Skip to main content

Host response profile of human brain proteome in toxoplasma encephalitis co-infected with HIV

Abstract

Background

Toxoplasma encephalitis is caused by the opportunistic protozoan parasite Toxoplasma gondii. Primary infection with T. gondii in immunocompetent individuals remains largely asymptomatic. In contrast, in immunocompromised individuals, reactivation of the parasite results in severe complications and mortality. Molecular changes at the protein level in the host central nervous system and proteins associated with pathogenesis of toxoplasma encephalitis are largely unexplored. We used a global quantitative proteomic strategy to identify differentially regulated proteins and affected molecular networks in the human host during T. gondii infection with HIV co-infection.

Results

We identified 3,496 proteins out of which 607 proteins were differentially expressed (≥1.5-fold) when frontal lobe of the brain from patients diagnosed with toxoplasma encephalitis was compared to control brain tissues. We validated differential expression of 3 proteins through immunohistochemistry, which was confirmed to be consistent with mass spectrometry analysis. Pathway analysis of differentially expressed proteins indicated deregulation of several pathways involved in antigen processing, immune response, neuronal growth, neurotransmitter transport and energy metabolism.

Conclusions

Global quantitative proteomic approach adopted in this study generated a comparative proteome profile of brain tissues from toxoplasma encephalitis patients co-infected with HIV. Differentially expressed proteins include previously reported and several new proteins in the context of T. gondii and HIV infection, which can be further investigated. Molecular pathways identified to be associated with the disease should enhance our understanding of pathogenesis in toxoplasma encephalitis.

Background

Toxoplasma encephalitis (TE) is a major complication of central nervous system caused by Toxoplasma gondii, an obligate intracellular protozoan. TE presents with highly varied neurological symptoms with both focal and diffuse neurological abnormalities. The risk of developing TE is high in immunocompromised individuals including patients with HIV infection, solid organ transplantation and those receiving immunosuppressive therapy, mostly due to recrudescence of latent infection [13].

Various diagnostic tests have been in practice for the diagnosis of toxoplasmosis in serum, CSF and brain tissues [3]. Conventionally, radiologic techniques such as CT scan and MRI scan are used for visualizing the lesions produced in TE [4]. Enzyme-linked immunosorbent assays to measure IgG and IgM levels against T. gondii in serum and urine are also routinely used in diagnostics [5]. Advanced diagnostic procedures including loop mediated isothermal amplification reactions and PCR-based assays are also used for the detection of T. gondii with a high degree of sensitivity [6].

The life cycle of T. gondii is divided into sexual and asexual phases. Sexual reproduction occurs in cats, which are the definitive hosts for this parasite. Gametocytes present in the intestinal epithelium of cats are fertilized and infectious oocysts are shed through feces. These oocysts sporulate in the environment and contaminate soil, water and food, through which infection is transmitted to warm blooded animals including humans. These warm blooded animals act as intermediate hosts, in which asexual reproduction occurs in the form of actively replicating tachyzoites. Host immune response induces conversion of tachyzoites to semi-dormant bradyzoites, which form tissue cysts. The life cycle of T. gondii is completed when tissue cysts are ingested again by cats [7].

Around one-third of the general population worldwide is considered to be infected with T. gondii, most of whom are asymptomatic [7]. The prevalence of infection depends on the climatic conditions and the age of the population. Studies in India have shown that ~25% of general population is seropositive for T. gondii IgG antibodies [3]. Transmission of T. gondii to humans may occur from an infected mother to the fetus or orally by consumption of either sporulated oocysts from contaminated soil, food or water or bradyzoites from undercooked or uncooked meat. Upon gaining entry into the human body, T. gondii gets converted into tachyzoite stage, crosses the gastrointestinal barrier to disseminate throughout the body including immune privileged sites such as brain, retina and fetus. Tachyzoites invade the astrocytes and microglial cells to breach the blood-brain barrier and form tissue cysts in the brain [8]. In immunocompromised individuals with <200 CD4+ cells per μl, the conversion of bradyzoites to tachyzoites leads to TE [1].

During acute infection and reactivation of latent T. gondii infection, the immune system challenges the parasite by producing interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α) and inducible nitric oxide synthase (iNOS). CD4+ T cells, CD8+ T cells and natural killer cells produce IFN-γ and TNF-α following T. gondii infection. IL-12, IL-6 and IL-4 also were shown to prevent formation of cysts and tachyzoite proliferation, probably through the maintenance of IFN-γ and TNF-α mediated resistance [9]. IFN- γ also induces the expression of vascular cell adhesion molecule-1 (VCAM-1), which enhances the recruitment of CD4+ T cells and CD8+ T cells to the site of infection [10]. Molecular mechanisms underlying the host-pathogen interactions have been studied using mouse models and cell lines with respect to TE. Though substantial information was provided by these studies, there still remain unanswered questions that emphasize the need for further studies at the molecular level in humans [11, 12]. Therefore, we carried out a global unbiased quantitative proteomic analysis of brain tissues from TE patients co-infected with HIV by iTRAQ labeling and high-resolution mass spectrometry using LTQ-Orbitrap Velos mass spectrometer. Differentially expressed proteins identified from TE brain tissues co-infected with HIV included several novel proteins along with a few proteins, which were reported earlier. We also validated three differentially expressed proteins using immunohistochemistry (IHC) and found that their expression was consistent with the mass spectrometry results.

Results and discussion

We carried out a quantitative proteomic analysis of the frontal lobe brain tissues from TE patients co-infected with HIV and uninfected control subjects using iTRAQ labeling followed by SCX fractionation and high resolution Fourier transform mass spectrometry. The mass spectra were searched using SEQUEST search algorithm against Human RefSeq protein database version 52, which comprised of 33,987 protein sequences. In total, we identified 3,496 proteins of which 607 proteins were differentially expressed (≥1.5-fold). Among the differentially expressed proteins, 293 and 314 proteins were found to be overexpressed and downregulated, respectively, in brain tissues of TE patients in comparison to control brain tissues (Additional file 1: Table S1). A partial list of overexpressed and downregulated proteins is provided in Tables 1 and 2, respectively. Representative MS/MS spectra of selected proteins are provided in Figure 1. We have deposited the mass spectrometry data to Human Proteinpedia and PRIDE repositories [13, 14].

Table 1 A partial list of upregulated regulated proteins in toxoplasma encephalitis
Table 2 A partial list of downregulated regulated proteins in toxoplasma encephalitis
Figure 1
figure 1

Representative MS/MS spectra of peptides of differentially expressed proteins: A) Major histocompatibility complex, class I, B (HLA-B) protein is upregulated 4.4-fold in toxoplasma encephalitis brain tissues when compared to controls. B) Alpha-1 antitrypsin (SERPINA1) protein is upregulated by 3.0-fold in toxoplasma encephalitis brain tissues when compared with control. C) Ras homolog family member G (RHOG) protein is downregulated by 2.5-fold in toxoplasma encephalitis brain tissues when compared with control. D) CD9 molecule (CD9) is downregulated by 2.0-fold in toxoplasma encephalitis brain tissues when compared with control.

Categorization of differentially expressed proteins based on Gene Ontology annotation

We categorized differentially expressed proteins based on their biological processes and molecular function using annotations in Human Protein Reference Database (HPRD; http://www.hprd.org) [15, 16], which follows Gene Ontology (GO) standards. Proteins involved in important biological processes such as cell communication (23%), metabolism/energy pathways (14%), transport (13%), cell growth/ maintenance (10%) and immune response (5%) were found to be altered in response to TE co-infected with HIV (Figure 2A). Classification of differentially expressed proteins based on their molecular functions enriched for proteins with catalytic activity (5%), transporter activity (9%), GTPase activity (5%), and cell adhesion activity (4%) (Figure 2B).

