Meizothrombin: Zymogen or Proteinase? Slow, Ligand-Dependent Equilibration Between Equally Populated Zymogen-Like and Proteinase-Like Forms Explains Its Selectively Anticoagulant Function

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 533-533
Author(s):  
Harlan Bradford ◽  
Sriram Krishnaswamy

Abstract Abstract 533 Prothrombinase activates prothrombin by catalyzing its ordered proteolysis at Arg320 followed by cleavage at Arg271. Initial cleavage at Arg320 yields the proteinase meizothrombin (mIIa), which accumulates abundantly as an intermediate before its conversion to thrombin (IIa). Although mIIa is a proteinase, it only acts on a limited subset of substrates cleaved by IIa. mIIa is considered an anticoagulant proteinase because it functions efficiently in protein C activation while exhibiting poor clotting activity or reactivity towards antithrombin III. This limited substrate repertoire of mIIa has remained enigmatic and is generally considered to lie in the retention of covalent linkage to the fragment 1.2 (F12) domain allowing for membrane binding. Instead, our recent findings with IIa, illustrating its ligand dependent interconversions between zymogen-like and proteinase-like states, predict that covalent linkage of F12 to the proteinase domain in mIIa would impart it with zymogen-like properties. We produced stable and active mIIa (mIIaQQQ) using a recombinant prothrombin variant in which the bonds susceptible to autolysis were rendered uncleavable by substitution of Arg at 155, 271 and 284 with Gln. Fluorescence stopped flow studies were pursued with the probes dansyl arginine 3-ethyl piperidine amide (DAPA) or Nα-dansyl-(p-guanidino)-L-phenylalanine-piperidide (I-2581) to characterize the binding of ligands to the active site of mIIaQQQ and IIa. Binding to IIa was rapid and consistent with a rate limiting, bimolecular interaction between probe and the active site of the proteinase. In contrast, traces with mIIaQQQ were distinctly biphasic with ∼50% of the fluorescence change occurring on the millisecond timescale followed by a slow second phase (∼50%) that occurred over several seconds. Global fitting indicated that the findings were consistent with a pre-equilibrium between two forms of mIIa, one which binds the active site probe with μM affinity and a second that binds with nM affinity. The two forms interconvert with forward and reverse rate constants of ∼2 s−1. We surmise that these reflect zymogen-like and proteinase-like forms that are equally populated and interconvert slowly with each other in a ligand-dependent fashion. Accordingly, the distributions of the two forms could be altered by ligands established to affect the transition of IIa between zymogen-like and proteinase-like states. The equilibrium distribution was altered to favor the zymogen-like form by decreasing Na+ to 0 at constant ionic strength. In contrast, soluble thrombomodulin (sTM) drove the equilibrium towards the proteinase-like state in a manner consistent with a 1:1 interaction between mIIaQQQ and sTM. Surprisingly, the pre-equilibrium was heavily dependent on covalent linkage with fragment 1 (F1) or its structural integrity. Proteolytic removal of F1, chelation of Ca2+ with EDTA or elimination of 4-carboxyglutamic acid modifications had a profound effect on forcing the enzyme into the proteinase-like state. Thus, the equilibrium distribution of mIIa between zymogen-like and proteinase-like forms is affected by F1 and its Ca2+-stabilized conformation despite the fact that this domain is expected to be distant from the catalytic site. Our findings shed unexpected light into the mechanisms underlying the peculiar activity profile of mIIa relative to IIa. Its ability to interconvert slowly and reversibly between equally populated zymogen-like and proteinase-like states lies at the heart of its properties. By driving it to proteinase, thrombomodulin imparts full activity to mIIa allowing for efficient function in the anticoagulant pathway. In contrast, more weakly binding substrates, inhibitors or ligands will be less effective at perturbing the equilibrium thereby allowing mIIa to persist in blood with reduced activity towards procoagulant substrates. The F1 domain participates in an unexpected way in enforcing these unique features of mIIa. By virtue of its essential role in modulating the equilibrium distribution between zymogen-like and proteinase-like states, we document a new function for F1 in its role as a zymogenizer of mIIa. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 122-122
Author(s):  
Joseph A. Micucci ◽  
Parvathi Kamath ◽  
Anuja Khan ◽  
Paul E. Bock ◽  
Sriram Krishnaswamy

