scholarly journals Tpr Deficiency Disrupts Erythroid Maturation With Impaired Chromatin Condensation in Zebrafish Embryogenesis

Author(s):  
Shuang Wu ◽  
Kai Chen ◽  
Tao Xu ◽  
Ke Ma ◽  
Lei Gao ◽  
...  

Vertebrate erythropoiesis involves nuclear and chromatin condensation at the early stages of terminal differentiation, which is a unique process to distinguish mature erythrocytes from erythroblasts. However, the underlying mechanisms of chromatin condensation during erythrocyte maturation remain elusive. Here, we reported a novel zebrafish mutantcas7 with erythroid maturation deficiency. Positional cloning showed that a single base mutation in tprb gene, which encodes nucleoporin translocated promoter region (Tpr), is responsible for the disrupted erythroid maturation and upregulation of erythroid genes, including ae1-globin and be1-globin. Further investigation revealed that deficient erythropoiesis in tprbcas7 mutant was independent on HIF signaling pathway. The proportion of euchromatin was significantly increased, whereas the percentage of heterochromatin was markedly decreased in tprbcas7 mutant. In addition, TPR knockdown in human K562 cells also disrupted erythroid differentiation and dramatically elevated the expression of globin genes, which suggests that the functions of TPR in erythropoiesis are highly conserved in vertebrates. Taken together, this study revealed that Tpr played vital roles in chromatin condensation and gene regulation during erythroid maturation in vertebrates.

Blood ◽  
2005 ◽  
Vol 105 (4) ◽  
pp. 1448-1455 ◽  
Author(s):  
Loïc Garçon ◽  
Catherine Lacout ◽  
Fédor Svinartchouk ◽  
Jean-Pierre Le Couédic ◽  
Jean-Luc Villeval ◽  
...  

Abstract Growth factor independence-1B (Gfi-1B) is a transcription factor with a highly conserved transcriptional repressor snail–Gfi-1 (SNAG) domain and 6 zinc-finger domains at the N- and C-terminus, respectively. Disruption of the Gfi-1B gene is lethal in the embryo with failure to produce definitive enucleated erythrocytes. In this study, we analyzed the role of Gfi-1B in human erythropoiesis. We observed an increase of Gfi-1B expression during erythroid maturation of human primary progenitor cells. We studied the consequences of variations in Gfi-1B expression in 2 transformed cell lines (K562 and UT7 cells), as well as in primary CD36+/GPA– progenitors. A knock-down of Gfi-1B delayed the terminal differentiation of K562 and primary cells. Forced expression of Gfi-1B in UT7 and K562 cells led to an arrest of proliferation and an induction of erythroid differentiation. Enforced expression of Gfi-1B in primary cells at the colony-forming units-erythroid (CFU-E) stage led to a partial glycophorin A (GPA) induction after erythropoietin (EPO) withdrawal but failed to protect cells from apoptosis. Deletion of the SNAG repressor domain abolished Gfi-1B–induced erythroid maturation, strongly suggesting that Gfi-1B acts in the late stage of erythroid differentiation as a transcriptional repressor.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2320-2320
Author(s):  
Chong Yang ◽  
Toshio Suda ◽  
Xiaoxuan Lin ◽  
Mitsuhiro Endo

