Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Amy E. Schmidt is active.

Publication


Featured researches published by Amy E. Schmidt.


Trends in Cardiovascular Medicine | 2003

Structure–Function Relationships in Factor IX and Factor IXa

Amy E. Schmidt; S. Paul Bajaj

Factor IX (FIX) consists of an N-terminal gamma-carboxyglutamic acid (Gla) domain followed by two epidermal growth factor (EGF)-like domains, and the C-terminal serine protease domain. During physiologic coagulation, one of the activators of FIX is the FVIIa/tissue factor (TF) complex. In this reaction, the Gla and EGF1 domains of FIX are thought to interact with TF. The FIXa that is generated then combines with FVIIIa on the platelet surface to activate FX in the coagulation cascade. In this assembly, the protease domain and possibly the EGF2 domain of FIXa are thought to provide the primary specificity in binding to FVIIIa. Disruption of the interaction of FIX/FX with TF and of the FIXa:FVIIIa interface may provide a pharmacologic target as an alternative strategy for the development of antithrombotic agents.


Journal of Biological Chemistry | 2006

High Resolution Structures of p-Aminobenzamidine- and Benzamidine-VIIa/Soluble Tissue Factor UNPREDICTED CONFORMATION OF THE 192-193 PEPTIDE BOND AND MAPPING OF Ca2+, Mg2+, Na+, AND Zn2+ SITES IN FACTOR VIIa

S.P. Bajaj; Amy E. Schmidt; Sayeh Agah; Madhu S. Bajaj; Kaillathe Padmanabhan

Factor VIIa (FVIIa) consists of a γ-carboxyglutamic acid (Gla) domain, two epidermal growth factor-like domains, and a protease domain. FVIIa binds seven Ca2+ ions in the Gla, one in the EGF1, and one in the protease domain. However, blood contains both Ca2+ and Mg2+, and the Ca2+ sites in FVIIa that could be specifically occupied by Mg2+ are unknown. Furthermore, FVIIa contains a Na+ and two Zn2+ sites, but ligands for these cations are undefined. We obtained p-aminobenzamidine-VIIa/soluble tissue factor (sTF) crystals under conditions containing Ca2+, Mg2+, Na+, and Zn2+. The crystal diffracted to 1.8Å resolution, and the final structure has an R-factor of 19.8%. In this structure, the Gla domain has four Ca2+ and three bound Mg2+. The EGF1 domain contains one Ca2+ site, and the protease domain contains one Ca2+, one Na+, and two Zn2+ sites. 45Ca2+ binding in the presence/absence of Mg2+ to FVIIa, Gla-domainless FVIIa, and prothrombin fragment 1 supports the crystal data. Furthermore, unlike in other serine proteases, the amide N of Gly193 in FVIIa points away from the oxyanion hole in this structure. Importantly, the oxyanion hole is also absent in the benzamidine-FVIIa/sTF structure at 1.87Å resolution. However, soaking benzamidine-FVIIa/sTF crystals with d-Phe-Pro-Arg-chloromethyl ketone results in benzamidine displacement, d-Phe-Pro-Arg incorporation, and oxyanion hole formation by a flip of the 192-193 peptide bond in FVIIa. Thus, it is the substrate and not the TF binding that induces oxyanion hole formation and functional active site geometry in FVIIa. Absence of oxyanion hole is unusual and has biologic implications for FVIIa macromolecular substrate specificity and catalysis.


Journal of Biological Chemistry | 2005

Crystal structure of Kunitz domain 1 (KD1) of tissue factor pathway inhibitor-2 in complex with trypsin. Implications for KD1 specificity of inhibition

