Acute Myeloid Leukemia Cells Require STAT5 for Survival.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1616-1616
Author(s):  
Martin Carroll ◽  
Tae Kon Kim ◽  
Kenichi Higashino ◽  
Alan M. Gewirtz

Abstract Signal transducer and activator of transcription (STAT) family proteins play crucial roles in the cytokine signaling pathways which regulate survival and proliferation of normal hematopoietic cells. However, the role of STAT proteins in regulating survival in leukemia remains poorly defined. STAT3 and STAT5, for example, have been reported to be constitutively activated in acute myeloid leukemia (AML) cells, however the physiologic significance of this activation is unknown. In order to better understand the role of STAT3 and STAT5 in AML biology, we studied their expression, activation, and requirement for cell growth in several AML cell lines and primary AML cells collected from patients at the University of Pennsylvania Cancer Center. We first confirmed the activation of STAT3 and STAT5 in primary AML cells by western blotting. An analysis of AML patient samples revealed elevated levels of constitutive STAT3 phosphorylation in 6 of 7 patient samples and constitutive STAT5 phosphorylation in 8 of 9 patient samples. In addition, 6 AML cell lines (K562, HL-60, MOLM-14, U937, KG-1, NB4) displayed constitutive STAT3 and STAT5 activation. In order to evaluate the functional significance of constitutive activation of STAT3 and STAT5 in AML cells, we designed and synthesized short interfering RNAs (siRNAs) to silence the expression of these proteins. For initial characterization, the siRNAs were delivered to MOLM-14 cells using an AMAXA nucleofector device (AMAXA, Inc. Gaithersburg, MD)(Program O-17/Solution V). Nucleofected siRNA diminished STAT3 expression by 85% at 24 hour but had little effect on cell proliferation (13%±3% decrease at 24 hour, 8%±4% at 48 hour, 5%±1% at 72 hour) compared to control siRNA treated cells. In contrast, STAT5 siRNA decreased STAT5 expression 80% at 24 hour compared to control treated cells but inhibited cell proliferation by 19%±1% at 24 hour, 22%±1% at 48 hour, 16%±3% at 72 hour in comparison to control siRNA treated cells suggesting a more important role for STAT5 in regulating cell proliferation. To study the effect of these siRNA molecules in primary AML cells, we first determined our ability to nucleofect primary cells by examining delivery efficiency of fluorescein labeled siRNA. Four different patient samples were evaluated and the mean ± SD of cells successfully transfected was 52%±12. Transfection of multiple AML patient samples with STAT3 siRNA decreased STAT3 expression but led to only modest decrease (6–18% at 48 hour, 7–36% at 72 hour) in AML cell survival. However, transfection of cells with STAT5 siRNA, but not control siRNA, led to a consistent decrease (25–54% at 48 hour, 21–60% at 72 hour) in AML cell survival. The decrease in survival was proportional to the transfection efficiency in the different samples. These results provide the first evidence that STAT5 expression and activation is necessary for the survival of primary AML cells. In fact, the data suggests a greater role of STAT5 in survival of primary cells than in survival of AML cell lines. Accordingly, STAT5 appears to be a legitimate target for the treatment of AML.

2020 ◽  
Vol 40 (4) ◽  
Author(s):  
Yi Xiao ◽  
Taoran Deng ◽  
Xi Ming ◽  
Jinhuang Xu

