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. 134, No. Supplement_1 ( 2019-11-13), p. 4650-4650
    Abstract: Introduction. Hematopoietic cell kinase (HCK) belongs to the Src kinase family (SFK) involved in the oncogenic process and hematological malignancy. Some SFK inhibitors are currently under investigation in clinical trials for leukemia after demonstrating efficacy in patients with solid tumors. We have previously reported that HCK is overexpressed in leukemic cells and its inhibition by lentivirus resulted in reduction of cell growth and increased cell death (Roversi et al. BBA Mol Basis Dis. 2017, 1863(2):450-61). In light of the genomic and molecular diversity of myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML), the development of chemical compounds specific for new molecular targets is currently an important subject. Aims. To investigate the in vitro and in vivo effects of a new chemical compound targeting HCK inhibition (iHCK), alone or in combination with the most used drugs for treatment of MDS and AML (Azacytidine - Aza - or Cytarabine - Ara-C). Methods. After iHCK development, we tested its activity alone or in combination with Aza or Ara-C in CD34+ cells isolated from AML patients (n=5) as well as in a panel of myeloid leukemia cell lines (KG1, HL-60, HEL and K562). Additionally, we tested the iHCK in normal and malignant cells cultured in a 3D bioscaffold obtained by decellularization of bovine bone marrow (Bianco et al. Biomat Sci 2019, 7(4):1516-28), in order to mimic the bone marrow niche. After informed written consent and approval of the Ethical Committee of University of Campinas (CAAE 1000.0.146.00-11), in accordance to the Helsinki Declaration, CD34+ cells were isolated from bone marrows of healthy donors (HD), MDS and AML patients and were treated with iHCK or vehicle (DMSO) in liquid culture, for three days. Meanwhile, HS-5 mesenchymal cells were cultured into the 3D bioscaffold. iHCK or vehicle treated CD34+ cells were introduced into the 3D bioscaffold containing HS-5 and evaluated after 7 and 14 days, by light microscopy (hematoxilin and eosin regular staining) and immunohistochemistry (expression of CD34 and CD90 antigens). NOD.CB17-Prkdcscid/J mice received 2 Gy irradiation followed by transplantation with caudal intravenous injection of leukemia cells obtained from hCG-PML-RARα transgenic mice. After acute promyelocytic leukemia (APL) establishment, animals were treated or not with intraperitoneally iHCK and peripheral blood was collected for hematological analysis and protein was extracted from spleen and bone marrows for Western Blot analysis. ANOVA and Student's T-Test were used. Results.In leukemia cell lines and primary cells, the combinatory treatment of iHCK and Cytarabine (1μM) or 5-Azacitidine (1μM) demonstrated synergistic effects, compared to either drug alone, on the reduction of growth and induction of cell death (P 〈 0.001; Figure 1). Further, Western blot revealed increased BAX expression and decreased BCL-XL expression. Moreover, iHCK treatment was able to reduce the activation of oncogenic pathways, MAPK/ERK and PI3K/AKT, leading to severe reduction of ERK, AKT and p70S6 phosphorylation. Treatment with iHCK reduced CD34+ MDS and AML cells proliferation cultured into the 3D bioscaffold but had no effect upon normal CD34+cells. In vivo analysis showed that APL mice treated with iHCK (5μM) for 48h had reduced leukocyte number compared to APL mice treated with vehicle (13.2±1.1 vs 49.4±18.8; P 〈 0.001). No alterations in hemoglobin levels and platelet were found. Likewise, the in vivo iHCK (2.5μM, 5.0μM or 10.0μM) treatment decreased the phosphorylation of ERK, AKT and P70S6K proteins of leukemic cells (Figure 2). Conclusion.The iHCK pharmacological inhibitor has an antiproliferative activity in leukemic cells without altering cell death and survival rate of normal cells, demonstrating on-target malignant cell killing activity as a single agent or in combination with Azacytidine (Aza) or Cytarabine (Ara-C). 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: 2019
    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. 132, No. Supplement 1 ( 2018-11-29), p. 3830-3830