Amy E. Schmidt; Hitendra S. Chand; Dulio Cascio; Walter Kisiel; S. Paul Bajaj

Kunitz domain 1 (KD1) of tissue factor pathway inhibtor-2 inhibits trypsin, plasmin, and factor VIIa (FVIIa)/tissue factor with Ki values of 13, 3, and 1640 nm, respectively. To investigate the molecular specificity of KD1, crystals of the complex of KD1 with bovine β-trypsin were obtained that diffracted to 1.8 Å. The P1 residue Arg-15 (bovine pancreatic trypsin inhibitor numbering) in KD1 interacts with Asp-189 (chymotrypsin numbering) and with the carbonyl oxygens of Gly-219 and Oγ of Ser-190. Leu-17, Leu-18, Leu-19, and Leu-34 in KD1 make van der Waals contacts with Tyr-39, Phe-41, and Tyr-151 in trypsin, forming a hydrophobic interface. Molecular modeling indicates that this complementary hydrophobic patch is composed of Phe-37, Met-39, and Phe-41 in plasmin, whereas in FVIIa/tissue factor, it is essentially absent. Arg-20, Tyr-46, and Glu-39 in KD1 interact with trypsin through ordered water molecules. In contrast, insertions in the 60-loop in plasmin and FVIIa allow Arg-20 of KD1 to directly interact with Glu-60 in plasmin and Asp-60 in FVIIa. Moreover, Tyr-46 in KD1 electrostatically interacts with Lys-60A and Arg-60D in plasmin and Lys-60A in FVIIa. Glu-39 in KD1 interacts directly with Arg-175 of the basic patch in plasmin, whereas in FVIIa, such interactions are not possible. Thus, the specificity of KD1 for plasmin is attributable to hydrophobic and direct electrostatic interactions. For trypsin, hydrophobic interactions are intact, and electrostatic interactions are weak, whereas for FVIIa, hydrophobic interactions are missing, and electrostatic interactions are partially intact. These findings provide insight into the protease selectivity of KD1.


Journal of Biological Chemistry | 2002

Thermodynamic linkage between the S1 site, the Na+ site, and the Ca2+ site in the protease domain of human activated protein C (APC) - Sodium ion in the APC crystal structure is coordinated to four carbonyl groups from two separate loops

Amy E. Schmidt; K. Padmanabhan; M.C Underwood; Wolfram Bode; Timothy Mather; S.P. Bajaj

The serine protease domain of activated protein C (APC) contains a Na+ and a Ca2+ site. However, the number and identity of the APC residues that coordinate to Na+ is not precisely known. Further, the functional link between the Na+ and the Ca2+ site is insufficiently defined, and their linkage to the substrate S1 site has not been studied. Here, we systematically investigate the functional significance of these two cation sites and their thermodynamic links to the S1 site. Kinetic data reveal that Na+ binds to the substrate-occupied APC withK d values of ∼24 mm in the absence and ∼6 mm in the presence of Ca2+. Sodium-occupied APC has ∼100-fold increased catalytic efficiency (∼4-fold decrease in K m and ∼25-fold increase in k cat) in hydrolyzing S-2288 (H-d-Ile-Pro-Arg-p-nitroanilide) and Ca2+ further increases thisk cat slightly (∼1.2-fold). Ca2+binds to the protease domain of APC with K d values of ∼438 μm in the absence and ∼105 μmin the presence of Na+. Ca2+ binding to the protease domain of APC does not affect K m but increases the k cat ∼10-fold, and Na+ further increases this k cat∼3-fold and decreases the K m value ∼3.7-fold. In agreement with the K m data, sodium-occupied APC has ∼4-fold increased affinity in binding top-aminobenzamidine (S1 probe). Crystallographically, the Ca2+ site in APC is similar to that in trypsin, and the Na+ site is similar to that in factor Xa but not thrombin. Collectively, the Na+ site is thermodynamically linked to the S1 site as well as to the protease domain Ca2+ site, whereas the Ca2+ site is only linked to the Na+site. The significance of these findings is that under physiologic conditions, most of the APC will exist in Na2+-APC-Ca2+ form, which has 110-fold increased proteolytic activity.