Abstract Adult erythropoiesis involves a series of well-coordinated events resulting in the production of mature red blood cells. One of such events is the mitochondria clearance, which is known to occur cell-autonomously via autophagy-dependent mechanisms. Interestingly, we identified a sequential changes in the transcriptional pattern during terminal erythroid differentiation based on the expression of several macroautophagy (e.g. Atg3, Atg5, Atg7 and Atg10) and non-canonical mitophagy (e.g. Pink1, Park2, Bnip3l/Nix, P62 and Ulk1) genes. Hence we hypothesize that the progressive reduction in mitochondria during terminal erythroid differentiation is directed by distinct transcriptionally-regulated programs. Notably, we revealed a gradual reduction of the expression of lysosome related genes (e.g. Lamp1, CD63, and Atp6v) and lysosomal activities from early to late stages of terminal differentiation. On the other hand, P62-Pink1-Parkin mediated ubiquitin proteasome degradation of mitochondria proteins seems to be more prominent during late stage erythropoiesis. Hence our data suggest that mitochondria clearance is predominantly mediated by canonical autophagy during early stages of terminal differentiation, whereas non-canonical mitophagy pathway seem to play a more predominant role to regulate late stages erythroid maturation. Next, we discovered mitochondria transfer activities from erythroblasts to their niche. In the context of erythropoiesis, macrophages are known to interact closely with erythroblasts to provide a specialized niche for erythroid precursors to proliferate, differentiate and enucleate. We showed defective erythropoiesis after macrophage depletion in the bone marrow. Subsequently, we identified a tendency for early erythroblasts to associate with macrophages and isolated those erythroblasts from mito-dendra2 mice with trackable mitochondria to establish a murine primary cell co-culture system. Then we report mitochondria transfer activities in the erythroid niche via different modes including direct uptake, micro-vesicle transfer and tunnelling nanotubes (TNT). Interestingly, interchangeable structures between micro-vesicles and TNTs have been observed, suggesting an interplay between cytoskeleton and membrane lipid molecules in the mitochondria transfer mechanisms. Furthermore, mitochondria transfer activities have also been observed in the co-culture of mito-dendra2 erythroid cells with a macrophage cell line, RAW cells, and are significantly enhanced by the activation of the RAW cells via Tfe3 activation. In summary, our findings may provide insight into the mitochondria clearance machineries that mediates erythroid maturation to fulfil the clinical demand for large scale transfusable blood cell production in vitro. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Author(s):  
Ileana Cantú ◽  
Harmen J.G. van de Werken ◽  
Nynke Gillemans ◽  
Ralph Stadhouders ◽  
Steven Heshusius ◽  
...  

ABSTRACTKrüppel-like factor 1 (KLF1) is an essential transcription factor for erythroid development, as demonstrated by Klf1 knockout mice which die around E14 due to severe anemia. In humans, >65 KLF1 variants, causing different erythroid phenotypes, have been described. The Klf1 Nan variant, a single amino acid substitution (p.E339D) in the DNA binding domain, causes hemolytic anemia and is dominant over wildtype KLF1. Here we describe the effects of the Nan variant during fetal development. We show that Nan embryos have defects in erythroid maturation. RNA-sequencing of the Nan fetal liver cells revealed that Exportin 7 (Xpo7) was among the ~780 deregulated genes. This nuclear exportin is implicated in terminal erythroid differentiation; in particular it is involved in nuclear condensation. Indeed, KLF1 Nan fetal liver cells had larger nuclei and reduced chromatin condensation. Knockdown of XPO7 in wildtype erythroid cells caused a similar phenotype. We conclude that reduced expression of XPO7 is partially responsible for the erythroid defects observed in Nan erythroid cells.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1331-1331
Author(s):  
Yanming Li ◽  
Shuge Liu ◽  
Hongzhu QU ◽  
Yadong Yang ◽  
Nan Ding ◽  
...  

