Thromboxane synthase: structure and function of protein and gene

https://doi.org/10.1016/S0090-6980(02)00045-XGet rights and content

Introduction

Thromboxane A synthase (TXAS) catalyzes roughly equal amounts of two distinct reactions with PGH2 as substrate [1] (, ).PGH2TXA2PGH2→12-HHT+MDAThe first reaction is an isomerization of PGH2 to form thromboxane A2, the second a fragmentation of PGH2 to form 12-HHT and malondialdehyde. TXA2 has been implicated as an autocrine or paracrine lipid mediator in a wide variety of pathophysiological processes [2]. TXA2 generally acts via the TXA2 receptor, a protein which contains seven transmembrane domains and which is coupled to G-protein signaling pathways [3], [4]. TXAS activity was first described in platelets [5] and the enzyme was later purified to homogeneity as a 60 kDa hemoprotein with spectroscopic characteristics of a cytochrome P450 [6]. The assignment of TXAS to the P450 superfamily was confirmed when the cDNA was cloned and the functional importance of the cysteine ligand to the heme was demonstrated [7], [8], [9]. The focus of this review is on more recent studies of TXAS mechanism, kinetics, structure–function relationships, gene structure, and control of gene expression. The reader is encouraged to consult several excellent reviews in the past several years for more comprehensive coverage of other aspects of TXAS [10], [11], [12], [13], [14].

Section snippets

Heme environment

Initial work with TXAS purified from platelets established that the heme in the resting enzyme is in the ferric low spin state and that ligand-dependent shifts in the heme absorbance spectrum resemble those observed in other P450s [6]. The recent development of a bacterial system for large-scale expression of TXAS has permitted more detailed and extensive spectroscopic characterization of the heme environment [15]. The EPR spectrum of recombinant TXAS revealed signals for low spin ferric heme

Reaction mechanism and kinetics

Analysis of spectral shifts observed upon binding of PGH2 analogs that have a carbon substituted for one of the oxygens in the endoperoxide function (U44069 and U46619) led to the conclusion that the TXAS heme iron interacts with the oxygen attached to C-9 of PGH2 [16]. This interaction of ferric TXAS heme with the C-9 endoperoxide oxygen is the first step in the reaction mechanism proposed by Hecker and Ullrich ([16], Fig. 1). Stopped-flow kinetic studies with U44069 and recombinant TXAS

Structure–function studies

A structural model for TXAS has been constructed based on crystallographic data for P450BM3 [21]. This structural model has guided mutagenic studies targeted at residues predicted to be involved with heme or substrate binding [22], [23]. The relative positions of the mutated residues in the structural model are illustrated in Fig. 2. In agreement with an earlier report [9], the proximal heme ligand C480 was found to be critical for activity, with all substitutions at that position resulting in

Human TXAS variants

Eight missense mutations have been found in the TXAS gene in a sample of 200 normal volunteers [24]. This natural mutagenic study revealed R61H, D161E, N246S, L357V, Q417E, E450K, T451N, and R466Q substitutions. The activities of the mutant enzymes were not examined directly, but no clinical abnormalities were evident. Each of these TXAS variants involve residues which are at, or close to, the protein surface and well away from the active site in the TXAS structural model (Fig. 3). Of

Integration of TXAS into eicosanoid pathway

TXA2 synthesis in vivo obviously involves coordinated action of a series of enzymes, including phospholipases which release arachidonate from esterified precursors, PGH synthase-1 or -2 which convert arachidonate into PGH2, and TXAS itself. PGH2 can be converted into a number of other prostanoid signaling mediators besides TXA2, and the cellular controls which determine the actual profile of prostanoids produced in a given cell under a particular set of circumstances are beginning to be

Organization of the human TXAS gene

The human TXAS is a single copy gene, located at chromosome 7q34-35; isolation of the complete TXAS genomic DNA was hampered by its large size [28], [29]. A 193-kb contiguous DNA containing the entire human TXAS gene was mapped from a combination of yeast artificial chromosome and bacteriophage P1 libraries [30]. As shown in Fig. 4, the TXAS gene contains 13 exons with five large introns; two of 45-kb and one each of 35-, 25- and 17-kb, making it the largest cytochrome P450 gene characterized

