Next Article in Journal
New Milk Protein-Derived Peptides with Potential Antimicrobial Activity: An Approach Based on Bioinformatic Studies
Next Article in Special Issue
Farm Animal Serum Proteomics and Impact on Human Health
Previous Article in Journal
High-Resolution Imaging Reveals New Features of Nuclear Export of mRNA through the Nuclear Pore Complexes
Previous Article in Special Issue
Quantitative Proteomic Analysis of Serum Proteins from Oral Cancer Patients: Comparison of Two Analytical Methods
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Immunoproteome of Aspergillus fumigatus Using Sera of Patients with Invasive Aspergillosis

by
Emylli D. Virginio
1,
Paula H. Kubitschek-Barreira
1,
Marjorie Vieira Batista
2,3,
Marcelo R. Schirmer
4,
Eliana Abdelhay
4,
Maria A. Shikanai-Yasuda
2,5 and
Leila M. Lopes-Bezerra
1,*
1
Laboratory of Cellular Mycology and Proteomics, Biology Institute, University of Rio de Janeiro State (UERJ), Rio de Janeiro 20550-013, Brazil
2
Laboratory of Immunology (LIM 48), Clinics Hospital and Department of Infectious and Parasitic Diseases, Faculty of Medicine, University of São Paulo, São Paulo 05403-000, Brazil
3
Clinics of Infectious Diseases, Clinics Hospital, Faculty of Medicine, University of São Paulo, São Paulo 05403-900, Brazil
4
National Cancer Institute, Center for Bone Marrow Transplants, Rio de Janeiro 20230-130, Brazil
5
Department of Infectious and Parasitic Diseases, Faculty of Medicine, University of São Paulo, São Paulo 05403-000, Brazil
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2014, 15(8), 14505-14530; https://doi.org/10.3390/ijms150814505
Submission received: 13 May 2014 / Revised: 24 July 2014 / Accepted: 8 August 2014 / Published: 20 August 2014
(This article belongs to the Special Issue Advances in Proteomic Research)

Abstract

:
Invasive aspergillosis is a life-threatening lung or systemic infection caused by the opportunistic mold Aspergillus fumigatus. The disease affects mainly immunocompromised hosts, and patients with hematological malignances or who have been submitted to stem cell transplantation are at high risk. Despite the current use of Platelia Aspergillus as a diagnostic test, the early diagnosis of invasive aspergillosis remains a major challenge in improving the prognosis of the disease. In this study, we used an immunoproteomic approach to identify proteins that could be putative candidates for the early diagnosis of invasive aspergillosis. Antigenic proteins expressed in the first steps of A. fumigatus germination occurring in a human host were revealed using 2-D Western immunoblots with the serum of patients who had previously been classified as probable and proven for invasive aspergillosis. Forty antigenic proteins were identified using mass spectrometry (MS/MS). A BLAST analysis revealed that two of these proteins showed low homology with proteins of either the human host or etiological agents of other invasive fungal infections. To our knowledge, this is the first report describing specific antigenic proteins of A. fumigatus germlings that are recognized by sera of patients with confirmed invasive aspergillosis who were from two separate hospital units.

1. Introduction

Invasive aspergillosis is a life-threatening lung or systemic infection that primarily affects hematological patients under chemotherapy and hematopoietic stem cell transplant (HSCT) patients [1]. The infection is fatal in 30%–90% of the patients, including those given treatment [2]. The main etiological agent of invasive aspergillosis is the opportunistic mold Aspergillus fumigatus, which is responsible for 90% of aspergillus infections [3].
A confirmed diagnosis of invasive aspergillosis remains challenging and is frequently not achieved until necropsy. The isolation of aspergilli from cultures lacks sensitivity and, therefore, is ineffective for the diagnosis of invasive aspergillosis; blood cultures are rarely positive even in patients with confirmed invasive aspergillosis [4,5]. Moreover, the isolation of aspergilli in blood cultures or insputum samples does not necessarily indicate the presence of the invasive disease. Positive results usually represent only colonization due the high colonization rate in immunocompromised patients; thus, false-positive results due environmental contamination are frequent [5,6].
The “gold standard” for the diagnosis of invasive aspergillosis remains histopathological examination or biopsy; however, this often requires invasive procedures to obtain tissue for the examination. In most cases, the aggressiveness of the underlying disease, as well as the toxic effects of the hematological therapies, make this type of examination impossible in critically ill patients [3,7,8].
Currently, the routine techniques used for the diagnosis of invasive aspergillosis are computational tomography (CT) and the ELISA test for galactomannan (GM) (PlateliaAspergillus—BioRad, Hercules, CA, USA); these are considered along with microbiological findings and the clinical signs and symptoms of the patient [9,10]. The GM molecule is an immunodominant cell wall polysaccharide of Aspergillus and Penicillium species that is released during fungus growth [11,12]. Although it provides a fast serological result, the efficacy of the GM test remains controversial and varies depending on the clinic or health center, as previously reviewed by Xavier et al. [13]. False-positives have also been reported, for example, following treatment with a beta-lactam antibiotic; however, recent reports suggest that the new preparations of piperacillin–tazobactam do not test positive with galactomannan. Cross-reactions with fungi, such as Fusarium spp., Penicillium, Cladosporium and Histoplasma have also been reported [14,15,16,17]. The mean specificity of the test is 85% and the sensitivity varies from 29% to 100% [9,13].
The difficulties in reaching an early and precise diagnosis are also true for other invasive fungal infections. To define and classify the main invasive fungal infections in immunocompromised patients, the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group created and revised the definitions for clinical and epidemiological research. According to the definitions, invasive fungal infections are classified as “proven”, “probable”, or “possible” [5,18]. Thus, there remains an urgent need to develop new diagnostic tools to prevent the onset of the disease.
The sequencing of the A. fumigatus genome and the advances in the proteomic field have made it feasible to study and identify putative candidates for the immunodiagnosis of invasive aspergillosis. Few antigens specific for allergic bronchopulmonary aspergillosis (ABPA), aspergilloma, and invasive aspergillosis are known and/or being evaluated for diagnosis [19]. Furthermore, some studies have already shown the potential of some proteins as biomarkers for the immunodiagnosis of invasive aspergillosis; however, none of these came to a clinical trial [20,21,22,23,24].
In this context, the aim of this study was to investigate the antigenic proteins revealed by patients’ sera using cell wall extracts of A. fumigatus germlings in an attempt to find putative candidates for the diagnosis of invasive aspergillosis.