Abstract Tripartite motif (TRIM) 31 is a member of TRIM family and exerts oncogenic role in the progression and drug resistance of several cancers. However, little is known about the relevance of TRIM31 in acute myeloid leukemia (AML). Herein, we investigated the role of TRIM31 in AML. We examined the expression levels of TRIM31 in the blood samples from 34 patients with AML and 34 healthy volunteers using qRT-PCR. The mRNA levels of TRIM31 in human bone marrow stromal cells (HS-5) and five AML cell lines were also detected. Loss/gain-of-function assays were performed to assess the role of TRIM31 in AML cells proliferation, apoptosis and sensitivity to daunorubicin. The expression levels of pro-caspase 3, cleaved caspase 3, Wnt3a, β-catenin, cyclin D1 and c-Myc were measured using Western blot. TRIM31 expression levels were significantly up-regulated in AML patients and cell lines. Knockdown of TRIM31 suppressed cell proliferation and promoted apoptosis in AML-5 and U937 cells. The IC50 of daunorubicin was significantly decreased in TRIM31 siRNA (si-TRIM31) transfected cells. Oppositely, induced cell proliferation and decreased cell apoptosis were observed in pcDNA-3.1-TRIM31 transfected cells. Furthermore, knockdown of TRIM31 suppressed the activation of Wnt/β-catenin pathway in AML cells. Activation of Wnt/β-catenin pathway by LiCl abolished the effects of si-TRIM31 on cell proliferation, apoptosis and sensitivity to daunorubicin in AML cells. In conclusion, the results indicated that TRIM31 promoted leukemogenesis and chemoresistance to daunorubicin in AML. The oncogenic role of TRIM31 in AML was mediated by the Wnt/β-catenin pathway. Thus, TRIM31 might serve as a therapeutic target for the AML treatment.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2951-2951
Author(s):  
Raoul Tibes ◽  
Ashish Choudhary ◽  
Amanda Henrichs ◽  
Sadia Guled ◽  
Irma Monzon ◽  
...  

Abstract In order to improve treatment strategies for Acute Myeloid Leukemia (AML), we adapted a functional genomics approach using RNAi screening to identify molecular targets that are vital to the growth of AML. Herein we report the first large-scale kinome gene silencing screen in AML. A high throughput RNAi screen was developed for the efficient siRNA transfection of AML cell lines. Eight commercially available cationic lipid-based transfection reagents were tested for their ability to transfect several AML cell lines with siRNA. These extensive transfection optimization experiments identified two AML cells lines TF-1 and ML4 with up to 95–100 and 70–75% transfection efficiency respectively. Two independent replicate kinome screens were performed on both cell lines using a siRNA library targeting 572 kinase genes with 2 siRNA/gene. At 96 hours post transfection, cell proliferation was assessed and the B-score method was used to background correct and analyze the screening data. Several siRNA to specific kinases were identified that significantly inhibit cell proliferation of up to ~40–88%. Hits were defined at two thresholds: siRNA having a B-score of <−2 providing a statistically significance of p<0.05 (confidence of > 95%) and a cutoff B-score of <−1.5 providing greater than 87% confidence for each siRNA hit. Two different kinases (2 siRNA/gene/screen) were identified as major growth regulating kinases in TF1 cells with all 4 siRNA/gene having a B-score <−2. For two additional kinases, 3/4 siRNA for each gene had a Bscore <−2. Expanding the cutoff to a B-score <−1.5 three further kinases were targeted by at least 3/4 siRNA/gene. Similar analysis using the same criteria for ML4 cells identified one kinase targeted by 3/4 siRNA at a B-score <−2, seven kinases with 2/4 siRNA <−2 and two kinases with 3/4 siRNA/gene at a B-score of <−1.5. Common hits for both cell lines with at least 6/8 siRNA per gene from 4 screens performing at a B-score <−2 identified two kinases, one of them PLK1. Applying a B-score threshold of <−1.5, we identified five kinases for which at least 5/8 siRNA/gene from 4 screens met these criteria. Kinases/genes will be presented at the meeting.Confirmation of gene silencing and validation of growth response is currently underway for a subset of genes. Among the strongest hits are siRNA targeting PLK1, as well as siRNA targeting three other kinase-genes involved in regulating cell cycle progression and checkpoints and gene ontology (GO) analysis showed enrichment in cell cycle and cell cycle-checkpoint processes. Inhibitors against PLK1 and other kinase hits identified in the screen are in (pre)-clinical development and if confirmed, our experiments provide a strong rational to test these in AML. The application of RNAi based screening is useful in the identification of genes important in AML proliferation, which could serve as targets for therapeutic intervention and guide AML drug development. Furthermore, results from these types of functional genomics approaches hold promise to be rapidly translated into clinical application.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 915-915
Author(s):  
Stuart A Rushworth ◽  
Lyubov Zaitseva ◽  
Megan Y Murray ◽  
Matthew J Lawes ◽  
David J MacEwan ◽  
...  

