Next Article in Journal
The Mitochondrial m.3243A>G Mutation on the Dish, Lessons from In Vitro Models
Previous Article in Journal
Sodium Butyrate Induces Mitophagy and Apoptosis of Bovine Skeletal Muscle Satellite Cells through the Mammalian Target of Rapamycin Signaling Pathway
Previous Article in Special Issue
Long COVID as a Tauopathy: Of “Brain Fog” and “Fusogen Storms”
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Blood Proteome Profiling Reveals Biomarkers and Pathway Alterations in Fragile X PM at Risk for Developing FXTAS

1
Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
2
Genome Center, Bioinformatics Core, University of California Davis, Davis, CA 95616, USA
3
Genome Center, Proteomics Core, Genome and Biomedical Sciences Facility, University of California Davis, Davis, CA 95616, USA
4
Division of Biostatistics, School of Medicine, University of California Davis, Davis, CA 95616, USA
5
MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA
6
Department of Pediatrics, University of California Davis Medical Center, Sacramento, CA 95817, USA
7
Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, Sacramento, CA 95817, USA
8
Department of Psychology, University of California Davis, Davis, CA 95616, USA
9
Department of Psychology, University of Maryland, College Park, MD 20742, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(17), 13477; https://doi.org/10.3390/ijms241713477
Submission received: 15 July 2023 / Revised: 18 August 2023 / Accepted: 21 August 2023 / Published: 30 August 2023
(This article belongs to the Special Issue Genomics in Neurodegenerative Diseases)

Abstract

:
Fragile X-associated Tremor/Ataxia Syndrome (FXTAS) is a neurodegenerative disorder associated with the FMR1 premutation. Currently, it is not possible to determine when and if individual premutation carriers will develop FXTAS. Thus, with the aim to identify biomarkers for early diagnosis, development, and progression of FXTAS, along with associated dysregulated pathways, we performed blood proteomic profiling of premutation carriers (PM) who, as part of an ongoing longitudinal study, emerged into two distinct groups: those who developed symptoms of FXTAS (converters, CON) over time (at subsequent visits) and those who did not (non-converters, NCON). We compared these groups to age-matched healthy controls (HC). We assessed CGG repeat allele size by Southern blot and PCR analysis. The proteomic profile was obtained by liquid chromatography mass spectrometry (LC-MS/MS). We identified several significantly differentiated proteins between HC and the PM groups at Visit 1 (V1), Visit 2 (V2), and between the visits. We further reported the dysregulated protein pathways, including sphingolipid and amino acid metabolism. Our findings are in agreement with previous studies showing that pathways involved in mitochondrial bioenergetics, as observed in other neurodegenerative disorders, are significantly altered and appear to contribute to the development of FXTAS. Lastly, we compared the blood proteome of the PM who developed FXTAS over time with the CSF proteome of the FXTAS patients recently reported and found eight significantly differentially expressed proteins in common. To our knowledge, this is the first report of longitudinal proteomic profiling and the identification of unique biomarkers and dysregulated protein pathways in FXTAS.

