their ratio in both RA/RARS and more advanced disease, similar to those of the whole cohort (Table 2, green). On the other hand, some parameters were affected by ESA only in one subgroup (Table 2, blue): The ESA effect on CD4+CD25+ cells was evident only in patients with advanced disease (Table 2, blue). ESA affected CD4+ and CD8+ cell stimulation (CD69) in RA/RARS, similar to that seen in the whole cohort (Table 2, blue). Of note, in more advanced disease, CD4+ and CD8+ cells achieved stimulation in the MDS group not treated with ESA, with no difference between MDS and MDS+EPO. This finding needs to be further addressed in larger cohorts and with additional markers of activation. Conclusions: MDS patients display T-cell abnormalities that are improved upon EPO treatment. MDS is a heterogeneous disease where the immune system both affects and is affected by the disease. As such, treatment with ESAs might ameliorate not only the anemia, but also the immune deficiencies and perhaps the disease itself. Future studies will clarify the immunomodulatory role of ESA in the various stages of MDS. 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: 2016
    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...