Abstract Introduction Despite recent significant progress in the understanding of the biology of acute myeloid leukemia (AML) the clinical outcomes for the majority of patients diagnosed with AML presently remain poor. Consequently, there is an urgent need to identify pharmacological strategies in AML, which are not only effective but can be tolerated by the older, less well patient. Recently our group and others have shown that there is high Bruton’s Tyrosine Kinase (BTK) phosphorylation and RNA expression in AML. Moreover, our recent study described for the first time that ibrutinib and BTK-targeted RNA interference reduced factor-induced proliferation of both AML cell lines and primary AML blasts, as well as reducing AML blast adhesion to bone marrow stromal cells. Inhibition of BTK has been shown to regulate chronic lymphocytic leukemia, mantle cell lymphoma and multiple myeloma cell migration by inhibiting SDF1 (stromal derived factor 1) induced CXCR4 regulated cell trafficking. Here we report that in human AML ibrutinib in addition functions in a similar way to inhibit SDF1/CXCR4-mediated AML migration at concentrations achievable in vivo. Methods To investigate the role of BTK in regulating AML migration we used both pharmacological inhibitor ibrutinib and genetic knockdown using a lentivirus mediated BTK targeted miRNA in primary AML blasts and AML cell lines. We examined migration of AML blasts and AML cells to SDF-1 using Transwell permeable plates with 8.0µM pores. Western blotting was used to examine the role of SDF-1 in regulating BTK, AKT and MAPK activation in primary AML blasts. Results We initially examined the expression of CXCR4 in human AML cell lines and found that 4/4 cell lines were positive for CXCR4 expression. Next we examined the effects of ibrutinib on the migration of the AML cell lines U937, MV4-11, HL60 and THP-1 in response to SDF1. We found that ibrutinib can inhibit the migration of all AML cell lines tested. We tested the in-vitro activity of ibrutinib on SDF-1 induced migration in a spectrum of primary AML blasts from a wide age spectrum of adult patients and across a range of WHO AML subclasses and found that ibrutinib significantly inhibits primary AML blast migration (n=12). Next we found that ibrutinib can inhibit SDF-1 induced BTK phosphorylation and downstream MAPK and AKT signalling in primary AML blast. Finally to eliminate the problems associated with off target ibrutinib activity we evaluated migration of AML cells lines using genetic inhibition of BTK. The introduction of BTK-specific miRNA dramatically inhibited the expression of BTK in THP-1 and HL60 and reduced SDF1 mediated migration confirming that BTK is involved in regulating AML migration in response to SDF1. Conclusions These results reported here provide a molecular mechanistic rationale for clinically evaluating BTK inhibition in AML patients and suggests that in some AML patients the blasts count may initially rise in response to ibrutinib therapy, analgous to similar clinical observations in CLL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Author(s):  
Huan Cai ◽  
Makoto Kondo ◽  
Lakshmi Sandhow ◽  
Pingnan Xiao ◽  
Anne-Sofie Johansson ◽  
...  

Impairement of normal hmatopoiesis and leukemia progression are two well-linked processes during leukemia development and controlled by the bone marrow (BM) niche. Extracellular matrix proteins including laminin are important BM niche components. However, their role in hematopoiesis regeneration and leukemia is unknown. Laminin α4 (Lama4), a major receptor-binding chain of several laminins, is altered in BM niches in mice with acute myeloid leukemia (AML). So far, the impact of Lama4 on leukemia progression remains unknown. We here report that Lama4 deletion in mice resulted in impaired hematopoiesis regeneration following irradiation-induced stress, which is accompanied with altered BM niche composition and inflammation. Importantly, in a transplantation-induced MLL-AF9 AML mouse model, we demonstrate accelerated AML progression and relapse in Lama4-/-mice. Upon AML exposure, Lama4-/- mesenchymal stem cells (MSCs) exhibited dramatic molecular alterations including upregulation of inflammatory cytokines that favor AML growth. Lama4-/- MSCs displayed increased anti-oxidant activities and promoted AML stem cell proliferation and chemoresistance to cytarabine, which was accompanied by increased mitochondrial transfer from the MSCs to AML cells and reduced reactive oxygen species in AML cells in vitro. Similarly, we detected lower levels of reactive oxygen species in AML cells from Lama4-/- mice post-cytarabine treatment. Notably, LAMA4 inhibition or knockdown in human MSCs promoted human AML cell proliferation and chemoprotection. Together, our study for the first time demonstrates a critical role of Lama4 in impeding AML progression and chemoresistance. Targeting Lama4 signaling pathways may offer potential new therapeutic options for AML.