Journal of Biological Chemistry | 1999

Cloning and Characterization of the EAP30 Subunit of the ELL Complex That Confers Derepression of Transcription by RNA Polymerase II

Amy E. Schmidt; Trissa Miller; Susan L. Schmidt; Ramin Shiekhattar; Ali Shilatifard

The product of the human oncogene ELLencodes an RNA polymerase II transcription factor that undergoes frequent translocation in acute myeloid leukemia (AML). In addition to its elongation activity, ELL contains a novel type of RNA polymerase II interaction domain that is capable of repressing polymerase activity in promoter-specific transcription. Remarkably, the ELL translocation that is found in patients with AML results in the deletion of exactly this functional domain. Here we report that the EAP30 subunit of the ELL complex has sequence homology to the Saccharomyces cerevisiae SNF8, whose genetic analysis suggests its involvement in the derepression of gene expression. Remarkably, EAP30 can interact with ELL and derepress ELL’s inhibitory activity in vitro. This finding may reveal a key role for EAP30 in the pathogenesis of human leukemia.


Journal of Biological Chemistry | 2011

Engineering Kunitz Domain 1 (KD1) of Human Tissue Factor Pathway Inhibitor-2 to Selectively Inhibit Fibrinolysis PROPERTIES OF KD1-L17R VARIANT

Madhu S. Bajaj; Godwin I. Ogueli; Yogesh Kumar; Kanagasabai Vadivel; Gregory Lawson; Sreejesh Shanker; Amy E. Schmidt; S. Paul Bajaj

Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accordingly, it has been proposed for use as an anticoagulant. Full-length TFPI-2 or its isolated first Kunitz domain (KD1) also inhibits plasmin; therefore, it has been proposed for use as an antifibrinolytic agent. However, the anticoagulant properties of TFPI-2 or KD1 would diminish its antifibrinolytic function. In this study, structure-based investigations and analysis of the serine protease profiles revealed that coagulation enzymes prefer a hydrophobic residue at the P2′ position in their substrates/inhibitors, whereas plasmin prefers a positively charged arginine residue at the corresponding position in its substrates/inhibitors. Based upon this observation, we changed the P2′ residue Leu-17 in KD1 to Arg (KD1-L17R) and compared its inhibitory properties with wild-type KD1 (KD1-WT). Both WT and KD1-L17R were expressed in Escherichia coli, folded, and purified to homogeneity. N-terminal sequences and mass spectra confirmed proper expression of KD1-WT and KD1-L17R. Compared with KD1-WT, the KD1-L17R did not inhibit factor XIa, plasma kallikrein, or factor VIIa/tissue factor. Furthermore, KD1-L17R inhibited plasmin with ∼6-fold increased affinity and effectively prevented plasma clot fibrinolysis induced by tissue plasminogen activator. Similarly, in a mouse liver laceration bleeding model, KD1-L17R was ∼8-fold more effective than KD1-WT in preventing blood loss. Importantly, in this bleeding model, KD1-L17R was equally or more effective than aprotinin or tranexamic acid, which have been used as antifibrinolytic agents to prevent blood loss during major surgery/trauma. Furthermore, as compared with aprotinin, renal toxicity was not observed with KD1-L17R.


Transfusion | 2015

An alternative method to dithiothreitol treatment for antibody screening in patients receiving daratumumab.

Amy E. Schmidt; Scott A. Kirkley; Nisha Patel; Debra Masel; Renee Bowen; Neil Blumberg; Majed A. Refaai

1. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care 2012;35 Suppl 1: S64–71. 2. Davey RJ. The blood centre as a community health resource. Vox Sang 2006;91:206–13. 3. Kessler DA, Ortiz C, Grima K, et al. Cardiovascular disease risk assessment and prevention in blood donors. Transfusion 2012;52:2174–82. 4. Gore MO, Eason SJ, Colby RA, et al. Glycated hemoglobin in 14,850 adolescent blood donors: a pilot screening program. Diabetes Care 2014;37:e3–4.l 5. Lenhard MJ, Maser RE, Kolm P, et al. Screening blood donors for diabetes: analysis of use, accuracy, and cost. Transfusion 2013;53:2776–81.