Abstract PDCD2 was first identified in human in 1995, which is highly homologous to a rat programmed cell death related gene Rp8. Nonetheless, up to now, there have been only 43 papers about PDCD2 published. Therefore, the role of PDCD2 in erythroid differentiation remains largely unknown. TF-1 cell is a erythroleukemia erythroblast, which is a good system for investigating the proliferation and differentiation of myeloid progenitor cells for its responsiveness to multiple cytokines.To study the function of PDCD2 in erythroid differentiation, we performed PDCD2 knockdown in TF-1 cells. The expression of PDCD2 was significantly decreased at both mRNA and protein level (Fig.1A, B). The expression of globin genes were detected by real-time qPCR, and the results indicated that ebsilon-, gamma-, beta-globin genes expression increased after PDCD2 knockdown in TF-1 cells (Fig.1C). Also, the expression of CD71 and CD235a at cell surface was measured by FACS, and our data showed that the percentage of CD71+CD235a+ cells was significantly increased by PDCD2 knockdown in TF-1 cells (Fig.1D). Furthermore, we induced TF-1 cells to erythroid differentiation by removing GM-CSF and adding EPO in culture medium. Both globin genes expression at mRNA level and CD71+CD235a+ expression at cell surface were measured at different time points during induction. With the progression of induction, expression of globin genes and the percentage of CD71+CD235a+ cells were increased in both control and PDCD2 knockdown cells (Fig.2). However, at same time points, increased expression of globin genes were detected in PDCD2 knockdown cells, compared to control cells (Fig.2A, B). Similar pattern was also observed in terms of the expression of CD71+CD235a+ at cell surface (Fig.2C). GATA-1 and KLF1 are the two most important transcription factors during erythroid differentiation. We detected the expression of the two TFs after PDCD2 knockdown in TF-1 cells. We found that expression of GATA-1 and KLF1 was augmented after PDCD2 knockdown (Fig.3A, B), and the augmentation remained during the induced differentiation (Fig.3C, D). To get a further insight of how PDCD2 regulates erythrioid differentiation, we performed a PDCD2 ChIP assay in normal TF-1 cells. Our data showed that there was an enrichment of PDCD2 at GATA-1 promoter region (data not shown). Since there is no evidence indicating that PDCD2 is a transcription factor, we assumed that PDCD2 bind at DNA sequence indirectly through interacting with other proteins. We thus performed co-IP assay of HDAC8 and HDAC1 in TF-1 cells and found that PDCD2 interacted with HDAC8 (data not shown). These data indicate that PDCD2 knockdown promoted induced erythroid differentiation in TF-1 cells. The promotion may be partially explained by increased expression of GATA-1 and KLF1. PDCD2 interacting with HDAC8 and binding at GATA-1 promoter region may play a role during erythroid differentiation. Figure 1 Figure 1. Figure 2 Figure 2. Figure 3 Figure 3. Disclosures No relevant conflicts of interest to declare.


2013 ◽  
Vol 382 (1-2) ◽  
pp. 127-136 ◽  
Author(s):  
Yan-Ni Ma ◽  
Ming-Tai Chen ◽  
Zhi-Kui Wu ◽  
Hua-Lu Zhao ◽  
Hai-Chuan Yu ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2323-2323
Author(s):  
Tzu-Fang Lou ◽  
Shuguang Ma ◽  
Ashley Williams ◽  
Betty S. Pace

Abstract The control of human hemoglobin gene switching from fetal γ-globin to adult β-globin is important in erythroid maturation and treatment approaches for sickle cell disease. Drugs that reverse the γ to β-globin ratio (γ/β) have been used effectively in clinical settings. Interaction between globin genes and the locus control region is a widely accepted mechanism for competitive γ-gene silencing. Studies in K562 cells are limited and often not correlated with in vivo response in part due to a lack of β-globin expression. Therefore, we tested a KU812 erythroleukemia cell line containing active γ- and β-globin genes, as an in vitro model for screening HbF inducers. Cell viability was monitored by trypan blue exclusion and globin mRNA measured in K562 cells (γ-globin) and KU812 cells (γ and β globin) by quantitative-PCR (q-PCR). Treatments with the histone deacetylase (HDAC) inhibitors: sodium butyrate (NaB, 2mM), trichostatin A (TSA, 0.2–0.5μM), suberoylanilide hydroxamic acid (SAHA, 2.5–5μM) or STI571 (0.5μM) were completed. In addition, hydroxyurea (HU, 100μM) was tested as an HbF inducer control. Results: KU812 cells grown in suspension in IMDM and 10% fetal bovine serum showed viability and proliferative capacity similar to K562 cells. Drug concentrations for SAHA and TSA were decreased to attain acceptable cell viability for 48hr inductions. Controls studies in K562 cells treated with NaB and TSA showed 60–80% viability and 3.2-fold increased γ-globin expression normalized by GAPD (Gγ/GAPD, See Table). For the new agents SAHA (0–5μM) and STI571 (0–2μM) dose response studies showed 62% and 66% viability in K562 cells at 50μM, and a concomitant increase in γ/GAPD mRNA from 1.7 to 40-fold respectively. For KU812 cells at steady-state we observed 40-fold higher γ vs. β globin mRNA. Inductions with NaB, TSA and SAHA produced increased γ/β ratios up to 4.5-fold along with β-gene repression; a scenario desirable in sickle cell patients. Although STI571 induced γ-globin 8-fold this effect was countered by 14-fold β-globin induction to produce a net reversal of the γ/β ratio (0.58). Control studies with HU showed on average an 1.4-fold increase in γ/β globin by 48 hrs suggesting that modest changes in this competitive ratio is sufficient to achieve significant clinical benefits. These data support KU812 cells as a good model for testing competitive γ-globin activation by drug inducers, which could not be ascertained in K562 cells. We plan to correlate: γ/β ratios, HbF protein (by ELISA; Bethyl Lab. INC., Montgomery, TX, and 3) histone H3 and H4 acetylation levels (by western blot) in K562 and KU812 cells. Preliminary ELISA data showed 2.4 to 6.4-fold increase in HbF protein by NaB and SAHA respectively in K562 cells. This data combined with the favorable γ/β globin ratio observed with SAHA suggests this agent might be an efficacious HbF inducer in vivo. Summary: The γ/β globin mRNA ratio was determined in KU812 cells to establish a better measure of drug-mediated HbF induction in vivo. We will screen novel agents for therapeutic potential. KU812 cells might also serve as a good model to study the elusive mechanisms involved in the γ to β globin switch during erythroid maturation. K562 KU812 1TSA 0.5μM; 2TSA 0.2μM; 3SAHA 5μM;†SAHA 2.5μM;* p<0.05 HbF Inducer Drug conc. γ/GAPD Fold Inc. γ/β Fold Inc. None 0 1.0 1.0 NaB 2mM 3.2* 4.51* TSA 0.2–0.5 μM 2.11* 1.612 SAHA 2.5–5 μM 1.73 3.75†* STI571 M μ 0.5 40.0* 0.58* HU μ 100M 1.3* 1.35*