Abstract The binding of ligands to anion binding exosite I (ABEI) and exosite II (ABEII) on prothrombin (II) derivatives plays an integral role in regulating their function. These exosites are located on opposite faces of the proteinase domain and exhibit unique binding specificities. Fragment 1.2 (F12) binds to ABEII and acts as a zymogen-promoting allosteric ligand. Conversely, Na+ and active site ligands stabilize the proteinase state. Here, we investigated the allosteric linkage between ABEI and the Na+ binding site, active site or ABEII using multiple ABEI ligands and prothrombin derivatives differentially poised along the zymogen to proteinase continuum. Prethrombin 2 (P2) represents the most zymogen-like state that differs from thrombin (IIa) because it is not cleaved at the R320 site. To mimic the zymogen-like character of P2 in a cleaved IIa molecule, residues responsible for N-terminal insertion and proteinase formation (IVE) were swapped with TAT to produce IIaTAT, a mutant with vastly diminished proteolytic activity. Alanine was substituted with the catalytic serine residue in IIa (IIaS195A) to represent the proteinase without a ligand at its active site. The thermodynamics of interactions between the thrombin inhibitor and ABEI ligand hirugen (Hir) and the various reference states was assessed using isothermal titration calorimetry (ITC). Titration of Hir into P2, IIaTAT or IIaS195A revealed thermodynamically more favorable binding to proteinase-like IIaS195A in comparison to zymogen-like P2 or IIaTAT. Binding of Hir to IIaS195A was affected by the concentration of Na+ at constant ionic strength. Global analysis done in the presence of increasing concentrations of Na+ revealed a 5-fold increase in Hir binding affinity when IIaS195A is ligated with Na+; demonstrating positive allosteric linkage between ABEI and Na+ binding. Using a truncated Staphylocoagulase variant that binds ABEI without N-terminus insertion (SC13-325), we found that SC13-325 binding alone promoted active site opening and fluorescent inhibitor incorporation in zymogen-like P2. These data reinforce the observation that ABEI ligands promote a proteinase-like state in prothrombin derivatives through positive allosteric linkage with the Na+ binding and active sites. Interestingly, inhibitor incorporation and ITC studies both showed that SC13-325 interacts poorly with II, but strongly with P2 despite both species being zymogens. These findings imply that the ABEII ligand F12, which is produced upon cleavage of II at R271 to form P2, may display negative allosteric linkage with ABEI. Titration of SC13-325 into pre-formed complexes of P2/F12 revealed a drastic reduction in affinity of SC13-325 for P2 when F12 is bound to ABEII. Thus, F12 binding at ABEII negatively affects ABEI binding. Further studies used soluble thrombomodulin (sTM) as the most physiologically pertinent ligand for ABEI. ITC and global analysis of the binding of F12 to P2 in the presence of different concentrations of sTM revealed a ~1200-fold decrease in binding affinity and enthalpy for either the binding of F12 to P2 bound to sTM or the binding of sTM to P2 bound to F12. These data illustrate the strong negative linkage associated with the binding of protein ligands to ABEI and ABEII which yields the appearance of competitive and mutually exclusive binding at the two sites despite the fact that they are on opposite sides of the proteinase domain. Allosteric linkage between ligand binding at the two exosites is centered on the ability of F12 binding to ABEII and favor zymogen-like forms and ligand binding to ABEI to favor proteinase-like forms. Thus, allosteric linkage among exosites is vital to the interconversion of prothrombin species along the zymogen to proteinase spectrum. These ligand-dependent conformational shifts and associated changes in function are likely to greatly contribute to the dynamic roles that IIa plays during coagulation. Disclosures No relevant conflicts of interest to declare.


2004 ◽  
Vol 382 (2) ◽  
pp. 751-757 ◽  
Author(s):  
Pakorn WINAYANUWATTIKUN ◽  
Albert J. KETTERMAN