Figure 2
figure 2

Gene Ontology (GO) analysis of differentially regulated proteins: A) Biological processes-based analysis showed majority of the proteins involved in processes such as cell communication, growth and transport. B) Molecular function-based analysis revealed a majority of the proteins with unknown functions. However, a significant number were seen with transporter and catalytic activities among others.

Functional annotation of differentially expressed proteins in toxoplasma encephalitis

Pathway analysis of the differentially expressed proteins in TE co-infected with HIV brain tissue samples using the Pathway Architect module in GeneSpring (version 12.6) revealed the enrichment of several proteins involved in pathways such as neural transmission across chemical synapse, neurotransmitter release cycle, antigen processing and presentation along with complement and coagulation cascade pathway. In addition, several key modulators of the brain-derived neurotrophic factor (BDNF) signaling pathway were found to be differentially regulated, including aldo-keto reductase (AKR1C1), alkaline phosphatase (ALPL), secreted phosphoprotein (SPP1), heat shock protein (HSPA1A), chromogranin B (CHGB), ras homolog family member (RHOG), regulating synaptic membrane exocytosis (RIMS3), inositol polyphosphate-1-phosphatase (INPP1), integrin, alpha 2 (ITGA2), filamin C (FLNC) and polymerase I transcript release factor (PTRF) [17]. RHOG, which was found to be downregulated in TE compared to control tissues, is involved in mediating BDNF-induced neurite outgrowth [18]. It has been reported that HIV-1 promotes neuronal injury by reducing the length of neuronal processes through reduction in the mature BDNF levels in neurons [19]. Therefore, differential expression of several important molecules in BDNF signaling pathway may be the cause of cognitive aberration in HIV-associated TE (Figure 3).

Figure 3
figure 3

Biological network analysis of differentially expressed proteins using GeneSpring identified deregulated BDNF pathway: Nineteen proteins involved in the BDNF pathway were differentially expressed including the downregulation of the BDNF receptor protein – NTRK2. Proteins involved in BDNF mediated lamellipodia formation and neurite outgrowth were downregulated whereas proteins involved in BDNF mediated synaptic plasticity were overexpressed.

Biological significance of the differentially expressed human proteins

Differentially expressed proteins identified in the current study covers a diverse spectrum of functions including transmission across chemical synapses, antigen processing and presenting, neurotransmitter release cycle and interferon signaling. We identified proteins including guanine nucleotide binding protein (GNAO1), major histocompatibility complex class II protein (HLA-DRB1), heat shock 70 kDa protein 1A/1B, extracellular protein laminin (LAMA2), signal transducer and activation of transcription (STAT1), which have been previously reported to be associated with TE among the upregulated proteins. Calcium signaling by multiple G–protein effectors has been reported to be altered by inflammatory mediators, in inflammatory conditions in central nervous system (CNS) including infection, trauma, stroke, neurodegeneration and autoimmunity [20]. HLA-DRB1 is known to be a risk factor in HIV-1 patients for developing opportunistic infections, especially TE [21]. In addition, heat shock 70 kDa protein (HSPA1B) polymorphism has been shown to be associated with risk for schizophrenia, positive symptoms as well as clinical outcome [22, 23]. LAMA2 is an extracellular matrix protein, which regulates and promotes oligodendrocyte development, whereas STAT1 is actively modulated by T. gondii increasing the likelihood of its survival within the host [2426].

Host immune response proteins

Among the differentially expressed proteins, those involved in host immune response, such as complement proteins and major histocompatibility proteins were identified in this study. We detected several proteins reported in interferon-gamma signaling to be overexpressed, including guanylate binding protein (GBP1), major histocompatibility complex protein (HLA-B), intercellular adhesion molecule (ICAM1), ubiquitin-like modifier (ISG15) and STAT1. Microtubule-associated protein tau (MAPT), C-reactive protein (CRP) along with STAT1, involved in IL-2, IL-5 and IL-6 signaling pathways, were also found to be overexpressed in TE brain tissues. In patients with T. gondii and HIV infection, marked decrease in CD4+ T cells has been reported and often there is no significant increase in the antibody titer, as observed during aggravation of infection [27]. Furthermore, in T cells from HIV patients with toxoplasmosis, secretion of both interferon-gamma and IL-2 has been known to be altered [28]. Persistence of toxoplasma infection is contributed by aberrant production of these vital immune cytokines. Therefore, with the progressive depletion of the T cell dependent protective mechanisms in HIV-1, especially CD8+ T cells, probability of developing toxoplasma infection increases. However, in our study we observed overexpression of proteins associated with interferon-gamma and IL-2 signaling pathways, indicating restored immune response in the patients. Similar restoration of immune response has been reported by other studies in patients previously subjected to antiretroviral therapy [29, 30].

Other proteins involved in interferon signaling that were found to be overexpressed include beta-2 microglobulin (B2M), major histocompatibility complex protein (HLA-G), interferon-induced protein with tetratricopeptide repeats (IFIT1 and IFIT3) and myxovirus (influenza virus) resistance 1 interferon-inducible protein (MX1). B2M is important in diagnosis of HIV and its rise in HIV patients is considered as an indicator of HIV progression [31]. Increased HLA-G level is known to be associated with congenital transmission of T. gondii whereas, increased expression of IFIT1 has been reported in HIV-1 infected astrocytes [32, 33]. Overexpression of B2M, IFIT3, MX1 and STAT1 has been reported in the brains of neuro-AIDS patients, including those on combination antiretroviral therapy and also in non-human primate model of neuro-AIDS [34]. JAK/STAT pathway is known to be modulated by toxoplasma and involved in the pathogenesis of depression [35, 36]. It has also been shown that toxoplasmosis is associated with depression along with other psychiatric manifestations [37].

Proteins in neurotransmitter release cycle and transmission across chemical synapses

In this study, several proteins playing an important role in neurotransmitter release cycle and transmission across chemical synapses were found to be differentially expressed. These include actin (ACTN2), calcium channel voltage-dependent gamma subunit 2 (CACNG2), DnaJ protein homolog (DNAJC5), gamma-aminobutyric acid receptors (GABBR1, GABBR2, GABRA1 and GABRG2), guanine nucleotide binding proteins (GNG13 and GNG4), glutamate receptors (GRIA3 and GRIN1), monoamine oxidase A (MAOA), member RAS oncogene family (RAB3A), solute carrier family proteins (SLC17A7, SLC32A1 and SLC6A1), syntaxins (STX1A and STXBP1), synapsins (SYN1, SYN2 and SYN3) and synaptotagmin (SYT1). Among these, CACNG2 has been reported to be associated with schizophrenia [38, 39]. DNAJC5 anchored to synaptic vesicles, along with exocrine, endocrine and neuroendocrine secretory granules, has been shown to have anti-neurodegenerative properties [40]. Interestingly, evidence of GABAergic dysfunction in lateral cerebella, due to reduced protein levels of both receptors GABBR1 and GABBR2, has been reported in subjects of schizophrenia, bipolar disorder and major depression [41]. Also, it has been shown that mutations in GABRA1 lead to epileptic encephalopathies and mutations in GABRG2 have been associated with generalized epilepsy [4244]. Among overexpressed, glutamate receptors GRIA3 has a positive association in women patients with schizophrenia patients [45]. MAOA is known to catalyze the oxidative deamination of amines and has been implicated in antisocial behavior [46]. Alterations in solute carrier family proteins (SLC17A7, SLC32A1 and SLC6A1) implicating impairment of synaptic transmission may contribute in schizophrenia pathology [47]. Among syntaxin proteins (STX1A and STXBP1), STXBP1 was known to be a key molecule in ion channel regulation and synaptic exocytosis and has been reported widely in cases of migraine in different geographical settings [48]. Mutations in STXBP1 gene are responsible for infantile epileptic encephalopathy-4 [49, 50]. SYN1 gene has been reported as a susceptible gene for autism-spectrum disorder (ASD). Knockout mice study revealed that lack of Syn1, Syn2 and Syn3 cause cognitive impairment and activates strong epileptic phenotype in mice (Syn1 and/or Syn2) [51]. Synaptotagmin-1 (SYT1) triggers neurotransmitter release at synapse by binding to calcium (Ca2+) [52]. Therefore, differential expression of neurotransmitter release cycle and transmission across chemical synapses may predispose the patient suffering from TE infection to seizure activity and cognitive impairments. Moreover, a number of studies connect T. gondii with higher incidence of schizophrenia [53, 54].

