首页 | 本学科首页   官方微博 | 高级检索  
   检索      


Correlation of Fragile Histidine Triad (Fhit) Protein Structural Features with Effector Interactions and Biological Functions
Authors:Flavia Pichiorri  Hiroshi Okumura  Tatsuya Nakamura  Preston N Garrison  Pierluigi Gasparini  Sung-Suk Suh  Teresa Druck  Kelly A McCorkell  Larry D Barnes  Carlo M Croce  and Kay Huebner
Institution:Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University and Comprehensive Cancer Center, Columbus, Ohio 43210 and the §Department of Biochemistry, University of Texas Health Science Center, San Antonio, Texas 78229
Abstract:We have previously shown that Fhit tumor suppressor protein interacts with Hsp60 chaperone machinery and ferredoxin reductase (Fdxr) protein. Fhit-effector interactions are associated with a Fhit-dependent increase in Fdxr stability, followed by generation of reactive oxygen species and apoptosis induction under conditions of oxidative stress. To define Fhit structural features that affect interactions, downstream signaling, and biological outcomes, we used cancer cells expressing Fhit mutants with amino acid substitutions that alter enzymatic activity, enzyme substrate binding, or phosphorylation at tyrosine 114. Gastric cancer cell clones stably expressing mutants that do not bind substrate or cannot be phosphorylated showed decreased binding to Hsp60 and Fdxr and reduced mitochondrial localization. Expression of Fhit or mutants that bind interactor proteins results in oxidative damage and accumulation of cells in G2/M or sub-G1 fractions after peroxide treatment; noninteracting mutants are defective in these biological effects. Gastric cancer clones expressing noncomplexing Fhit mutants show reduction of Fhit tumor suppressor activity, confirming that substrate binding, interaction with heat shock proteins, mitochondrial localization, and interaction with Fdxr are important for Fhit tumor suppressor function.Fhit protein is a powerful tumor suppressor that is frequently lost or reduced in cancer cells because of rearrangement of the exquisitely DNA damage-sensitive fragile FHIT gene. Restoration of Fhit expression suppresses tumorigenicity of cancer cells of various types, and the ability to induce apoptosis in cancer cells in vitro is reduced by specific Fhit mutations (1, 2).Through studies of signal pathways affected by Fhit expression, by searches for Fhit protein effectors, and by in vitro analyses of Fhit activity, we and others have defined Fhit enzymatic activity in vitro (3), apoptotic activity in cells and tumors (46), and most recently identification of a Fhit protein complex that affects Fhit stability, mitochondrial localization, and interaction with ferredoxin reductase (Fdxr)5 (7). The complex includes Hsp60 and Hsp10 that mediate Fhit stability and may affect import into mitochondria, where Fhit interacts with Fdxr, which is responsible for transferring electrons from NADPH to cytochrome P450 via ferredoxin. Virally mediated Fhit restoration in Fhit-deficient cancer cells increases production of intracellular reactive oxygen species (ROS), followed by increased apoptosis of cancer cells under oxidative stress conditions; conversely, Fhit-negative cells escape apoptosis, likely carrying oxidative DNA damage that contributes to accumulation of mutations.The Fhit protein sequence, showing high homology to the histidine triad (HIT) family of proteins, suggested that the protein product would hydrolyze diadenosine tetraphosphate or diadenosine triphosphate (Ap3A) (8), and in vitro studies showed that Ap3A was cleaved into ADP and AMP by Fhit. The catalytic histidine triad within Fhit was essential for catalytic activity (3), and a Fhit mutant that substituted Asn for His at the central histidine (H96N mutant) was catalytically inactive, although it bound substrate well (3). Early tumor suppression studies showed that cancer cells stably transfected with wild type (WT) or H96N mutant Fhit were suppressed for tumor growth in nude mice. This suggested the hypothesis that the Fhit-substrate complex sends the tumor suppression signal (9, 10). To test this hypothesis, a series of FHIT alleles was designed to reduce substrate-binding and/or hydrolytic rates and was characterized by quantitative cell-death assays on cancer cells virally infected with each allele. The allele series covered defects as great as 100,000-fold in kcat and increases as large as 30-fold in Km. Mutants with 2–7-fold increases in Km had significantly reduced apoptotic indices and the mutant with a 30-fold increase in Km retained little apoptotic function. Thus, the proapoptotic function of Fhit, which is likely associated with tumor suppressor function, is limited by substrate binding and is unrelated to substrate