1. Introduction

The prevalence of various neurodegenerative diseases, such as Alzheimer’s dementia and Parkinson’s disease, has risen in recent years among many populations due to the increase in the aging population. Developing effective treatments for these complex disorders is challenging due to the complex underlying molecular mechanisms involved, the lack of biomarkers for early diagnosis, the broad spectrum of symptoms, limited natural history, data, and the difficulty in conducting clinical trials with small patient populations. Identifying biomarkers and changes in the associated pathways, particularly in assays, that can quickly and objectively indicate changes in disease pathology is crucial for improving patient outcomes.
Fragile X-associated Tremor/Ataxia Syndrome (FXTAS) is a late-onset neurodegenerative disorder with an average age of onset of 62 that affects carriers of a premutation (PM) allele (55–200 CGG repeats) in the fragile X messenger ribonucleoprotein 1 (FMR1) gene, usually presenting with a more severe clinical phenotype in males, likely due to the presence of a second X chromosome in females [1,2]. The high prevalence of the premutation allele among the general population (1:430 males and 1:110–200 females) leads to an estimate of approximately 1.5 million individuals in the general US population who are at risk for FMR1 premutation associated disorders over their life spans. In addition, among the PM population, an estimated 8–16% of females and 40–60% of males are at risk of developing FXTAS [2,3].
Currently, there is no effective specific treatment for FXTAS, and the motor/cognitive symptoms progressively worsen over time, causing reduced quality of life, increased medical expenses, and eventually premature death. FXTAS is clinically distinguished by the presence of intention tremor, cerebellar ataxia, global brain atrophy and white matter disease, autonomic dysfunction, progressive Parkinsonism, and ubiquitin-positive intranuclear inclusions in brain astrocytes, neurons, and Purkinje cells [4]. It is caused by the expanded CGG repeats (55–200 CGG) in the 5′UTR of the FMR1 gene. In those with the normal FMR1 gene, the number of CGG repeats lies between 5 and 44, while individuals carrying alleles with a repeat expansion greater than 200 develop fragile X syndrome (FXS), the most common form of intellectual disability and monogenic cause of autism spectrum disorder (ASD) [5]. At the molecular level, the eight- to tenfold increase in the level of FMR1 mRNA in a PM containing the expanded CGG repeats [6] leads to RNA toxicity and ultimately to neurodegeneration. Three main mechanisms have been proposed to explain the pathogenesis of FXTAS, including the sequestration of CGG-binding proteins amplified by the elevated levels of FMR1 mRNA, the production of toxic FMRPolyG proteins due to RAN translation, and the chronic activation of the DNA damage response [7,8].
Mass spectrometry (MS)-based proteomics, which involves the advance of data mining and bioinformatic analysis to examine protein structure and function, can be used as an effective technology to quickly analyze large amounts of clinical and biological information within a given sample [9]. Recent advances in proteomic profiling technology and processing have also made it possible to efficiently analyze hundreds of proteins, precisely obtain a snapshot of the altered pathways in an organism and identify biomarkers for disease development and progression [10]. Although these MS-based proteomic workflows for biomarker discovery and profiling are well established, studies focused on blood proteome profiling and, importantly, on samples collected at different time points have not been carried out in PM at risk of FXTAS.
Recently, Ma and colleagues (2019) performed LC-MS/MS-based proteomics of the intranuclear inclusion isolated from postmortem FXTAS brain tissue. Their work highlighted the presence of more than 200 proteins within the inclusions, including a high abundance of SUMO2 and p62/sequestosome-1 (p62/SQSTM1), supporting a model where the inclusion formation results from increased protein loads and elevated oxidative stress [11]. Later, based on these observations, a proteomic profile was characterized in the FXTAS cortex as compared to those obtained from healthy controls (HC) [12]. Specifically, a significant decrease in the abundance of proteins including tenascin-C (TNC), cluster of differentiation 38 (CD38), and phosphoserine aminotransferase 1 (PSAT1) was observed in these samples. In addition, the authors confirmed the significantly high abundance of novel neurodegeneration-related proteins and of the small ubiquitin-like modifier 1/2 (SUMO1/2) in the FXTAS cortex as compared to HC [12]. Finally, a recent study reported changes in the levels of many proteins, including amyloid-like protein 2, contactin-1, afamin, cell adhesion molecule 4, NPC intracellular cholesterol transporter 2, and cathepsin, by comparing the cerebrospinal fluid (CSF) proteome of FXTAS patients with the CSF of HC patients. Changes in acute phase response signaling, liver X receptor/retinoid X receptor (LXR/RXR) activation, and farnesoid X receptor (FXR)/RXR activation pathways were observed [13].
Importantly, no study evaluating predictive biomarkers by blood proteomic alterations in PM, who developed symptoms of FXTAS over time has been reported to date. Here, we present our findings on global profiling derived from male participants enrolled in an ongoing longitudinal study carried out at the UC Davis MIND Institute. The participants have been followed for at least two longitudinal time points: Visit 1 (V1) and Visit 2 (V2). At each time point, neuroimaging, neuropsychological, and molecular measurements, as well as medical and neurological examinations, were collected. A fraction of the premutation participants, all symptom-free or not meeting criteria for FXTAS diagnosis at the time of enrollment (V1), developed symptoms later on (V2) that warranted a diagnosis of FXTAS during the study and were defined as converters (CON). The remaining premutation participants who did not develop symptoms that warranted a diagnosis of FXTAS by the time of the follow-up visit at (V2) are here defined as non-converters (NCON). In the current work, we performed the blood proteome profiling of PM, including CON and NCON, at both V1 and V2 and compared it to HC. We identified a number of potential predictive proteomic biomarkers for early diagnosis, as they showed significant changes in expression levels over time only in the converter group, and we also reported the altered protein pathways among the groups, suggesting their role in the pathogenies of the disorder.

2. Results

2.1. Demographics

DNA testing confirmed the presence of a premutation allele in the PM group, with the participants who converted at V2 (CON; n = 17) and PM who did not convert at V2 (NCON; n = 19), and the absence of a premutation allele in the healthy control (HC; n = 12) group. Participant ethnicity did not differ significantly between the three groups. CGG repeat numbers were significantly lower in healthy controls compared to the other two groups (p < 0.001 in both comparisons) but not significantly different between CON and NCON. Healthy controls were significantly younger than non-converters (p = 0.0319), as reported in Table 1.

2.2. Differential Protein Expression between Healthy Control and Premutation Groups

To identify biomarkers potentially associated with the development and progression of FXTAS, we compared the blood proteomic profile of HC to the PM, including CON and NCON. The groups display a separation trend, as shown in Figure 1. A sparse partial least squares discriminant analysis (s-PLSDA) was performed, which showed that all samples from each group aggregated, and the separation between groups indicated differences in the proteomic characteristics between PM and HC and between CON and NCON. A total of 79 proteins were identified by s-PLSDA analysis to be features that separate the groups. Out of these, 78 were among the list of significantly differentially expressed proteins in differential expression analysis using limma. Their expression profile is summarized in Table 2 and Figure 2.

