Endothelial Cell-Selective Adhesion Molecule (ESAM) Is Required for the Ontogeny of Definitive Hematopoietic System in Mice

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3874-3874
Author(s):  
Tomoaki Ueda ◽  
Takafumi Yokota ◽  
Yasuhiro Shingai ◽  
Yukiko Doi ◽  
Tomohiko Ishibashi ◽  
...  

Abstract We previously reported that endothelial cell-selective adhesion molecule (ESAM), which was initially identified as an endothelial cell-specific antigen, is an effective lifelong hematopoietic stem cell (HSC) marker in mice and humans (Yokota Blood 2009; Ishibashi Exp Hematol 2016). Prior to the advent of the first definitive HSC, ESAM was already expressed on hemogenic endotherium in the developing aorta of murine embryos. We also reported that ESAM expression on HSCs is functionally important for adult hematopoiesis because ESAM deficiency causes life-threatening myelo-suppression, especially severe anemia, after administration of 5-fluorouracil (5-FU) (Sudo J Immunol 2012, PLoS One 2016). Collective data obtained from the genotyping of newborn ESAM knockout (KO) mice suggested that the number of homozygous (homo) ESAM KO mice was about half of that expected as per the Mendelian ratio. The functional significance of ESAM in the development of hematopoiesis, however, has yet to be determined. Thus, in the present study we have analyzed how ESAM deletion affects hematopoietic development in fetuses of ESAM KO mice. Unexpectedly, the frequency and the size of ESAM homo KO fetuses were comparable to those of wildtype (WT) or heterozyqous KO littermates at embryonic day (E) 14.5. However, we found that the liver of ESAM homo KO fetuses contained significantly fewer mononuclear cells. FACS analyses revealed that all the tested hematopoietic cell populations, including lineage- Sca1+cKitHigh (LSK) and LSK CD150+ CD48- HSCs, B220+ B cells, Gr1+ myeloid cells, and Ter119+ erythroid cells, were significantly decreased in the ESAM homo KO fetal liver. Erythroid differentiation was thought to be delayed in ESAM homo KO fetuses because Ter119+ mature erythroid cells significantly decreased whereas CD71+ Ter119- immature cells significantly increased. HSC-enriched LSK cells from E14.5 ESAM homo KO mice produced fewer numbers of blood cells in MS5 co-culture than those from the others, particularly B-lineage cells, suggesting that the growth and differentiation potential of HSCs is impaired in the absence of ESAM. Although ESAM-deficient fetuses grew without an apparent malfunction in developing organs until E14.5, we found that life-threatening events occurred in 3 days following E14.5. Approximately half of homo KO fetuses exhibited severe anemia at E15.5 and died before E17.5. Quantitative real-time PCR analyses from E16.5 ESAM KO homo fetal livers revealed a significant reduction in messenger RNA (mRNA) levels for adult globins (α and β major). In addition, the mRNA level for an erythroid-specific isoenzyme of 5-aminolevulinic acid synthase 2 (ALAS2), the first and rate-limiting enzyme in the heme biosynthesis pathway, was also found to be reduced in the liver of E16.5 ESAM KO homo fetuses. To learn more about molecular mechanisms involved in the developmental failure of hematopoiesis in ESAM KO fetuses, we performed RNA sequencing (RNA-seq) of LSK cells sorted from E14.5 WT and ESAM KO homo mice. We found that, while transcripts for embryonic globins (ζ and Ey) remained substantially, those for adult globins (α, β major, and β minor) were markedly down-regulated in ESAM-KO HSCs. The results suggested that ESAM deficiency disturbs the globin switch from embryonic to adult type. ALAS2 was insufficiently induced in LSK cells of ESAM KO fetal livers, which presumably results in defects in heme biosynthesis. During the embryonic development, rapid and explosive production of erythroid cells is imperative to support the growth and survival of fetuses. To meet the physiological requirement, definitive erythropoiesis occurs in the developing liver and replaces primitive erythropoiesis. Our data suggest that ESAM expression is indispensable for the development of definitive erythropoiesis. In conclusion, we have revealed that ESAM plays a critical role in the development of definitive hematopoiesis. Approximately half of ESAM KO homo fetuses died between E15.5 and E17.5, at least partly due to the delay of adult hemoglobin synthesis in the absence of ESAM. Disclosures Yokota: SHIONOGI & CO., LTD.: Research Funding. Doi:Yakult Honsha Co.,Ltd.: Speakers Bureau. Shibayama:Novartis Pharma: Honoraria, Research Funding, Speakers Bureau; Celgene: Honoraria, Research Funding, Speakers Bureau; Takeda: Speakers Bureau; Chugai Pharmaceutical: Speakers Bureau; Ono Pharmaceutical: Speakers Bureau. Kanakura:Chugai Pharmaceutical: Research Funding; Pfizer: Research Funding; Shionogi: Research Funding; Kyowa Hakko Kirin: Research Funding; Fujimotoseiyaku: Research Funding; Toyama Chemical: Research Funding; Bristol Myers: Research Funding; Alexionpharma: Research Funding; Nippon Shinyaku: Research Funding; Astellas: Research Funding; Eisai: Research Funding.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 814-814
Author(s):  
Peng Huang ◽  
Scott A. Peslak ◽  
Xianjiang Lan ◽  
Eugene Khandros ◽  
Malini Sharma ◽  
...  