Blood ◽  
2000 ◽  
Vol 95 (12) ◽  
pp. 3765-3770 ◽  
Author(s):  
Jan-Jacob Schuringa ◽  
Albertus T. J. Wierenga ◽  
Wiebe Kruijer ◽  
Edo Vellenga

Abstract To explore the activation patterns of signal transducer and activator of transcription 3 (Stat3) in acute myeloid leukemia (AML), we examined whether the phosphorylation of tyrosine705 (Tyr705) and serine727 (Ser727) residues was abnormally regulated in cells from patients with AML. In 5 of 20 (25%) patients with AML, Stat3 was constitutively phosphorylated on Tyr705 and Ser727, which were not further up-regulated by treatment with IL-6. Furthermore, Stat3 was constitutively bound to the IRE response element in these cells as determined by electrophoretic mobility shift assay, and stimulation with IL-6 did not result in increased DNA binding. Interestingly, AML cells with constitutive Stat3 activation also secreted high levels of IL-6 protein. Treating these AML cells with anti-IL-6 resulted in restored IL-6–inducible Stat3 phosphorylation on both Tyr705 and Ser727 with low or undetectable basal phosphorylation levels in unstimulated cells. In contrast, treatment with anti-IL-1 did not result in altered Stat3 phosphorylation patterns. The constitutive IL-6 expression was associated with elevated levels of suppressor of cytokine signaling-1 (SOCS-1) and SOCS-3 mRNA expression, which were not down-regulated by anti-IL-6. These data indicate that the constitutive Stat3 activation in the investigated AML blasts is caused by high IL-6 secretion levels, thus stimulating the Jak/Stat pathway in an autocrine manner, a paracrine manner, or both.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3612-3612 ◽  
Author(s):  
Kaikai Huang ◽  
Bingjie Ding ◽  
Qingxiu Zhong ◽  
Xuejie Jiang ◽  
Xiaodong Li ◽  
...  

Abstract Hh/IGF-1R/PI3K/Akt/MRP1 pathway induce refractory acute myeloid leukemia Background: Although a great progress has made in the treatment of acute myeloid leukemia(AML), chemoresistance and relapse due to multidrug resistance (MDR) remains a therapeutic challenge. We have proven that LBH589 combined with Bortezomib had synergistic effects on proliferation, apoptosis and sensitivity to cytotoxic drugs via AKT and NF-κB pathways in chemoresistant HL60/ADR cells and refractory AML primary cells. Using gene chip analysis, we found that among those disregulated pathways, Hedgehog(Hh) signaling pathway was probably the upstream paths which could regulate others. Aims: The purpose of the study was to investigate the expression of Hh pathway in AML including primary cells and cell lines, analyze the relationship between Hh expression and clinical prognosis. Also investigate the biological effect of Hh pathway inhibitor NVP-LDE225 in reversing drug-resistance in HL60/adriamycin-resistant(ADR) cells and refractory AML primary cells and the underlying mechanisms. Besides, animal experiments were done to verify the results in vitro. Methods: Western blot assay were used to determine the protein expression of Ptch–Smo–Gli-1–Shh in HL60 and HL60/ADR cell lines and refractory or non-refractory primary AML cells. Kaplan–Meier curves were used to estimate Relapse-free survival(RFS) and Over-all survival(OS). HL60/ADR cells and refractory primary cells were treated with adriamycin or combined with NVP-LDE225 concentration among 0-10µM). Proliferation were evaluated by 3-(4,5)-dimethylthiahiazo (-z-yl)-3,5-di-phenytetrazoliumromide (MTT) assay and cell apoptosis were analyzed by Annexin V-FITC/PI staining through flow-cytometry(FCM). Intercellular adriamycin accumulation(MFI) were analyzed by FCM. The changes in protein levels of Gli-1–IGF-1R–p-IGF-1R–IRS-1–Akt–p-Akt–MRP-1–Bcl-2 were detected by Western blot. Besides, we used a nude mouse xenograft model to verify the anti-proliferative effects in vivo. Results: We found that the refractory primary AML cells and HL60/ADR cells correlated with higher activation of the Hh pathway, however in non-refractory primary cells or chemosensitive cell lines HL60, such activation was less pronounced. Higher protein expression of this pathway was related to higher recurrence rate and associated with poor relapse free survival (RFS) and poor overall survival (OS)(P=0.002)(a). NVP-LDE225, a potent and selective Hh inhibitor, significantly reverted resistance of adriamycin(reversal fold was 3.75), induced cell apoptosis(P=0.007), increased the intracellular adriamycin accumulation(P=0.000), decreased protein expression of p-IGF-1R, IRS-1, p-Akt, Bcl-2, MRP1 also Gli-1(b). These effects were likely to be mediated via inhibition of IGF-1R/PI3K/Akt/MRP1 pathway. Besides, in a AML xenograft mouse model, NVP-LDE225 combined with ADM resulted in significant tumor regression(P=0.002) (c). Conclusions: These findings provided evidence that targeting the Hh pathway might be a therapeutic avenue for overcoming MDR resistance in myeloid leukemia. Keywords: Hedgehog signaling, acute myeloid leukemia, IGF-1R/PI3K/Akt signaling Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1536-1536 ◽  
Author(s):  
Huilin Huang ◽  
Hengyou Weng ◽  
Xi Qin ◽  
Boxuan Simen Zhao ◽  
Lou Dore ◽  
...  