2. Results and Discussion

2.1. Western Immunoblots and Antigenic Proteins Identified

In recent decades, invasive fungal infections (IFI) have been considered the most important cause of morbidity and mortality in severely immunosuppressed patients. Although candidiasis remains the most frequent IFI in critically ill patients, aspergillosis and mucormycosis have also emerged as significant causes of morbidity and mortality. HSCT recipients and patients with prolonged neutropenia represent the main risk group for invasive aspergillosis [25]. In these patients, A. fumigatus is by far the most important etiological agent of invasive aspergillosis, especially in HSCT patients with acute leukemia (5% to 25%) and in some solid organ transplantation patients [3,7,26].
As mentioned previously, the actual diagnostic methods lack specificity and sensitivity for the early diagnosis of invasive aspergillosis. In this context, many efforts have been undertaken to identify new molecular tools that could reduce this difficulty. Immunoproteomic-based antigen identification is a convenient tool that is widely used to indicate putative candidates for the molecular diagnosis of fungal infections, including invasive aspergillosis [23,24,27,28]. Germlings are cells in an early stage of growth, and surface proteins in this morphotype may play an essential role in the fungal-host interaction [29,30]. In addition, the cell surface location of these proteins makes germlings more easily recognized by the host immune system [12]. Thus, proteins present in the A. fumigatus germling cell wall can represent important putative antigenic markers for the early diagnostic of invasive aspergillosis.
In this study, the antigenic profile of cell surface proteins of A. fumigatus germlings (GT6 h) were identified through an immunoproteomic approach. The 2-DE profile of the GT6 h extract, obtained as previously described [31], is shown in Figure 1. All antigenic proteins identified in this study, as well as their molecular mass, isoelectric points and functions, are listed in Table 1. The Western immunoblot analysis using the distinct pools of human sera, which were typed following the EORTC/MSG criteria as proven/Hospital 1; proven/Hospital 2 and probable, are shown in Figure 2A–C; the correspondent antigenic proteins recognized by each pool of sera are listed in Table 2.
As control, the GT6 h extract was probed with sera from patients with underlying diseases similar to those found in the invasive aspergillosis proven patients. It is important to note that these patients did not receive antifungical therapy, did not develop any fungal disease, and survived for at least one month (data not shown). The immunoblot performed with the control sera revealed positive spots that corresponded to ten antigenic proteins (Table 2). Some of these proteins had already been described as A. fumigatus antigens in other studies using the sera of immunized rabbits, mice, and patients with a clinical suspicion of allergic bronchopulmonary aspergillosis [20,21,23,28,32], suggesting that they could be putative biomarkers for aspergillosis. However, our data suggest that these antigens can cross-react with the control pool of sera, indicating that they are unspecific for diagnostic purposes.
An important feature on diagnostic tests is their discriminate capacity among pathologies that can be clinically similar. Some studies demonstrate that the diagnosis of invasive aspergillosis can be confused with a range of other invasive fungal infections, such as paracoccidioidomycosis, fusariosis and mucormycosis [29,30,31,32,33]. This scenario emphasizes the need for more selective diagnostic methods for the diagnosis of invasive fungal infections, including invasive aspergillosis. In this context, we also tested a pool of sera from patients with other invasive mycoses (Figure 2D) including histoplasmosis, fusariosis, cryptococcosis and paracoccidioidomycosis. Positive spots correspondent to twenty-two proteins were revealed with this pool of sera (other mycoses) (Table 2). This cross-reactivity observed suggest that these proteins lack specificity for diagnostic purposes of invasive aspergillosis; these were not considered for further analysis.
Figure 1. Proteomic profile 2-DE of TG6 h cell wall extract of A. fumigatus. Seventy-five mg of proteins were fractionated on pH 3–11 non-linear gradient 18-cm IPG strips followed by 12% homogenous 2-D SDS PAGE. Proteins were visualized via silver staining. The identified antigenic spots are indicated using Arabic numbers or by Letters. IEF = Isoelectric Focusing. IPG = Immobilized pH gradient. NL = Non linear.
Figure 1. Proteomic profile 2-DE of TG6 h cell wall extract of A. fumigatus. Seventy-five mg of proteins were fractionated on pH 3–11 non-linear gradient 18-cm IPG strips followed by 12% homogenous 2-D SDS PAGE. Proteins were visualized via silver staining. The identified antigenic spots are indicated using Arabic numbers or by Letters. IEF = Isoelectric Focusing. IPG = Immobilized pH gradient. NL = Non linear.
Ijms 15 14505 g001
Table 1. All antigenic proteins identified via mass spectrometry (MS/MS) in the cell surface extracts of A. fumigatus.
Table 1. All antigenic proteins identified via mass spectrometry (MS/MS) in the cell surface extracts of A. fumigatus.
SpotCov. (95%)Accession NumberEC NumberTheorical Mr/PIOrfOrganismNameFunctionPeptidesConf.Sequence
14.60Q4WC603.2.1.2158.148/4.93AFUA_8G05610A. fumigatusProbable β-glucosidase btgEDegradation of cellulosic biomass299EPGQFGVER
99VYSTDCNSLEYIGEAAR
85.02Q4X1G36.3.5.5129.214/5.87AFUA_2G10070A. fumigatusCarbamoyl-phosphate synthase, large subunitNitrogen compound metabolic process499FAESVGYPVLVR
99QIALLVGSTEDDVR
99AAESVGYPIIVR
99LADEVYYLPVTPEYVTHVIER
11, 12, 13, 14, 158.94Q4X0G7-93.198/6.51AFUA_2G13530A. fumigatusTranslation elongation factor EF-2 subunit, putativeTranslation elongation factor activity699GHVYSEEQRPGTPLFNVK
99ALGDVQVYPDR
99AYLPVNESFGFNGDLR
99DLEEDHAGVPLR
99VNFTIEEIR
99FSVSPVVQR
13, 155.45B0XND2-81.445/5.74AFUB_004530A. fumigatusPolyadenylate-binding proteinRNA-binding399NIDQEVTDEEFR
98NLTDDVDDEKLR
99SLGYAYVNYNNTADGER
249.66Q6MYM4-80.04/5.08AfA5C5.047A. fumigatusHeat shock protein Hsp88, putativeResponse to stress599FIAGPIVQR
99KNELESTIYELR
99LDLPGPEEKPR
99STPTLVGFGTR
99TLSFTLNQDEAIAR
14.16P40292-80.64/4.94AFUA_5G04170A. fumigatusHeat shock protein 90-Heat shock protein hsp1 (Asp f 12)Promotes maturation, structural maintenance and proper regulation of specific target proteins involved for instance in cell cycle control and signal transduction899ADLINNLGTIAR
99GVVDSEDLPLNLSR
99HFSVEGQLEFR
99IILHLKDEQTDYLNESR
99RAPFDLFETK
99TGQFGWSANMER
98LGIHEDAQNR
96DFELEETEEEKAER
267.83Q4WDH16.3.4.1386.42/5.31AFUA_6G04730A. fumigatusBifunctional purine biosynthetic protein Ade1, putativede novo IMP biosynthetic process, purine nucleobase biosynthetic process499EGEVVYQVGELKPR
99GLAHITGGGLVENVPR
99HNIPTAAYQNFYEYEPAR
99VIASTATASTLEEALR
35, 36, 378.77Q4WLN14.2.1.3 85.53/6.26 AFUA_6G12930A. fumigatusMitochondrial aconitate hydratase, putativeMitochondrial genome maintenance699FTGEYDAVPATAR
99HLGGLAIITR
99LQRPLTYAEK
99QHIGDFAR
99SLFTVTPGSEQIR
99WVVIGDWNYGEGSSR
46, 4712.66Q4WJ30-69.66/5.08AFUA_1G07440A. fumigatusMolecular chaperone Hsp70ATP and nucleotide binding; protein refolding699ATAGDTHLGGEDFDNR
99DAGLIAGLNVLR
99FELTGIPPAPR
99SSVHEIVLVGGSTR
99TTPSFVAFTDTER
96LVNHFVNEFKR
5911.37Q4WMB7-53.56/4.58AFUA_6G10470A. fumigatusZinc finger protein ZPR1Cellular response to starvation; regulation of mitotic cell cycle499DIILESFECEHCGHR
99FTTVEGLLTEIR
99GESQLTTVEGVIQR
99YTLDVENEEDFQR
61, 627.98Q4WCM2-66.97/5.30AFUA_8G03930A. fumigatusHsp70 chaperone (HscA), putativeProtein refolding; ATP-binding499AVITVPAYFNDNQR
99DAGAIAGLNVLR
99QQLESYISR
99SQVDEIVLVGGSTR
63, 64, 6514.01Q4X1H5-74.46/6.02AFUA_2G09960A. fumigatusMitochondrial Hsp70 chaperone (Ssc70), putativeProtein refolding; protein targeting to mitochondrion799DAGQIAGLNVLR
99IVQHTNGDAWVEAR
99LLGNFQLVGIPPAHR
99NAVVTVPAYFNDSQR
99SQLESLVEPLINR
99TTPSVVAFAQDGER
98YSPSQIGGFILQK
7110.47A1D3E6-46.64/4.69NFIA_016350A. fischerianusProtein phosphatase 2C, putative Protein dephosphorylation399ISAAGGFVDFGR
99NQFEETPDNYDLENDR
99VANGDGPCAPPEYAEFR
8.35Q4WU69-54.25/4.50AFUA_5G07390A. fumigatus60S ribosome biogenesis protein Sqt1, putative Structural constituent of ribosome399GEYVVTAGLDGR
99VEFLQTNLAALASR
96DERPVLPQSYESNPQPK
8.98Q4WTN7-48.34/4.43AFUA_5G05540A. fumigatusNucleosome assembly protein Nap1, putativeNucleosome assembly399EESLDHATAASLFAR
99SSGYIESLPAPVR
99MEYLDRPGFR