Reactivation of fetal hemoglobin in adult red blood cells benefits patients with sickle cell disease and β-thalassemia. BCL11A is one of the predominant repressors of fetal γ-globin transcription and stands as an appealing target for therapeutic genome manipulation. However, pharmacologic perturbation of BCL11A function or its co-regulators remains an unmet challenge. Previously, we reported the discovery of the erythroid-enriched protein kinase HRI as a novel regulator of γ-globin transcription and found that HRI functions in large part via controlling the levels of BCL11A transcription (Grevet et al., Science, 2018). However, the specific mechanisms underlying HRI-mediated modulation of BCL11A levels remain unknown. To identify potential HRI-controlled transcription factors that regulate BCL11A, we performed a domain-focused CRISPR screen that targeted the DNA binding domains of 1,447 genes in the human erythroid cell line HUDEP2. Activating transcription factor 4 (ATF4) emerged as a novel γ-globin repressor. Prior studies reported that ATF4 production is under positive influence of HRI. Specifically, HRI phosphorylates translation factor EIF2α which in turn augments translation of ATF4 mRNA. As expected, HRI deficiency reduced ATF4 protein amounts in HUDEP2 and primary erythroid cells. We further found that the degree of γ-globin reactivation was similar in ATF4 and HRI-depleted cells. ATF4 ChIP-seq in both HUDEP2 and primary erythroblast identified 4,547 and 3,614 high confidence binding sites, respectively. Notably, we did not observe significant enrichment of ATF4 binding or even the presence of an ATF4 consensus motif at the γ-globin promoters, suggesting that ATF4 regulates the γ-globin genes indirectly. However, ATF4 specifically bound to one of the three major BCL11A erythroid enhancers (+55) in both cell types. This was the sole binding site within the ~0.5Mb topologically associating domain that contains the BCL11A gene. Eliminating this ATF4 motif via CRISPR guided genome editing lowered BCL11A mRNA levels and increased γ-globin transcription. Capture-C showed that ATF4 knock-out or removal of the ATF4 site at the BCL11A (+55) enhancer decreased chromatin contacts with the BCL11A promoter. Forced expression of BCL11A largely restored γ-globin silencing in cells deficient for ATF4 or lacking the ATF4 motif in the BCL11A (+55) enhancer. An unexplained observation from our prior study was that HRI loss did not significantly lower Bcl11a levels in murine erythroid cells. Therefore, we mutated the analogous ATF4 motif in the Bcl11a enhancer in the murine erythroid cell line G1E. Unlike in human cells, Bcl11a mRNA synthesis was decreased only very modestly, and there was no effect on the murine embryonic globin genes whose silencing requires Bcl11a. This suggests that the species specific regulation of BCL11A by HRI results from divergent functional roles of ATF4 binding at the BCL11A (+55) enhancer. In sum, our studies uncover a major pathway that extends linearly from HRI to ATF4 to BCL11A to γ-globin. Moreover, these results further support HRI as a pharmacologic target for the selective regulation of BCL11A and γ-globin. Disclosures Blobel: Pfizer: Research Funding; Bioverativ: Research Funding.