Validation of differentially expressed proteins by immunohistochemistry

Differentially expressed proteins identified using high resolution mass spectrometry from frontal lobe tissues of TE were validated using IHC. For this, we selected three candidates - HLA-B, SERPINA1 and RHOG - for IHC validation based on the molecular function of the proteins and its significance to the disease manifestation (Figure 4).

Figure 4
figure 4

Immunohistochemistry-based validation of differentially expressed proteins in toxoplasma encephalitis co-infected with HIV: A) Histiocytic elements in the toxoplasma lesion strongly expressed HLA-B, while the pale zones represent the necrotic acellular zones, B) Higher magnification highlights strong expression of HLA-B in the perivascular histiocytes, C) The expression of RHOG is marginally downregulated in the histiocytes and newly formed capillary endothelia, D) Control brain has low expression of alpha-1 antitrypsin (SERPINA1) in glial cells and a necrotic vessel wall, E) In the case of toxoplasma encephalitis the glial cells, the micro vessels and histiocytes expressed alpha-1 antitrypsin strongly unlike the control.

Major histocompatibility complex, class I (HLA-B)

HLA-B protein known to be present on surface of nucleated cells is involved in antigen presentation to cytotoxic T cells. Earlier studies have shown that infection with T. gondii elicits HLA-B expression in dendritic cells, astrocytes and macrophages [55]. Toxoplasma antigens induce significant HLA-B expression in murine TE brain tissue, which may play role in Toxoplasma antigen presentation evoking active immune response [56]. We found upregulation of HLA-B in HIV-associated TE brain tissues. Immunohistochemical staining showed strong HLA-B expression in histiocytic elements of toxoplasma lesion, while the pale zones represent the necrotic acellular zones. Strong expression of HLA-B was observed in the perivascular histiocytes at higher magnification.

Alpha-1 antitrypsin (SERPINA1)

Alpha-1 antitrypsin (SERPINA1) is a member of the serpin family. Polymorphisms in the SERPINA1 gene have been reported to be associated with fibromyalgia and mood disorders [57]. Hypersialylated SERPINA1 has been reported to be a protein of significant diagnostic value for early detection of Parkinson’s dementia [58]. It has also been associated with other neurological disorders like Alzheimer’s disease and schizophrenia [59, 60]. It is interesting to note that serine protease inhibitor neuroserpin regulates the growth and maturation of hippocampal neurons [61] dysfunction of hippocampus is one of the critical factors in the pathogenesis of schizophrenia [62]. Apart from this alpha-1 antitrypsin blocks the processing of group antigens (Gag) and envelop proteins (Env), which results in suppression of HIV-1 morphogenesis [63]. Immunohistochemical labeling showed strongly expressed alpha-1 antitrypsin in the glial cells, the micro vessels and histiocytes of toxoplasma encephalitis brain tissues unlike in the control brain tissues, which showed low expression in glial cells and the necrotic vessel wall.

Ras homolog family member G (RHOG)

RHOG is mainly involved in actin cytoskeleton rearrangement, cell migration and protection of cell from anoikis [64, 65]. Recent studies unraveled the role of RHOG in pathogenesis of bacterial infections [66]. It has been detected in rat hippocampus, cerebellum and specific expression was observed in oligodendrocytes and neuronal cells of brain [67, 68]. As reported by multiple studies RHOG has significant role in various neurological processes including neurite extension, spine morphogenesis, neuronal progenitor cell proliferation and axonal outgrowth [69, 70]. We observed down-regulation of RHOG in TE brain tissues co-infected with HIV. Immunohistochemical labeling showed the marginal downregulation of RHOG expression in the histiocytes and newly formed capillary endothelia.

Conclusions

TE is a principal opportunistic infection of the CNS in individuals infected with HIV. The molecular profile associated with pathological progression of TE has not been explored using a global proteomics platform. Utilizing an iTRAQ-based quantitative proteomic approach, we identified a subset of proteins, which are differentially expressed in TE co-infected with HIV brain tissues. Our data provides insights into molecular profiles of TE, which can be investigated through several hypotheses. We found differential regulation of key proteins involved in host-immune response and neurotransmitter release cycle, neuronal growth, energy metabolism and calcium transport. Increase in the neurotransmission across synapses may predispose the patients to seizure activity and cognitive impairment in the clinical evolution of TE. We also identified overexpression of proteins, which may have a role in the development of schizophrenia among TE patients. Upregulation of caveolar proteins and intercellular adhesion molecules indicate their possible role along with gp41 protein from HIV in mediating the entry of the pathogens by breaching blood-brain barrier.

Methods

Sample collection

The study was approved by scientific ethics committee of National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India. Brain tissue samples from the lesion site within the frontal lobe from five confirmed cases of cerebral toxoplasmosis associated with HIV-1 infection were collected during autopsy and divided into symmetric halves. One half was processed for histopathological studies including immunohistochemistry for toxoplasma and macrophage response (CD 68), while the other half was used for proteomic study. All tissues included in the study revealed features of toxoplasma encephalitis in the cortical ribbon and subcortical zone with areas of necrosis, inflammation and variable CD 68 positive histocytic response along with the presence of toxoplasma organisms (both bradyzoites and tachyzoites). All cases were positive for HIV-1 on serological testing and confirmed to be HIV-1 clade C by clade specific PCR developed at Jawaharlal Nehru Centre for Advanced Scientific Research in Bangalore and validated. Five uninfected control brain tissues from the similar age and sex group were collected from victims of road traffic accidents within 10-19 h post mortem during autopsy (Additional file 2: Tables S2 and S3). Fifteen toxoplasma encephalitis frontal cortex brain tissue samples including five samples taken for proteomics study from age group 1 to 54 years and one uninfected control sample were included for immunohistochemistry validation. The autopsy was conducted within 3-19 hours and bodies were kept at 4°C after death. The control tissues were confirmed to be negative for HIV-1 and toxoplasma infection. The samples were obtained from the archives of Human Brain Tissue Repository, Department of Neuropathology, NIMHANS, Bangalore.