2.3. Identification of Proteomic Biomarkers of FXTAS

From this untargeted proteomic profiling, we identified 227 proteins that showed significant changes in expression (adjusted p < 0.05) in pairwise comparisons of the CON as compared to the NCON at V1 (Table 3) and 196 proteins at V2 (Table 4). Between the CON and NCON, we observed 67 proteins that were consistently differentially expressed (adjusted p < 0.05) at V1 and kept changing at V2 (Table 5). While comparing the visits, we identified 170 differentially expressed (adjusted p < 0.05) proteins between V1 and V2 in the converter group, suggesting their role as biomarkers for the progression of FXTAS (Table 6).

2.4. Protein, Lipids, and Amino-Acid Pathways Altered in Individuals Who Developed FXTAS over Time

We further identified the pathways that are altered from V1 to V2 in CON and NCON, including protein lipids and amino acids. Upon examination of protein pathways that were altered between visits in NCON and CON (Figure 3), we found that pathways associated with cell signaling, immune function, cellular organization growth and proliferation, and inflammatory response were those that were more significantly altered from V1 to V2 in the CON group. Similarly, when investigating the protein pathways altered between NCON and CON at V1 or V2, we found that pathways related to synapse signaling (retrograde endocannabinoid signaling pathway) and lipid metabolism were more significantly altered between NCON and CON at V2 (Figure 4). Interestingly, when investigating the list of consistently differentially expressed proteins between CON and NCON groups at V1 and V2, we observed that the pathways related to neurodegeneration are ranked among the top enriched pathways, including the pathways of neurodegeneration, Huntington’s disease, and Alzheimer’s disease (Figure 5), which provides confidence that the potentially relevant biomarkers may be among these proteins. From the gene ontology point of view, the proteins that are consistently differentially expressed between CON and NCON at both visits are enriched in mitochondrial functions, protein synthesis machinery, and transport, as well as positive regulation of the BMP signaling pathway (Figure 6). These suggest the association of this list of proteins with FXTAS development, similar to other neurodegenerative disorders. Further, upon development of FXTAS at V2 (Figure 7), we observed a high level of dysregulation in retrograde endocannabinoid signaling pathways, mRNA surveillance pathways, cancer, cGMP-PKG signaling, calcium, sphingolipid, and lipid pathways, as observed in other neurodegenerative disorders such as Alzheimer’s disease, dementia, and Parkinsonism [14]. Further investigating the lipid and amino-acid metabolism, we detected various associated proteins that were differentially expressed in CON as compared to NCON at V2, suggesting their role in the progression of FXTAS (Figure 8).

2.5. Differentially Expressed Common Proteins Identified from CSF and Blood Proteomic Profiling

We compared the blood proteome profile of CON at V2 with the recently reported cerebrospinal fluid (CSF) proteome of FXTAS patients. The CSF proteome identified 414 proteins, out of which 46 were identified to be significantly altered between FXTAS patients and controls [13]. In the present study of the blood proteome, we identified a total of 2166 proteins, of which 97 were found to be common with the CSF proteome, and eight proteins were significantly altered in both studies, including Complement C3, Alpha-2-HS-glycoprotein, Pigment epithelium-derived factor, Inter-alpha-trypsin inhibitor heavy chain H2, Retinol-binding protein 4, Alpha-2-macroglobulin, Prothrombin, and Lumican (Figure 9).