1997 ◽  
Vol 273 (5) ◽  
pp. H2490-H2497 ◽  
Author(s):  
Tatjana R. Faruqi ◽  
Serpil C. Erzurum ◽  
F. Takao Kaneko ◽  
Paul E. Dicorleto

Polyinosinic-polycytidylic acid [poly(I-C)] is a synthetic double-stranded RNA (dsRNA) that simulates a viral-infected state in cells. It has been shown that viral infection, as well as poly(I-C), stimulates leukocyte adhesion to endothelial cell (EC) monolayers and that this is mediated through the surface expression of the adhesion molecules E-selectin, vascular cell adhesion molecule 1 (VCAM-1), and intercellular adhesion molecule 1. We have tested the involvement of nitric oxide (NO) in poly(I-C)-induced monocytic cell adhesion to human vascular EC. Using primary cultured EC for these studies, we confirmed the results from previous reports that these cells have higher basal levels of NO production than passaged cells. Poly(I-C)-induced monocytic cell adhesion to primary EC was concentration-dependently inhibited by 40–74% by the nitric oxide synthase (NOS) inhibitor NG-methyl-l-arginine (l-NMA), as well as three other NOS inhibitors, without significantly affecting interleukin-1β-induced adhesion.l-NMA inhibited poly(I-C)-induced surface expression of E-selectin and VCAM-1 by 25 and 45%, respectively, and mRNA levels of E-selectin and VCAM-1 by 62 and 74%, respectively. Primary EC transiently transfected with a plasmid containing an E-selectin promoter-driven luciferase reporter gene showed that l-NMA treatment reduced poly(I-C)-induced E-selectin promoter activity to basal levels. Electrophoretic mobility shift analysis indicated that poly(I-C)-induced nuclear factor-κB (NF-κB) binding to a radiolabeled oligonucleotide corresponding to the consensus NF-κB binding domain of the E-selectin promoter was decreased byl-NMA pretreatment. Hence, NO appears to augment E-selectin gene expression in response to poly(I-C) at the transcriptional level in vascular EC. Collectively, these data support the hypothesis that NO augments poly(I-C)-induced EC activation. These data suggest a novel role for NO as a response mediator in dsRNA-induced leukocyte adhesion to EC.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2094-2094
Author(s):  
Aurelie Desgardin ◽  
Tatiana Abramova ◽  
Jenny Lin ◽  
Eun-Hee Shim ◽  
John M Cunningham

Abstract Abstract 2094 Krüppel-like factor 1 (KLF1) is a zinc finger-encoding transcription factor that recognizes CACC elements, and is essential for maximal erythroid-specific gene transcription. Several critical mechanisms dependent on KLF1 and required for gene activation have been elucidated, predominantly using the beta-globin locus. KLF1 has been associated with the ordered recruitment of SWI/SNF and RNA polymerase-II complexes, necessary for chromatin remodeling and gene transcription respectively. KLF1 has also been reported to influence erythroid-specific heme biosynthesis. Studies in KLF1-null fetal erythroblasts and a KLF-1 deficient cell line have demonstrated that mRNA levels of the first three enzymes of the biosynthetic pathway are underrepresented. However, although in vitro studies of the rate-limiting enzymes ALAS2 and PBGD suggested a potential regulatory role for KLF1, in vivo studies failed to validate these findings. ALAD is the second enzyme of the pathway. Complete loss of ALAD expression in erythroid cells results in catastrophic events during zebrafish ontogeny. Interestingly, no human erythropoietic defect has been reported as a consequence of aberrant ALAD expression. To extend the analysis of KLF1's regulation of heme biosynthesis, we evaluated KLF1 binding of enzyme regulatory sequences by EMSA and ChIP studies, identifying a KLF1 binding CACC element in the erythroid-specific ALAD promoter. This regulatory element was transactivated specifically by a KLF1 transgene in KLF1-deficient cells. Using a unique 4-OH-Tamoxifen (4-OHT) mediated KLF1-inducible erythroid cell line (K1-ERp), we identified KLF1 as an essential, and early (within 2 hours of induction) activator of transcription of the endogenous ALAD, but not ALAS2 or PBGD genes. Further studies in K1-ERp cells, including DNAseI hypersensitivity and ChIP assays revealed that KLF1 occupancy at the erythroid-specific ALAD promoter triggers a series of molecular events including histone modifications, and enhanced recruitment of the sequence-specific transcription factors, GATA-1, NF-E2 and the TAL-1/SCL multiprotein complex. Importantly, we identified differences in the kinetics of recruitment of the closely related histone acetyltransferases proteins CBP and p300 and the SWI/SNF ATPase Brg1. The latter complex was recruited subsequent to KLF1 binding, although the ALAD promoter was already DNAseI hypersensitive. These results suggest strongly that KLF1 plays a major role in the regulation of heme biosynthesis in erythroid cells. Furthermore, our data challenges a model in which an identical temporal cascade of molecular events are required for transcription at KLF1-dependent promoters. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4295-4295
Author(s):  
Valeria Visconte ◽  
Steffan T. Nawrocki ◽  
Bartlomiej P Przychodzen ◽  
Kevin R. Kelly ◽  
Mei Yin ◽  
...  