hydrolysis (11).Fhit, a homodimeric protein of 147 amino acids, is a target of tyrosine phosphorylation by the Src family protein kinases, which can phosphorylate Tyr-114 of Fhit in vitro and in vivo (12). After co-expression of Fhit with the Elk tyrosine kinase in Escherichia coli to generate phosphorylated forms of Fhit, unphosphorylated, mono-, and diphosphorylated Fhit were purified, and enzyme kinetics studies showed that monophosphorylated Fhit exhibited monophasic kinetics with Km and kcat values ∼2- and ∼7-fold lower, respectively, than for unphosphorylated Fhit. Diphosphorylated Fhit exhibited biphasic kinetics; one site had Km and kcat values ∼2- and ∼140-fold lower, respectively, than for unphosphorylated Fhit; the second site had a Km ∼60-fold higher and a kcat ∼6-fold lower than for unphosphorylated Fhit (13). Thus, it was possible that the alterations in Km and kcat values for phosphorylated forms of Fhit might favor formation and lifetime of the Fhit-Ap3A complex and enhance tumor suppressor activity (see
Fhit forms
Kinetic parameters
% Sub-G1
Direct binding
Subcellular location
Co-IP in vivo
8-OHdG
Apoptosis
Tumor suppressor
Km (mm)kcat (s–1)A549MKN74Hsp60FdxrHsp60Fdxr
Fhit WT 1.6 +/– 0.19 2.7 +/– 0.95 43 24 Yes Yes Cyt & mito Yes Yes Yes Yes Yes
Catalyt mutants
   H96D Up 2-fold Down >2 × 104 29 NT NT NT Cyt & mito Yes Yes NT Yes NT
   H96N Up 2-fold Down >5 × 105 31 14.4 NT NT Cyt & mito Yes Yes Yes Yes Yes
Loop mutants
   Y114A Up 23-fold Down 2-fold 3.7 NT NT NT Cyt +/– +/– +/– No No
   Y114D NT NT 2.9 6 NT NT Cyt +/– +/– No –/+
   Y114E NT NT NT NT NT NT Cyt & mito –/+ –/+ No NT
   Y114F Up 5-fold Up 1.1-fold 11.5 3 NT NT Cyt & mito –/+ –/+ No No
   Y114W Up 5-fold Up 1.4-fold NT NT NT NT Cyt & mito –/+ NT NT
   del113–117 Up 10-fold Down 38-fold 5 NT NT NT NT NT NT No NT
Other mutants
   L25W Up 7-fold Down 4-fold 15 NT NT NT Cyt NT –/+
   I10W,L25W Up 32-fold Down 6-fold 11 NT NT NT NT NT NT NT NT NT
   F5W Up 3.3 fold NT NT 5 NT NT NT NT NT +/– No NT
Purified pFhit
   pFhit Down 0.4-fold Down 7-fold NA NA –/+ Yes NA NA NA NA NA NA
   ppFhit Down 0.4-fold Down > 100-fold NA NA –/+ Yes NA NA NA NA NA NA
Up 60-fold Down 6-fold
Open in a separate windowTo explore the in vivo importance of the Tyr-114 phosphorylation site and define Fhit-mediated signaling events, Semba et al. (14) compared the differential biological effects of Ad-FHIT WT and Ad-FHIT Tyr-114 mutant expression in human lung cancer cells. Caspase-dependent apoptosis was effectively induced only by WT Fhit protein. However, the biological significance of phosphorylation at Tyr-114 has been difficult to study because the endogenous phosphorylated forms have very short half-lives; activation of epidermal growth facto receptor family members induces Fhit phosphorylation by Src and proteasome degradation of phosphorylated Fhit (15).Although there are possible connections among the various pathways known to be altered in Fhit-deficient cells, apoptosis, DNA damage-response checkpoint activation, ROS production, and related biological effects of Fhit loss or overexpression, details of the pathway(s) leading from Fhit overexpression to cell death and tumor suppression have not been delineated. Now that a Fhit signaling complex has been identified, we set out to examine which structural features of Fhit protein might participate in individual steps of the pathway leading from Fhit overexpression through complex formation, subcellular localization, interaction with mitochondrial Fdxr, DNA damage induction, cell cycle changes, apoptosis, and ultimately tumor suppression. The underlying hypotheses were as follows: substrate-binding mutants would behave similarly to WT; nonsubstrate-binding mutants would be defective in some step of the pathway, perhaps complexing with heat shock proteins or Fdxr or perhaps induction of DNA damage; and Tyr-114 mutants, which also affect formation or stability of the enzyme-substrate complex, would also be defective in executing some step of the Fhit overexpression pathway to cell death. One goal was to identify specific mutants that exhibited deficiency in specific steps of the pathway, so that such mutants could be used to dissect each step in more detail. Using in vitro Fhit and Fhit-effector protein interactions, we aimed to determine the following: 1) which proteins of the complex interact directly with Fhit, and 2) the biological role of these interactions in vivo. Using cancer cells expressing exogenous WT and mutant Fhit proteins, we were able to examine the structural features of Fhit that affect the direct interaction with its effectors, participate in ROS production, and are necessary for tumor suppression activity.
Keywords:
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号