3. Discussion

The identification of protein biomarkers and altered molecular pathways in FXTAS is a crucial requirement for both the research and clinical communities as it improves our ability to identify individuals most at risk for the disease as well as to create novel targeted therapies. There are multiple proteins and pathways that were found to be highly implicated in FXTAS. SUMO2 and p62/sequestosome-1 (p62/SQSTM1) proteins have been observed to accumulate in intranuclear inclusions isolated from postmortem FXTAS brain tissue [11], while tenascin-C (TNC), cluster of differentiation 38 (CD38), and phosphoserine aminotransferase 1 (PSAT1) have been observed in FXTAS cortex [12]. Furthermore, it is worth noting that previous studies have examined the proteomic profile of cerebrospinal fluid (CSF) in individuals with Fragile X-associated Tremor/Ataxia Syndrome (FXTAS), highlighting alterations in proteins and pathways when compared to healthy controls [13]. However, to the best of our knowledge, our study represents the first longitudinal investigation of blood proteomic changes specifically in PM, some of whom exhibit progressive symptoms of FXTAS over time. These findings provide valuable insights into the potential role of these proteomic alterations as biomarkers for early diagnosis, disease progression, and the overall development of FXTAS.
We observed a number of important proteins altered between HC and PM, including both CON and NCON (Table 2). Further, we found that a number of those proteins associated with various important pathways are dysregulated between CON and NCON at V1 (Table 3), V2 (Table 4), and even between visits (Table 5 and Table 6). Interestingly, most of these significantly dysregulated proteins are linked to essential pathways and reported to be involved in the development of other age-related neurodegenerative disorders like Alzheimer’s disease, dementia, and Parkinsonism.
In our previous study, we reported lipid and amino acid metabolism dysregulation along with mitochondrial dysfunction in individuals developing FXTAS over time. Specifically, we reported on the clear involvement of different types of lipids in FXTAS and provided evidence of the role that their dysregulation plays in the development and progression of FXTAS [15,16]. Specifically, we have identified altered sphingolipid metabolic pathways, including increased levels of sphingosine, sphinganine, and ceramides, in PM who developed FXTAS over time. Further, we reported on decreased levels of the hexosylceramides and lactosylceramides (LCER), both implicated in neuroinflammatory diseases and mitochondrial dysfunction [17,18], common features observed in FXTAS. In this study, we confirmed and validated the previous finding as we observed abrupted sphingolipids and amino acid metabolism (Figure 8) along with mitochondrial dysfunction in PM, including both CON and NCON at the protein level (Figure 6).
Indeed, proteomic profiles clearly show a different protein signature among the groups (CON vs. NCON at both V1 and V2), and enrichment pathway analysis demonstrates the involvement of key pathways, including lipids, mitochondria, neurodegeneration, and others, as illustrated in Figure 5, Figure 6, Figure 7 and Figure 8. Among these proteins, the cytochrome c oxidase subunit Va (COX5A) and the mitochondrial electron transport chain associated protein MT-CO2 were differentially expressed in the CON group (Table 6). As COX5A is involved in maintaining normal mitochondrial function and plays a vital role in aging-related cognitive deterioration via BDNF/ERK1/2 regulation [19], it could represent a potential target for anti-senescence drugs. The mitochondrially encoded cytochrome C oxidase II (MT-CO2) is located in the mitochondrial inner membrane is part of the respiratory chain complex IV, which is defective in individuals with FXTAS. It is a biomarker of Huntington’s disease [20] and associated with cerebellar ataxia and neuropathy [21], both clinical features observed in FXTAS. Further, a recent metabolomic study of patients with mitochondrial disease demonstrated elevated acylcarnitine levels, suggesting that an altered fatty acid oxidation pathway may represent a downstream mitochondrial respiratory chain dysfunction [22]. Interestingly, we reported high levels of plasma acylcarnitines in the CON group but not in the NCON group [15].
Neural degeneration is a key contributor to the development of neurodegenerative disorders, and we observed a differential expression of the VASP protein in CON. Downregulation of VASP leads to neuronal cell death through an apoptotic pathway and is implicated in the establishment and maintenance of the axonal structure; changes in the expression level can trigger neuronal degeneration [23]. In addition, we identified the RNA and mRNA protein pathway dysregulation in CON at V2 (Figure 7), including snRNA-associated Sm-like protein (LSm3), a critical activating factor for mRNA removal in eukaryotic cells participating in RNA metabolism, silencing, and degradation. Abnormal expression of LSM3 has been found to be associated with mild cognitive impairment (MCI) and Alzheimer’s disease (AD) [24].
In one of the recent studies, Abbasi and colleagues characterized the cerebrospinal fluid (CSF) proteome of FXTAS patients and reported 317 proteins, among which the expression levels of 38 were significantly altered between FXTAS patients and controls [13]. We looked at the overlap with our dataset of 2069 identified proteins and found 97 proteins in common, along with eight significantly altered proteins in both studies (Figure 9). Retinol-binding protein 4 (RBP4) is one of the 8 proteins altered in the CSF as well as in the blood of individuals who developed FXTAS over time. RBP4 is the sole specific transport protein for vitamin A (retinol), and it has been reported that RBP4 can directly induce retinal neurodegeneration in mice through microglia [25]. In the CON group, we also observed increased levels of the C3 protein, a key component of the complement cascade signaling pathway and of the immune system that plays a crucial role in inflammation and host defense [26]. Overactivation of C3 has also been reported in AD, leading to neuronal damage [27], suggesting its contribution to neurodegeneration in various neurological diseases, including AD and FXTAS. Our findings demonstrate that overactivation of C3 could be contributing to neurodegeneration and that perhaps blocking C3 function could be protective and might lead to the development of strategies for future target treatments.
Among the other proteins, which were commonly differentially expressed in this study using blood from FXTAS (CON V2) and in the study using CSF, were the pigment epithelium-derived factor (PEDF), a unique neurotrophic protein that decreases with aging, the acute-phase protein alpha-2 macroglobulin (A2M), which is a significant component of the innate immune system; the serine protease inhibitor (SERPIN), associated with diverse thrombosis disorders, the inter-alpha-trypsin inhibitor heavy chain 2 (ITIH2), the small leucine-rich proteoglycan (LUM), a member of the small leucine-rich proteoglycan family playing a role in cancer, adhesion, and migration [28,29,30]. These neurodegeneration-associated proteins have also been linked to inflammatory processes [28,31], which are observed in FXTAS pathogenesis and may be promising target pathways for pharmacology.
Finally, blood-brain barrier (BBB) abnormalities have been reported across multiple neurodegenerative disorders such as vascular dementia, MS, Lewy body disease, and spinal muscular atrophy and may contribute to the neurological pathology that often enhances neurodegenerative disorders [32,33]. The CSF/serum quotient of albumin (QAlb) is an indirect measurement of the permeability of the BBB, and [13] highlighted the significant correlations between patients’ QAlb and their respected CGG repeat length and FXTAS rating scale score. They suggested that the observed higher QAlb levels in their study and also in the CON group from our study presented here were associated with a more severe clinical phenotype and proposed dysregulations in BBB permeability as a clinical prognostic measure for disease severity for patients diagnosed with FXTAS. Of relevance, our findings that disruption in protein levels and associated pathways, detected in both blood and CSF, argue in favor of the use of a less invasive tissue, blood, to be utilized to identify molecular biomarkers, predictors of disease development, severity, and progression.
One of the limitations of this study is the small sample sizes; however, it is important to acknowledge that FXTAS is a disease that has been understudied and is not common, making it challenging to obtain a larger sample pool. Despite these obstacles, longitudinal and additional studies with a larger sample size should be conducted to confirm our findings, identify the most robust and predictive biomarkers, and gain further insight into the disease pathogenesis. Despite these limitations, our study offers valuable information on the proteomic differences between PM, who developed the disorder over time and controls, which can lead to more comprehensive research into the disease’s underlying mechanisms and potential therapeutic interventions.