Abstract Autophagy is a conserved mechanism of protein degradation that plays a physiologic role in iron homeostasis. The RARS subtype of MDS is characterized by inefficient erythropoiesis and the presence of erythroblasts with iron-laden mitochondria (ring sideroblasts, RS). Patients with RARS exhibit an accumulation of iron due to the inability of their erythroid precursors to properly process iron intended for hemoglobin synthesis. RARS erythroid cells display increased autophagy compared to cells from healthy subjects and mice lacking the essential autophagy genes Atg1 and Atg7 develop progressive anemia due to the inefficient removal of defective mitochondria. We previously showed that the presence of SF3B1 mutations, which are frequent in RARS patients, are associated with the presence of increased mitochondrial iron content and low levels of apoptosis. This finding suggests that cell death-independent mechanisms may temper excess iron by triggering the clearance of mitochondria during erythroid maturation. RNA sequencing analyses have shown that the autophagy pathway is upregulated in SF3B1MUT cells. We investigated the role of mitochondrial autophagy in the alleviation of iron-related damage in RARS erythroid cells and the potential therapeutic benefit of autophagy-stimulating agents for the selective improvement of erythropoiesis and iron homeostasis in SF3B1MUT RARS cells. Transmission electron microscopy (TEM) demonstrated an accumulation of iron in the mitochondria of SF3B1MUT (K700E, n=4) compared to SF3B1WT RARS erythroblasts. Flow cytometry confirmed increased mitochondrial iron in SF3B1MUT (n=10) compared to SF3B1WT (n=10) RARS cells (82% ± 10 vs. 24% ± 5; P = 0.004). Autophagic vacuolization of the cytoplasm and an increased number of autophagosomes were found in SF3B1MUT (K700E) compared to SF3B1WT erythroblasts (n=2). The mRNA expression levels of numerous autophagy genes were elevated in SF3B1MUT vs. SF3B1WT RARS cells: ATG complexes (ATG2A/B, FC=2; ATG4A, FC=2; ATG9A, FC=5; ATG4C, P=0.05; ATG18 (FC=4.8; P= 0.02), autophagy-initiating kinases (ULK1, FC=2; ULK3, FC=3.9; P=0.05), and cathepsins involved in late stage autophagy (CTSL1, FC=20; CTSD, FC=5.8; P=0.05; CTSB, FC=2.1; CTSE, FC=5.9; CTSD, FC=2; P=0.01). qRT-PCR confirmed that SF3B1MUT cells expressed elevated mRNA levels of selected genes. We next administered 3 FDA-approved drugs with established autophagy-stimulating properties [temsirolimus, metformin, arsenic trioxide (ATO)] to a transgenic SF3B1 mouse model (SF3B1+/-) that exhibits anemia, dysplasia, and RS (Visconte, J Hematol Oncol 2014) to evaluate drug-mediated improvement of erythropoiesis and autophagic clearance of excess iron. Eight-month-old SF3B1+/- mice (n = 10 per group) were treated with vehicle control and the following: temsirolimus (10 mg/kg i.p. 5d/week for 2 wk), metformin (250 mg/kg/d, gavage for 2 wk), and ATO (10 mg/kg i.p. 5 d/wk for 2 wk). All agents were well tolerated and triggered morphologic features of autophagy including increased autophagosome-like structures by TEM. No effects on BM cellularity and/or dysplasia were noted, although changes in the morphology of myeloid cells (numerous swollen nuclei) were detected by Wright stain. Mitochondria that were engulfed in autophagosomes were frequently seen. After 2 weeks, temsirolimus-treated SF3B1+/- mice showed an incremental increase in hemoglobin by 1.2 g/dL (7.9 vs. 6.6 g/dL; P=0.07) and in mean corpuscular volume (43.5 vs. 42.4 fL; P=0.08). Erythropoiesis was improved as shown by increased levels of CD71-positive cells by immunohistochemistry in cells post-temsirolimus treatment compared to pre-treatment (10-20% vs. 5%). Increased dividing erythroid cells with binucleation and budding were also observed in cells following temsirolimus treatment. These hematologic changes were not detected with ATO or metformin. In sum, our data support a role for activated autophagy in the pathogenesis of RARS and indicate that stimulating autophagy with approved existing drugs or novel investigational drugs may yield therapeutic benefit in SF3B1MUT RARS patients. Disclosures Kelly: Pharmacyclics: Consultancy, Speakers Bureau; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Speakers Bureau. Advani:Pfizer: Consultancy, Research Funding. Carraway:Novartis: Membership on an entity's Board of Directors or advisory committees; Incyte: Membership on an entity's Board of Directors or advisory committees; Baxalta: Speakers Bureau; Amgen: Membership on an entity's Board of Directors or advisory committees; Celgene Corporation: Research Funding, Speakers Bureau.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3200-3200
Author(s):  
Chun-Nam Lok ◽  
Prem Ponka