Glutathione S-transferases (GSTs) are dimeric proteins that play a major role in cellular detoxification. The GSTs in mosquito Anopheles dirus species B, an important malaria vector in South East Asia, are of interest because they can play an important role in insecticide resistance. In the present study, we characterized the Anopheles dirus (Ad)GST D3-3 which is an alternatively spliced product of the adgst1AS1 gene. The data from the crystal structure of GST D3-3 shows that Ile-52, Glu-64, Ser-65, Arg-66 and Met-101 interact directly with glutathione. To study the active-site function of these residues, alanine substitution site-directed mutagenesis was performed resulting in five mutants: I52A (Ile-52→Ala), E64A, S65A, R66A and M101A. Interestingly, the E64A mutant was expressed in Escherichia coli in inclusion bodies, suggesting that this residue is involved with the tertiary structure or folding property of this enzyme. However, the I52A, S65A, R66A and M101A mutants were purified by glutathione affinity chromatography and the enzyme activity characterized. On the basis of steady-state kinetics, difference spectroscopy, unfolding and refolding studies, it was concluded that these residues: (1) contribute to the affinity of the GSH-binding site (‘G-site’) for GSH, (2) influence GSH thiol ionization, (3) participate in kcat regulation by affecting the rate-limiting step of the reaction, and in the case of Ile-52 and Arg-66, influenced structural integrity and/or folding of the enzyme. The structural perturbations from these mutants are probably transmitted to the hydrophobic-substrate-binding site (‘H-site’) through changes in active site topology or through effects on GSH orientation. Therefore these active site residues appear to contribute to various steps in the catalytic mechanism, as well as having an influence on the packing of the protein.


2019 ◽  
Vol 48 (18) ◽  
pp. 5933-5939 ◽  
Author(s):  
Sonja Pullen ◽  
Somnath Maji ◽  
Matthias Stein ◽  
Sascha Ott

Terminal ligand fixation by covalent linkage to the bridging bdt ligand hinders ligand rotations.


Blood ◽  
1989 ◽  
Vol 73 (6) ◽  
pp. 1540-1544 ◽  
Author(s):  
DM Monroe ◽  
DM McCord ◽  
MN Huang ◽  
KA High ◽  
RL Lundblad ◽  
...  

Abstract Factor IX Hilo is a variant factor IX molecule that has no detectable coagulant activity. The defect in factor IX Hilo arises from a point mutation in the gene such that in the protein Arg180 is converted to a Gln. Activation of factor IX Hilo by factor Xla was monitored using the fluorescent active site probe p-aminobenzamidine. Normal factor IX showed complete activation in one hour as determined by measuring the increase in fluorescence when p-aminobenzamidine bound to activated factor IX. Factor IX Hilo showed no increase in fluorescence even after 24 hours, indicating that the active site was not exposed. Polyacrylamide gel electrophoresis showed that factor IX Hilo was cleaved to a light chain plus a larger peptide with a molecular weight equivalent to a heavy chain covalently linked to an activation peptide. Amino terminal amino acid sequencing of factor IX Hilo cleaved by factor Xla showed cleavage only at Arg145-Ala146, indicating that the Gln180-Val181 bond was not cleaved and that the active site was thus not exposed. The presence of factor IX Hilo in patient plasma was responsible for the patient having a very long ox brain prothrombin time characteristic of severe hemophilia Bm. Patient plasma had an ox brain prothrombin time of 100 seconds using a Thrombotest kit, significantly prolonged over the normal control value of 45 seconds. When factor IX Hilo was depleted from patient plasma using an immunoaffinity column, the ox brain prothrombin time decreased to 41 seconds. When factor IX Hilo was added back to depleted patient plasma, to normal plasma depleted of factor IX by the same affinity column, or to plasma from a CRM- hemophilia B patient, the ox brain prothrombin time was significantly prolonged. We conclude that the Arg180 to Gln mutation in factor IX Hilo results in a molecule that cannot be activated by factor Xla. Further, our data suggest that the mutation results in a molecule that interacts with components of the extrinsic pathway to give a prolonged ox brain prothrombin time.


2020 ◽  
Vol 21 (7) ◽  
pp. 2519
Author(s):  
Yuji Nozaki ◽  
Jinhai Ri ◽  
Kenji Sakai ◽  
Kaoru Niki ◽  
Masanori Funauchi ◽  
...  

Thrombomodulin (TM) is a single transmembrane, multidomain glycoprotein receptor for thrombin, and is best known for its role as a cofactor in a clinically important natural anticoagulant pathway. In addition to its anticoagulant function, TM has well-defined anti-inflammatory properties. Soluble TM levels increase significantly in the plasma of septic patients; however, the possible involvement of recombinant human soluble TM (rTM) transduction in the pathogenesis of lipopolysaccharide (LPS)-induced nephrotoxicity, including acute kidney injury (AKI), has remained unclear. Mice were injected intraperitoneally with 15 mg/kg LPS. rTM (3 mg/kg) or saline was administered to the animals before the 3 and 24 h LPS-injection. At 24 and 48 h, blood urea nitrogen, the inflammatory cytokines in sera and kidney, and histological findings were assessed. Cell activation and apoptosis signal was assessed by Western blot analysis. In this study using a mouse model of LPS-induced AKI, we found that rTM attenuated renal damage by reducing both cytokine and cell activation and apoptosis signals with the accumulation of CD4+ T-cells, CD11c+ cells, and F4/80+ cells via phospho c-Jun activations and Bax expression. These findings suggest that the mechanism underlying these effects of TM may be mediated by a reduction in inflammatory cytokine production in response to LPS. These molecules might thereby provide a new therapeutic strategy in the context of AKI with sepsis.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1592-1592 ◽  
Author(s):  
Lee A. Honigberg ◽  
Ashley M. Smith ◽  
Jun Chen ◽  
Patti Thiemann ◽  
Erik Verner

Abstract There is increasing evidence indicating that B-cell-receptor (BCR) signaling is required for survival of non-Hodgkin’s lymphoma (NHL) cells. Bruton’s tyrosine kinase (Btk) is required for BCR signaling and mutations that inactivate human Btk cause X-linked-agammaglobulinemia, a B-cell immunodeficiency. Although Btk functions selectively in B cells, the Btk active site is structurally similar to the active site in several Src and Abl kinases and as a result, there have been few highly selective small molecule inhibitors of Btk. We have developed a series of covalent Btk inhibitors that target Cys-481 in Btk and this approach results in increased potency and selectivity over related kinases that lack a Cys residue at this position (ChemMedChem 15:58). PCI-32765 is a Cys-481 targeting Btk inhibitor that has been optimized for potency, selectivity and pharmacokinetics. In cellular assays, PCI-32765 inhibits BCR-stimulus induced phosphorylation of Phospholipase-C-gamma, a Btk substrate, as well as downstream phosphorylation of Erk (IC50 < 100 nM). In addition, PCI-32765 induces apoptosis and inhibits proliferation in a subset of NHL cell lines including DHL-4, DHL-6, WSU-DLCL2, OCI-Ly10 and DOHH2 (IC50s = 0.6–1.6uM). We have used RNAi knockdown in DOHH2 cells as an independent method to confirm that Btk is required for lymphoma cell proliferation. In vivo, orally dosed PCI-32765 (50mg/kg) inhibits growth of DOHH2 and WSU-DLCL2 xenografts. PCI-32765 also prevents disease progression in a mouse collagen-induced arthritis model (12.5mg/kg PO), indicating that other B cell lineage diseases are sensitive to Btk inhibition. In order to further characterize the selectivity and in vivo potency of PCI-32765, we have developed PCI-33380, an active-site probe consisting of a covalent Btk inhibitor linked to the fluorophore Bodipy-FL. PCI-33380 binds to Btk and can be detected by flow cytometry or by denaturing gel electrophoresis of cell lysates. In cell lysates, the probe labels a single predominant band of the same molecular weight as Btk and this band is absent in cells from xid mice. Labeling of this band is inhibited (IC50=10nM) by a brief pre-treatment of cells with PCI-32765, indicating that the probe can be used to assess occupancy of Btk by a covalent inhibitor. We have used the probe to quantitate the inhibition of Btk by PCI-32765 in vivo. A single oral dose of PCI-32765 (10mg/kg) delivered to mice leads to rapid and complete inhibition of Btk in spleen. In addition, a single oral dose of PCI-32765 fully inhibits Btk in xenograft tumors and peripheral blood cells and this inhibition is maintained for up to 24hr. The Btk probe provides pharmacodynamic measurements that may allow optimization of dosing and schedule for in vivo studies and we are currently adapting the probe assays for use in monitoring the inhibition of Btk in human clinical trials.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3591-3591
Author(s):  
Xiaoyan Du ◽  
Timothy Myles ◽  
John Morser ◽  
Lawrence L. Leung

Abstract Abstract 3591 Poster Board III-528 Introduction Prochemerin is a 163 amino acid precursor protein with a C-terminal domain highly susceptible to proteolysis. Its chemotactic activity is unmasked upon C-terminal cleavage by proteases of the coagulation, fibrinolytic and inflammatory systems. Here, we studied whether thrombin is able to cleave prochemerin to generate active forms of chemerin. Methods The 15mer peptide prochemerin C-terminal sequence (YFPGQFAFSKALPRS) or prochemerin was incubated with thrombin at different concentrations and times. The reactions were stopped by addition of PPACK before determining their chemotactic activity in a transwell assay using CMKLR1-transfected cells and their mass by mass-spectrometry. Results Thrombin (0-100 nM) dose-dependently cleaved 15mer to 14mer (YFPGQFAFSKALPR). Over a longer reaction time, the 10mer (YFPGQFAFSK) was also detected. The 15mer was almost inert in the chemotaxis assay but thrombin-cleaved 15mer caused migration of CMKLR1 transfectants. The 14mer and 10mer at 1 μM induced CMKLR1 cell migration at a rate of 3200 and 2800 cells/ml, but 1 μM 15mer did not induce any chemotaxis. Thrombin can also cleave the 14mer to a 10mer as determined by mass spectrometry. Using selected thrombin mutants for the Na+ binding site (E229K) and the active site YPPW-insertion loop (W50A), we found that thrombin hydrolysis of the 15mer was dependent on the Na+ bound “fast” form of thrombin and active site topology. The 10mer could be further activated by carboxypeptidase N (CPN) by removing the C-terminal lysine, whereas the C-terminal arginine of 14mer could not be cleaved by CPN, which did not affect its activity. Full-length prochemerin was also activated by thrombin (100nM) and the chemotactic activity further increased by CPN (50nM) about 6 fold. Conclusions Prochemerin is a new substrate for thrombin. Thrombin-cleaved chemerins are active chemoattractants in chemotaxis. This extends the molecular link between blood coagulation and CMKLR1-mediated immune responses. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2215-2215
Author(s):  
Harlan Bradford ◽  
Sriram Krishnaswamy

Abstract Abstract 2215 The proteolytic activation of prothrombin catalyzed by prothrombinase is a paradigm for zymogen activation resulting from ordered cleavage at multiple sites. Initial cleavage at Arg320 forms meizothrombin (mIIa). The associated conversion of zymogen to proteinase is instrumental in facilitating further processing at Arg271 to yield thrombin. A full kinetic explanation for this process remains obscure and controversial because of the limitations of standard kinetic approaches. We now uncover novel facets of this reaction by rapid kinetic studies to approximate single catalytic turnover. Product formation was measured continuously by stopped flow using Dansyl arginine (3-ethyl-1,5-pentanediyl) amide (DAPA) or discontinuously using rapid quenching and analysis by SDS-PAGE or peptidyl substrate cleavage following rapid mixing of preformed prothrombinase (0.3 μM) with prothrombin (0.3 μM). Prothrombin cleavage, assessed discontinuously, was essentially complete within 0.2 seconds. The results indicated initial cleavage at Arg320 to form mIIa followed by subsequent cleavage at Arg271 to produce thrombin. However, product formation measured continuously with DAPA, yielded a pronounced lag and proceeded ∼20-30-fold more slowly. The intermediate, mIIa, which is expected to bind DAPA was invisible to the continuous measurements. Analysis was further simplified using a recombinant prothrombin variant, which is exclusively cleaved at Arg320 to produce mIIa and not processed further. Continuous detection of proteinase formation by stopped flow proceeded ∼30-fold more slowly than cleavage at Arg320 measured discontinuously. These findings indicate that rapid cleavage at the Arg320 site is followed by an unexpectedly slow reaction (t½ ∼ 0.5–1 s) in which the cleaved product matures to form a competent active site. This conclusion was further tested employing stable mIIa prepared by the action of Ecarin on recombinant prothrombin variants that were not degraded further even without occluding the active site. Stopped flow kinetic studies for the binding of DAPA to these variants revealed markedly biphasic traces. The data could be globally analyzed according to a two step mechanism with an initial slow equilibrium between zymogen-like and proteinase forms in which only the proteinase form can bind the active site ligand. The zymogen- like and proteinase forms were approximately equally populated and interconverted slowly (t½ ∼ 0.5 s). These findings independently corroborate the conclusions from the kinetic studies of prothrombin cleavage. Accordingly, inclusion of this slow step could explain profiles of prothrombin depletion, transient formation of mIIa and the final appearance of thrombin seen in the action of prothrombinase on prothrombin. Zymogen-like mIIa accumulates at much higher concentrations than would be predicted from knowledge of the kinetics of the individual cleavage steps because its slow maturation to proteinase is required for further cleavage at Arg271. Thus, the rate-limiting maturation of mIIa to proteinase plays a dominant role in regulating thrombin formation. Furthermore, while mIIa is a poor catalyst for many of substrates of thrombin, it retains the ability to bind thrombomodulin and function in the anticoagulant pathway. As a result of its unexpectedly zymogen-like character and slow conversion to proteinase, mIIa produced as an intermediate by prothrombinase would be resistant to inhibition in plasma and thereby potentially be dispersed by flowing blood to exert its selectively anticoagulant functions distant from its site of production. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1693-1693 ◽  
Author(s):  
Angela C Court Recart ◽  
Anil Sadarangani ◽  
Daniel Goff ◽  
Alice Y Shih ◽  
Russell Wall ◽  
...  

Abstract Abstract 1693 The aim of this study is to develop clinical strategies that will HALT progression of CML by reducing leukemia stem cell (LSC) burden using a clinical grade JAK2 inhibitor, SAR302503 (SAR503, Sanofi, Cambridge, MA), alone or in combination with a potent BCR-ABL inhibitor, dasatinib. For this, CML patient samples in blast crisis phase (BC CML) were subjected to immunomagnetic bead CD34 selection or FACS Aria ll sorted to obtain leukemic progenitors (LSC/CD34+CD38+Lin−). Malignant progenitors were then transplanted into neonatal RAG2−/−gc−/− mice, and 8 weeks post-transplant, mice were treated with SAR503, dasatinib and vehicle for 14 days. Following treatment, hematopoietic tissues were analyzed for human engraftment by FACS analysis. Our results revealed that single agent experiments with SAR503 had a cytostatic rather than a cytoreductive effect on BC LSC. The treatment alone (60 mg/kg twice daily administered by oral gavage) did not significantly reduce leukemic progenitor burden in the liver, spleen, bone marrow and peripheral blood. Conversely, combination therapy with SAR503 and dasatinib (50mg/kg/day) significantly reduced LSC progenitors in all tissues examined. Interestingly, we observed that dasatinib alone therapy reduced the LSC burden in the liver, spleen, and peripheral blood, but the bone marrow retained a significant population of BC LSC. Also we found that the GMP population, previously shown to be enriched for BC LSC (Jamieson et al NEJM 2004; Abrahamsson et al PNAS 2009), was preferentially localized in the bone marrow. As shown by our laboratory and others, LSC therapeutic resistance may be influenced by extrinsic cues provided by the niche (e.g. promoting quiescence). Because quiescence has been implicated in driving tyrosine kinase inhibitor resistance and LSC survival and because the bone marrow retains a resistant population, we decide to perform secondary transplantation experiments to determine relapse potential (self-renewal). LSC progenitors were isolated by immunomagnetic bead selection of human CD34+ cells from marrows and spleens of treated mice. After serially transplanting an equal number of this cells into secondary recipients, we observed a significant reduction in LSC serial transplantation only following combination treatment, suggesting that the combination therapy can abolish LSC self-renewal capacity and thereby potentially prevent relapse. To validate drug exposure, we have been performing both genomic and nanoproteomic analysis. Regarding the proteomics validation studies, we analyzed sorted LSC derived from spleen (pooled 5 mice per group) that were treated with vehicle or SAR503 for 14 days. The analysis was performed to detect status of p-JAK2, JAK2, p-STAT5 and B2-microglobulin (loading control). We observed a down regulation on the levels of p-JAk2 (active site Tyr 1007–08) and p-Stat5 (active site Tyr 694) (35% and 42% respectively), while no changes are observed for total JAK2 protein or B2M between both conditions. The full transcriptome sequencing, on sorted LSC treated with SAR503 alone and in combination with dasatinib, identified specific isoform changes in the JAK/STAT pathway that could be used as biomarkers of response and could explain the synergistic effect of the combination therapy. We have also characterized, at an isoform level, biomarkers of resistance that could explain relapse of disease after single agent therapy and we are currently validating these findings. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document