4. Materials and Methods

4.1. Study Participants

As part of a continuing longitudinal study, male participants PM, over the age of 45 years and male participants with non-carrier age-matched healthy controls (HC) were recruited as detailed in [34]. All participants were white in ethnicity, with the exception of three Hispanic participants in the HC group, one in the CON group, and none in the NCON group. The studies and all protocols were carried out in accordance with the Institutional Review Board at the University of California, Davis. All participants gave written informed consent before participating in the study, in line with the Declaration of Helsinki. FXTAS stage scoring was based on the clinical descriptions as previously described [35]. Three categories were used in the diagnosis of FXTAS as explained in Zafarullah and Tassone [36] and termed “definite”, “probable” and “possible FXTAS”. Three age-matched groups were included in this study: CON, NCON, and HC. Using the data from two brain scans, neurological assessment, FXTAS stage, and CGG repeat length, 17 participants were classified as “CON” as they developed clear FXTAS symptomology and thus met criteria of diagnosis between visits (FXTAS stage score was 0–1 at V1 and ≥2 at V2); 19 were defined as “NCON” because they continued to show no signs of FXTAS at V2 (FXTAS stage score was 0–1 at both V1 and V2); and 12 as HC (normal FMR1 alleles/non-PM). The stages of FXTAS range from no tremor at stage 0 to significant tremor that interferes with activities of daily living and intermittent falls at stage 3 [35].

4.2. CGG Repeat Length

Genomic DNA (gDNA) was isolated from 5 mL of peripheral blood leukocytes using the Gentra Puregene Blood Kit (Qiagen, Hilden, Germany). CGG repeat allele size and methylation status were assessed using a combination of Southern blot and PCR analysis. Details of the protocols are as previously reported [37,38].

4.3. Sample Handling and Preparation

Peripheral blood was collected in cell preparation tube (CPT) vacutainers with sodium citrate (Becton Dickinson, Singapore) and centrifuged according to the manufacturer’s recommendations for separating mononuclear cells from whole blood. PBMCs were washed with Dulbecco’s phosphate-buffered saline (PBS) and frozen in RPMI 1640 media with 10% fetal bovine serum and 10% dimethyl sulfoxide. Frozen, isolated PBMCs were quickly thawed in a 37 °C water bath, transferred to a 1.5 mL tube, and spun for 20 min to pellet the cells. The freezing medium was removed, and proteins were extracted in 5% SDS in 50 mM triethyl ammonium bicarbonate (TEAB). Protein concentration was determined by BCA assay (Pierce, Appleton, WI, USA), and 150 ug of proteins was digested on an S-Trap™ (ProtiFi, New York, NY, USA) Digestion column plate. Initially, 10 mM dithiothreitol (DTT) was added, incubated at 50 °C for 10 min, and rested at room temperature for 10 min. Next, 5 mM iodoacetamide (IAA) was added and incubated at room temperature for 30 min in the dark. The samples were acidified with 12% phosphoric acid, followed by the addition of freshly made S-trap buffer (90% methanol, 100 mM TEAB, pH 7.1), and mixed immediately by inversion.
The entire acidified lysate buffer mix was transferred to the S-trap plate and pushed through with a Tecan Resolvex A200 (Tecan, Männedorf, Switzerland) until all the solution passed through. Columns were washed with 400 μL of S-trap buffer. Trypsin enzyme digest buffer was carefully added (1:25 enzyme: total protein in 120 μL of 50 mM TEAB, pH 8.0) to the column. After two hours of incubation at 37 °C, the same amount of trypsin and TEAB was added to the S-trap as a boost step, and the reaction continued overnight at 37 °C. The following day, peptides were eluted from the S-trap. Peptide elution steps included 80 μL of 50 mM TEAB (pH 8.0) and 80 μL of 0.5% formic acid 80 μL of the solution containing 50% acetonitrile and 0.5% formic acid. The final pooled elution was dried down in a speed vacuum. Peptides were resuspended in 0.1% TFA and 2% ACN and quantified using the Pierce™ Quantitative Fluorometric Peptide Assay (Thermo Fisher Scientific, Waltham, MA, USA).