Abstract In erythroid cells iron uptake from transferrin (Tf) is utilized largely for heme synthesis. Here we provide evidence that Tf receptor (TfR) expression and cellular uptake of iron from Tf is stimulated by enhanced heme synthesis. Incubation of murine erythroleukemia (MEL) cells with 5-aminolevulinic acid (ALA) resulted in an increase in TfR expression accompanied by enhanced uptake of iron from Tf and incorporation of iron into heme. ALA-mediated enhancement of TfR mRNA expression was completely prevented by succinylacetone, an inhibitor of ALA dehydratase, and N-methylprotoporphyrin, an inhibitor of ferrochelatase, indicating that the effect of ALA required its metabolism to heme. Treatment of cells with ALA was associated with enhanced iron regulatory protein-2 (IRP-2) binding activity, which could be blocked by inhibitors of heme synthesis and supplementation of the culture medium with a permeable iron chelate or Tf. In all cases, IRP-2 activities were correlated exactly with TfR mRNA levels. Thus, in addition to the previously characterized transcriptional up-regulation of TfR expression in differentiating erythroid cells, increased TfR expression mediated by enhanced heme biosynthesis may ensure sufficient iron availability for optimal heme synthesis and prevent possible protoporphyrin accumulation under conditions of inadequate iron supply.


2000 ◽  
Vol 278 (5) ◽  
pp. G734-G743 ◽  
Author(s):  
Shigeyuki Kawachi ◽  
Stephen Jennings ◽  
Julian Panes ◽  
Adam Cockrell ◽  
F. Stephen Laroux ◽  
...  

The objectives of this study were to quantify cytokine mRNA levels and endothelial cell adhesion molecule message and protein expression in healthy wild-type and interleukin-10-deficient (IL-10−/−) mice that develop spontaneous and chronic colitis. We found that colonic message levels of IL-1, IL-6, tumor necrosis factor-α, interferon-γ, lymphotoxin-β, and transforming growth factor-β were elevated in colitic mice 10- to 35-fold compared with their healthy wild-type controls. In addition, colonic message levels of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and mucosal addressin cell adhesion molecule-1 (MAdCAM-1) were found to be increased 10-, 5-, and 23-fold, respectively, in colitic IL-10−/−mice compared with their wild-type controls. Immunoradiolabeling as well as immunohistochemistry revealed large and significant increases in vascular surface expression of colonic ICAM-1, VCAM-1, and MAdCAM-1 in the mucosa as well as the submucosa of the colons of colitic mice. These data are consistent with the hypothesis that deletion of IL-10 results in the sustained production of proinflammatory cytokines, leading to the upregulation of adhesion molecules and infiltration of mononuclear and polymorphonuclear leukocytes into the cecal and colonic interstitium.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3555-3555
Author(s):  
John D Belcher ◽  
Fuad Abdulla ◽  
Chunsheng Chen ◽  
Ping Zhang ◽  
Dormarie Rivera-Rodriguez ◽  
...  