    Abstract: Coordinated arrangement of bone marrow (BM) microenvironment is essential for the maintenance of hematopoietic stem cells and their progenitors (HSPC), and for the continuous supply of peripheral blood (PB) cells. Quiescence and/or mobilization of these BM cells into the circulation are tightly regulated however non-physiological or stress conditions, such as infections, can accelerate them. Our previous findings have shown that polyphenols from green tea modulate the myeloid population (Gr-1+Mac-1+) of BM, spleen and PB of mice challenged by lipopolysaccharide (LPS). We have also observed a reduction in the circulating levels of thrombin-antithrombin complex (TAT) and soluble endothelial protein C receptor (sEPCR) of these mice. It has been reported that thrombin induces the rapid HSPC mobilization through coagulation thrombin/PAR-1 axis, and quiescence is maintained across the APC/EPCR/PAR-1 axis (Nat. Med. 2015, 21:1307-17). In this context, our goal was to investigate the effects of polyphenols on the Lin-Sca-1+c-Kit+ (LSK) primitive stem cell population and mature populations including: T and B lymphocytes, monocytes, granulocytes and erythrocytes (Lin+). The expression of PAR-1 and EPCR in these cells, and the levels of sEPCR in the BM fluid were also evaluated. The 250 mg/kg dose of polyphenol from green tea extract was given once every 7 days orally (gavage) to mice (n=6/group) challenged with i.p. injection of 100µg LPS. Control group received vehicle only. After 24h, mice were sacrificed; BM and PB were collected for the assays. LPS injection induced an increase in circulating LSK, without affecting the pool of these cells in BM. Treatment with polyphenols prevented recruitment of LSK into the PB, and did not alter the percentage of BM LSK. Corroborating these findings, polyphenols also reduced the number of immature progenitors (CFU-C) in the PB, evaluated by the clonal culture assay. In addition, polyphenols did not affect the increase in the number of Lin+ cells and circulating leukocytes (total WBC) induced by LPS injection. We also observed that LPS induced a rapid increase in the number of PB LSK and Lin+ cells expressing PAR-1, while reducing BM LSK expressing EPCR. Polyphenols did not affect PAR-1 expression in both populations, but partially recovered expression of EPCR in BM LSK. Polyphenols also reduced the increased levels of sEPCR induced by LPS in the BM niche. Taken together, our results demonstrate that polyphenols promote an effect on the quiescence/mobilization of both the most primitive hematopoietic cells and their progenitors, but does not affect mature populations. The reduction in vascular permeability observed in the BM of mice using Evans blue reinforces this action of polyphenols. It appears that the effect of polyphenols is thrombin dependent; although polyphenols did not affect the expression of PAR-1, they modulated EPCR, mainly in the medullary microenvironment with maintenance of barrier integrity and consequent cellular quiescence. In addition, polyphenols appear to have an anti-inflammatory effect, possibly triggered by their effect on the EPCR. Disclosures De Paula: Hematology and Transfusion Medicine Center, University of Campinas: Employment. Shiraishi:Hematology and Transfusion Medicine Center, University of Campinas: Employment. Queiroz:University of Campinas: Employment.
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 4996-4996
    Abstract: The mechanism underlying quiescence and/or mobilization of hematopoietic stem cells and their bone marrow progenitors (HSPC) into circulation are tightly regulated for the continuous supply of peripheral blood cells; however, non-physiological or stress conditions, such as infections, can accelerate these mechanisms. Our results have shown that polyphenols modulate quiescence/mobilization of HSPC, but do not affect mature populations. Thrombin has been reported to induce the rapid HSPC mobilization through coagulation thrombin/PAR-1 axis, and quiescence is maintained across the APC/EPCR/PAR-1 axis (Nat. Med. 2015, 21:1307-17). Our objective was to investigate the effect of polyphenols on thrombin/PAR-1 and APC/EPCR/PAR-1 axis. C57BL/6J mice (6-8 weeks old) were treated with polyphenols from green tea extract (250 mg/kg body weight) orally (gavage) once every seven days and injected (i.p.) at day 7 with lipopolysaccharide (LPS) (100μg;Sigma) (n=6). The control group received vehicle and was injected with LPS (n=6). After 24h of LPS injection, mice were anesthetized for blood collection, and then sacrificed for bone marrow collection. PAR-1 and EPCR expression, nitric oxide (NO) production and endothelial nitric oxide synthase (eNOS) phosphorylation were evaluated in HSPC by flow cytometry. The functional ability of HSC was assessed by competitive repopulation assay. Vascular permeability was studied using Evans blue. After LPS injection, mice showed reduced expression of EPCR in bone marrow LSK parallel to an increase of PAR-1 expression in circulating immature and mature cells. Treatment of these mice with polyphenols partially prevented the reduced expression of EPCR in bone marrow LSK (13±3 vs 54±12; p 〈 0.05), but did not affect the increased PAR-1 expression in circulating immature and mature cells. Evans blue assay revealed a reduction in the vascular permeability of the bone marrow