Protein extraction and normalization

Around 100 mg of frontal lobe brain tissue from each individual were homogenized by crushing in liquid nitrogen with the aid of a mortar and pestle. Once the tissues were ground to fine powder, around 1 ml of 0.5% SDS was added and homogenized. Tissue lysate was transferred to sterile microcentrifuge tubes and centrifuged at 14,000 × g for 30 min at 4°C. Clear supernatant was collected in separate microcentrifuge tubes. Total protein estimation was carried out using BCA protein assay (Pierce, Thermo Scientific). Normalization of protein amounts was confirmed using 10% SDS-PAGE. Equivalent amounts of protein from each of the five TE with HIV-1 samples were pooled and similar processing was done for control samples.

iTRAQ labeling and strong cation exchange chromatography

Pooled protein samples from each condition were reduced, alkylated and digested with trypsin. iTRAQ labeling (Applied Biosystems, cat# - 4352135) of peptides was carried out according to manufacturer’s protocol as described earlier [71]. Briefly, 200 μg of protein from each condition was treated with 4 μl of reducing agent (tris (2-carboxyethyl) phosphine) at 60°C for 1 hour and alkylation of cysteine residues was carried out by incubating the samples with 2 μl of methyl methanosulfonate for 10 minutes at room temperature. Alkylated proteins were then digested using sequencing grade trypsin (Promega, cat# - V511A) with an enzyme to substrate ratio of 1:20 (w/w) at 37°C for 16 hours. Peptides from each condition were split into two equal halves and iTRAQ labeling was carried out in replicates. Peptides from control samples was labeled using iTRAQ reagent yielding reporter ions 114 and 115, peptides from TE with HIV-1 patient samples was labeled with iTRAQ reagents yielding reporter ions 116 and 117. Labeling was carried out by incubating the peptide-label mixture at room temperature for one hour, the reaction was quenched by addition of 150 μl milliQ water followed by incubation at room temperature for one hour.

iTRAQ labeled peptides from control and TE with HIV-1 were pooled, vacuum dried and reconstituted in 5 mM potassium phosphate buffer, 25% acetonitrile (pH 2.8) (solvent A). Strong cation exchange chromatography was carried out as described earlier [72]. The peptides were fractionated on polysulfoethyl A column (PolyLC, Columbia, MD) (100 × 2.1 mm, 5 μm particles with 300 Å pores) Agilent 1200 infinity series HPLC system for a period of 50 minutes using a gradient of increasing salt concentration of up to 350 mM KCl in solvent A. The fractions collected were pooled based on SCX profile into 21 fractions, dried and reconstituted in 40 μl of 0.1% formic acid. Samples were desalted using C18 (3 M Empore high-performance extraction disks) stage tip dried and stored in -20°C till mass spectrometric analysis.

LC-MS/MS analysis

LC-MS/MS analysis was carried out on LTQ Orbitrap Velos ETD – mass spectrometer (Thermo Scientific, Bremen, Germany) interfaced with Proxeon nanoLCII (Thermo Scientific, Bremen, Germany). Peptides were analyzed on a Reversed Phase Liquid Chromatography (RPLC). The RPLC system equipped with a pre-column (2 cm, 5 μ – 100 Å) and an analytical column (12 cm, 3 μ – 100 Å) made with magic AQ C18 material (Michrom Bioresources, Inc, Auburn, CA) packed in-house. Further, the peptides are sprayed using 10 μ nano electro spray emitter tip (New Objective, Woburn, MA) fixed to an NSI source. The peptides were loaded on the pre column using solvent A (0.1% formic acid, 5% acetonitrile and resolved on the analytical column using a linear gradient of 8-30% solvent B (95% acetonitrile, 0.1% formic acid) for 75 minutes at a constant flow rate of 0.35 μl/min. The spray voltage and heated capillary temperature were set to 2.2 kV and 250°C, respectively. The MS spectra were acquired in a data-dependent manner in the m/z range of 350 to 1800. From each MS survey scan, 15 most intense precursor ions were selected for fragmentation. MS and MS/MS scans were acquired in an Orbitrap mass analyzer and the peptides were fragmented by higher energy collision dissociation with normalized collision energy of 41%. MS scans were acquired at a resolution of 60,000 at 400 m/z, while MS/MS scans were acquired at a resolution of 15,000. The automatic gain control (AGC) for full FT MS was set to 1 million ions and for FT MS/MS was set to 0.1 million ions with a maximum time of accumulation of 200 ms and 500 ms, respectively.

Data analysis

The mass spectrometry data obtained was searched against the human RefSeq 52 proteins using Proteome Discoverer, version 1.3.0.339 (Thermo Scientific, Bremen, Germany) workflow. The workflow consisted of spectrum selector, reporter ion quantifier and SEQUEST nodes along with peptide validator node. Search parameters included trypsin as the enzyme with 1 missed cleavage; oxidation of methionine was set as dynamic modification, while methylthio modification of cysteine and iTRAQ modification at N-terminus of the peptide and lysine were set as static modifications. Precursor and fragment mass tolerance were set to 20 ppm and 0.1 Da, respectively with a signal to noise ratio of 1.5 for a precursor mass range of 350-10,000 Da. The raw data obtained was searched against decoy database to calculate 1% false discovery rate cut-off score. Spectra that matched to the contaminants and those that did not pass the 1% FDR threshold were not considered for analysis.

Bioinformatics analysis

Bioinformatics analysis was carried out to categorize proteins based on biological processes, cellular component and molecular function using annotations in Human Protein Reference Database (HPRD) [15, 16], which is in compliance with gene ontology (GO) standards. The differentially expressed proteins having ≥1.5 fold change were taken as input and biological network was generated using Pathway Architect module of GeneSpring version 12.6 (Agilent Biosystems, Santa Clara, CA). We also integrated the interaction data from NetPath (http://www.netpath.org) and HPRD (http://www.hprd.org) in the analysis using GeneSpring software as described earlier [17, 72, 73].

Immunohistochemical labeling

Validation of differentially expressed proteins in TE co-infected with HIV-1 was carried out by immunohistochemical staining for 15 toxoplasma encephalitis cases and 6 controls using commercially available antibodies. Formalin fixed and paraffin embedded autopsy tissues were collected and cut into 5 μm thick sections on glass slides. These slides were subjected for deparaffinization and rehydration. Endogenous peroxidase activity was quenched by 3% H2O2 for 20 min at room temperature. Heat induced antigen retrieval was carried out using pressure cooker by placing the tissue sections in citrate buffer (pH 6.0). The tissue sections were incubated with 3% skimmed milk in PBS, pH 7.4 at room temperature followed by incubation with primary antibodies at following dilutions – anti-major histocompatibility complex, class I, B (dilution 1:200, cat # sc-55582, Santa Cruz), anti-alpha-1-antitrypsin (dilution 1:2000, cat # HPA00927-100 μl, Sigma-Aldrich), anti-rho-related GTP-binding protein RhoG (dilution 1:50, cat # HPA039871-100UL, Sigma-Aldrich) for two hours at room temperature. In parallel with test slides, appropriate negative and positive controls were also subjected for incubation at room temperature, followed by incubation with pre-diluted secondary antibody conjugated with poly HRP (catalog # QD630-XAKE) from BioGenex. The reaction was visualized with chromogen substrate DAB/H2O2 as per manufacturer’s instructions. The sections were counterstained with hematoxylin. The immunolabeled sections were examined and staining pattern, intensity, subcellular localization were visually scored by two expert neuropathologists - AM and SKS (NIMHANS).