We have previously shown that heme, a damage-associated molecular pattern (DAMP), released from sickle RBCs interacts with the innate immune toll-like receptor 4 (TLR4) on endothelium and blood cells to activate cell signaling. This activates the pro-inflammatory transcription factor NF-κB leading to the production of cytokines and adhesion molecules that promote inflammation, coagulation, and vaso-occlusion (VO). Previous studies in our lab have demonstrated that inhibition of TLR4 in Townes-SS mice with TAK242 reduces microvascular stasis in response to hemin, hypoxia/reoxygenation and LPS. To further delineate the role of TLR4 in sickle cell disease (SCD), we bred a global Tlr4-/- deficiency state into Townes-AA mice expressing normal human adult hemoglobin A and Townes-SS mice expressing sickle hemoglobin S. These Tlr-/- Townes mice were backcrossed 10 generations to homogenize their genetic background with our Tlr4+/+ Townes colony. Townes-SS Tlr4-/- mice developed less microvascular stasis than Townes-SS Tlr4+/+ mice in response to challenges with heme infusion (4% vs 30% at 1 hr, p<0.05, Fig 1A); LPS infusion (13% vs 36% at 1 hr, p<0.05, Fig 1B); and hypoxia/reoxygenation (3% vs 20% at 1 hr, p<0.05, Fig 1C). We next measured the complete blood counts, serum chemistries, and urine analytes in the Tlr4+/+ and -/- mice. We also quantified mRNA levels via qPCR for liver and kidney cytokines, adhesion molecule, heme oxygenase 1 (HO-1) and coagulation factors in all four genotypes (AA Tlr4+/+; AA Tlr4-/-; SS Tlr4+/+; SS Tlr4-/-). Hemoglobin levels in SS mice were lower than AA mice, but there were no differences between Tlr4+/+ and Tlr4-/- mice. WBC counts were greater in SS than AA mice, but there were no differences between Tlr4+/+ and Tlr4-/- mice. Bilirubin and liver function tests (LFTs) were higher in SS than AA, but there were no differences between Tlr4+/+ and Tlr4-/- mice. In the liver, adhesion molecules and HO-1 were higher in SS than AA, but there were no differences between Tlr4+/+ and Tlr4-/- mice. To define a potential mechanism for decreased microvascular stasis in response to hemin infusion in SS Tlr4-/-, we measured cytokines and adhesion molecule expression after heme challenge. When SS Tlr4+/+ mice were infused with hemin, we found increased expression of pro-inflammatory cytokines and cell adhesion molecules. SS Tlr4-/- mice infused with hemin had reduced expression of cytokines and cell adhesion molecules compared to SS Tlr4+/+ mice; most notably in IL-6 in the liver, adhesion molecules VCAM-1 and ICAM-1 in the liver and kidneys (p<0.05), and P-selectin and von Willebrand factor in the lungs (Fig 2). Our data thus far indicate that TLR4 of the innate immune system plays a critical role in VO and inflammation in challenged SS mice. We speculate that modulation of TLR4 with targeted inhibitors would be beneficial for SCD patients. Disclosures Belcher: Mitobridge, an Astellas Company: Consultancy, Research Funding. Vercellotti:Mitobridge, an Astellas Company: Consultancy, Research Funding.


1998 ◽  
Vol 5 (2-3) ◽  
pp. 179-188 ◽  
Author(s):  
MICHAEL J EPPIHIMER ◽  
J A N I C E RUSELL ◽  
R O B E R T LANGLEY ◽  
G I N A VALLIEN ◽  
DONALD C ANDERSON ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document