4.4. Liquid Chromatography Mass Spectrometry (LC-MS/MS)

LC separation was carried out on a Dionex Nano Ultimate 3000 (Thermo Scientific) with a Thermo Easy-Spray source fitted with a PepSep emitter. The digested peptides were reconstituted in 2% acetonitrile/0.1% trifluoroacetic acid, and 5 µL of each sample was loaded onto a Thermo Scientific PepMap 100 C18 5 μm 0.3 mm × 5 mm reverse phase trap, where they were desalted online before being separated on a PepSep 8 cm ID 150 1.5 μm reverse phase column. Peptides were eluted using a 90 min gradient with a flow rate of 0.500 μL/min. The samples were run on an Orbitrap Exploris 480 (Thermo Scientific) in data-independent acquisition (DIA) mode; mass spectra were acquired using a collision energy of 30, resolution of 30 K, maximum inject time mode on auto, and an AGC target of 1000%, using an isolation window of 45.7 Da in the m/z range 350–1200 m/z. Raw spectrometry data and analysis are available from the Massive and Proteome Exchange repositories using the respective ID numbers (MSV000092680, PXD044608).

4.5. Data Analysis

DIA data were analyzed using Spectronaut 15 (Biognosys Schlieren, Schlieren, Switzerland), using the direct DIA workflow with the default settings. Briefly, trypsin/P-Specific was set for the enzyme, allowing two missed cleavages. Fixed modifications were set for carbamidomethyl, and variable modifications were set to acetyl (protein N-term) and oxidation. For DIA search identification, PSM and Protein Group FDR were set at 0.01%. A minimum of 2 peptides per protein group were required for quantification. A report was exported from Spectronaut using the reporting feature and imported into SimpliFi (https://simplifi.protifi.com/) for QC and statistical analysis (Protifi, Farmingdale, NY, USA).
For the age and CGG repeats, the p-values are from an ANOVA F-test followed by Tukey HSD pairwise comparisons. Differential expression analyses were conducted using limma-voom. For comparisons between PM and HC participants at baseline, the model used in limma included PM/HC as the only factor. For analyses of PM among participants at V1 and V2, the model used in limma included factors for conversion status, time, and the interaction between conversion status and time, and estimates and standard errors of log fold changes were adjusted for within-participant correlations. Multiple testing corrections were carried out using the Benjamini-Hochberg (BH) approach. Pathway enrichment analysis was carried out using the Wilcoxon rank-sum test on the raw p-values from the differential expression analysis on individual comparisons. Pathway enrichment analysis was carried out using Fisher’s exact test on the overlapping list of proteins that are significantly different between CON and NCON at V1 and at V2 using the BH adjusted p-value cutoff of 0.05. Pathway enrichment visualization uses the continuous raw p-values from the enrichment analysis. sPLS-DA analysis was carried out using the R package mixOmics version 3.17 [39].

5. Conclusions

Currently, there is no effective treatment for FXTAS, and the only options available focus on managing the symptoms. So, a deep understanding of the FXTAS pathogenesis requires the identification of proteins that can be used to understand the altered pathways, serve as biomarkers for early identification of the most at-risk carriers to develop the syndrome, and lead towards the development of targeted therapeutics. However, the investigation of neurodegenerative disorders, including FXTAS, is limited by the availability of accessible sample types. In this study, by using a unique approach of high-throughput mass spectrometry proteomic profiling of blood samples from PM, including longitudinal analysis, we identified a unique set of potential proteomic biomarkers for early diagnosis of FXTAS. In addition, we also observed a significant dysregulation in various protein pathways involved in cellular function and inflammatory responses. These identified pathways may be valuable for the development of effective drugs and therapeutics for this devasting neurodegenerative disorder.

Author Contributions

Conceptualization, M.Z., R.H., S.M.R. and D.H.; methodology, M.Z., M.R.S., B.S.P. and F.T.; software, J.L. and B.P.D.-J.; validation, M.Z. and F.T.; formal analysis, J.L. and B.P.D.-J.; investigation, M.Z., M.R.S. and B.S.P.; resources, F.T.; data curation, J.L. and B.P.D.-J.; writing—original draft preparation, M.Z.; writing—review and editing, M.Z., J.L., M.R.S., B.S.P., B.P.D.-J., R.H., S.M.R., D.H. and F.T.; supervision, F.T.; project administration, F.T.; funding acquisition, R.H., S.M.R., D.H. and F.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by NIH Grant R01NS110100 to DH and SMR. LC/MS was supported by NIH grant 1S10OD026918-01A1. LC/MS was supported by NIH grant 1S10OD026918-01A1.

Institutional Review Board Statement

The studies and all protocols were carried out in accordance with the Institutional Review Board at the University of California, Davis. All participants gave written informed consent before participating in the study, in line with the Declaration of Helsinki.

Informed Consent Statement

Informed consent was obtained from all individual participants involved in the study.

Data Availability Statement

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Acknowledgments

We thank the participants of the community-based studies who donated their time and samples for this study. This paper is dedicated to the memory of Matteo.

Conflicts of Interest

M.Z.: No disclosures to report. J.L.: No disclosures to report. M.S.: No disclosures to report. B.P.: No disclosures to report. B.J.: No disclosures to report. R.J.H. has received funding from the Azrieli Foundation, Zynerba Pharmaceuticals, and Tetra Pharmaceuticals for treatment studies in fragile X syndrome. She has also received funding from NICHD (HD036071) to study FXTAS. S.M.R.: No disclosures to report. D.H.: UC Davis has received funding for Hessl’s consulting from Zynerba, Tetra, Healx, and Ovid for fragile X syndrome clinical trials: F.T.: Has received the funding from Azrieli Foundation and Zynerba for studies in fragile X syndrome.