Data availability

We submitted the mass spectrometry data to Human Proteinpedia to make it available to the public [13]. The study design, experimental details, list of proteins and peptides identified in the study can be accessed using the following URL: http://www.humanproteinpedia.org/data_display?exp_id=00707. The mass spectrometry proteomics data have also been deposited to the ProteomeXchange Consortium [14] via the PRIDE partner repository with the dataset identifier PXD000261.

Abbreviations

CT:

Computed tomography

CSF:

Cerebrospinal fluid

CNS:

Central nervous system

FDR:

False discovery rate

iTRAQ:

Isobaric tag for relative and absolute quantitation

IHC:

Immunohistochemistry

HPLC:

High performance liquid chromatography

HIV:

Human immunodeficiency virus

MRI:

Magnetic resonance imaging

PCR:

Polymerase chain reaction

SCX:

Strong cation exchange

TE:

Toxoplasma encephalitis.

References

  1. Tan IL, Smith BR, von Geldern G, Mateen FJ, McArthur JC: HIV-associated opportunistic infections of the CNS. Lancet Neurol. 2012, 11: 605-617. 10.1016/S1474-4422(12)70098-4

    Article  PubMed  Google Scholar 

  2. Hakko E, Ozkan HA, Karaman K, Gulbas Z: Analysis of cerebral toxoplasmosis in a series of 170 allogeneic hematopoietic stem cell transplant patients. Transpl Infect Dis. 2013, 15: 575-580. 10.1111/tid.12138

    Article  CAS  PubMed  Google Scholar 

  3. Mittal V, Ichhpujani RL: Toxoplasmosis - an update. Trop Parasitol. 2011, 1: 9-14. 10.4103/2229-5070.72109

    Article  PubMed Central  PubMed  Google Scholar 

  4. Kumar GG, Mahadevan A, Guruprasad AS, Kovoor JM, Satishchandra P, Nath A, Ranga U, Shankar SK: Eccentric target sign in cerebral toxoplasmosis: neuropathological correlate to the imaging feature. J Magn Reson Imaging. 2010, 31: 1469-1472. 10.1002/jmri.22192

    Article  PubMed Central  PubMed  Google Scholar 

  5. Bhattacharyya S, Khurana S, Dubey ML: Anti-Toxoplasma gondii antibody detection in serum and urine samples by enzyme-linked immunosorbent assay in HIV-infected patients. Indian J Pathol Microbiol. 2013, 56: 20-23. 10.4103/0377-4929.116143

    Article  PubMed  Google Scholar 

  6. Cao L, Cheng R, Yao L, Yuan S, Yao X: Establishment and application of a loop-mediated isothermal amplification method for simple, specific, sensitive, and rapid detection of toxoplasma gondii. J Vet Med Sci. 2013, 76: 9-14.

    Article  PubMed Central  PubMed  Google Scholar 

  7. Montoya JG, Liesenfeld O: Toxoplasmosis. Lancet. 2004, 363: 1965-1976. 10.1016/S0140-6736(04)16412-X

    Article  CAS  PubMed  Google Scholar 

  8. Feustel SM, Meissner M, Liesenfeld O: Toxoplasma gondii and the blood-brain barrier. Virulence. 2012, 3: 182-192. 10.4161/viru.19004

    Article  PubMed Central  PubMed  Google Scholar 

  9. Suzuki Y: Host resistance in the brain against Toxoplasma gondii. J Infect Dis. 2002, 185 (Suppl 1): S58-S65.

    Article  CAS  PubMed  Google Scholar 

  10. Wang X, Michie SA, Xu B, Suzuki Y: Importance of IFN-gamma-mediated expression of endothelial VCAM-1 on recruitment of CD8+ T cells into the brain during chronic infection with Toxoplasma gondii. J Interferon Cytokine Res. 2007, 27: 329-338. 10.1089/jir.2006.0154

    Article  PubMed  Google Scholar 

  11. Sullivan WJ, Jeffers V: Mechanisms of Toxoplasma gondii persistence and latency. FEMS Microbiol Rev. 2012, 36: 717-733. 10.1111/j.1574-6976.2011.00305.x

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  12. Andenmatten N, Egarter S, Jackson AJ, Jullien N, Herman JP, Meissner M: Conditional genome engineering in Toxoplasma gondii uncovers alternative invasion mechanisms. Nat Methods. 2013, 10: 125-127.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  13. Kandasamy K, Keerthikumar S, Goel R, Mathivanan S, Patankar N, Shafreen B, Renuse S, Pawar H, Ramachandra YL, Acharya PK, Ranganathan P, Chaerkady R, Prasad TSK, Pandey A: Human Proteinpedia: a unified discovery resource for proteomics research. Nucleic Acids Res. 2009, 37: D773-D781. 10.1093/nar/gkn701

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  14. Vizcaino JA, Cote RG, Csordas A, Dianes JA, Fabregat A, Foster JM, Griss J, Alpi E, Birim M, Contell J, O’Kelly G, Schoenegger A, Ovelleiro D, Perez-Riverol Y, Reisinger F, Rios D, Wang R, Hermjakob H: The PRoteomics IDEntifications (PRIDE) database and associated tools: status in 2013. Nucleic Acids Res. 2013, 41: D1063-D1069. 10.1093/nar/gks1262

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Access guide to human proteinpedia. Current Protocols in Bioinformatics. 2013, Chapter 1: Unit 1 21, John Wiley & Sons, Inc,

  16. Goel R, Harsha HC, Pandey A, Prasad TSK: Human protein reference database and human proteinpedia as resources for phosphoproteome analysis. Mol Biosyst. 2012, 8: 453-463. 10.1039/c1mb05340j

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  17. Sandhya VK, Raju R, Verma R, Advani J, Sharma R, Radhakrishnan A, Nanjappa V, Narayana J, Somani BL, Mukherjee KK, Pandey A, Christopher R, Prasad TSK: A network map of BDNF/TRKB and BDNF/p75NTR signaling system. J Cell Commun Signal. 2013, 7: 301-307. 10.1007/s12079-013-0200-z

    Article  PubMed Central  PubMed  Google Scholar 

  18. Namekata K, Watanabe H, Guo X, Kittaka D, Kawamura K, Kimura A, Harada C, Harada T: Dock3 regulates BDNF-TrkB signaling for neurite outgrowth by forming a ternary complex with Elmo and RhoG. Genes Cells. 2012, 17: 688-697. 10.1111/j.1365-2443.2012.01616.x

    Article  CAS  PubMed  Google Scholar 

  19. Bachis A, Avdoshina V, Zecca L, Parsadanian M, Mocchetti I: Human immunodeficiency virus type 1 alters brain-derived neurotrophic factor processing in neurons. J Neurosci. 2012, 32: 9477-9484. 10.1523/JNEUROSCI.0865-12.2012

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  20. Hamby ME, Coppola G, Ao Y, Geschwind DH, Khakh BS, Sofroniew MV: Inflammatory mediators alter the astrocyte transcriptome and calcium signaling elicited by multiple G-protein-coupled receptors. J Neurosci. 2012, 32: 14489-14510. 10.1523/JNEUROSCI.1256-12.2012

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  21. Habegger de Sorrentino A, Lopez R, Motta P, Marinic K, Sorrentino A, Iliovich E, Rubio AE, Quarleri J, Salomon H: HLA class II involvement in HIV-associated Toxoplasmic encephalitis development. Clin Immunol. 2005, 115: 133-137. 10.1016/j.clim.2005.01.003

    Article  CAS  PubMed  Google Scholar 

  22. Pae CU, Drago A, Kim JJ, Mandelli L, De Ronchi D, Serretti A: The impact of heat shock protein 70 gene variations on clinical presentation and outcome in schizophrenic inpatients. Neuropsychobiology. 2009, 59: 135-141. 10.1159/000218075

    Article  CAS  PubMed  Google Scholar 

  23. Pae CU, Kim TS, Kwon OJ, Artioli P, Serretti A, Lee CU, Lee SJ, Lee C, Paik IH, Kim JJ: Polymorphisms of heat shock protein 70 gene (HSPA1A, HSPA1B and HSPA1L) and schizophrenia. Neurosci Res. 2005, 53: 8-13. 10.1016/j.neures.2005.05.004

    Article  CAS  PubMed  Google Scholar 

  24. Scheele S, Nystrom A, Durbeej M, Talts JF, Ekblom M, Ekblom P: Laminin isoforms in development and disease. J Mol Med (Berl). 2007, 85: 825-836. 10.1007/s00109-007-0182-5

    Article  CAS  Google Scholar 

  25. Schneider AG, Abi Abdallah DS, Butcher BA, Denkers EY: Toxoplasma gondii triggers phosphorylation and nuclear translocation of dendritic cell STAT1 while simultaneously blocking IFNγ-induced STAT1 transcriptional activity. PLoS One. 2013, 8: e60215- 10.1371/journal.pone.0060215

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. Collazo CM, Yap GS, Hieny S, Caspar P, Feng CG, Taylor GA, Sher A: The function of gamma interferon-inducible GTP-binding protein IGTP in host resistance to Toxoplasma gondii is Stat1 dependent and requires expression in both hematopoietic and nonhematopoietic cellular compartments. Infect Immun. 2002, 70: 6933-6939. 10.1128/IAI.70.12.6933-6939.2002

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  27. Derouin F, Leport C, Pueyo S, Morlat P, Letrillart B, Chene G, Ecobichon JL, Luft B, Aubertin J, Hafner R, Vildé JL, Salamon R: Predictive value of Toxoplasma gondii antibody titres on the occurrence of toxoplasmic encephalitis in HIV-infected patients. ANRS 005/ACTG 154 Trial Group. AIDS. 1996, 10: 1521-1527. 10.1097/00002030-199611000-00010

    Article  CAS  PubMed  Google Scholar 

  28. Sarciron ME, Gherardi A: Cytokines involved in Toxoplasmic encephalitis. Scand J Immunol. 2000, 52: 534-543. 10.1046/j.1365-3083.2000.00817.x

    Article  CAS  PubMed  Google Scholar 

  29. Alfonzo M, Blanc D, Troadec C, Huerre M, Eliaszewicz M, Gonzalez G, Koyanagi Y, Scott-Algara D: Temporary restoration of immune response against Toxoplasma gondii in HIV-infected individuals after HAART, as studied in the hu-PBMC-SCID mouse model. Clin Exp Immunol. 2002, 129: 411-419. 10.1046/j.1365-2249.2002.01941.x

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  30. Lejeune M, Miró JM, De Lazzari E, García F, Claramonte X, Martínez E, Ribera E, Arrizabalaga J, Arribas JR, Domingo P, Ferrer E, Plana M, Valls ME, Podzamczer D, Pumarola T, Jacquet A, Mallolas J, Gatell JM, Gallart T, : Restoration of T cell responses to toxoplasma gondii after successful combined antiretroviral therapy in patients with AIDS with previous toxoplasmic encephalitis. Clin Infect Dis. 2011, 52: 662-670. 10.1093/cid/ciq197

    Article  CAS  PubMed  Google Scholar 

  31. Chitra P, Bakthavatsalam B, Palvannan T: Beta-2 microglobulin as an immunological marker to assess the progression of human immunodeficiency virus infected patients on highly active antiretroviral therapy. Clin Chim Acta. 2011, 412: 1151-1154. 10.1016/j.cca.2011.01.037

    Article  CAS  PubMed  Google Scholar 

  32. Robert-Gangneux F, Gangneux JP, Vu N, Jaillard S, Guiguen C, Amiot L: High level of soluble HLA-G in amniotic fluid is correlated with congenital transmission of Toxoplasma gondii. Clin Immunol. 2011, 138: 129-134. 10.1016/j.clim.2010.12.004

    Article  CAS  PubMed  Google Scholar 

  33. Kim SY, Li J, Bentsman G, Brooks AI, Volsky DJ: Microarray analysis of changes in cellular gene expression induced by productive infection of primary human astrocytes: implications for HAD. J Neuroimmunol. 2004, 157: 17-26. 10.1016/j.jneuroim.2004.08.032

    Article  CAS  PubMed  Google Scholar 

  34. Winkler JM, Chaudhuri AD, Fox HS: Translating the brain transcriptome in neuroAIDS: from non-human primates to humans. J Neuroimmune Pharmacol. 2012, 7: 372-379. 10.1007/s11481-012-9344-5

    Article  PubMed Central  PubMed  Google Scholar 

  35. Malemud CJ, Miller AH: Pro-inflammatory cytokine-induced SAPK/MAPK and JAK/STAT in rheumatoid arthritis and the new anti-depression drugs. Expert Opin Ther Targets. 2008, 12: 171-183. 10.1517/14728222.12.2.171

    Article  CAS  PubMed  Google Scholar 

  36. Denkers EY, Bzik DJ, Fox BA, Butcher BA: An inside job: hacking into Janus kinase/signal transducer and activator of transcription signaling cascades by the intracellular protozoan Toxoplasma gondii. Infect Immun. 2012, 80: 476-482. 10.1128/IAI.05974-11

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  37. Henriquez SA, Brett R, Alexander J, Pratt J, Roberts CW: Neuropsychiatric disease and Toxoplasma gondii infection. Neuroimmunomodulation. 2009, 16: 122-133. 10.1159/000180267

    Article  CAS  PubMed  Google Scholar 

  38. Liu YL, Fann CS, Liu CM, Chen WJ, Wu JY, Hung SI, Chen CH, Jou YS, Liu SK, Hwang TJ, Hsieh MH, Chang CC, Yang WC, Lin JJ, Chou FH, Faraone SV, Tsuang MT, Hwu HG: RASD2, MYH9, and CACNG2 genes at chromosome 22q12 associated with the subgroup of schizophrenia with non-deficit in sustained attention and executive function. Biol Psychiatry. 2008, 64: 789-796. 10.1016/j.biopsych.2008.04.035

    Article  CAS  PubMed  Google Scholar 

  39. Yang HC, Liu CM, Liu YL, Chen CW, Chang CC, Fann CS, Chiou JJ, Yang UC, Chen CH, Faraone SV, Tsuang MT, Hwu HG: The DAO gene is associated with schizophrenia and interacts with other genes in the Taiwan Han Chinese population. PLoS One. 2013, 8: e60099- 10.1371/journal.pone.0060099

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  40. Johnson JN, Ahrendt E, Braun JE: CSPalpha: the neuroprotective J protein. Biochem Cell Biol. 2010, 88: 157-165. 10.1139/O09-124

    Article  CAS  PubMed  Google Scholar 

  41. Fatemi SH, Folsom TD, Thuras PD: Deficits in GABA(B) receptor system in schizophrenia and mood disorders: a postmortem study. Schizophr Res. 2011, 128: 37-43. 10.1016/j.schres.2010.12.025