2.71Q4WH995.3.4.1 56.19/4.58AFUA_2G06150A. fumigatusProtein disulfide isomerase Pdi1, putative Cell redox homeostasis; glycerol ether metabolic process199AANDVFTSFAESQR
73, 747.11Q4WXF15.4.2.157.45/5.44AFUA_3G09290A. fumigatusPhosphoglycerate mutase, 2,3-bisphosphoglycerate-independentGlucose catabolic process399VQDNDTLFFFNYR
99EIGIGEIATVVGR
99EITQLLGDYDR
15.26Q4WGP12.3.1.1252.03/6.26AFUA_7G05720A. fumigatusPyruvate dehydrogenase complex, dihydrolipoamide acetyltransferase component, putativeAcetyl-CoA biosynthetic process from pyruvate599FTAVINPPQAAILAVGTTR
99LQPSLDREPNISPAAK
99NVHSLGLSSISNQIK
99VPAVNSSWR
99ENPHFFVSTTLSVTK
114, 11722.60Q96X304.2.1.1147.31/5.39AFUA_6G06770A. fumigatusEnolase (Asp f 22)Glycolysis;regulation of vacuole fusion, non-autophagic699AIVPSGASTGQHEAHELR
99DSYADNWGVMVSHR
99GNPTVEVDVVTETGLHR
99GVPLYAHISDLAGTK
99SGETEDVTIADIAVGLR
99TSDFQIVGDDLTVTNPGR
119, 12012.75Q4WS303.4.24.6453.27/5.90AFUA_1G14200A. fumigatusMitochondrial processing peptidase beta subunit, putativeMetalloendopeptidase activity599ASILLSLDGTTAVAEDIGR
99ITEKDVMDFANR
99LCYNVSAAEVER
99LNDLVHFALR
99TPEFIGSEIR
1204.70Q5AZS8-49.75/9.88AN6202.2A. nidulansRL3_NEUCR 60S ribosomal protein L3Structural constituent of ribosome199DEMIDVIAVTKGHGFQGVTSR
13125.66Q4WT692.7.2.344.76/6.31AFUA_1G10350A. fumigatusPhosphoglycerate kinasePhosphoglycerate kinase activity899ALESPSRPFLAILGGSK
99ASGGQVILLENLR
99FHPEEEGSYKDEEGK
99FHPEEEGSYKDEEGKK
99GLTALGDIYINDAFGTAHR
99IGNSLFDEAGSK
99IVLPVDYITADKFSADAK
99YSLKPVVPELEK
13214.52Q4WDF5-54.18/7.18AFUA_6G04570A. fumigatusTranslation elongation factor eEF-1 subunit gamma, putative Translation elongation factor activity799AVVPSPVFAEEAIK
99EYPHVDGHVFK
99HLTANTYLVGER
99ITLADYFGASLLTR
99TKQDYAAILR
98QDYAAILR
97LYGLPENGR
1415.65Q4WEU3 1.10.2.248.09/8.89AFUA_5G04210A. fumigatusUbiquinol-cytochrome C reductase complex core protein 2, putativeUbiquinolcytochrome-C reductase activity299ATQGFSQVR SNIAIVGSGSSTAEVSR
1782.99B0XM32-56.40/6.84AFUB_000800A. fumigatusCytochrome P450Oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen199LLSDQFAGFPSVNSR
176, 18022.15Q4WQK8-34.99/6.06AFUA_4G13170A. fumigatusG-protein comlpex beta subunit CpcBCell signaling599VDELKPEFIEK
99HLYSLHAGDEIHALVFSPNR
99LWELATGETTR
99TFVGHTSDVLSVSFSADNR
99TLIIWNLTR
A3.29Q4WGN63.6.3.-117.77/5.84AFUA_7G05660A. fumigatusTranslation elongation factor eEF-3Translation elongation factor activity399FLDNVIQHVVHYER
99TFEGGVVIITHSR
97LEEFGFLR
B5.87Q4WX09-71.15/6.50AFUA_3G07810A. fumigatusSuccinate dehydrogenase subunit Sdh1, putativeEectron transport chain; tricarboxylic acid cycle399AHHTVLATGGYGR
99KPHGEINLGYR
99GIIAYNQEDGTLHR
C5.45Q4X1P0-61.95/5.53AFUA_2G09290A. fumigatusAntigenic mitochondrial protein HSP60, putativeCellular response to temperature stimulus; protein refolding299AITLQDKFENLGAR
99ISAVQDIIPALEASTTLR
D20.81Q4WV253.6.3.1455.62/5.30AFUA_5G10550A. fumigatusATP synthase subunit betaATP catabolic process999DTGAPIKIPVGPGTLGR
99FTQAGSEVSALLGR
99IPVGPGTLGR
99IVGEEHYAVATR
99IVNVTGDPIDER
99LVLEVSQHLGENVVR
99VALTGLTIAEYFR
99VALVFGQMNEPPGAR
99VVDLLAPYAR
E23.15Q4WX433.6.4.1345.78/5.05AFUA_3G08160A. fumigatusATP-dependent RNA helicase eIF4A Complex eIF4F subunit-involved in the “cap” recognition; necessary to mRNA binding to ribossome799ALQEGPQVVVGTPGR
99DFTVSAMHGDMEQAQR
99GCQALILAPTR
99GVAINFVTADDVR
99GVYAYGFERPSAIQQR
99MFILDEADEMLSR
99VLIATDLLAR
F14.41Q4WNQ8-49.37/5.79AFUA_4G06620A. fumigatusGlutamate dehydrogenase -Glu/Leu/Phe/Val dehydrogenaseOxidoreductase activity599AANAGGVAVSGLEMAQNSAR
99FLGFEQIFK
99VVWEDDNHQVQINR
99YIEGARPWVHVGK
99EIGFLFGQYR
G18.06Q4WY39 4.1.2.1339.79/5.55AFUA_3G11690A. fumigatusFructose-bisphosphate aldolase, class IIFructose-bisphosphate aldolase activity; zinc ion binding699ASIAGSIAAAHYIR
99KSGVIVGDDVLR
99LFEYAQEK
99RVQVALEDFNTAGQL
99SGVIVGDDVLR
99VNLDTDMQYAYMSGVR
I6.37Q4WQ26-42.35/5.68AFUA_4G11330A. fumigatusAha1 domain familyATPase activator activity-Response to stress399QNWDVYYVR
VAVNTTTVTASDEFR
QNWDVYYVR
18.71Q4WQK36.3.1.239.90/5.48AFUA_4G13120A. fumigatusGlutamine synthetaseGlutamate-ammonia ligase activity-Glutamine biosynthetic process599DIVEAHYR
FSYGVADR
GDWNGAGLHTNVSTAATR
GGFPGAQGPYYCGVGTGK
HNEHIAVYGEGNEER
K23.86Q8TGG6-48.29/6.69AfA14E5.05A. fumigatusElongation factor TuTranslation elongation factor activity-Protein biosynthesis799AGDNSGLLLR
GITISTAHIEFSTDSR
GLANFLEYGAIDKAPEER
HYAHVDCPGHADYIK
TADEAADLSFPDGDQSR
THHPVAAEAGQR
TKPHVNIGTIGHVDHGK
L4.44Q4WJ751.2.4.141.48/6.36AFUA_1G06960A. fumigatusPyruvate dehydrogenase E1 component subunit alphaPyruvate dehydrogenase (acetyl-transferring) activity-Glycolytic process299ILFEDIYVR
SIIGELLGR
N14.76Q4WEU5-52.11/8.69AFUA_5G04230A. fumigatusCitrate synthaseCitrate (Si)-synthase activity - Tricarboxylic acid cycle/Cellular carbohydrate metabolic process499CLVWEGSVLDSEEGIR
FIEELIDR
ALGAPIERPK
ALGVLPQLIIDR
DLSAEWAAR
FIEELIDR
VIGEVTLDQAYGGAR
O5.02Q4WWD53.-.-.-53.02/5.47AFUA_3G05450A. fumigatusGlutamate carboxypeptidase, putativeCarboxypeptidase, Hydrolase Protease299EHLDLPPVVIAR
QVDELSNSFIDR
P8.85Q4WYW41.1.1.8656.35/9.32AFUA_3G14490A. fumigatusKetol-acid reductoisomeraseKetol-acid reductoisomerase activity-branched-chain amino acid biosynthetic process399DQGLNVIVGVR
EVYSDLYGER
TLYFSHGFSPVFK
Cov. = Coverage; EC number = Enzyme Commission number; Mr = Molecular weight range in kDa; PI = Isoelectric point; Conf. = Confidence.
Figure 2. 2-D Western immunoblot of proteins extracts of A. fumigatus germlings from the pool of patient’s sera classified as (A) proven/hospital 1; (B) probable; (C) proven/hospital 2; or (D) other-mycosis. The identified antigenic proteins are indicated with the accession number (UniProtKB).
Figure 2. 2-D Western immunoblot of proteins extracts of A. fumigatus germlings from the pool of patient’s sera classified as (A) proven/hospital 1; (B) probable; (C) proven/hospital 2; or (D) other-mycosis. The identified antigenic proteins are indicated with the accession number (UniProtKB).
Ijms 15 14505 g002
Table 2. Antigenic proteins revealed using the different pools of sera.
Table 2. Antigenic proteins revealed using the different pools of sera.
SpotNameProven-Hospital 1Proven-Hospital 2ProbableOther MycosesControl
1Probable β-glucosidase btgEXXXXX
8Carbamoyl-phosphate synthase, large subunitX
11, 12, 13, 14, 15Translation elongation factor EF-2 subunit, putativeX
13, 15Polyadenylate-binding proteinX
24Heat shock protein Hsp88, putativeXXXX
Heat shock protein 90–Heat shock protein hsp1 (Asp f 12)XXX
26Bifunctional purine biosynthetic protein Ade1, putativeX
35, 36, 37Mitochondrial aconitate hydratase, putativeX
46, 47Molecular chaperone Hsp70XXXX
59Zinc finger protein ZPR1XXX
61, 62Hsp70 chaperone (HscA), putativeXXX
63, 64, 65Mitochondrial Hsp70 chaperone (Ssc70), putativeXXX
71Protein phosphatase 2C, putativeXXXX
60S ribosome biogenesis protein Sqt1, putativeXXXX
Nucleosome assembly protein Nap1, putativeXXXX
Protein disulfide isomerase Pdi1, putativeXXXX
73, 74Phosphoglycerate mutase, 2,3-bisphosphoglycerate-independentX
Pyruvate dehydrogenase complex, dihydrolipoamide acetyltransferase component, putativeX
114, 117Enolase (Asp f 22)XXX
119, 120Mitochondrial processing peptidase β subunit, putativeXXX
120RL3_NEUCR 60S ribosomal protein L3XXX
131Phosphoglycerate kinaseXXX
132,133Translation elongation factor eEF-1 subunit γ, putativeXXX
140, 141Ubiquinol-cytochrome C reductase complex core protein 2, putativeX
178Cytochrome P450X
176, 180G-protein comlpex beta subunit CpcBX
ATranslation elongation factor eEF-3XX
BSuccinate dehydrogenase subunit Sdh1, putativeX
CAntigenic mitochondrial protein HSP60, putativeX
DATP synthase subunit βX
EATP-dependent RNA helicase eIF4AX
FGlutamate dehydrogenase—Glu/Leu/Phe/Val dehydrogenaseXX
GFructose-bisphosphate aldolase, class IIXX
IAha1 domain familyX
IGlutamine synthetaseX
KElongation factor TuX
LPyruvate dehydrogenase E1 component subunit αX
NCitrate synthaseX
OGlutamate carboxypeptidase, putativeX
PKetol-acid reductoisomeraseX
A total of fourteen antigenic proteins were exclusively revealed by sera of patients with proven aspergillosis, as shown in Table 2 (grey lines). Among these, four proteins were also recognized by pool of sera classified as probable by the EORTC/MSG criteria. Five out of fourteen proteins were positively recognized by the pool of patients with proven aspergillosis from both Hospital 1 and 2. Some of these identified antigens had also been described in other reports based on assays with the sera of immunized rabbits, mice and of patients with the clinical suspicion of allergic bronchopulmonary aspergillosis [20,21,23,28,32]. To our knowledge, this work is the first to describe four antigens: eEF-3, eIF4A, cytochrome P450 and Ade1, which are putative candidates for diagnostic utility.