2020 ◽  
Vol 4 (7) ◽  
pp. 1464-1477 ◽  
Author(s):  
Emilie-Fleur Gautier ◽  
Marjorie Leduc ◽  
Meriem Ladli ◽  
Vincent P. Schulz ◽  
Carine Lefèvre ◽  
...  

Abstract Murine-based cellular models have provided and continue to provide many useful insights into the fundamental mechanisms of erythropoiesis, as well as insights into the pathophysiology of inherited and acquired red cell disorders. Although detailed information on many aspects of these cell models is available, comprehensive proteomic data are lacking. This is a critical knowledge gap, as proteins are effectors of most biologic processes. To address this critical unmet need, proteomes of the murine cell lines Friend erythroleukemia (MEL), GATA1 erythroid (G1ER), and embryonic stem cell–derived erythroid progenitor (MEDEP) and proteomes of cultured murine marrow–derived erythroblasts at different stages of terminal erythroid differentiation were analyzed. The proteomes of MEDEP cells and primary murine erythroid cells were most similar, whereas those of MEL and G1ER cells were more distantly related. We demonstrated that the overall cellular content of histones does not decrease during terminal differentiation, despite strong chromatin condensation. Comparison of murine and human proteomes throughout terminal erythroid differentiation revealed that many noted transcriptomic changes were significantly dampened at the proteome level, especially at the end of the terminal differentiation process. Analysis of the early events associated with induction of terminal differentiation in MEDEP cells revealed divergent alterations in associated transcriptomes and proteomes. These proteomic data are powerful and valuable tools for the study of fundamental mechanisms of normal and disordered erythropoiesis and will be of broad interest to a wide range of investigators for making the appropriate choice of various cell lines to study inherited and acquired diseases of the erythrocyte.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1479-1479 ◽  
Author(s):  
Alireza Ghamari ◽  
Gabriela Pregerning ◽  
Ernest Fraenkel ◽  
Alan B. Cantor