    Article  PubMed Central  PubMed  Google Scholar 

  42. Bouthour W, Leroy F, Emmanuelli C, Carnaud M, Dahan M, Poncer JC, Levi S: A human mutation in Gabrg2 associated with generalized epilepsy alters the membrane dynamics of GABAA receptors. Cereb Cortex. 2012, 22: 1542-1553. 10.1093/cercor/bhr225

    Article  PubMed  Google Scholar 

  43. Allen AS, Berkovic SF, Cossette P, Delanty N, Dlugos D, Eichler EE, Epstein MP, Glauser T, Goldstein DB, Han Y, Heinzen EL, Hitomi Y, Howell KB, Johnson MR, Kuzniecky R, Lowenstein DH, Lu YF, Madou MR, Marson AG, Mefford HC, Esmaeeli Nieh S, O'Brien TJ, Ottman R, Petrovski S, Poduri A, Ruzzo EK, Scheffer IE, Sherr EH, Yuskaitis CJ, : De novo mutations in epileptic encephalopathies. Nature. 2013, 501: 217-221. 10.1038/nature12439

    Article  CAS  PubMed  Google Scholar 

  44. Eugene E, Depienne C, Baulac S, Baulac M, Fritschy JM, Le Guern E, Miles R, Poncer JC: GABA(A) receptor gamma 2 subunit mutations linked to human epileptic syndromes differentially affect phasic and tonic inhibition. J Neurosci. 2007, 27: 14108-14116. 10.1523/JNEUROSCI.2618-07.2007

    Article  CAS  PubMed  Google Scholar 

  45. Magri C, Gardella R, Valsecchi P, Barlati SD, Guizzetti L, Imperadori L, Bonvicini C, Tura GB, Gennarelli M, Sacchetti E, Barlati S: Study on GRIA2, GRIA3 and GRIA4 genes highlights a positive association between schizophrenia and GRIA3 in female patients. Am J Med Genet B Neuropsychiatr Genet. 2008, 147B: 745-753. 10.1002/ajmg.b.30674

    Article  CAS  PubMed  Google Scholar 

  46. Fergusson DM, Boden JM, Horwood LJ, Miller AL, Kennedy MA: MAOA, abuse exposure and antisocial behaviour: 30-year longitudinal study. Br J Psychiatry. 2011, 198: 457-463. 10.1192/bjp.bp.110.086991

    Article  PubMed Central  PubMed  Google Scholar 

  47. Eastwood SL, Harrison PJ: Decreased expression of vesicular glutamate transporter 1 and complexin II mRNAs in schizophrenia: further evidence for a synaptic pathology affecting glutamate neurons. Schizophr Res. 2005, 73: 159-172. 10.1016/j.schres.2004.05.010

    Article  CAS  PubMed  Google Scholar 

  48. Tropeano M, Wöber-Bingöl C, Karwautz A, Wagner G, Vassos E, Campos-de-Sousa S, Graggaber A, Zesch HE, Kienbacher C, Natriashvili S, Kanbur I, Wöber C, Collier DA: Association analysis of STX1A gene variants in common forms of migraine. Cephalalgia. 2012, 32: 203-212. 10.1177/0333102411433300

    Article  PubMed  Google Scholar 

  49. Depienne C, Gourfinkel-An I, Baulac S, LeGuern E: Genes in infantile epileptic encephalopathies. Jasper’s Basic Mechanisms of the Epilepsies. Edited by: Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV. 2012, Bethesda (MD): National Center for Biotechnology Information (US), 4,

    Google Scholar 

  50. Vatta M, Tennison MB, Aylsworth AS, Turcott CM, Guerra MP, Eng CM, Yang Y: A novel STXBP1 mutation causes focal seizures with neonatal onset. J Child Neurol. 2012, 27: 811-814. 10.1177/0883073811435246

    Article  PubMed  Google Scholar 

  51. Greco B, Manago F, Tucci V, Kao HT, Valtorta F, Benfenati F: Autism-related behavioral abnormalities in synapsin knockout mice. Behav Brain Res. 2013, 251: 65-74.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  52. Fernández-Chacón R, Königstorfer A, Gerber SH, García J, Matos MF, Stevens CF, Brose N, Rizo J, Rosenmund C, Südhof TC: Synaptotagmin I functions as a calcium regulator of release probability. Nature. 2001, 410: 41-49. 10.1038/35065004

    Article  PubMed  Google Scholar 

  53. Mortensen PB, Norgaard-Pedersen B, Waltoft BL, Sorensen TL, Hougaard D, Yolken RH: Early infections of Toxoplasma gondii and the later development of schizophrenia. Schizophr Bull. 2007, 33: 741-744. 10.1093/schbul/sbm009

    Article  PubMed Central  PubMed  Google Scholar 

  54. Schwarcz R, Hunter CA: Toxoplasma gondii and schizophrenia: linkage through astrocyte-derived kynurenic acid?. Schizophr Bull. 2007, 33: 652-653. 10.1093/schbul/sbm030

    Article  PubMed Central  PubMed  Google Scholar 

  55. Dzierszinski F, Pepper M, Stumhofer JS, LaRosa DF, Wilson EH, Turka LA, Halonen SK, Hunter CA, Roos DS: Presentation of Toxoplasma gondii antigens via the endogenous major histocompatibility complex class I pathway in nonprofessional and professional antigen-presenting cells. Infect Immun. 2007, 75: 5200-5209. 10.1128/IAI.00954-07

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  56. Schluter D, Lohler J, Deckert M, Hof H, Schwendemann G: Toxoplasma encephalitis of immunocompetent and nude mice: immunohistochemical characterisation of Toxoplasma antigen, infiltrates and major histocompatibility complex gene products. J Neuroimmunol. 1991, 31: 185-198. 10.1016/0165-5728(91)90040-E

    Article  CAS  PubMed  Google Scholar 

  57. Schmechel DE, Edwards CL: Fibromyalgia, mood disorders, and intense creative energy: A1AT polymorphisms are not always silent. Neurotoxicology. 2012, 33: 1454-1472. 10.1016/j.neuro.2012.03.001

    Article  CAS  PubMed  Google Scholar 

  58. Jesse S, Lehnert S, Jahn O, Parnetti L, Soininen H, Herukka S-K, Steinacker P, Tawfik S, Tumani H, von Arnim CAF, Neumann M, Kretzschmar HA, Kulaksiz H, Lenter M, Wiltfang J, Ferger B, Hengerer B, Otto M: Differential sialylation of serpin A1 in the early diagnosis of Parkinson’s disease dementia. PLoS One. 2012, 7: e48783- 10.1371/journal.pone.0048783

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  59. Maes OC, Kravitz S, Mawal Y, Su H, Liberman A, Mehindate K, Berlin D, Sahlas DJ, Chertkow HM, Bergman H, Melmed C, Schipper HM: Characterization of alpha1-antitrypsin as a heme oxygenase-1 suppressor in Alzheimer plasma. Neurobiol Dis. 2006, 24: 89-100. 10.1016/j.nbd.2006.06.009

    Article  CAS  PubMed  Google Scholar 

  60. Yang Y, Wan C, Li H, Zhu H, La Y, Xi Z, Chen Y, Jiang L, Feng G, He L: Altered levels of acute phase proteins in the plasma of patients with schizophrenia. Anal Chem. 2006, 78: 3571-3576. 10.1021/ac051916x

    Article  CAS  PubMed  Google Scholar 

  61. Lee TW, Montgomery JM, Birch NP: The serine protease inhibitor neuroserpin regulates the growth and maturation of hippocampal neurons through a non-inhibitory mechanism. J Neurochem. 2012, 121: 561-574. 10.1111/j.1471-4159.2011.07639.x