2.2. BLAST Analysis

The fourteen antigens revealed from the immunoproteome of the sera from patients with proven invasive aspergillosis (n = 12) were selected as putative candidates for the diagnosis of invasive aspergillosis. Their protein sequences were compared with human proteins via BLAST analyses to ensure their potential specificity for A. fumigatus and cross-reactivity with human proteins. Our results showed that only two antigenic proteins, cytochrome P450 and eEF-3, had no homology with human proteins.
As mentioned previously, the diagnosis of invasive aspergillosis can be confused with a range of other invasive fungal infections [33,34,35,36,37]. In this context, we also compared (via BLAST analysis) the sequences of the two above-described proteins with proteins of Rizophus spp. and other fungi from the Mucorales order; Penicillium spp., Paracoccidioides brasiliensis, Fusarium spp., and Paecilomyces spp., as described in the methodology section. The results shown in Table 3 indicate that both cytochrome P450 and eEF-3 can be putative markers for the selective diagnosis of A. fumigatus infections.
Table 3. BLAST analysis of the two main antigens identified against the protein sequences of etiological agents of other invasive fungal infections.
Table 3. BLAST analysis of the two main antigens identified against the protein sequences of etiological agents of other invasive fungal infections.
MicroorganismsParametersCytochrome P450eEF-3
MucoralesScore56.223.9
E-value8 × 10−112.2
Identity25%26%
Protein homology (organism)Cytochrome P450 51 (Cunninghamella elegans)Glyceraldehyde-3-phosphate dehydrogenase (Rhizomucor miehei)
PenicilliumScore45.127.3
E-value6 × 10−70.57
Identity23%56%
Protein homology (organism)Eburicol 14-α-demethylase (Penicillium chrysogenum)Peroxisomal biogenesis factor 6 (Penicillium chrysogenum)
P. brasiliensisScore48.126.6
E-value0.640.51
Identity56%33%
Protein homology (organism)Translation factor GUF1Probable Xaa-Pro aminopeptidase PADG
RhizopusScore19.643.5
E-value8.82.7
Identity67%26%
Protein homology (organism)Rhizopuspepsin-2 (Rhizopus niveus)Peptidyl-prolyl cis–trans isomerase cyp11 (Rhizopus delemar)
FusariumScore211112
E-value5 × 10−636 × 10−6
Identity32%35%
Protein homology (organism)Cytochrome P450 503A1 (Fusarium proliferatum)Iron-sulfur clusters transporter ATM1 (Fusarium graminearum)
PaecilomycesScore--
E-value--
Identity--
Protein homology (organism)No matchNo match
The cytochrome P450 superfamily is made up of monooxygenases that play key roles in a range of biochemical processes from catalysis to xenobiotic detox and degradation; cytochrome P450 is found in every living form [38]. In general, cytochrome P450 isoforms have being described as essential for the membrane ergosterol biosynthesis, and some isoforms are involved in the production of aflatoxin in A. parasiticus [39,40,41]. In A. fumigatus, triazole resistance is often related to mutations in a gene that encodes a cytochrome P450 isoform, the cyp51 gene [42,43,44]. Although the secondary structures of the proteins of the cytochrome P450 superfamily are well conserved, there is a low homology among the primary amino acid sequences of different species [45,46,47,48]. These data are consistent with the result of our BLAST analysis that shows the low homology of the identified A. fumigatus cytochrome P450 found in this study with proteins of other fungi (Table 3). The cytochrome P450 identified in this study is predicted in the A. fumigatus genome but has no characterized function. To our knowledge, this is the first report showing the antigenic diagnostic potential of an A. fumigatus cytochrome P450.
The most promising antigen was the translation elongation factor eEF-3. This protein showed the lowest sequence homology in the BLAST analysis (Table 3). The translation process functions in a series highly regulated steps that are catalyzed by the eukaryotic initiation factors [49]. In general, the process is highly conserved from bacteria to mammals: the eEF-1 is incumbent on delivering the aminoacyl-tRNA to the ribosomal A-site [50], and the eEF-2 has a translocase activity [51]. However, another factor is required in fungi (an ATPase factor, namely eEF3). This requirement is unique in fungi ribosomes. This fungal-specific protein is absent in mammalian cells and has already being described by our group as a putative drug target in A. fumigatus [27]. The eEF-3 is an ATPase of the ATP binding cassette (ABC) family member [52]. The majority of this superfamily’s members are integral membrane transporters that are involved in the import or export of diverse substrates across lipid bilayers [53]. However, eEF-3 lacks the transmembrane domain because it is a soluble factor with two ABC domains arranged in tandem. One of these domains carries a unique chromodomain-like insertion that is hypothesized to play a significant role in its binding to the ribosome [54]. A recent study showed that mutations in the chromodomain-like insertion of eEF-3 resulted in reduced growth rate and slower translation elongation. These mutations also compromised the ribosome-stimulated ATPase activity of eEF3, strongly suggesting that it exerts an allosteric effect on the hydrolytic activity of eEF3 [55]. These features contributed to the overexpression of eEF-3 in the first steps of A. fumigatus filamentation (germlings), strengthening the hypothesis that this protein may be a good drug target [31].
Our previous studies showed that this protein was found to be overexpressed up to eight-fold on the surface of the germlings compared with mature A. fumigatus hyphae [31]. In this study, the eEF-3 factor was identified as an antigenic protein of A. fumigatus recognized by the sera of patients with proven invasive aspergillosis. Taken together, these observations strongly suggest that in addition to being a putative drug target, the identified A. fumigatus eEF-3 factor can also be a promising candidate for the diagnosis of invasive aspergillosis.

3. Experimental Section

3.1. Fungal Strain and Culture Conditions

The A. fumigatus strain used in this study was AF293, which was originally isolated at autopsy from a patient with IPA and kindly provided by Dr. Scott Filler of Harbor-UCLA Medical Center, University of California, CA, USA.
A. fumigatus was first grown in Sabouraud Agar (Difco, Detroit, MI, USA) roux flask for 7 days at 37 °C. The conidia were than harvested using a cell scraper in the presence of PBS-Tween 20 (0.01%). This suspension was vacuum-filtered using a Büchner filler with a nylon membrane (Sefar Nitex 03-28/17, 7, Sefar Inc., Heiden, Switzerland) to remove hyphae fragments. A ratio of 107 conidia/mL was then incubated in Sabouraud Broth (Difco, Maryland, MD, USA) in a 500-mL flask on a shaker at 37 °C and 150 rpm for 6 h to obtain the conidia germlings.

3.2. Preparation of Germiling Conidia Protein Extract (GTM6 h)

Conidia germling cells were submitted to chemical extraction [56] using protein extraction buffer containing Tris–HCl 25 mM, DTT 2 mM, PMSF 1 mM and EDTA 5 mM, pH 8.5. The conidia germling cells were incubated with the protein extraction buffer in a ratio of 0.7 g of cells (wet weight) per 5 mL of buffer for 2 h at 4 °C under gentle agitation. The proteins extracted using this process were separated via centrifugation. The extract was precipitated with trichloroacetic acid/acetone [57] and re-suspended in rehydration buffer containing 7 M urea, 2 M thiourea and CHAPS 4%. The protein concentration was determined using the Bradford method (Bio-Rad, Hercules, CA, USA) according to the manufacturer’s recommendations. The absence of membrane leakage and consequently intracellular proteins or material derived from dead cells, in this type of extraction have been previously described [27].

3.3. Patients and Control Subjects

All of the serum samples of patients were obtained with informed patient consent and the permission of the local human ethics committee. All serum samples were classified according to the EORTC/MSG criteria [18]. Three serum samples of patients clinically diagnosed as proven and thirteen serum samples of patients clinically diagnosed as probable were obtained from the Bone Marrow Transplant Center of the National Institute of Cancer (INCA-Brazil), henceforth referred to as Hospital 1. More information about the characteristics of the patients from Hospital 1 is shown in Table 4. Nine serum samples of patients classified as “proven” for invasive aspergillosis were obtained from the Hospital das Clínicas of the Faculty of Medicine from the University of São Paulo (USP-Brazil), henceforth referred to as Hospital 2. Serum samples from patients with other fungal infections viz. histoplasmosis (n = 1), fusariosis (n = 3), cryptococcosis (n = 1) and paracoccidioidomycosis (n = 1) were also provided by Hospital 2. These patients had also underlying diseases similar to those found in the aspergillosis cases. As a negative control, sera from six patients with underlying diseases similar to the aspergillosis cases, such as acute myeloid leukemia (n = 2), non-Hodgkin lymphoma (n = 2), multiple myeloma (n = 1) and myelodysplastic syndrome (n = 1), were also provided by Hospital 2. These patients did not receive antifungal treatment, presented no colonization by any fungal species and survived for at least 30 days. More information about the characteristics of the patients from Hospital 2 is shown in Table 5. The serum samples were pooled for the immunoproteome assays as follows: proven/hospital 1, proven/hospital 2, probable or other-mycosis.
Table 4. Additional information about patients from Hospital 1.
Table 4. Additional information about patients from Hospital 1.
Patient Hospital 1GenderAgeUnderlying DiseaseHistopathologyEORTC/MSG Classification
1M10ALL/HSCT-Probable
2F5MDS/HSCT-Probable
3F39MDS/HSCT-Probable
4F22HL/HSCT-Probable
5M16ALL/HSCT-Probable
6M34HL/HSCT-Probable
7M15ALL/HSCT-Probable
8F53CML/HSCT-Probable
9M20ALL/HSCT-Probable
10M53AA/HSCT-Probable
11M50AML/HSCT-Probable
12M9ALL/HSCT-Probable
13F7ALL/HSCT-Probable
14F29NHL/HSCTA, fumigates (lung biopsy)Proven
15F11AML/HSCTA, fumigates (lung biopsy)Proven
16F28AML/HSCTA, flavus (lung biopsy)Proven
EORTC/MSG = European Organization for Research and Treatment of Cancer (EORTC), Mycoses Study Group (MSG); ALL = Acute Lymphoblastic Leukemia; HSCT = Hematopoietic Stem Cell Transplantation; MDS = Myelodysplastic Syndrome; HL = Hodgkin Lymphoma; CML = Chronic Myeloid Leukemia; AA = Aplastic Anemia; AML = Acute Myeloid Leukemia; NHL = non-Hodgkin Lymphoma.
Table 5. Additional information about patients from Hospital 2.
Table 5. Additional information about patients from Hospital 2.
Patient Hospital 2GenderAgeUnderlying DiseaseHistopathologyEORTC/MSG Classification
1F19AMLAspergillus sp. (necropsy)Proven
2F28AML/HSCTAspergillus sp. (necropsy)Proven
3F50NHL/HSCTAspergillus sp. (lung biopsy)Proven
4F58ALLAspergillus sp. (necropsy)Proven
5M26ALLAspergillus sp. (laryngeal biopsy)Proven
6M58Lymphoma/HSCTAspergillus sp. (sinus biopsy)Proven
7M39AMLAspergillus sp. (sinus biopsy)Proven
8M59NHLAspergillus sp. (lung biopsy)Proven
9F9Fulminant hepatitis/SOTAspergillus sp. (lung biopsy and necropsy)Proven
10M35AA/HSCTFusarium sp. (blood culture and skin biopsy)Proven
11M17AA/HSCTFusarium sp. (blood culture and skin biopsy)Proven
12M51NHL/HSCTFusarium sp. (blood culture)Proven
13F49NoHistoplasma sp. (lymph node biopsy and immuno-histochemistry)Proven
14M41NoParacoccidioides sp. (tracheal secretion culture and direct mycroscopy of palatum)Proven
15F18SELCryptococcus neoformans var. gattii (bronchoalveolar lavage culture)Proven
AML = Acute Myeloid Leukemia; HSCT = Hematopoietic Stem Cell Transplantation; NHL = non-Hodgkin Lymphoma; ALL = Acute Lymphoblastic Leukemia; AA = Aplastic Anemia; SEL = Systemic lupus erythematosus.

3.4. 2-D SDS PAGE

The focusing was performed using 75 or 400 μg of GT6h protein and IPG strips (Immobiline DryStrip 3–11 NL, 18 cm) with the addition of 1.2% DeStreak and 1% IPG buffer 3–11 (GE Healthcare, Piscataway, NJ, USA). Immobilized pH-gradient strips were reduced (1.5% w/v dithioerythritol) and alkylated (2.5% w/v iodocetamide) in equilibration buffer (6 M urea, 50 mM Tris–HCl, pH 6.8, 30% glycerol, 2% SDS). Equilibrated strips were run on homogeneous 12% polyacrylamide gels using a Protean II XL cell electrophoresis system (Bio-Rad, Hercules, CA, USA). The analytic gels were stained with silver [58], and preparative gels were stained using colloidal Coomassie [59] for protein identification.

3.5. Western Immunoblot

For the immunoblottings, the resolved proteins were transferred to nitrocellulose membranes using a Trans-Blot Cell system (Bio-Rad). The transblotted proteins on the membrane were checked with Ponceau, and each membrane was blocked with 5% skim milk solution in 50 mM Tris and 150 mM NaCl containing 0.1% of Tween-20 (TBS-T). Then, the membranes were washed with 1% skim milk solution in TBS-T and incubated separately with each primary antibody (pools of sera: proven/hospital 1, proven/hospital 2, probable, other-mycosis, control) diluted in TBS-T at a 1:500 ratio for two hours at 4 °C under gentle agitation. The membranes were washed with 1% fat free milk solution in TBS-T (as above) and incubated with the secondary antibody (anti-human IgG peroxidase conjugated) (Sigma Co., St Louis, MO, USA) diluted in TBS-T at a 1:1000 ratio for two hours at 4 °C under gentle agitation. After washing with TBS, the membranes were incubated with the ECL Prime Western Blotting Detection Reagent (GE Healthcare, Menlo Park, CA, USA) according to the manufacturer’s recommendations, and the antigenic spots were visualized using a Molecular Imaging ChemiDoc XRS system (Bio-Rad, Hercules, CA, USA).

3.6. Protein Identification

Spots of interest were manually excised from the preparative 2-DE gels. These spots were destained, shrunk, vacuum-dried, as described elsewhere [27] and then, were incubated with 12.5 ng/μL sequencing grade trypsin (Promega, Madison, WI, USA) overnight at 37 °C. After digestion, the supernatants were separated and the peptides were extracted twice into 0.5% trifluoroacetic acid/50% acetonitrile and once into 100% acetonitrile. These extracts were pooled, and their volumes were vacuum-dried. The derived concentrated peptide suspension for each spot of interest was spotted on a MALDI target plate, mixed with a saturated solution of matrix α-cyano-4-hydroxytrans-cinnamic acid (Sigma Co., St Louis, MO, USA) and allowed to air-dry at room temperature. The samples were analyzed with a 5800 AB-SCIEX MALDI-TOF/TOF mass spectrometer (Applied Biosystems, Foster City, CA, USA) in automated mode. A MALDI MS spectrum was acquired from each spot (800 shots/spectrum), and 10 precursor peaks with a signal-to-noise ratio greater than 40 in at least two consecutive fractions were automatically selected for MS/MS analysis (4000 shots/spectrum). A collision energy of 1 keV was used with air as the collision gas. All mass spectra were externally calibrated using the mass standards kit for the 4700 proteomics analyzer (Applied Biosystems, Foster City, CA, USA). The spectra were searched against an in-house database constructed using “A. fumigatus” as the selection criteria in Protein Pilot software using the Paragon algorithm (Applied Biosystems, Foster City, CA, USA). The name of the ORF (open reading frame) from A. fumigatus was found in the UniProt (Universal Protein Resource) server using the UniProt Knowledge/Swiss-Prot database.

3.7. Homology Analysis

The sequences of the antigenic proteins were aligned and compared using the protein BLAST tool of the NCBI database (http://blast.ncbi.nlm.nih.gov). The sequences of the identified A. fumigatus proteins were compared with sequences of human proteins and with proteins from other microorganisms. The selected microorganisms for comparison in the BLAST analyses are the etiological agents of mycosis that can be confused (diagnostically) with invasive aspergillosis (Rizophus spp. and other fungi of the Mucorales order, Penicillium spp., Paracoccidioides brasilienisis, Fusarium spp. and Paecilomyces spp.). The proteins with identity values lower than 40% and E-values higher than 1 × 10−50 were identified to have no homology.

4. Conclusions

Two antigenic proteins of A. fumigatus are described in this work as putative candidates for the immunodiagnostic of invasive aspergillosis: cytochrome P450 and eEF-3. These proteins presented no homology with human proteins and low homology with etiological agents of other IFIs. Among these, the elongation factor eEF-3 identified in A. fumigatus germlings is the most promising candidate once it shows the lowest homology with proteins of other fungal species that cause infections, which could be misdiagnosed with invasive aspergillosis.

Acknowledgments

This work was supported by Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq)—grant 306582/2011-2, Fundação Carlos Chagas de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ)—grant E-26/102.819/2012 and Fundação de Amparo à Pesquisa do Estado de São Paulo(FAPESP)—grant 2012/50212-1. LMLB is a research fellow of CNPq and Faperj.
We would like to thank Aline Penna-de-Carvalho, technician of the Anatomy Department from Instituto de Biologia Roberto Alcântara Gomes for the help using the Molecular Imaging ChemiDoc XRS system (Bio-Rad) and Constancia Lima Diogo for the preparation of the bank of serum at Hospital 2.

Author Contributions

Emylli Dias Virginio had performed all experiments and data analysis.
Paula H. Kubitschek-Barreira contributed with the MS/MS data acquisition and protein identification analysis.
Marcelo R. Schirmer and Eliana Abdelhay contributed with all serum samples from Hospital 1 (proven and probable aspergillosis patients) and, are responsible for the clinical data of these groups of patients.
Maria Aparecida Shikanai-Yasuda and Marjorie V. Batista had collected and classified all serum samples from Hospital 2 (proven aspergillosis patients, patients with other mycoses and negative control group) and, are responsible for the clinical data of these groups of patients.
Leila M. Lopes-Bezerra is the intellectual mentor of this work and therefore, had contributed with the experimental design and revision of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Patterson, T.F. Risk stratification for invasive aspergillosis: Early assessment of host susceptibility. Med. Mycol. 2009, 47, S255–S260. [Google Scholar] [CrossRef]
  2. Steinbach, W.J.; Stevens, D.A.; Denning, D.W.; Moss, R.B. Advances against aspergillosis. Clin. Infect. Dis. 2003, 37, S155–S156. [Google Scholar] [CrossRef]
  3. Denning, D.W. Invasive aspergillosis. Clin. Infect Dis. 1998, 26, 781–805. [Google Scholar]
  4. Duthie, R.; Denning, D.W. Aspergillus fungemia: Report of two cases and review. Clin. Infect. Dis. 1995, 20, 598–605. [Google Scholar] [CrossRef]
  5. Ascioglu, S.; Rex, J.H.; de Pauw, B.; Bennett, J.E.; Bille, J.; Crokaert, F., Denning; Donnelly, J.P.; Edwards, J.E.; Erjavec, Z.; et al. Invasive fungal infections cooperative group of the European Organization for research and treatment of cancer; mycoses study group of the National Institute of allergy and infectious diseases. Defining opportunistic invasive fungal infections in immunocompromised patients with cancer and hematopoietic stem cell transplants: An international consensus. Clin. Infect. Dis. 2002, 34, 7–14. [Google Scholar] [CrossRef]
  6. Soubani, A.O.; Khanchandani, G.; Ahmed, H.P. Clinical significance of lower respiratory tract Aspergillus culture in elderly hospitalized patients. Eur. J. Clin. Microbiol. Infect. Dis. 2004, 23, 491–494. [Google Scholar] [CrossRef]
  7. Latgé, J.P. Aspergillus fumigatus and Aspergillosis. Clin. Microbiol. Rev. 1999, 12, 310–350. [Google Scholar]
  8. Thornton, C.R. Detection of invasive Aspergillosis. Adv. Appl. Microbiol. 2010, 70, 187–216. [Google Scholar] [CrossRef]
  9. Mennink-Kersten, M.A.; Donnelly, J.P.; Verweij, P.E. Detection of circulating galactomannan for the diagnosis and management of invasive aspergillosis. Lancet Infect. Dis. 2004, 4, 349–357. [Google Scholar] [CrossRef]
  10. Chamilos, G.; Kontoyiannis, D.P. Defining the diagnosis of invasive aspergillosis. Med. Mycol. 2006, 44, 163–172. [Google Scholar] [CrossRef]
  11. Beauvais, A.; Schmidt, C.; Guadagnini, S.; Roux, P.; Perret, E.; Henry, C.; Paris, S.; Mallet, A.; Prévost, M.C.; Latgé, J.P. An extracellular matrix glues together the aerial-grown hyphae of Aspergillus fumigatus. Cell Microbiol. 2007, 9, 1588–1600. [Google Scholar] [CrossRef]
  12. Fontaine, T.; Simenel, C.; Dubreucq, G.; Adam, O.; Delepierre, M. Molecular organization of the alkali-insoluble fraction of Aspergillus fumigatus cell wall. J. Biol. Chem. 2000, 275, 27594–27607. [Google Scholar]
  13. Xavier, M.O.; Pasqualotto, A.C.; Cardoso, I.C.; Severo, L.C. Cross-reactivity of Paracoccidioides brasiliensis, Histoplasma capsulatum, and Cryptococcus species in the commercial Platelia Aspergillus enzyme immunoassay. Clin. Vaccine Immunol. 2009, 16, 132–133. [Google Scholar] [CrossRef]
  14. Adam, O.; Aupérin, A.; Wilquin, F.; Bourhis, J.H.; Gachot, B.; Chachaty, E. Treatment with piperacillin-tazobactam and false-positive Aspergillus galactomannan antigen test results for patients with hematological malignancies. Clin. Infect. Dis. 2004, 38, 917–920. [Google Scholar] [CrossRef]
  15. Mikulska, M.; Furfaro, E.; del Bono, V.; Raiola, A.M.; Ratto, S.; Bacigalupo, A.; Viscoli, C. Piperacillin/tazobactam (Tazocin™) seems to be no longer responsible for false-positive results of the galactomannan assay. J. Antimicrob. Chemother. 2012, 67, 1746–1748. [Google Scholar] [CrossRef]
  16. Tortorano, A.M.; Esposto, M.C.; Prigitano, A.; Grancini, A.; Ossi, C.; Cavanna, C.; Cascio, G.L. Cross-reactivity of Fusarium spp. in the Aspergillus Galactomannan enzyme-linked immunosorbent assay. J. Clin. Microbiol. 2012, 50, 1051–1053. [Google Scholar] [CrossRef]
  17. Wheat, L.J.; Hackett, E.; Durkin, M.; Connolly, P.; Petraitiene, R.; Walsh, T.J.; Knox, K.; Hage, C. Histoplasmosis-associated cross-reactivity in the BioRad Platelia Aspergillus enzyme immunoassay. Clin. Vaccine Immunol. 2007, 14, 638–640. [Google Scholar] [CrossRef]
  18. De Pauw, B.; Walsh, T.J.; Donnelly, J.P.; Stevens, D.A.; Edwards, J.E.; Calandra, T.; Pappas, P.G.; Maertens, J.; Lortholary, O.; Kauffman, C.A.; et al. Revised definitions of invasive fungal disease from the European Organization for research and treatment of cancer/invasive fungal infections cooperative group and the National Institute of allergy and infectious diseases mycoses study group (EORTC/MSG) consensus group. Clin. Infect. Dis. 2008, 46, 1813–1821. [Google Scholar] [CrossRef]
  19. Kniemeyer, O.; Lessing, F.; Brakhage, A.A. Proteome analysis for pathogenicity and new diagnostic markers for Aspergillus fumigatus. Med. Mycol. 2009, 47, S248–S254. [Google Scholar] [CrossRef]
  20. Singh, B.; Oellerich, M.; Kumar, R.; Kumar, M.; Bhadoria, D.P.; Reichard, U.; Gupta, V.K.; Sharma, G.L.; Asif, A.R. Immuno-reactive molecules identified from the secreted proteome of Aspergillus fumigatus. J. Proteome Res. 2010, 9, 5517–5529. [Google Scholar] [CrossRef]
  21. Singh, B.; Sharma, G.L.; Oellerich, M.; Kumar, R.; Singh, S.; Bhadoria, D.P.; Katyal, A.; Reichard, U.; Asif, A.R. Novel cytosolic allergens of Aspergillus fumigatus identified from germinating conidia. J. Proteome Res. 2010, 9, 5530–5541. [Google Scholar] [CrossRef]
  22. Kumar, A.; Ahmed, R.; Singh, P.K.; Shukla, P.K. Identification of virulence factors and diagnostic markers using immunosecretome of Aspergillus fumigatus. J. Proteomics 2011, 74, 1104–1112. [Google Scholar] [CrossRef]
  23. Asif, A.R.; Oellerich, M.; Amstrong, V.W.; Gross, U.; Reichard, U. Analysis of the cellular Aspergillus fumigatus proteome that reacts with sera from rabbits developing an acquired immunity after experimental aspergillosis. Electrophoresis 2010, 31, 1947–1958. [Google Scholar] [CrossRef]
  24. Shi, L.N.; Li, F.Q.; Huang, M.; Lu, J.F.; Kong, X.X.; Wang, S.Q.; Shao, H.F. Immunoproteomics based identification of thioredoxin reductase GliT and novel Aspergillus fumigatus antigens for serologic diagnosis of invasive aspergillosis. BMC Microbiol. 2012, 12, 11. [Google Scholar] [CrossRef]
  25. Sipsas, N.V.; Kontoyiannis, D.P. Invasive fungal infections in patients with cancer in the Intensive Care Unit. Int. J. Antimicrob. Agents 2012, 39, 464–471. [Google Scholar] [CrossRef]
  26. Denning, D.W. Aspergillus species. In Principles and Practice of Infectious Diseases, 5th ed.; Mandell, G.L., Bennett, J.E., Dolin, R., Eds.; Churchill Livingstone: Philadelphia, PA, USA, 2000; Volume 2, pp. 2674–2685. [Google Scholar]
  27. Pitarch, A.; Jiménez, A.; Nombela, C.; Gil, C. Decoding serological response to Candida cell wall immunome into novel diagnostic, prognostic, and therapeutic candidates for systemic candidiasis by proteomic and bioinformatic analyses. Mol. Cell. Proteomics 2006, 5, 79–96. [Google Scholar]
  28. Lai, H.Y.; Tam, M.F.; Tang, R.B.; Chou, H.; Chang, C.Y.; Tsai, J.J.; Shen, H.D. cDNA cloning and immunological characterization of a newly identified enolase allergen from Penicillium citrinum and Aspergillus fumigatus. Int. Arch. Allergy Immunol. 2002, 127, 181–190. [Google Scholar] [CrossRef]