References

  1. Cabal-Herrera, A.M.; Tassanakijpanich, N.; Salcedo-Arellano, M.J.; Hagerman, R.J. Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS): Pathophysiology and Clinical Implications. Int. J. Mol. Sci. 2020, 21, 4391. [Google Scholar] [CrossRef] [PubMed]
  2. Rodriguez-Revenga, L.; Madrigal, I.; Pagonabarraga, J.; Xunclà, M.; Badenas, C.; Kulisevsky, J.; Gomez, B.; Milà, M. Penetrance of FMR1 Premutation Associated Pathologies in Fragile X Syndrome Families. Eur. J. Hum. Genet. 2009, 17, 1359–1362. [Google Scholar] [CrossRef] [PubMed]
  3. Jacquemont, S.; Hagerman, R.J.; Leehey, M.A.; Hall, D.A.; Levine, R.A.; Brunberg, J.A.; Zhang, L.; Jardini, T.; Gane, L.W.; Harris, S.W.; et al. Penetrance of the Fragile X–Associated Tremor/Ataxia Syndrome in a Premutation Carrier Population. JAMA 2004, 291, 460–469. [Google Scholar] [CrossRef] [PubMed]
  4. Salcedo-Arellano, M.J.; Dufour, B.; McLennan, Y.; Martinez-Cerdeno, V.; Hagerman, R. Fragile X Syndrome and Associated Disorders: Clinical Aspects and Pathology. Neurobiol. Dis. 2020, 136, 104740. [Google Scholar] [CrossRef]
  5. Hagerman, R.J.; Berry-Kravis, E.; Hazlett, H.C.; Bailey, D.B.; Moine, H.; Kooy, R.F.; Tassone, F.; Gantois, I.; Sonenberg, N.; Mandel, J.L.; et al. Fragile X Syndrome. Nat. Rev. Dis. Primers 2017, 3, 17065. [Google Scholar] [CrossRef]
  6. Tassone, F.; Hagerman, R.J.; Taylor, A.K.; Gane, L.W.; Godfrey, T.E.; Hagerman, P.J. Elevated Levels of FMR1 mRNA in Carrier Males: A New Mechanism of Involvement in the Fragile-X Syndrome. Am. J. Hum. Genet. 2000, 66, 6. [Google Scholar] [CrossRef]
  7. Hagerman, R.J.; Hagerman, P. Fragile X-Associated Tremor/ataxia Syndrome—Features, Mechanisms and Management. Nat. Rev. Neurol. 2016, 12, 403–412. [Google Scholar] [CrossRef]
  8. Zafarullah, M.; Tang, H.-T.; Durbin-Johnson, B.; Fourie, E.; Hessl, D.; Rivera, S.M.; Tassone, F. FMR1 Locus Isoforms: Potential Biomarker Candidates in Fragile X-Associated Tremor/ataxia Syndrome (FXTAS). Sci. Rep. 2020, 10, 11099. [Google Scholar] [CrossRef]
  9. Al-Amrani, S.; Al-Jabri, Z.; Al-Zaabi, A.; Alshekaili, J.; Al-Khabori, M. Proteomics: Concepts and Applications in Human Medicine. World J. Biol. Chem. 2021, 12, 57–69. [Google Scholar] [CrossRef]
  10. Cui, M.; Cheng, C.; Zhang, L. High-Throughput Proteomics: A Methodological Mini-Review. Lab. Investig. 2022, 102, 1170–1181. [Google Scholar] [CrossRef]
  11. Ma, L.; Herren, A.W.; Espinal, G.; Randol, J.; McLaughlin, B.; Martinez-Cerdeño, V.; Pessah, I.N.; Hagerman, R.J.; Hagerman, P.J. Composition of the Intranuclear Inclusions of Fragile X-Associated Tremor/Ataxia Syndrome. Acta Neuropathol. Commun. 2019, 7, 143. [Google Scholar] [CrossRef] [PubMed]
  12. Holm, K.N.; Herren, A.W.; Taylor, S.L.; Randol, J.L.; Kim, K.; Espinal, G.; Martiínez-Cerdeño, V.; Pessah, I.N.; Hagerman, R.J.; Hagerman, P.J. Human Cerebral Cortex Proteome of Fragile X-Associated Tremor/Ataxia Syndrome. Front. Mol. Biosci. 2020, 7, 600840, Corrigendum in Front. Mol. Biosci. 2021, 8, 695407. [Google Scholar]
  13. Abbasi, D.A.; Nguyen, T.T.A.; Hall, D.A.; Robertson-Dick, E.; Berry-Kravis, E.; Cologna, S.M. Correction to: Characterization of the Cerebrospinal Fluid Proteome in Patients with Fragile X-Associated Tremor/Ataxia Syndrome. Cerebellum 2022, 21, 99–100. [Google Scholar] [CrossRef] [PubMed]
  14. Estes, R.E.; Lin, B.; Khera, A.; Davis, M.Y. Lipid Metabolism Influence on Neurodegenerative Disease Progression: Is the Vehicle as Important as the Cargo? Front. Mol. Neurosci. 2021, 14, 788695. [Google Scholar] [CrossRef]
