Indoleamine 2,3-dioxygenase (IDO) catalyzes the oxidation of L-tryptophan to N-formyl kynurenine. In contrast to the wide spectrum of P450 monooxygenases, IDO is one of the only two heme-based dioxygenases in humans. Despite decades of effort, its dioxygenase mechanism remains elusive. IDO is an immunosuppressive enzyme, which plays an important role in allowing cancer cells to escape from immune surveillance. Recently, it has attracted a great deal of attention due to the recognition of its potential as a therapeutic target for cancer. Hence, there is a critical need for the delineation of the dioxygenase and inhibitory mechanisms of the two human isoforms of IDO, named hIDO1 and hIDO2. The long term goal of our program is (i) to define the molecular mechanism of heme-based dioxygenases, filling in the knowledge gap in heme oxygen chemistry, and (ii) to delineate the antitumor effect of IDO1/IDO2 inhibitors, aiding in the definition of IDO-linked cancer physiology. The objective of this application is to characterize the molecular properties of hIDO1 and hIDO2, in order to define their catalytic and inhibitory mechanisms. Our central hypothesis is that the catalytic and inhibitory mechanisms of the two IDO isoforms are distinct. The rationale is that the successful completion of this project will offer strong, conceptual and evidence-based guidelines for the development of IDO- targeted intervention against cancer. Thus, the proposed research is relevant to that part of NIH's mission that pertains to the development of fundamental knowledge that will potentially help to reduce the burdens of human disability. Guided by strong preliminary data, our hypothesis will be tested by the pursuit of two specific aims: (i) define the dioxygenase mechanisms of hIDO1 and hIDO2, and (ii) identify the inhibition mechanisms of hIDO1 and hIDO2. To achieve our objective we will employ a multi-faceted approach with a complementary set of spectroscopic techniques (Raman, UV-Vis, FTIR, EPR, MS and X-ray crystallography), combined with computational methodologies (MD and QM/MM) and mutagenesis. The proposed project is innovative because (i) the availability of both hIDO1 and hIDO2 places us in a unique position for carrying out the proposed comparative studies, and (ii) the unique fast-mixing/freeze-quenching techniques developed in our lab enable effective structural characterization of key enzymatic intermediates that are inaccessible in other labs. The proposed research is significant because the successful completion of this project will lay a solid foundation for the future design of novel therapeutic strategies targeting IDO, as well as to advance fundamental understanding of heme-based dioxygenase chemistry.

Public Health Relevance

The proposed studies are focused on an important, but poorly understood, area of research dealing with two isoforms of indoleamine 2,3-dioxygenase, which play an essential role in allowing cancer cells to escape from immune surveillance. The proposed research is relevant to public health, because a detailed understanding of catalytic and inhibitory mechanisms of the two isoforms of the enzyme should provide a foundation for the development of their inhibitors for pharmaceutical intervention against cancer.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM086482-04
Application #
8451545
Study Section
Macromolecular Structure and Function A Study Section (MSFA)
Program Officer
Anderson, Vernon
Project Start
2010-06-01
Project End
2014-04-30
Budget Start
2013-05-01
Budget End
2014-04-30
Support Year
4
Fiscal Year
2013
Total Cost
$318,807
Indirect Cost
$126,755
Name
Albert Einstein College of Medicine
Department
Physiology
Type
Schools of Medicine
DUNS #
110521739
City
Bronx
State
NY
Country
United States
Zip Code
10461
Lewis-Ballester, Ariel; Forouhar, Farhad; Kim, Sung-Mi et al. (2016) Molecular basis for catalysis and substrate-mediated cellular stabilization of human tryptophan 2,3-dioxygenase. Sci Rep 6:35169
Álvarez, Lucía; Lewis-Ballester, Ariel; Roitberg, Adrián et al. (2016) Structural Study of a Flexible Active Site Loop in Human Indoleamine 2,3-Dioxygenase and Its Functional Implications. Biochemistry 55:2785-93
Malachowski, William P; Winters, Maria; DuHadaway, James B et al. (2016) O-alkylhydroxylamines as rationally-designed mechanism-based inhibitors of indoleamine 2,3-dioxygenase-1. Eur J Med Chem 108:564-576
Ahn, Young O; Lee, Hyun Ju; Kaluka, Daniel et al. (2015) The two transmembrane helices of CcoP are sufficient for assembly of the cbb3-type heme-copper oxygen reductase from Vibrio cholerae. Biochim Biophys Acta 1847:1231-9
Kaluka, Daniel; Batabyal, Dipanwita; Chiang, Bing-Yu et al. (2015) Spectroscopic and mutagenesis studies of human PGRMC1. Biochemistry 54:1638-47
Ishigami, Izumi; Hikita, Masahide; Egawa, Tsuyoshi et al. (2015) Proton translocation in cytochrome c oxidase: insights from proton exchange kinetics and vibrational spectroscopy. Biochim Biophys Acta 1847:98-108
Egawa, Tsuyoshi; Haber, Jonah; Fee, James A et al. (2015) Interactions of Cu(B) with Carbon Monoxide in Cytochrome c Oxidase: Origin of the Anomalous Correlation between the Fe-CO and C-O Stretching Frequencies. J Phys Chem B 119:8509-20
Hayden, Eric Y; Kaur, Prerna; Williams, Thomas L et al. (2015) Heme Stabilization of ?-Synuclein Oligomers during Amyloid Fibril Formation. Biochemistry 54:4599-610
Ahn, Young O; Mahinthichaichan, Paween; Lee, Hyun Ju et al. (2014) Conformational coupling between the active site and residues within the K(C)-channel of the Vibrio cholerae cbb3-type (C-family) oxygen reductase. Proc Natl Acad Sci U S A 111:E4419-28
Sabat, Joseph; Egawa, Tsuyoshi; Lu, Changyuan et al. (2013) Catalytic intermediates of inducible nitric-oxide synthase stabilized by the W188H mutation. J Biol Chem 288:6095-106

Showing the most recent 10 out of 29 publications