  29. Rohde, M.; Schwienbacher, M.; Nikolaus, T.; Heesemann, J.; Ebel, F. Detection of early phase specific surface appendages during germination of Aspergillus fumigatus conidia. FEMS Microbiol. Lett. 2002, 206, 99–105. [Google Scholar] [CrossRef]
  30. Rhodes, C.J. Aspergillus fumigatus: Growth and virulence. Med. Mycol. 2006, 44, 77–81. [Google Scholar] [CrossRef]
  31. Kubitschek-Barreira, P.H.; Curty, N.; Neves, G.W.; Gil, C.; Lopes-Bezerra, L.M. Differential proteomic analysis of Aspergillus fumigatus morphotypes reveals putative drug targets. J. Proteomics 2013, 78, 522–534. [Google Scholar] [CrossRef]
  32. Denikus, N.; Orfaniotou, F.; Wulf, G.; Lehmann, P.F.; Monod, M.; Reichard, U. Fungal antigens expressed during invasive aspergillosis. Infect. Immun. 2005, 73, 4704–4713. [Google Scholar] [CrossRef]
  33. Lamaris, G.A.; Chamilos, G.; Lewis, R.E.; Safdar, A.; Raad, II.; Kontoyiannis, D.P. Scedosporium infection in a tertiary care cancer center: A review of 25 cases from 1989–2006. Clin. Infect. Dis. 2006, 43, 1580–1584. [Google Scholar] [CrossRef]
  34. Cortez, K.J.; Roilides, E.; Quiroz-Telles, F.; Meletiadis, J.; Antachopoulos, C.; Knudsen, T.; Buchanan, W.; Milanovich, J.; Sutton, D.A.; Fothergill, A.; et al. Infections caused by Scedosporium spp. Clin. Microbiol. Rev. 2008, 21, 157–197. [Google Scholar] [CrossRef]
  35. Mori, Y.; Nagasaki, Y.; Kamezaki, K.; Takenaka, K.; Iwasaki, H.; Harada, N.; Miyamoto, T.; Abe, Y.; Shimono, N.; Akashi, K.; et al. High incidence of false-positive Aspergillus galactomannan test in multiple myeloma. Am. J. Hematol. 2010, 5, 449–451. [Google Scholar]
  36. Morrissey, C.O. Advancing the field: Evidence for new management strategies in invasive fungal infections. Curr. Fungal. Infect. Rep. 2013, 7, 51–58. [Google Scholar] [CrossRef]
  37. Vidovic, A.; Arsic-Arsenijevic, V.; Tomin, D.; Djunic, I.; Jakovic, R.; Loncar, Z.; Barac, A. Proven invasive pulmonary mucormycosis successfully treated with amphotericin B and surgery in patient with acute myeloblastic leukemia: A case report. J. Med. Case Rep. 2013, 7, 263. [Google Scholar] [CrossRef]
  38. Crešnar, B.; Petrič, S. Cytochrome P450 enzymes in the fungal kingdom. Biochim. Biophys. Acta 2010, 1814, 29–35. [Google Scholar]
  39. Ehrlich, K.C.; Chang, P.K.; Yu, J.; Cotty, P.J. Aflatoxin biosynthesis cluster gene cypA is required for G aflatoxin formation. Appl. Environ. Microbiol. 2004, 70, 6518–6524. [Google Scholar] [CrossRef]
  40. Wen, Y.; Hatabayashi, H.; Arai, H.; Kitamoto, H.K.; Yabe, K. Function of the cypX and moxY genes in aflatoxin biosynthesis in Aspergillus parasiticus. Appl. Environ. Microbiol. 2005, 71, 3192–3198. [Google Scholar] [CrossRef]
  41. Lepesheva, G.I.; Waterman, M.R. Sterol 14-α-demethylase cytochrome P450 (CYP51), a P450 in all biological kingdoms. Biochim. Biophys. Acta 2007, 1770, 467–477. [Google Scholar]
  42. Verweij, P.E.; Mellado, E.; Melchers, W. Multiple-triazole-resistant aspergillosis. N. Engl. J. Med. 2007, 356, 1481–1483. [Google Scholar] [CrossRef]
  43. Mavridou, E.; Bruggemann, R.J.; Melchers, W.J.; Verweij, P.E.; Mouton, J.W. Impact of cyp51A mutations on the pharmacokinetic and pharmacodynamics properties of voriconazole in a murine model of disseminated aspergillosis. Antimicrob. Agents Chemother. 2010, 54, 4758–4764. [Google Scholar] [CrossRef]
  44. Kuipers, S.; Brüggemann, R.J.; de Sévaux, R.G.; Heesakkers, J.P.; Melchers, W.J.; Mouton, J.W.; Verweij, P.E. Failure of posaconazole therapy in a renal transplant patient with invasive aspergillosis due to Aspergillus fumigatus with attenuated susceptibility to posaconazole. Antimicrob. Agents Chemother. 2011, 55, 3564–3566. [Google Scholar] [CrossRef]
  45. Van den Brink, H.M.; van Gorcom, R.F.; van den Hondel, C.A.; Punt, P.J. Cytochrome P450 enzyme systems in fungi. Fungal Genet. Biol. 1998, 23, 1–17. [Google Scholar] [CrossRef]
  46. Nelson, D.R. Cytochrome P450 and the individuality of species. Arch. Biochem. Biophys. 1999, 369, 1–10. [Google Scholar] [CrossRef]
  47. Nelson, D.R. Cytochrome P450 nomenclature. Methods Mol. Cell. Biol. 1998, 107, 15–24. [Google Scholar]
  48. Nelson, D.R. Metazoan cytochrome P450 evolution. Comp. Biochem. Physiol. C Pharmacol. Toxicol. Endocrinol. 1998, 121, 15–22. [Google Scholar]
  49. Hinnebusch, A.G.; Dever, T.E.; Asano, K. Translational Control in Biology and Medicine; Sonenberg, N., Hershey, J.W.B., Mathews, M.B., Eds.; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, NY, USA, 2007; pp. 225–268. [Google Scholar]
  50. Carvalho, M.D.; Carvalho, J.F.; Merrick, W.C. Biological characterization of various forms of elongation factor 1 from rabbit reticulocytes. Arch. Biochem. Biophys. 1984, 234, 603–611. [Google Scholar] [CrossRef]
  51. Skogerson, L.; Moldave, K. Characterization of the interaction of aminoacyltransferase II with ribosomes. Binding of transferase II and translocation of peptidyl transfer ribonucleic acid. J. Biol. Chem. 1968, 243, 5354–5360. [Google Scholar]
  52. Liu, M.; Gelli, A. Elongation factor 3, EF3, associates with the calcium channel Cch1 and targets Cch1 to the plasma membrane in Cryptococcus neoformans. Eukaryot. Cell 2008, 7, 1118–1126. [Google Scholar] [CrossRef]
  53. Hollenstein, K.; Dawson, R.J.; Locher, K.P. Structure and mechanism of ABC transporter proteins. Curr. Opin. Struct. Biol. 2007, 17, 412–418. [Google Scholar] [CrossRef]
  54. Uritani, M.; Miyazaki, M. Characterization of the ATPase and GTPase activities of the elongation factor 3 (EF-3) from yeasts. J. Biochem. 1988, 103, 522–530. [Google Scholar]
  55. Sasikumar, A.N.; Kinzy, T.G. Mutations in the chromodomain-like insertion of translation elongation factor 3 compromise protein synthesis through reduced ATPase activity. J. Biol. Chem. 2014, 289, 4853–4860. [Google Scholar] [CrossRef]
  56. López-Ribot, J.L.; Alloush, H.M.; Masten, B.J.; Chaffin, W.L. Evidence for presence in the cell wall of Candida albicans of a protein related to the hsp70 family. Infect. Immun. 1996, 64, 3333–3340. [Google Scholar]
  57. Yuan, S.Y. Protein kinase signalling in the modulation of microvascular permeability. Vascul. Pharmacol. 2002, 39, 213–223. [Google Scholar] [CrossRef]
  58. Pardo, M.; Ward, M.; Bains, S.; Molina, M.; Blackstock, W.; Gil, C.; Nombela, C. A proteomic approach for the study of Saccharomyces cerevisiae cell wall biogenesis. Electrophoresis 2000, 21, 3396–3410. [Google Scholar] [CrossRef]
  59. Pitarch, A.; Sanchez, M.; Nombela, C.; Gil, C. Sequential fractionation and two-dimensional gel analysis unravels the complexity of the dimorphic fungus Candida albicans cell wall proteome. Mol. Cell. Proteomics 2002, 1, 967–982. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Virginio, E.D.; Kubitschek-Barreira, P.H.; Batista, M.V.; Schirmer, M.R.; Abdelhay, E.; Shikanai-Yasuda, M.A.; Lopes-Bezerra, L.M. Immunoproteome of Aspergillus fumigatus Using Sera of Patients with Invasive Aspergillosis. Int. J. Mol. Sci. 2014, 15, 14505-14530. https://doi.org/10.3390/ijms150814505

AMA Style

Virginio ED, Kubitschek-Barreira PH, Batista MV, Schirmer MR, Abdelhay E, Shikanai-Yasuda MA, Lopes-Bezerra LM. Immunoproteome of Aspergillus fumigatus Using Sera of Patients with Invasive Aspergillosis. International Journal of Molecular Sciences. 2014; 15(8):14505-14530. https://doi.org/10.3390/ijms150814505

Chicago/Turabian Style

Virginio, Emylli D., Paula H. Kubitschek-Barreira, Marjorie Vieira Batista, Marcelo R. Schirmer, Eliana Abdelhay, Maria A. Shikanai-Yasuda, and Leila M. Lopes-Bezerra. 2014. "Immunoproteome of Aspergillus fumigatus Using Sera of Patients with Invasive Aspergillosis" International Journal of Molecular Sciences 15, no. 8: 14505-14530. https://doi.org/10.3390/ijms150814505

Article Metrics

Back to TopTop