Journal of Biological Chemistry | 2007

Substitution of the Gla Domain in Factor X with That of Protein C Impairs Its Interaction with Factor VIIa/Tissue Factor LACK OF COMPARABLE EFFECT BY SIMILAR SUBSTITUTION IN FACTOR IX

Matthew Ndonwi; George J. Broze; Sayeh Agah; Amy E. Schmidt; S. Paul Bajaj

We previously reported that the first epidermal growth factor-like (EGF1) domain in factor X (FX) or factor IX (FIX) plays an important role in the factor VIIa/tissue factor (FVIIa/TF)-induced coagulation. To assess the role of γ-carboxyglutamic acid (Gla) domains of FX and FIX in FVIIa/TF induced coagulation, we studied four new and two previously described replacement mutants: FXPCGla and FIXPCGla (Gla domain replaced with that of protein C), FXPCEGF1 and FIXPCEGF1 (EGF1 domain replaced with that of protein C), as well as FXPCGla/EGF1 and FIXPCGla/EGF1 (both Gla and EGF1 domains replaced with those of protein C). FVIIa/TF activation of each FX mutant and the corresponding reciprocal activation of FVII/TF by each FXa mutant were impaired. In contrast, FVIIa/TF activation of FIXPCGla was minimally affected, and the reciprocal activation of FVII/TF by FIXaPCGla was normal; however, both reactions were impaired for the FIXPCEGF1 and FIXPCGla/EGF1 mutants. Predictably, FXIa activation of FIXPCEGF1 was normal, whereas it was impaired for the FIXPCGla and FIXPCGla/EGF1 mutants. Molecular models reveal that alternate interactions exist for the Gla domain of protein C such that it is comparable with FIX but not FX in its binding to FVIIa/TF. Further, additional interactions exist for the EGF1 domain of FX, which are not possible for FIX. Importantly, a seven-residue insertion in the EGF1 domain of protein C prevents its interaction with FVIIa/TF. Cumulatively, our data provide a molecular framework demonstrating that the Gla and EGF1 domains of FX interact more strongly with FVIIa/TF than the corresponding domains in FIX.


Journal of Clinical Apheresis | 2017

Extracorporeal photopheresis practice patterns: An international survey by the ASFA ECP subcommittee

Nancy M. Dunbar; Jay S. Raval; Andrew D. Johnson; Cori Abikoff; Jill Adamski; Laura Cooling; Brenda J. Grossman; Haewon C. Kim; Marisa B. Marques; Shanna Morgan; Amy E. Schmidt; Steven R. Sloan; Leon Su; Zbigniew M. Szczepiorkowski; F. Bernadette West; Edward C.C. Wong; Jennifer Schneiderman

Although many apheresis centers offer extracorporeal photopheresis (ECP), little is known about current treatment practices.


Seminars in Thrombosis and Hemostasis | 2015

Platelet Transfusion and Thrombosis: More Questions than Answers.

Amy E. Schmidt; Majed A. Refaai; Neil Blumberg

Platelets perform a vital role in hemostasis and their role in inflammation is becoming increasingly evident. Blood transfusion is the most common procedure performed in hospitals and platelet transfusions comprise a significant proportion. Over the past few decades, retrospective studies and randomized clinical trials have demonstrated that blood transfusion is more harmful than previously thought and is associated with numerous complications, such as transfusion-associated lung injury, transfusion-associated cardiac overload, transfusion-associated immune modulation, and infectious diseases such as human immunodeficiency virus, hepatitis C virus, and hepatitis B virus. Recent data suggest an association between platelet transfusion and thrombosis. This review will highlight the mechanistic issues that may be relevant to the epidemiologic associations of platelet transfusion with thrombosis and mortality in critically ill patients.

Collaboration


Dive into the Amy E. Schmidt's collaboration.

Top Co-Authors

Avatar

Majed A. Refaai

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

S. Paul Bajaj

University of California

View shared research outputs
Top Co-Authors

Avatar

Neil Blumberg

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Scott A. Kirkley

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Debra Masel

University of Rochester

View shared research outputs
Top Co-Authors

Avatar

Joanna M. Heal

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jason H. Mendler

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Myra Coppage

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge