monox

8
Characterizing the novel protein p33MONOX Manisha Mishra Noriko Inoue Klaus Heese Received: 4 June 2010 / Accepted: 18 September 2010 / Published online: 14 December 2010 Ó Springer Science+Business Media, LLC. 2010 Abstract The novel protein p33MONOX (p33Monoox- ygenase) was over-expressed in neuroblastoma cells dem- onstrating its inhibitory effect on the phosphorylation of the App (amyloid precursor protein) and Bcl2 (B-cell lymphoma 2) proteins but mediating higher activation of Mapk1/3 (mitogen-activated protein kinase 1/3). We employed a variety of cell biology techniques to show the localization of p33MONOX to the cytoplasm of pyramidal neurons in the mouse brain hippocampus. We also carried out a yeast-two-hybrid screening plus co-immunoprecipi- tation and bio-informatics to determine COBRA1 (cofactor of BRCA1 (breast cancer type 1)), NOL12 (nucleolar protein 12), and PRNP (prion protein) as p33MONOX- interacting proteins. Bio-computational analyses revealed a flavine-containing monooxygenase (FMO)-1 motif, thus linking p33MONOX to a group of previously character- ized proteins, the MICALs (molecule interacting with CasL). Concluding, p33MONOX might regulate pre- and post-transcriptional control of dynamic processes related to growth cone guidance. Keywords Alzheimer’s disease Á Apoptosis Á Neurodegeneration Introduction Alzheimer’s Disease (AD) remains the most common cause of dementia in all age groups, characterized by progressive neurodegeneration and profound cognitive deficits [14]. Much of what is known about AD revolves around the amyloid precursor protein (APP) and presenilin-1 (PSEN1) and PSEN2 [5]. However, there remains a myriad of other proteins that could possibly play an equally crucial role in the development of AD. Although the etiology of sporadic AD is poorly understood, there is evidence that aberrant iron deposition, oxidative stress and mitochondria insufficiency play a role in the pathogenesis of sporadic AD and other aging-related neurodegenerative disorders. The excessive generation of free radicals may promote neurofibrillary tangle (NFT) formation as well as amyloid deposition in AD brains. Conversely, the neurotoxic effects of certain amyloid fragments may be mediated by free radical intermediates [68]. P33MONOX was discovered in our recent study on brain site-specific gene-expression analysis, when we com- pared the gene-expression pattern in the temporal and occipital lobe of early stage AD subjects with control patients [9]. In that study, p33MONOX, a novel gene with unknown functions as yet, was identified to be down-regu- lated in the occipital lobe of an early stage AD patient. In the current study, we characterized the potential biological sig- nificance of p33MONOX using molecular and cell biologi- cal as well as bio-computational analyses. Electronic supplementary material The online version of this article (doi:10.1007/s11010-010-0690-4) contains supplementary material, which is available to authorized users. M. Mishra Á K. Heese (&) Department of Molecular and Cell Biology, School of Biological Sciences, College of Science, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore, Singapore e-mail: [email protected] N. Inoue Medical Center for Translational Research, Osaka University Hospital, Suita, Osaka, Japan 123 Mol Cell Biochem (2011) 350:127–134 DOI 10.1007/s11010-010-0690-4

Upload: deodatus-kardo-girsang

Post on 16-Sep-2015

217 views

Category:

Documents


4 download

DESCRIPTION

Journal about monox

TRANSCRIPT

  • Characterizing the novel protein p33MONOX

    Manisha Mishra Noriko Inoue Klaus Heese

    Received: 4 June 2010 / Accepted: 18 September 2010 / Published online: 14 December 2010

    Springer Science+Business Media, LLC. 2010

    Abstract The novel protein p33MONOX (p33Monoox-

    ygenase) was over-expressed in neuroblastoma cells dem-

    onstrating its inhibitory effect on the phosphorylation of

    the App (amyloid precursor protein) and Bcl2 (B-cell

    lymphoma 2) proteins but mediating higher activation of

    Mapk1/3 (mitogen-activated protein kinase 1/3). We

    employed a variety of cell biology techniques to show the

    localization of p33MONOX to the cytoplasm of pyramidal

    neurons in the mouse brain hippocampus. We also carried

    out a yeast-two-hybrid screening plus co-immunoprecipi-

    tation and bio-informatics to determine COBRA1 (cofactor

    of BRCA1 (breast cancer type 1)), NOL12 (nucleolar

    protein 12), and PRNP (prion protein) as p33MONOX-

    interacting proteins. Bio-computational analyses revealed a

    flavine-containing monooxygenase (FMO)-1 motif, thus

    linking p33MONOX to a group of previously character-

    ized proteins, the MICALs (molecule interacting with

    CasL). Concluding, p33MONOX might regulate pre- and

    post-transcriptional control of dynamic processes related to

    growth cone guidance.

    Keywords Alzheimers disease Apoptosis Neurodegeneration

    Introduction

    Alzheimers Disease (AD) remains the most common cause

    of dementia in all age groups, characterized by progressive

    neurodegeneration and profound cognitive deficits [14].

    Much of what is known about AD revolves around the

    amyloid precursor protein (APP) and presenilin-1 (PSEN1)

    and PSEN2 [5]. However, there remains a myriad of other

    proteins that could possibly play an equally crucial role in the

    development of AD. Although the etiology of sporadic AD is

    poorly understood, there is evidence that aberrant iron

    deposition, oxidative stress and mitochondria insufficiency

    play a role in the pathogenesis of sporadic AD and other

    aging-related neurodegenerative disorders. The excessive

    generation of free radicals may promote neurofibrillary

    tangle (NFT) formation as well as amyloid deposition in AD

    brains. Conversely, the neurotoxic effects of certain amyloid

    fragments may be mediated by free radical intermediates

    [68]. P33MONOX was discovered in our recent study on

    brain site-specific gene-expression analysis, when we com-

    pared the gene-expression pattern in the temporal and

    occipital lobe of early stage AD subjects with control

    patients [9]. In that study, p33MONOX, a novel gene with

    unknown functions as yet, was identified to be down-regu-

    lated in the occipital lobe of an early stage AD patient. In the

    current study, we characterized the potential biological sig-

    nificance of p33MONOX using molecular and cell biologi-

    cal as well as bio-computational analyses.

    Electronic supplementary material The online version of thisarticle (doi:10.1007/s11010-010-0690-4) contains supplementarymaterial, which is available to authorized users.

    M. Mishra K. Heese (&)Department of Molecular and Cell Biology,

    School of Biological Sciences, College of Science,

    Nanyang Technological University, 60 Nanyang Drive,

    637551 Singapore, Singapore

    e-mail: [email protected]

    N. Inoue

    Medical Center for Translational Research,

    Osaka University Hospital, Suita, Osaka, Japan

    123

    Mol Cell Biochem (2011) 350:127134

    DOI 10.1007/s11010-010-0690-4

  • Materials and methods

    Reagents

    All reagents used for experiments were purchased from

    Sigma-Aldrich (Milwaukee, WI, USA) unless otherwise

    stated.

    Cell culture and transfection

    Rat B104 and human SHSY5Y neuroblastoma cells as well

    as rat PC12 cells (all from American Type Culture Col-

    lection (ATCC, Manassas, VA, USA)) were maintained in

    Dulbeccos Modified Eagle Medium (D-MEM/F12(1:1))

    plus 10% fetal bovine serum (FBS; Invitrogen, (Gibco),

    Carlsbad, CA, USA) at 37C in humidified 5% CO2/95%air. A p33Monox expression construct was generated by

    inserting rat p33Monox cDNA in-frame with the red fluo-

    rescent protein (DsRed) (pDsRed-Express-N1; BD Bio-

    sciences Clontech, Palo Alto, CA, USA) at the C-terminus

    of p33Monox (p33-CT-DsRed). B104 cells were tran-

    siently transfected with the p33-CT-DsRed expression

    vector using the Lipofectamine 2000 (Invitrogen) trans-

    fection reagent (according to the manufacturers protocol)

    and maintained in D-MEM medium containing 10% FBS

    at 37C. The transfected cells were then visualized byfluorescence microscopy (Nikon eclipse TE2000U, Nikon,

    Singapore). SHSY5Y, B104, and PC12 cells were stably

    transfected using a lentivirus expression system (p33 in

    EF.CMV.Gfp-Lenti-vector (elongation factor 1 alpha,

    cytomegalovirus promotors, green fluorescent protein;

    JHU-55, ATCC); co-expression of p33Monox and green

    Gfp) (control, mock-transfection) according to the manu-

    facturers protocol (Invitrogen) as briefly described in

    Supplementary materials and methods [10, 11].

    Western blot analyses

    Total protein cell lysates were subjected to western blot

    analyses as described previously (Supplementary materials

    and methods) [9, 10].

    Animal material, immunohistochemistry (IHC)

    and immunocytochemistry (ICC)

    Experimental methods, including the killing of animals,

    were performed in accordance with the International

    Guiding Principles for Animal Research (WHO) and

    approved by the local Institutional Animal Care & Use

    Committee (NTU-IACUC). Mouse tissues were isolated

    (C57BL/6J mice from the Animal Facility Centre at the

    National University (NUS) of Singapore) after humane

    killing of the animals using approved anaesthetic methods.

    Mouse brain perfusion, IHC, and ICC were performed

    as described previously (Supplementary materials and

    methods) [11].

    ProQuestTM two-hybrid-system with GatewayTM

    technology

    The two-hybrid-system is an in vivo yeast-based system

    that identifies the interaction between two proteins (here

    X = p33MONOX and Y = human brain cDNA library or

    COBRA1) by reconstituting an active transcription factor.

    The analysis was performed according to the manufac-

    turers protocol (Invitrogens brain ProQuestTM two-

    hybrid-system, Singapore) using p33MONOX as bait. In

    the ProQuestTM two-hybrid-system, in comparison to

    standard two-hybrid-systems, false positives are reduced

    because three independent transcription events (from dis-

    tinct promoters) must occur at independent chromosomal

    loci. Positive clones were confirmed by retransformation

    assays and protein co-immunoprecipitation (Co-IP, Sup-

    plementary materials and methods) [10].

    Results

    p33MONOX protein sequence analysis

    Bio-computational analyses of the p33MONOX protein

    sequence among the species of human, mouse, and rat

    showed high sequence similarity (Fig. 1) pointing to the

    possibility that p33MONOX plays a crucial role that is

    evolutionarily conserved.

    The most encouraging information about p33MONOX

    is the presence of a flavine-containing monooxygenase

    (FMO)-1 motif. The proteins comprising the FMO motif

    belong to a family of microsomal NADPH (nicotinamide

    adenine dinucleotide phosphate)- and oxygen-dependent

    flavoenzymes (with flavin adenine nucleotide (FAD) as a

    co-factor) that are distributed ubiquitously in mammalian

    species, and catalyze the oxidation of soft nucleophilic

    heteroatom centers in drugs, pesticides, and xenobiotics,

    using nucleotides as electron donors. FMO-1 catalyzes the

    N-oxygenation of secondary and tertiary amines. In some

    contexts, while performing the oxidation, they can generate

    reactive oxygen species (ROS) [1215]. Thus,

    p33MONOX protein is more likely to be a NADPH-

    dependent oxidoreductase.

    Sub-cellular localization and neuronal expression

    of p33Monox in the mouse brain

    We investigated the sub-cellular localization of p33Monox

    to obtain more information about its physiological role,

    128 Mol Cell Biochem (2011) 350:127134

    123

  • distribution, and site of activity in the cell. For this pur-

    pose, a p33Monox-DsRed fluorescent fusion protein was

    transiently expressed in B104 neuroblastoma cells. Using

    fluorescence microscopy, we show that p33Monox

    expression was confined to the neuronal cytoplasm thus

    confirming the bio-computational analysis data (Fig. 2a).

    Besides, ICC of nerve growth factor (Ngf)-differentiated

    PC12 cells also verified the localization of p33Monox in

    the cytoplasm with a substantial expression in the axonal

    growth cone (Fig. 2b).

    Furthermore, we conducted an IHC analysis of the

    mouse brain for the p33Monox protein expression and

    localization. This analysis revealed that p33Monox was

    physiologically expressed in neuronal pyramidal cells of

    the hippocampus and also in the neurons of the cortex

    (Fig. 3).

    p33Monox-mediated neuronal signaling

    To further corroborate the physiological significance of

    p33Monox, we analyzed its effect on the activation of

    pivotal proteins involved in neuronal survival and differ-

    entiation. Interestingly, upon over-expression in neuronal

    cells, p33Monox inhibited the phosphorylation of App

    (reduced Ab formation [16]) and Bcl2 (the functional sig-nificance of the dynamic phosphorylation status (regulated

    Fig. 1 Characteristic featuresof the p33MONOX protein

    sequence. Aligned protein

    sequences of p33MONOX in

    the human (H, black), mouse(M, blue), and rat (R, green)species, with red alphabetsdenoting the varying amino

    acid. There is evidently a high

    degree of conservation in the

    protein sequences amongst the

    species, suggesting a pivotal

    functional significance of

    p33MONOX. Bio-informatical

    analyses of p33MONOXs

    protein sequence revealed a

    potential flavine-containing

    monooxygenase (FMO)-1 motif

    and several Ser-/Thr-

    phosphorylation sites

    Mol Cell Biochem (2011) 350:127134 129

    123

  • by various kinases and protein phosphatase PP2A) has been

    discussed conflictive [1722]) while Erk1/2 (Mapk1/3) was

    activated (sustained phosphorylation is required for neu-

    ronal differentiation [23]) (Fig. 4a).

    p33MONOX interacts with COBRA1, NOL12,

    and PRNP

    Additional clarification about p33Monoxs potential cel-

    lular function was obtained by the yeast-two-hybrid screen

    combined with bio-informatic analyses (NCBI (National

    Center for Biotechnology information), EMBL-EBI

    (European Bioinformatics Institute), and SIB (Swiss Insti-

    tute of Bioinformatics, (Swiss-Prot & Tremble, ExPASy,

    and Proteomics tools)) databases were used). The analyses

    revealed that p33MONOX interacts with several proteins

    involved in the control of gene transcription, such

    as COBRA1 (the co-factor of BRCA1 is a newly charac-

    terized member of the negative elongation factor (NELF)

    complex; confirmed by Co-IP (Fig. 4b)) [2428], NOL12

    (nucleolar protein 12, also known as ribosomal RNA pro-

    cessing protein 17) [29, 30], and the prion protein (PRNP

    or PrP with isoform 2 as a potential growth suppressor

    that arrests the cell cycle at the G0/G1 phase) [3133]

    (Supplementary Tables 1 and 2).

    NOL12 itself interacts with several other proteins

    known to be pivotal regulators of gene transcription and

    cell cycle progression such as: SAP18 (or Sin3A-associated

    protein), CDK4 (cyclin-dependent kinase 4), SF3B3 (sub-

    unit 3 of the splicing factor 3b protein complex), and

    SLC25A38 (solute carrier family 25, member 38), a

    mitochondrial carrier protein that is widely expressed

    Fig. 2 Sub-cellular localizationof p33Monox. a B104 cellswere transfected with a

    p33-CT-DsRed expression

    vector as described in

    Supplementary materials and

    methods. Microscopic picture of

    a representative B104 cell under

    bright-field, red fluorescence(revealing cytoplasmic

    localization of p33Monox), and

    UV light (indicating nuclear

    DAPI staining), respectively;

    scale bar = 20 lm. b Co-ICCof Ngf-differentiated PC12 cells

    with p33Monox (green) andTuba1a (tubulin, red) or Syp(synaptophysin, red) asindicated. DAPI staining was

    used to indicate the nucleus.

    Representative pictures are

    shown. Pictures show the strong

    co-localization (merged, yellow)of p33Monox with tubulin in the

    cytoplasm and with Syp in the

    axonal growth cones (arrows).Scale bar = 50 lm

    130 Mol Cell Biochem (2011) 350:127134

    123

  • in the central nervous system [34]. Interestingly, by direct

    interaction NOL12 links p33Monox to SOD2 (superoxide

    dismutase 2). This protein is a member of the iron/

    manganese superoxide dismutase family that binds to the

    superoxide byproducts of oxidative phosphorylation and

    converts them to hydrogen peroxide and diatomic oxygen.

    Mutations in this gene have been associated with premature

    aging and sporadic motor neuron disease [35, 36] (further

    details in Supplementary Table 2).

    Discussion

    In the present study, we revealed the specific sites of

    p33Monox expression and its intensive localization in

    neural axonal growth cones. Apart from this, we observed

    that p33Monox showed an interesting inhibition of the

    phosphorylation of App and Bcl2 as well as an enhanced

    activation of Mapk1/3 [16, 20, 23]. We also found that

    p33Monox forms a complex with Cobra1, Nol12, and Prnp.

    Cobra1, also known as Nelf-b that is involved in control-

    ling axonal growth [25, 26], associates with the product of

    the breast cancer susceptibility gene Brca1, thus hinting at

    a plausible role of p33Monox in sequestering Cobra1 and

    thereby leading to profound effects on gene transcription

    signals during dynamic neurite outgrowth processes.

    Consequently, p33Monox can be likened to a group of

    cytosolic proteins, the MICALs (molecule interacting with

    CasL), which are also oxidoreductases that utilize FAD as a

    co-factor and communicate via ROS [3739]. During the

    past decade, we have begun to recognize that controlled

    production of ROS and regulated redox modifications of

    Fig. 3 Co-IHC analysis of p33Monox (green) and Mtap2 (red) inthe mouse brain revealed that p33Monox is expressed in the

    cytoplasm of pyramidal neurons in the hippocampus. Top left entirehippocampus formation including CA1, CA2, CA3 regions, and the

    dentate gyrus. Scale bar = 100 lm. Right enlarged CA1 regions.

    Scale bar = 20 lm. Bottom/left further magnified picture of the CA1region, clearly shows the cytoplasmic localization of p33Monox.

    Scale bar = 5 lm. Bottom/right representative picture taken form thecortex area thus indicating that p33Monox is generally expressed in

    neurons in the mouse brain. Scale bar = 20 lm

    Mol Cell Biochem (2011) 350:127134 131

    123

  • transcription factors or enzymes (such as kinases and

    phosphatases) are an essential part of signal transduction

    pathways [4042]. Similar to p33Monox, the MICALs are

    expressed in neuronal axons that associate with several

    cytoskeletal/-associated proteins, and are required for

    semaphorin-mediated repulsive axon guidance that is cru-

    cial for neuronal development. P33Monox may be another

    candidate for directly mediating the cytoskeletal alterations

    characteristic of semaphorin signaling and could be a novel

    target for the attenuation of axonal repulsion (Fig. 5).

    Given the presence of high amounts of ROS and other

    oxidants in the spinal cord after injury [43], and that aging

    and AD have previously been closely linked to the accu-

    mulation of oxidative stress [4446], regulation of redox

    signaling using antioxidants and specific enzyme inhibitors

    may be a powerful approach for encouraging neuronal

    regeneration [37]. The plausible link between MICAL,

    p33Monox, and SOD2 points to a role of p33Monox

    in controlling ROS that have been recently suggested to be a

    key factor in the cellular changes of an AD brain as several

    reports have suggested that mitochondrial abnormalities and

    oxidative stress play a role in sporadic AD [4749]. For

    instance, the heme-oxygenase-1 (HO-1), a member of the

    stress protein superfamily that operates with the NADPH

    cytochrome P450 reductase to oxidize heme, is widely

    accepted as a sensitive and fairly ubiquitous up-regulated

    marker of oxidative stress. It has also been shown to be

    consistently co-localized to NFTs and senile plaques in AD

    brains [5052].

    In conclusion, our data has shed new insights on the

    possible involvement of the novel protein p33Monox in the

    regulation of neuronal survival, differentiation, and axonal

    outgrowth and the connection among p33Monox, oxidative

    stress and AD will be a gripping topic for future investiga-

    tions, especially in view of potential antioxidant therapies.

    Acknowledgments This study was supported by an A*STAR grant(BMRC/04/1/22/19/360) to K.H. We thank Ms H.J. Tang and

    S. Yusof (both from the School of Biological Sciences, Nanyang

    Technical University) for technical assistance. We are particularly

    grateful to Prof. Dr. R. Li (Department of Molecular Medicine,

    Institute of Biotechnology, The University of Texas Health Science

    Center, 15355 Lambda Drive, San Antonio, TX, 78245-3207, USA)

    for providing us the anti-COBRA1 antibody.

    Fig. 4 a p33Monox inhibits the phosphorylation of pivotal signalingmolecules as indicated. Neuronal B104 cells were transfected with

    p33Monox as described in Supplementary materials and methods.

    Thereafter, the phosphorylation status was checked by western

    blotting. C control, non-transfected, GFP mock-transfection withGFP, p33Monox p33Monox-transfected. b p33MONOX Co-IP. Upontransfection of neuronal SHSY5Y cells, Co-IP was performed as

    described in Supplementary materials and methods to confirm the

    interaction between p33MONOX and COBRA1. Control Co-IP withunspecific serum

    Fig. 5 Schematic illustration of the potential action of p33MONOXin mediating neuronal survival, differentiation, and axonal outgrowth.

    In light of experimental and bio-informatical evidences it can be

    speculated that p33MONOX may act as platform to recruit down-

    stream effectors (e.g., COBRA1, NOL12, PRNP, SOD2, NF-jB,ROS, kinases, or phosphatases) to their site of action. The activity of

    these effectors could then be selectively modulated by redox

    modifications of key amino acid residues that could either be the

    direct effect of p33MONOXs monooxygenase activity or be the

    indirect consequence of a local increase in ROS. P33MONOX-

    mediated de-phosphorylation of App and Bcl2 as well as sustained

    phosphorylation of Mapk1/3 are required for the control of neuronal

    survival, differentiation, and growth-cone extension

    132 Mol Cell Biochem (2011) 350:127134

    123

  • References

    1. Welsh KA, Butters N, Hughes JP, Mohs RC, Heyman A (1992)

    Detection and staging of dementia in Alzheimers disease. Use of

    the neuropsychological measures developed for the Consortium

    to Establish a Registry for Alzheimers Disease. Arch Neurol

    49:448452

    2. Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R,

    Hansen LA, Katzman R (1991) Physical basis of cognitive

    alterations in Alzheimers disease: synapse loss is the major

    correlate of cognitive impairment. Ann Neurol 30:572580

    3. Heese K, Akatsu H (2006) Alzheimers diseasean interactive

    perspective. Curr Alzheimer Res 3:109121

    4. Mattson MP (2004) Pathways towards and away from Alzhei-

    mers disease. Nature 430:631639

    5. Selkoe DJ, Schenk D (2003) Alzheimers disease: molecular

    understanding predicts amyloid-based therapeutics. Annu Rev

    Pharmacol Toxicol 43:545584

    6. Butterfield DA (2002) Amyloid beta-peptide (1-42)-induced

    oxidative stress and neurotoxicity: implications for neurodegen-

    eration in Alzheimers disease brain. A review. Free Radic Res

    36:13071313

    7. Butterfield DA, Castegna A, Lauderback CM, Drake J (2002)

    Evidence that amyloid beta-peptide-induced lipid peroxidation

    and its sequelae in Alzheimers disease brain contribute to neu-

    ronal death. Neurobiol Aging 23:655664

    8. Pappolla MA, Chyan YJ, Omar RA, Hsiao K, Perry G, Smith

    MA, Bozner P (1998) Evidence of oxidative stress and in vivo

    neurotoxicity of beta-amyloid in a transgenic mouse model of

    Alzheimers disease: a chronic oxidative paradigm for testing

    antioxidant therapies in vivo. Am J Pathol 152:871877

    9. Yokota T, Mishra M, Akatsu H, Tani Y, Miyauchi T, Yamamoto

    T, Kosaka K, Nagai Y, Sawada T, Heese K (2006) Brain site-

    specific gene expression analysis in Alzheimers disease patients.

    Eur J Clin Invest 36:820830

    10. Heese K, Yamada T, Akatsu H, Yamamoto T, Kosaka K, Nagai

    Y, Sawada T (2004) Characterizing the new transcription regu-

    lator protein p60TRP. J Cell Biochem 91:10301042

    11. Nehar S, Mishra M, Heese K (2009) Identification and charac-

    terisation of the novel amyloid-beta peptide-induced protein p17.

    FEBS Lett 583:32473253

    12. Massey V (1995) Introduction: flavoprotein structure and mech-

    anism. FASEB J 9:473475

    13. Yeung CK, Lang DH, Thummel KE, Rettie AE (2000) Immu-

    noquantitation of FMO1 in human liver, kidney, and intestine.

    Drug Metab Dispos 28:11071111

    14. Ziegler DM (1990) Flavin-containing monooxygenases: enzymes

    adapted for multisubstrate specificity. Trends Pharmacol Sci

    11:321324

    15. Massey V (1994) Activation of molecular oxygen by flavins and

    flavoproteins. J Biol Chem 269:2245922462

    16. Lee MS, Kao SC, Lemere CA, Xia W, Tseng HC, Zhou Y, Neve

    R, Ahlijanian MK, Tsai LH (2003) APP processing is regulated

    by cytoplasmic phosphorylation. J Cell Biol 163:8395

    17. Ito T, Deng X, Carr B, May WS (1997) Bcl-2 phosphorylation

    required for anti-apoptosis function. J Biol Chem 272:1167111673

    18. Deng X, Ito T, Carr B, Mumby M, May WS Jr (1998) Reversible

    phosphorylation of Bcl2 following interleukin 3 or bryostatin 1 is

    mediated by direct interaction with protein phosphatase 2A.

    J Biol Chem 273:3415734163

    19. Deng X, Gao F, Flagg T, May WS Jr (2004) Mono- and multisite

    phosphorylation enhances Bcl2s antiapoptotic function and

    inhibition of cell cycle entry functions. Proc Natl Acad Sci USA

    101:153158

    20. Yamamoto K, Ichijo H, Korsmeyer SJ (1999) BCL-2 is phos-

    phorylated and inactivated by an ASK1/Jun N-terminal protein

    kinase pathway normally activated at G(2)/M. Mol Cell Biol

    19:84698478

    21. Chang BS, Minn AJ, Muchmore SW, Fesik SW, Thompson CB

    (1997) Identification of a novel regulatory domain in Bcl-X(L)

    and Bcl-2. EMBO J 16:968977

    22. Liu XA, Liao K, Liu R, Wang HH, Zhang Y, Zhang Q, Wang Q,

    Li HL, Tian Q, Wang JZ (2010) Tau dephosphorylation poten-

    tiates apoptosis by mechanisms involving a failed depho-

    sphorylation/activation of Bcl-2. J Alzheimers Dis 19:953962

    23. Kholodenko BN (2007) Untangling the signalling wires. Nat Cell

    Biol 9:247249

    24. McChesney PA, Aiyar SE, Lee OJ, Zaika A, Moskaluk C, Li R,

    El-Rifai W (2006) Cofactor of BRCA1: a novel transcription

    factor regulator in upper gastrointestinal adenocarcinomas. Can-

    cer Res 66:13461353

    25. Kramer PR, Wray S (2001) Nasal embryonic LHRH factor

    (NELF) expression within the CNS and PNS of the rodent. Brain

    Res Gene Expr Patterns 1:2326

    26. Kramer PR, Wray S (2000) Novel gene expressed in nasal region

    influences outgrowth of olfactory axons and migration of

    luteinizing hormone-releasing hormone (LHRH) neurons. Genes

    Dev 14:18241834

    27. Sun J, Blair AL, Aiyar SE, Li R (2007) Cofactor of BRCA1

    modulates androgen-dependent transcription and alternative

    splicing. J Steroid Biochem Mol Biol 107:131139

    28. Narita T, Yamaguchi Y, Yano K, Sugimoto S, Chanarat S, Wada

    T, Kim DK, Hasegawa J, Omori M, Inukai N, Endoh M, Yamada

    T, Handa H (2003) Human transcription elongation factor NELF:

    identification of novel subunits and reconstitution of the func-

    tionally active complex. Mol Cell Biol 23:18631873

    29. Suzuki S, Kanno M, Fujiwara T, Sugiyama H, Yokoyama A,

    Takahashi H, Tanaka J (2006) Molecular cloning and character-

    ization of Nop25, a novel nucleolar RNA binding protein, highly

    conserved in vertebrate species. Exp Cell Res 312:10311041

    30. Stelzl U, Worm U, Lalowski M, Haenig C, Brembeck FH,

    Goehler H, Stroedicke M, Zenkner M, Schoenherr A, Koeppen S,

    Timm J, Mintzlaff S, Abraham C, Bock N, Kietzmann S, Goedde

    A, Toksoz E, Droege A, Krobitsch S, Korn B, Birchmeier W,

    Lehrach H, Wanker EE (2005) A human proteinprotein inter-

    action network: a resource for annotating the proteome. Cell

    122:957968

    31. Juanes ME, Elvira G, Garcia-Grande A, Calero M, Gasset M (2009)

    Biosynthesis of prion protein nucleocytoplasmic isoforms by

    alternative initiation of translation. J Biol Chem 284:27872794

    32. Satoh J, Obayashi S, Misawa T, Sumiyoshi K, Oosumi K, Tab-

    unoki H (2009) Protein microarray analysis identifies human

    cellular prion protein interactors. Neuropathol Appl Neurobiol

    35:1635

    33. Haigh CL, Lewis VA, Vella LJ, Masters CL, Hill AF, Lawson

    VA, Collins SJ (2009) PrPC-related signal transduction is influ-

    enced by copper, membrane integrity and the alpha cleavage site.

    Cell Res 19:10621078

    34. Haitina T, Lindblom J, Renstrom T, Fredriksson R (2006)

    Fourteen novel human members of mitochondrial solute carrier

    family 25 (SLC25) widely expressed in the central nervous sys-

    tem. Genomics 88:779790

    35. Miao L, St Clair DK (2009) Regulation of superoxide dismutase

    genes: implications in disease. Free Radic Biol Med 47:344356

    36. Lynn S, Huang EJ, Elchuri S, Naeemuddin M, Nishinaka Y,

    Yodoi J, Ferriero DM, Epstein CJ, Huang TT (2005) Selective

    neuronal vulnerability and inadequate stress response in super-

    oxide dismutase mutant mice. Free Radic Biol Med 38:817828

    Mol Cell Biochem (2011) 350:127134 133

    123

  • 37. Terman JR, Mao T, Pasterkamp RJ, Yu HH, Kolodkin AL (2002)

    MICALs, a family of conserved flavoprotein oxidoreductases,

    function in plexin-mediated axonal repulsion. Cell 109:887900

    38. Massey V, Palmer G (1966) On the existence of spectrally dis-

    tinct classes of flavoprotein semiquinones. A new method for the

    quantitative production of flavoprotein semiquinones. Biochem-

    istry 5:31813189

    39. Ventura A, Pelicci PG (2002) Semaphorins: green light for redox

    signaling? Sci STKE 2002: pe44

    40. Kamata H, Hirata H (1999) Redox regulation of cellular signal-

    ling. Cell Signal 11:114

    41. Finkel T (1998) Oxygen radicals and signaling. Curr Opin Cell

    Biol 10:248253

    42. Meng TC, Fukada T, Tonks NK (2002) Reversible oxidation and

    inactivation of protein tyrosine phosphatases in vivo. Mol Cell

    9:387399

    43. Juurlink BH, Paterson PG (1998) Review of oxidative stress in

    brain and spinal cord injury: suggestions for pharmacological and

    nutritional management strategies. J Spinal Cord Med 21:309334

    44. Lovell MA, Ehmann WD, Butler SM, Markesbery WR (1995)

    Elevated thiobarbituric acid-reactive substances and antioxidant

    enzyme activity in the brain in Alzheimers disease. Neurology

    45:15941601

    45. Schuessel K, Leutner S, Cairns NJ, Muller WE, Eckert A (2004)

    Impact of gender on upregulation of antioxidant defence mecha-

    nisms in Alzheimers disease brain. J Neural Transm 111:11671182

    46. Nunomura A, Chiba S, Lippa CF, Cras P, Kalaria RN, Takeda A,

    Honda K, Smith MA, Perry G (2004) Neuronal RNA oxidation is

    a prominent feature of familial Alzheimers disease. Neurobiol

    Dis 17:108113

    47. Hirai K, Aliev G, Nunomura A, Fujioka H, Russell RL, Atwood

    CS, Johnson AB, Kress Y, Vinters HV, Tabaton M, Shimohama

    S, Cash AD, Siedlak SL, Harris PL, Jones PK, Petersen RB, Perry

    G, Smith MA (2001) Mitochondrial abnormalities in Alzheimers

    disease. J Neurosci 21:30173023

    48. Behl C (2005) Oxidative stress in Alzheimers disease: implica-

    tions for prevention and therapy. Subcell Biochem 38:6578

    49. Reddy PH (2006) Amyloid precursor protein-mediated free rad-

    icals and oxidative damage: implications for the development and

    progression of Alzheimers disease. J Neurochem 96:113

    50. Premkumar DR, Smith MA, Richey PL, Petersen RB, Castellani

    R, Kutty RK, Wiggert B, Perry G, Kalaria RN (1995) Induction of

    heme oxygenase-1 mRNA and protein in neocortex and cerebral

    vessels in Alzheimers disease. J Neurochem 65:13991402

    51. Schipper HM, Cisse S, Stopa EG (1995) Expression of heme

    oxygenase-1 in the senescent and Alzheimer-diseased brain. Ann

    Neurol 37:758768

    52. Smith MA, Kutty RK, Richey PL, Yan SD, Stern D, Chader GJ,

    Wiggert B, Petersen RB, Perry G (1994) Heme oxygenase-1 is

    associated with the neurofibrillary pathology of Alzheimers

    disease. Am J Pathol 145:4247

    134 Mol Cell Biochem (2011) 350:127134

    123

    Characterizing the novel protein p33MONOXAbstractIntroductionMaterials and methodsReagentsCell culture and transfectionWestern blot analysesAnimal material, immunohistochemistry (IHC) and immunocytochemistry (ICC)ProQuesttrade two-hybrid-system with Gatewaytrade technology

    Resultsp33MONOX protein sequence analysisSub-cellular localization and neuronal expression of p33Monox in the mouse brainp33Monox-mediated neuronal signalingp33MONOX interacts with COBRA1, NOL12, and PRNP

    DiscussionAcknowledgmentsReferences

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages true /GrayImageMinResolution 149 /GrayImageMinResolutionPolicy /Warning /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 150 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages true /MonoImageMinResolution 599 /MonoImageMinResolutionPolicy /Warning /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 600 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (None) /PDFXOutputConditionIdentifier () /PDFXOutputCondition () /PDFXRegistryName () /PDFXTrapped /False

    /CreateJDFFile false /Description > /Namespace [ (Adobe) (Common) (1.0) ] /OtherNamespaces [ > /FormElements false /GenerateStructure false /IncludeBookmarks false /IncludeHyperlinks false /IncludeInteractive false /IncludeLayers false /IncludeProfiles false /MultimediaHandling /UseObjectSettings /Namespace [ (Adobe) (CreativeSuite) (2.0) ] /PDFXOutputIntentProfileSelector /DocumentCMYK /PreserveEditing true /UntaggedCMYKHandling /LeaveUntagged /UntaggedRGBHandling /UseDocumentProfile /UseDocumentBleed false >> ]>> setdistillerparams> setpagedevice