Abstract Erythroid differentiation is controlled by the dynamic exchange of GATA family transcription factors. During early erythroid maturation, high GATA2 levels activate progenitor genes such as c-kit-, c-myc, and GATA2 itself. In contrast, GATA1 levels are low in early progenitor cells, but rise during terminal maturation. During this process GATA1 turns off GATA2 controlled early progenitor genes and activates terminal maturation genes, such as globin genes, heme biosynthesis enzymes, and iron transporters. This involves the exchange of GATA1 for GATA2 at key chromatin sites, the so-called "GATA factor switch". GATA factor switching is facilitated by the much shorter half-life of GATA2 (~30-60 min) compared to GATA1 (>4-6 hrs). We and others recently demonstrated that the E3 ubiquitin ligase adaptor protein FBW7 contributes to GATA2's relative instability. This prompted us to dissect the role of FBW7 during GATA switching and erythroid differentiation. We deleted the Fbw7 gene using CRISPR/Cas9 gene editing in the inducible G1-ER murine erythroid cell line. This resulted in the delayed clearance of GATA2 during differentiation. RNA-seq analysis at an early time points (7 hr) demonstrated impaired repression of GATA2 regulated genes and reduced activation of GATA1 target genes. Globally, altered gene expression was enriched for GATA factor switch genes. This ultimately resulted in delayed erythroid maturation. We also found that Fbw7 mRNA transcript levels increase during erythroid maturation in wild type cells. We identified a site ~40kb upstream of the Fbw7 gene transcriptional start site, which is itself a GATA factor switch site. We propose that FBW7 facilitates GATA factor switching by promoting the clearance of GATA2 from GATA factor switch sites. Moreover, we suggest that GATA factor switching at the Fbw7 locus itself reinforces the commitment of erythroid cells to terminal maturation, by enhancing the clearance of GATA2 and other Fbw7 progenitor target gene proteins such as c-Myc and c-Myb. As Fbw7 recognition of GATA2 requires phosphorylation of GATA2's degron motif, this suggests that signaling pathways, acting through Fbw7, may modulate erythroid maturation kinetics. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1117-1117
Author(s):  
Ramona Pop ◽  
Srijana Ranjit ◽  
Merav Socolovsky

Abstract The essential role of glycoprotein hormone erythropoietin (Epo) and its receptor, EpoR, in erythroid development is well established: both the EpoR−/− and Epo−/− mouse embryos die on embryonic day 13 (E13) due to failure of definitive erythropoiesis in fetal liver (Wu et al. 1995). It has been suggested that Epo’s principal role during erythropoiesis is to protect erythroid progenitors from apoptosis (Koury and Bondurant, Science 1990). Bcl-xL, an anti-apoptotic member of the bcl-2 family, is induced by EpoR signaling in erythroid cells via the Jak2/Stat5 pathway (Silva et al., Blood 1996; Socolovsky et al., Cell, 1999). Bcl-xL is essential for erythroid maturation: bcl-xL−/− embryos die in utero at the same stage as as EpoR−/− mice, lacking definitive erythropoiesis (Motoyama et al., Science 1995; J Exp Med, 1999). Recenlty, it has been shown that over-expression of bcl-xL in primary wild-type erythroblasts confers Epo independence on these cells in vitro and allows them to complete their differentiaion into red blood cells (Dolznig et al., Curr Biol, 2002). Here we reasoned that if the principal function of EpoR signaling is suppression of apoptosis via bcl-xL, it should be possible to rescue all aspects of erythroid differentiation in EpoR−/− fetal liver progenitors by retrovirally-transducing these cells with bcl-xL. We infected EpoR−/− fetal liver progenitors with bicistronic retroviral vectors expressing either bcl-xL or EpoR, each linked via an IRES sequence to a GFP reporter. Control EpoR−/− cells were infected with ‘empty’ bicistronic vector. Infection rates were in excess of 30% for all constructs, and transduced cells were identified for further analysis using GFP fluorescence. We examined terminal differentiation of the transduced EpoR−/− cells over the ensuing 48 hours, using several distinct assays, including their expression of the cell-surface differentiation markers CD71 and Ter119 by FACS, their ability to give rise to CFU-e colonies in semi-solid medium, their cell-cycle status using DNA content analysis and BrdU incorporation, and their maturation and hemoglobinization by diaminobenzidine staining and light microscopy. We found that EpoR−/− progenitors transduced with bcl-xL were protected from apoptosis, and underwent morphological changes characteristic of erythroid maturation, including decreasing cell size, nuclear condensation and expulsion, and accumulation of hemoglobin. These cells also upregulated the erythroid-specific cell surface marker Ter119. However, unlike EpoR−/− cells transduced with EpoR, bcl-xL -transduced cells did not express high levels of CD71, and failed to give rise to CFU-e colonies in semi-solid medium. Instead, they gave rise to small colonies of 6 cells or less. Cell cycle analysis showed that, throughout the 48 hours of erythroid terminal differentiation, the population of bcl-xL-transduced EpoR−/− cells had a lower fraction of cells in S-phase than control, EpoR-transduced EpoR−/− cells. The cell-cycle status of control, terminally-differentiating wild-type erythroid fetal liver progenitors was not altered by transduction with bcl-xL, excluding the possibility that it directly inhibits S-phase. Taken together our results indicate that bcl-xL does not rescue all aspects of erythroid differentiation in EpoR−/− erythroid progenitors. Specifically, the proliferative program during erythroid terminal differentiation is directly dependent on EpoR signaling, and is not simply a default pathway secondary to EpoR’s anti-apoptotic effect.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1203-1203
Author(s):  
Francine Rezzoug ◽  
Shelia Thomas ◽  
Donald Max Miller

Abstract Introduction Guanine-rich DNA sequences can form stable four stranded structures by folding of the DNA strands, forming stacks of G-tetrads called G-quadruplex. G-quadruplex forming sequences are found in eukaryotic telomeres, in promoter regions and in noncoding regions of the genome. Although the function of these structures has not yet been identified it has been suggested that they play a negative regulatory role at the transcription level. The most studied G-quadruplex sequences (AS1411 and PU27) show activity in reducing growth and invasiveness of malignant cells, as well as increasing cell death of a large array of tumors with promising therapeutic applications. We identified two G-quadruplex forming sequences in the human beta globin cluster (Hbd and Hbg2). We confirmed quadruplex formation by these sequences using circular dichroism spectroscopy. We also observed in Southern blot assay that these oligo-sequences bind to specific β-globin gene fragments after EcoRI digestion. Finally we demonstrate that neither Hbd nor Hbg2 (5 or 10µM) were growth inhibitory to four different leukemia cell lines after 6 days culture measured by MTT. Results The β-globin cluster contains 5 functional globin-like genes and long range regulator elements, the expression of each of these genes is tightly regulated during development (from embryo to fetus to adult), in cell type and in the course of differentiation/maturation of the erythroid lineage. Recent studies have demonstrated the importance of folding and looping of the DNA in the control of globin genes expression. We hypothesized that the G-quadruplex forming sequences within the β-globin gene cluster participate in regulation and splicing of the different globin genes during erythroid maturation. The erythroleukemia cell line K562 was used in this study because of its ability to differentiate into erythrocytes under Hemin stimulation. K562 cells were divided into 6 groups of treatment and cultured for 3 days in the presence or absence of Hemin (30µM) with or without either Hbd or Hbg2 oligonucleotides (5µM). Erythroid differentiation was evaluated by quantitative QRT-PCT for the expression of β-, γ- and ε-globin genes as well as α-globin and by flow-cytometry for the presence of β-, γ- and ε-globin; α-globin and CD235a (Glycophorin A) were used as controls. The data reveal a difference in gene expression for all globin genes when K562 were exposed to Hbd or Hbg2 only, interestingly, a significant up-regulation of β-globin was observed only in Hbg2 treated cells. Similarly in K562 induced to differentiate with Hemin show up-regulation of γ-, ε- and α- globin, the addition of Hbd or Hbg2 oligonucleotides to Hemin at the beginning of the culture slightly improve the expression of γ- and ε- however there was no difference for the expression of β- globin in the hemin +/- Hb oligonucleotides treated cells compared to control untreated cells. Flow-cytometry experiments confirmed the increase in the expression of each globin gene in all treatments compared to control. Altogether the data suggest a role for Hbd and Hbg2 in the regulation of β-globin gene transcription and therefore in the differentiation of K562 in erythroid lineage. Conclusions The implications of our findings are multiple. 1) the presence of G-quadruplex sequences in a gene complex not involved in growth regulation or oncogenesis; 2) to our knowledge this is the first time that G-quadruplex sequences are shown to specifically alter the expression of the gene in which they are located; and 3) the potential use of globin G-quadruplex to modify the globin gene expression therapeutically. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document