    Article  CAS  PubMed  Google Scholar 

  62. Tamminga CA, Stan AD, Wagner AD: The hippocampal formation in schizophrenia. Am J Psychiatry. 2010, 167: 1178-1193. 10.1176/appi.ajp.2010.09081187

    Article  PubMed  Google Scholar 

  63. Cordelier P, Strayer DS: Mechanisms of alpha1-antitrypsin inhibition of cellular serine proteases and HIV-1 protease that are essential for HIV-1 morphogenesis. Biochim Biophys Acta. 2003, 1638: 197-207. 10.1016/S0925-4439(03)00084-X

    Article  CAS  PubMed  Google Scholar 

  64. van Buul JD, Allingham MJ, Samson T, Meller J, Boulter E, Garcia-Mata R, Burridge K: RhoG regulates endothelial apical cup assembly downstream from ICAM1 engagement and is involved in leukocyte trans-endothelial migration. J Cell Biol. 2007, 178: 1279-1293. 10.1083/jcb.200612053

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  65. Yamaki N, Negishi M, Katoh H: RhoG regulates anoikis through a phosphatidylinositol 3-kinase-dependent mechanism. Exp Cell Res. 2007, 313: 2821-2832. 10.1016/j.yexcr.2007.05.010

    Article  CAS  PubMed  Google Scholar 

  66. Roppenser B, Roder A, Hentschke M, Ruckdeschel K, Aepfelbacher M: Yersinia enterocolitica differentially modulates RhoG activity in host cells. J Cell Sci. 2009, 122: 696-705. 10.1242/jcs.040345

    Article  CAS  PubMed  Google Scholar 

  67. Ishikawa Y, Katoh H, Nakamura K, Mori K, Negishi M: Developmental changes in expression of small GTPase RhoG mRNA in the rat brain. Brain Res Mol Brain Res. 2002, 106: 145-150. 10.1016/S0169-328X(02)00413-8

    Article  CAS  PubMed  Google Scholar 

  68. O’Kane EM, Stone TW, Morris BJ: Distribution of Rho family GTPases in the adult rat hippocampus and cerebellum. Brain Res Mol Brain Res. 2003, 114: 1-8. 10.1016/S0169-328X(03)00121-9

    Article  PubMed  Google Scholar 

  69. Fujimoto S, Negishi M, Katoh H: RhoG promotes neural progenitor cell proliferation in mouse cerebral cortex. Mol Biol Cell. 2009, 20: 4941-4950. 10.1091/mbc.E09-03-0200

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  70. Kim JY, Oh MH, Bernard LP, Macara IG, Zhang H: The RhoG/ELMO1/Dock180 signaling module is required for spine morphogenesis in hippocampal neurons. J Biol Chem. 2011, 286: 37615-37624. 10.1074/jbc.M111.268029

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  71. Kumar GS, Venugopal AK, Mahadevan A, Renuse S, Harsha HC, Sahasrabuddhe NA, Pawar H, Sharma R, Kumar P, Rajagopalan S, Waddell K, Ramachandra YL, Satishchandra P, Chaerkady R, Prasad TSK, Shankar K, Pandey A: Quantitative proteomics for identifying biomarkers for tuberculous meningitis. Clin Proteomics. 2012, 9: 12- 10.1186/1559-0275-9-12

    Article  PubMed Central  PubMed  Google Scholar 

  72. Puttamallesh VN, Sreenivasamurthy SK, Singh PK, Harsha HC, Ganjiwale A, Broor S, Pandey A, Narayana J, Prasad TSK: Proteomic profiling of serum samples from chikungunya-infected patients provides insights into host response. Clin Proteomics. 2013, 10: 14- 10.1186/1559-0275-10-14

    Article  PubMed Central  PubMed  Google Scholar 

  73. Kandasamy K, Mohan SS, Raju R, Keerthikumar S, Kumar GSS, Venugopal AK, Telikicherla D, Navarro JD, Mathivanan S, Pecquet C, Gollapudi SK, Tattikota SG, Mohan S, Padhukasahasram H, Subbannayya Y, Goel R, Jacob HKC, Zhong J, Sekhar R, Nanjappa V, Balakrishnan L, Subbaiah R, Ramachandra YL, Rahiman BA, Prasad TSK, Lin J-X, Houtman JCD, Desiderio S, Renauld J-C, Constantinescu SN, et al: NetPath: a public resource of curated signal transduction pathways. Genome Biol. 2010, 11: R3- 10.1186/gb-2010-11-1-r3

    Article  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

The study was supported by a research grant “DBT Programme Support on Neuroproteomics of Neurological Disorders” to IOB and NIMHANS by DBT, Government of India (BT/01/COE/08/05). Human brain tissues for the study were obtained from Human Brain Tissue Repository, a National Research Facility, Department of Neuropathology, NIMHANS, Bangalore, India. This publication was partly supported by a sub-award from The Johns Hopkins University, with funds provided from National Institute of Neurological Disorders and Stroke (NINDS) (Grant Number: 1RO1NS055628). The contents of the study are solely the responsibility of the authors and do not represent the official views of NINDS or JHU. We acknowledge Dr. G. Venkatasubramanian, Department of Psychiatry, NIMHANS, for his valuable input. Apeksha Sahu, Sreelakshmi K. Sreenivasamurthy and Gourav Dey are recipients of Junior Research Fellowship from the University Grants Commission (UGC), Government of India. Soujanya D. Yelamanchi is a recipient of INSPIRE Fellowship from the Department of Science and Technology (DST), Government of India. Anil K. Madugundu is the recipient of BINC-Junior Research Fellowship from DBT. Harsha Gowda is a Wellcome Trust-DBT India Alliance Early Career Fellow.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Akhilesh Pandey, Thottethodi Subrahmanya Keshava Prasad or Susarla Krishna Shankar.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

HG, PS, AP, AM, TSKP and SKS (NIMHANS) conceived and planned the study. PS, SKS (NIMHANS) and AM obtained frontal lobe brain tissue samples for this study. AS, SKS (IOB) and LDNL carried out sample preparation, fractionation and labeling. VNP carried out mass spectrometry analysis. AS, SK, LDNL and AKM analyzed mass spectrometry derived data. AS performed IHC validation; AM and SKS (NIMHANS) analyzed the IHC results; GD and AS prepared figures and tables. AS, SKS (IOB), SK, SDY, AKA and TSKP wrote the manuscript. AM, AP, HG, AS, PS, KKM and SKS (NIMHANS) provided critical inputs and revised the manuscript. All authors read and approved the final manuscript.

Electronic supplementary material

12014_2014_81_MOESM1_ESM.xlsx

Additional file 1: Table S1: A complete list of proteins along with peptides identified from toxoplasma encephalitis and control brain tissues. (XLSX 3 MB)

12014_2014_81_MOESM2_ESM.xlsx

Additional file 2: Table S2: Details of samples used for quantitative proteomic analysis of toxoplasma encephalitis. Table S3. Details of samples used for immunohistochemistry-based validation of differentially expressed molecules from quantitative proteomic analysis of toxoplasma encephalitis co-infected with HIV. (XLSX 13 KB)

Authors’ original submitted files for images

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sahu, A., Kumar, S., Sreenivasamurthy, S.K. et al. Host response profile of human brain proteome in toxoplasma encephalitis co-infected with HIV. Clin Proteom 11, 39 (2014). https://doi.org/10.1186/1559-0275-11-39

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1559-0275-11-39

Keywords