Abstract N 6-methyladenosine (m6A) modification is the most abundant internal RNA modification in eukaryotes. Recent studies have shown that the dynamic and reversible regulation of m6A modifications in mRNAs or non-coding RNAs plays critical roles in tissue development, stem cell self-renewal and differentiation, control of heat shock response, and circadian clock controlling, as well as in RNA metabolism and processing. However, little is known about the functions of m6A and m6A regulators in malignant hematopoiesis. METTL14 is a major m6A writer which together with METTL3 forms the core of the methyltransferase complex that catalyzes the conversion of adenosine (A) to m6A. Through qPCR assays, we found that METTL14 was aberrantly up-regulated in mononuclear cells (MNC) from acute myeloid leukemia (AML) patients with t(11q23), t(15;17), or t(8;21) relative to those from healthy donors. To investigate the pathological role of METTL14 in AML, we transduced lineage negative (Lin-) bone marrow (BM) progenitor cells from Mettl14fl/flCreERT mice with MLL-AF9, AML1-ETO9a, or PML-RARa fusion genes and performed colony-forming/replating assays with or without addition of 4-hydroxytamoxifen (4-OHT). Induction of genetic knockout of Mettl14 by 4-OHT treatment remarkably impaired the colony-forming ability of all these AML-related fusion genes after replating. After the first round of plating, we harvested MLL-AF9-transduced cells that were not treated with 4-OHT and transplanted them into lethally irradiated recipient mice. As expected, tamoxifen (TAM) treatment of transplanted mice significantly delayed leukemogenesis compared to mice treated with vehicle (MLL-AF9+TAM, with median survival of 91 days; MLL-AF9+vehicle, with median survival of 71 days; P=0.0012) (Fig.1A). In addition, specific knockdown of Mettl14 with shRNAs showed similar patterns to Mettl14 knockout. Thus our data demonstrate that Mettl14 is crucial for cell transformation and leukemogenesis. Further, to determine the role of Mettl14 in the maintenance of leukemia, we transduced leukemic BM cells from primary MLL-AF9 leukemic mice with shRNAs against Mettl14 or scramble shRNA and transplanted these cells into lethally irradiated recipient mice. Again, a significantly prolonged survival was observed in Mettl14 knockdown groups compared to that in the control group (MLL-AF9+shRNA1, with median survival of 32 days; MLL-AF9+shRNA2, with median survival of 32 days; MLL-AF9+shScramble, with median survival of 23.5 days; P< 0.001 for both knockdown groups) (Fig.1B). Noticeable, mice in Mettl14 knockdown groups showed less c-kit+ cells in BM than mice in the control group (Fig.1C). In addition to the mouse model, we used human leukemia cell lines to investigate the function of METTL14 in human AML cells. Silencing of METTL14 with shRNAs significantly inhibited cell viability, induced apoptosis as well as terminal differentiation of MONOMAC6 and NB4 cell lines (Fig.1D, E, F). Moreover, xenograft model showed that repression of METTL14 significantly inhibited the engraftment of MONOMAC6 cells and thus delayed the onset of leukemia in NSG-SGM3 (NSGS) immunodeficient mice (Fig.1G). Furthermore, knockdown of METTL14 sensitized MONOMAC cells to ATRA or PMA-induced differentiation. Taken together, our results support the oncogenic role of METTL14 in AML and highlight METTL14 as a novel therapeutic target in AML. Figure 1 Oncogenic roles of METTL14 in AML. Figure 1. Oncogenic roles of METTL14 in AML. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Banban Li ◽  
Ruinan Jia ◽  
Wei Li ◽  
Ying Zhou ◽  
Dongmei Guo ◽  
...  