TXAS gene expression

The TXAS gene is transcribed as a 2.1-kb mRNA (0.14-kb of 5′-untranslated region, 1.6-kb of coding region and 0.4-kb of 3′-untranslated region) in hematopoietic cells, such as platelets, macrophages, monocytes and leukocytes, as well as in various tissues, particularly in lung, kidney, liver, spleen, prostate, placenta and thymus [28], [33]. Single nucleotide polymorphisms were found in the promoter, protein coding region and 3′-untranslated region. In the promoter, a G to T mutation at −386 in

Transcriptional regulation of the TXAS gene

The transcriptional start site of human TXAS gene was identified in the same exon as the translational start site, and mapped 137–140 nucleotides upstream from the translational start site [28], [30]. The core promoter contains two putative elements, a TATA box and an initiator, located at −29/−26 and −3/+3, respectively (Fig. 5). The sequence of the pyrimidine-rich TXAS initiator, CACA+1TT, resembles a weak initiator consensus sequence PyPyA+1NT/APyPy (A is the transcriptional start site and

Acknowledgements

Work in the authors’ laboratories has been supported by United States National Institutes of Health grants GM52170, HL60625 and NS23327. We thank Drs. Ah-Lim Tsai and Masahiro Yaekashiwa for critical reading of the manuscript and Drs. Ke-He Ruan and Jiaxin Wu for help with the TXAS structural model.

First page preview

First page preview
Click to open first page preview

References (60)

  • A.L. Tsai et al.

    An improved sample packing device for rapid freeze-trap electron paramagnetic resonance spectroscopy kinetic measurements

    Anal. Biochem.

    (1998)
  • L.H. Wang et al.

    Identification of thromboxane A2 synthase active site residues by molecular modeling-guided site-directed mutagenesis

    J. Biol. Chem.

    (1996)
  • P.Y. Hsu et al.

    Identification of thromboxane synthase amino acid residues involved in heme-propionate binding

    Arch. Biochem. Biophys.

    (2000)
  • N. Ueno et al.

    Coupling between cyclooxygenase, terminal prostanoid synthase, and phospholipase A2

    J. Biol. Chem.

    (2001)
  • S.J. Baek et al.

    Genomic structure and polymorphism of the human thromboxane synthase-encoding gene

    Gene

    (1996)
  • R. Tazawa et al.

    Characterization of the complete genomic structure of human thromboxane synthase gene and functional analysis of its promoter

    Arch. Biochem. Biophys.

    (1996)
  • L. Zhang et al.

    Genomic organization, chromosomal localization, and expression of murine thromboxane synthase gene (Tbxas1)

    Genomics

    (1997)
  • Y. Tsujita et al.

    Role of a genetic variation in the promoter of human thromboxane synthase gene in myocardial infarction

    Atherosclerosis

    (2000)
  • L.H. Wang et al.

    Alternate splicing of human thromboxane synthase mRNA

    Arch. Biochem. Biophys.

    (1994)
  • B.A. Lewis et al.

    A functional initiator in the human beta-globin promoter

    J. Biol. Chem.

    (1995)
  • S.H. Orkin

    Transcription factors and hematopoietic development

    J. Biol. Chem.

    (1995)
  • R.A. Shivdasani et al.

    Transcription factor NF-E2 is required for platelet formation independent of the actions of thrombopoietin/MGDF in megakaryocyte development

    Cell

    (1995)
  • A. Kobayashi et al.

    Molecular cloning and functional characterization of a new cap’n’collar family transcritpion factor, Nrf3

    J. Biol. Chem.

    (1999)
  • Y. Ikeda et al.

    Suppression of rat thromboxane synthase gene transcription by peroxisome proliferator-activated receptor gamma in macrophages via an interaction with NRF2

    J. Biol. Chem.

    (2000)
  • M.L. Ogletree

    Overview of physiological and pathophysiological effects of thromboxane A2

    Fed. Proc.

    (1987)
  • R. Murray et al.

    Regulation of thromboxane receptor activation in human platelets

    Proc. Natl. Acad. Sci. USA

    (1989)
  • M. Hirata et al.

    Cloning and expression of cDNA for a human thromboxane A2 receptor

    Nature

    (1991)
  • P. Needleman et al.

    Identification of an enzyme in platelet microsomes which generates thromboxane A2 from prostaglandin endoperoxides

    Nature

    (1976)
  • Z. Xia et al.

    Expression of two different forms of cDNA for thromboxane synthase in insect cells and site-directed mutagenesis of a critical cysteine residue

    Biochem. J.

    (1993)
  • R.F. Shen et al.

    Thromboxanes: synthase and receptors

    J. Biomed. Sci.

    (1998)
  • Cited by (0)

    View full text