of LPS-injected mice treated with polyphenols (3.9±0.5 vs 2.1±0.1; p 〈 0.05). To assess whether polyphenols altered NO production, we measured NO levels and eNOS phosphorylation in immature LSK EPCRhigh (or LT-HSC) cells. NO production is activated by eNOS phosphorylation at Ser1177 and negatively regulated by eNOS phosphorylation at Thr495. LPS injection rapidly increased NO levels and eNOS phosphorylation at Ser1177 in bone marrow LSK of mice. Treatment of these mice with polyphenols reduced the percentage of bone marrow LSK EPCRhigh cells with higher intracellular NO (52±2.8 vs 28±5.6; p 〈 0.01) and increased eNOS phosphorylation at Thr495 in immature LSK. In order to evaluate the action of polyphenols on the functional ability of HSC, a competitive bone marrow repopulation assay was performed. Donor mice (C57BL/6J) received or not polyphenols followed by LPS injection (treated group: Polyphenols+LPS; control group: LPS), and bone marrow cells were transplanted (1:1) together with bone marrow cells of competitors (B6.SJL-PtprcaPepcb/BoyJ) in lethally irradiated recipients (B6.SJL-PtprcaPepcb/BoyJ). Mice were followed for 16 weeks and hematological analysis revealed no difference in circulating leukocytes, platelets or hemoglobin levels. Transplanted mice (recipients) presented a higher percentage of CD45+ cells from Polyphenols+LPS donors (33.7±13 vs 78.6±0.9; p 〈 0.05) in the peripheral blood, as well as increased number of T lymphocytes (6.7±4.5 vs 37.2±2.9; p 〈 0.05) and myeloid cells (68.5±1.7 vs 82.5±3.5; p 〈 0.05) from Polyphenols+LPS group. After 16 weeks, mice were euthanized and a higher percentage of LSK (or HSC) and LSK EPCRhigh (or LT-HSC) cells from Polyphenols+LPS donors were detected in the bone marrow, although only the percentage of LSK EPCRhigh was statistically different (0.0014±0.0001 vs 0.0032±0.001; p 〈 0.05). Taken together, our results indicate that polyphenols increased the functional ability of HSC in LPS-injected mice showing increased percentage of bone marrow LSK EPCRhigh cells, which are the most quiescent stem cells with strong self-renewal ability. Polyphenols reduced EPCR expression and NO production in immature cells of LPS-injected mice, exhibiting an anti-inflammatory effect that leads to the maintenance of barrier integrity and quiescence of cells, which was corroborated by reducing vascular permeability in the bone marrow. Thus, polyphenols appear to modulate quiescence/mobilization of HSPC through APC/EPCR/PAR-1 axis. 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: 2019
    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: Transplant Immunology, Elsevier BV, Vol. 81 ( 2023-12), p. 101919-
    Type of Medium: Online Resource
    ISSN: 0966-3274
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 2027651-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 554-554
    Abstract: Background: Primary Myelofibrosis (PMF) is a chronic myeloproliferative neoplasm (MPN) characterized by increased myeloid proliferation and associated with mutations that induce tyrosine-kinase activation mainly via JAK-STAT pathway, culminating in extensive bone marrow (BM) fibrosis in the course of disease progression. In contrast to the monoclonal origin of hematopoietic cells, fibroblasts proliferation is polyclonal, and mediators involved in fibrosis, neoangiogenesis and osteosclerosis seem to be involved in disease progression. Metformin (MTF) is a biguanide that exerts selective antineoplastic activity in a variety of malignancies, through its action on nutrients privation and hypoxia, leading to apoptosis. In JAK2-mutated cell lines, MTF reduced cell viability, proliferation and clonogenicity, while in Jak2V617F knock-in-induced mice, MTF reduced Ba/F3 JAK2V617F tumor burden and splenomegaly. These data suggest that MTF could have a therapeutic effect in PMF patients. Aims: To conduct an open label phase II study to evaluate MTF effects on BM fibrosis, inflammation mediators, JAK-STAT pathway activation and disease progression in PMF patients. Methods: PMF non-diabetic adults were eligible. Subjects with severe renal function impairment were not included. Patients received MTF (Glifage XR®) in rising doses until a maximum of 2500mg PO daily, according to tolerance. Primary endpoint was BM fibrosis reversion. Secondary endpoints included reduction of inflammation and downregulation of the JAK-STAT pathway. Clinical data was systematically compiled. Blood and BM samples were collected at the time points: pretreatment (0), 3 mo and 6 mo. Collagen was evaluated in BM biopsy specimens by Masson's trichrome stain: three representative areas from each slide were analyzed and the collagen/sample area was quantified using Image J software; the mean percentage of each slide was used for statistics. IL-6, IL-8 and TNF-α levels were analyzed in BM samples using multiplex assay. Phosphorylation status of intracellular proteins STAT3 and STAT5 was analyzed by flow cytometry and the percentage of cells was recorded using FlowJo software. In order to evaluate gene modulation following MTF exposure, samples at time points 0 and 6 mo were analyzed by PCR array for insulin signaling genes (PAHS-030Z, Qiagen). Genes with ±1.5 fold-change in both directions were selected for validation. For each experiment, statistical analysis was performed and a p value & lt;0.05 was considered statistically significant. Results were expressed as medians (min-max). This trial was approved by the Institutional and National Review Board; written informed consent was obtained from all subjects. Results: 11 patients (aged 40-84y) were included. Two subjects had early treatment discontinuation due to non-related causes. The median exposure to MTF was 10 mo (5-11) and the median dose was 2000mg/day (1500-2500mg). The most frequent adverse event was diarrhea (n=3). No life threatening event occurred. A reduction in BM collagen area percentage was observed comparing pretreatment biopsies (26.9% (14.8-53.1%)) versus 3 months (3.8% (2.3-4.0%), p=0.062) and versus 6 months of MTF use (0.84% (0.12-17.1%), p=0.125), however, this result was not statistically significant probably due to the low number of patients analyzed (n=5). IL-6, IL-8 and TNF-α levels did not differ between time points. Flow cytometry analysis demonstrated a trend in STAT3 phosphorylation decrease when comparing pretreatment samples versus 6 months of MTF use, though this result was not statistically significant (p=0.06). Mean fluorescence intensity for pSTAT3 was: pretreatment 10.53 ± 5.75, 3 mo 7.34 ± 2.4, 6 mo 5.41 ± 1.14; and for pSTAT5: pretreatment 14.03 ± 7.41; 3 mo 10.71 ± 7.74; 6 mo 6.03 ± 1.41. PCR array for insulin signaling genes showed 21 genes downregulated after 6 months of MTF treatment, including genes previously associated with MPN phenotype: INS (0 and 6 mo treatment fold-decrease: 0.18), NOS2 (0.24), VEGFA (0.34), LEP (0.34), IGFBP1 (0.38) and IRS2 (0.62). Conclusions: In this study, metformin showed to be a safe and well-tolerated drug. Our preliminary results demonstrated a trend in BM collagen reduction in PMF patients following metformin treatment. Downregulation of important genes associated with MPN phenotype was also noted. The trial is ongoing and these results will be validated at other time points for all subjects. Disclosures Pagnano: Abbvie: Consultancy; Pint Pharma: Consultancy; Sandoz: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    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. 134, No. Supplement_1 ( 2019-11-13), p. 5765-5765
    Abstract: Background: (-)-Epigallocatechin-3-gallate (EGCG) is a gallate ester obtained by the condensation of gallic acid with the (3R)-hydroxy group of (-)-epigallocatechin. This component, extracted from green tea, has multiple effects on signal transduction pathways and enzyme activities which could enhance apoptosis and suppress of cell proliferation, invasion, angiogenesis and metastasis in cancers. This study aims to evaluate the effect of EGCG in an experimental model of leukemia (PML-RARα mice). Methods: NOD.CB17-Prkdcscid/J mice (12-16 weeks old) received 2Gy irradiation followed by transplantation of leukemia cells obtained from hCG-PML-RARα transgenic mice by i.v. injection in the caudal vein. Establishment of disease was confirmed at day 12 through presence of leukocytosis ( 〉 30x103/µL), and/or anemia ( 〈 10g/dL), and/or thrombocytopenia ( 〈 500x103/µL), associated to the presence of blasts in blood. At 12th day, mice (n=10/group) were randomly selected to receive EGCG (25mg/kg/day) (Cayman Chemical Co., Michigan, USA) or vehicle i.p. for five consecutive days. Mice were then sacrificed and peripheral blood, bone marrow and spleens were collected for flow cytometry and western blot analysis. All experiments were approved by the Ethical Committee for Animal Experimentation of Institution (nº3995-1/A). Results: Hematological analysis revealed that EGCG treatment reversed leukocytosis (54.09±57.71 vs 11.45±16.08; p=0.0371), anemia (9.60±1.50 vs 11.32±1.36; p=0.0155) and thrombocytopenia (238.5±146.43 vs 475.8±247.91; p=0.0179) and prolonged survival of PML/RARα mice (13 vs 15 days; p=0.0017). Notably, EGCG reduced leukemia immature cells (CD45+CD34+) (8.04±2.49 vs 5.13±1.58; p=0.0060) and promyelocytes (CD45+CD117+) (73.54±12.85 vs 56.26±15.93; p=0.0157) in bone marrow of mice whereas increased mature myeloid cells (CD11b+Gr-1+) (6.15±3.00 vs 14.60±7.83; p=0.0051), possibly by inducing cellular differentiation. These results were corroborated by the reduction in promyelocytes (45.97±11.72 vs 30.29±11.01; p=0.0154), and the increase in neutrophils (CD45+Gr-1+) (38.20±14.34 vs 54.88±14.25; p=0.0178) and monocytes (CD45+CD11b+) (60.22±18.87 vs 76.79±15.59; p=0.0463) detected in peripheral blood. We then evaluated the effect of EGCG on cellular differentiation by studying degradation of PML/RARα oncoprotein. EGCG increased the percentage of cells with aggregated PML bodies stain in the bone marrow of PML-RARα mice, suggestive of higher degradation of oncoprotein, parallel to a reduction in PIN1 expression in bone marrow cells. Higher intracellular levels of reactive oxygen species (ROS) were also detected in leukemia immature cells (2101±1025 vs 3544±614; p=0.0051), promyelocytes (1765±1176 vs 3090±1282; p=0.0271) and neutrophils (1830±1093 vs 3532±1157; p=0.0033) of bone marrow. These results are consistent with literature data demonstrating that the ablation of PIN1 and/or induction of ROS could trigger PML/RARα degradation. EGCG has been reported to inhibit PIN1, a peptidyl isomerase overexpressed and/or over activated in human cancers, which is described as a key target in PML/RARα. Furthermore, apoptosis was detected in spleen cells of PML-RARα mice (5.97±4.19 vs 10.42±3.54; p=0.0197) in parallel to increased expression of BAX, reduced expression of BCL-2, and reduction of spleen weight (0.5587±0.05 vs 0.3949±0.10; p=0.0085). Conclusion: Collectively, our results support further evaluation of EGCG in clinical trials for acute myeloid leukemia. 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: 2019
    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: TAXON, Wiley, Vol. 71, No. 1 ( 2022-02), p. 178-198
    Abstract: The shortage of reliable primary taxonomic data limits the description of biological taxa and the understanding of biodiversity patterns and processes, complicating biogeographical, ecological, and evolutionary studies. This deficit creates a significant taxonomic impediment to biodiversity research and conservation planning. The taxonomic impediment and the biodiversity crisis are widely recognized, highlighting the urgent need for reliable taxonomic data. Over the past decade, numerous countries worldwide have devoted considerable effort to Target 1 of the Global Strategy for Plant Conservation (GSPC), which called for the preparation of a working list of all known plant species by 2010 and an online world Flora by 2020. Brazil is a megadiverse country, home to more of the world's known plant species than any other country. Despite that, Flora Brasiliensis , concluded in 1906, was the last comprehensive treatment of the Brazilian flora. The lack of accurate estimates of the number of species of algae, fungi, and plants occurring in Brazil contributes to the prevailing taxonomic impediment and delays progress towards the GSPC targets. Over the past 12 years, a legion of taxonomists motivated to meet Target 1 of the GSPC, worked together to gather and integrate knowledge on the algal, plant, and fungal diversity of Brazil. Overall, a team of about 980 taxonomists joined efforts in a highly collaborative project that used cybertaxonomy to prepare an updated Flora of Brazil, showing the power of scientific collaboration to reach ambitious goals. This paper presents an overview of the Brazilian Flora 2020 and provides taxonomic and spatial updates on the algae, fungi, and plants found in one of the world's most biodiverse countries. We further identify collection gaps and summarize future goals that extend beyond 2020. Our results show that Brazil is home to 46,975 native species of algae, fungi, and plants, of which 19,669 are endemic to the country. The data compiled to date suggests that the Atlantic Rainforest might be the most diverse Brazilian domain for all plant groups except gymnosperms, which are most diverse in the Amazon. However, scientific knowledge of Brazilian diversity is still unequally distributed, with the Atlantic Rainforest and the Cerrado being the most intensively sampled and studied biomes in the country. In times of “scientific reductionism”, with botanical and mycological sciences suffering pervasive depreciation in recent decades, the first online Flora of Brazil 2020 significantly enhanced the quality and quantity of taxonomic data available for algae, fungi, and plants from Brazil. This project also made all the information freely available online, providing a firm foundation for future research and for the management, conservation, and sustainable use of the Brazilian funga and flora.
    Type of Medium: Online Resource
    ISSN: 0040-0262 , 1996-8175
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2022
    detail.hit.zdb_id: 2081189-5
    detail.hit.zdb_id: 204216-2
    SSG: 12
    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...