BackgroundChemoresistance is emerging as a major barrier to successful treatment in acute myeloid leukemia (AML), and bone marrow stromal cells (BMSCs) protect leukemia cells from chemotherapy eventually leading to recurrence. This study was designed to investigate the role of p21-activated kinase 1 (PAK1) in AML progression and chemosensitivity, highlighting the mechanism of stroma-mediated chemoresistance.MethodsThe GEPIA and TCGA datasets were used to analyze the relationship between PAK1 mRNA expression and various clinical parameters of AML patients. Cell proliferation and apoptosis were examined to evaluate the role of PAK1 on chemosensitivity in AML by silencing PAK1 with shRNA or small molecular inhibitor. Human BMSC (HS-5) was utilized to mimic the leukemia bone marrow microenvironment (BMM) in vitro, and co-culture model was established to investigate the role of PAK1 in BMSC-mediated drug resistance.Resultsp21-activated kinase 1 high expression was shown to be associated with shorter overall survival in AML patients. The silence of PAK1 could repress cell proliferation, promote apoptosis, and enhance the sensitivity of AML cells to chemotherapeutic agents. More importantly, BMSCs induced PAK1 up-regulation in AML cells, subsequently activating the ERK1/2 signaling pathway. The effect of BMSC-mediated apoptotic-resistance could be partly reversed by knock down of PAK1.Conclusionp21-activated kinase 1 is a potential prognostic predictor for AML patients. PAK1 may play a pivotal role in mediating BMM-induced drug resistance, representing a novel therapeutic target in AML.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5048-5048
Author(s):  
Jie Jin ◽  
Shujuan Huang ◽  
Jiajia Pan

The tumor suppressor role of the metabolic genes is rarely reported. Here, we report that malate dehydrogenase 1 (MDH1) acts as an anti-oncogene in acute myeloid leukemia (AML) progression. In a screen of various leukemia cell lines, as well as peripheral blood mononuclear cells from patients with AML and healthy individuals, we observed that patients with AML had lower mRNA expression of MDH1 than healthy individuals. In addition, we found that overexpression of MDH1 in the AML cell lines MV4-11 and THP-1 in vitro promoted their differentiation, reduced their growth, induced apoptosis, and increased the proportion of cells in S phase, which were more sensitive to the chemotherapeutic drugs cytarabine and daunorubicin. Using an in vivo xenograft mouse model of AML, we found that disruption of MDH1 expression influenced tumor burden and mouse survival. These findings were consistent with an inverse relationship between MDH1 expression and overall survival in two independent cohorts of AML patients. We also identified changes in the expression of other genes, concomitant with reduced MDH1 expression in patients with AML, that largely explain the anti-oncogenic role of MDH1 in AML progression. Notably, downregulation of P53 and upregulation of the MAPK pathway in low MDH1-expressing MV4-11 cells were reversed upon overexpression of MDH1. In conclusion, we demonstrated that MDH1 plays a tumor-suppressing role in AML and can be used as a novel mimic drug therapy in the future. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document