  15. Zafarullah, M.; Palczewski, G.; Rivera, S.M.; Hessl, D.R.; Tassone, F. Metabolic Profiling Reveals Dysregulated Lipid Metabolism and Potential Biomarkers Associated with the Development and Progression of Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS). FASEB J. 2020, 34, 16676–16692. [Google Scholar] [CrossRef]
  16. Zafarullah, M.; Durbin-Johnson, B.; Fourie, E.S.; Hessl, D.R.; Rivera, S.M.; Tassone, F. Metabolomic Biomarkers Are Associated with Area of the Pons in Fragile X Premutation Carriers at Risk for Developing FXTAS. Front. Psychiatry 2021, 12, 691717. [Google Scholar] [CrossRef] [PubMed]
  17. Checa, A.; Bedia, C.; Jaumot, J. Lipidomic Data Analysis: Tutorial, Practical Guidelines and Applications. Anal. Chim. Acta 2015, 885, 1–16. [Google Scholar] [CrossRef]
  18. Novgorodov, S.A.; Riley, C.L.; Yu, J.; Keffler, J.A.; Clarke, C.J.; Van Laer, A.O.; Baicu, C.F.; Zile, M.R.; Gudz, T.I. Lactosylceramide Contributes to Mitochondrial Dysfunction in Diabetes. J. Lipid Res. 2016, 57, 546. [Google Scholar] [CrossRef]
  19. Xiyang, Y.-B.; Liu, R.; Wang, X.-Y.; Li, S.; Zhao, Y.; Lu, B.-T.; Xiao, Z.-C.; Zhang, L.-F.; Wang, T.-H.; Zhang, J. COX5A Plays a Vital Role in Memory Impairment Associated with Brain Aging via the BDNF/ERK1/2 Signaling Pathway. Front. Aging Neurosci. 2020, 12, 540040. [Google Scholar] [CrossRef]
  20. Alliance of Genome Resources. Available online: https://www.alliancegenome.org/ (accessed on 14 July 2023).
  21. Baty, K.; Farrugia, M.E.; Hopton, S.; Falkous, G.; Schaefer, A.M.; Stewart, W.; Willison, H.J.; Reilly, M.M.; Blakely, E.L.; Taylor, R.W.; et al. A Novel MT-CO2 Variant Causing Cerebellar Ataxia and Neuropathy: The Role of Muscle Biopsy in Diagnosis and Defining Pathogenicity. Neuromuscul. Disord. 2021, 31, 1186–1193. [Google Scholar] [CrossRef]
  22. Sim, K.G.; Carpenter, K.; Hammond, J.; Christodoulou, J.; Wilcken, B. Acylcarnitine Profiles in Fibroblasts from Patients with Respiratory Chain Defects Can Resemble Those from Patients with Mitochondrial Fatty Acid Beta-Oxidation Disorders. Metabolism 2002, 51, 366–371. [Google Scholar] [CrossRef] [PubMed]
  23. Franco, D.L.; Rezával, C.; Cáceres, A.; Schinder, A.F.; Ceriani, M.F. ENA/VASP Downregulation Triggers Cell Death by Impairing Axonal Maintenance in Hippocampal Neurons. Mol. Cell. Neurosci. 2010, 44, 154–164. [Google Scholar] [CrossRef] [PubMed]
  24. Tao, Y.; Han, Y.; Yu, L.; Wang, Q.; Leng, S.X.; Zhang, H. The Predicted Key Molecules, Functions, and Pathways That Bridge Mild Cognitive Impairment (MCI) and Alzheimer’s Disease (AD). Front. Neurol. 2020, 11, 233. [Google Scholar] [CrossRef] [PubMed]
  25. Xu, Z.; Bai, S.; Wu, H.; Fang, M. Elevated Retinal Retinol-Binding Protein 4 Levels in Diabetic Mice Can Induce Retinal Neurodegeneration through Microglia. Microsc. Res. Tech. 2023, 86, 223–231. [Google Scholar] [CrossRef]
  26. Westacott, L.J.; Wilkinson, L.S. Complement Dependent Synaptic Reorganisation during Critical Periods of Brain Development and Risk for Psychiatric Disorder. Front. Neurosci. 2022, 16, 840266. [Google Scholar] [CrossRef]
  27. Wu, T.; Dejanovic, B.; Gandham, V.D.; Gogineni, A.; Edmonds, R.; Schauer, S.; Srinivasan, K.; Huntley, M.A.; Wang, Y.; Wang, T.-M.; et al. Complement C3 Is Activated in Human AD Brain and Is Required for Neurodegeneration in Mouse Models of Amyloidosis and Tauopathy. Cell Rep. 2019, 28, 2111–2123.e6. [Google Scholar] [CrossRef]
  28. Lee, J.M.; Kim, S.R. Prothrombin Kringle-2, a Mediator of Microglial Activation: New Insight in Alzheimer’s Disease Pathogenesis. Neural Regen. Res. 2022, 17, 2675–2676. [Google Scholar]
  29. Rotunno, M.S.; Lane, M.; Zhang, W.; Wolf, P.; Oliva, P.; Viel, C.; Wills, A.-M.; Alcalay, R.N.; Scherzer, C.R.; Shihabuddin, L.S.; et al. Cerebrospinal Fluid Proteomics Implicates the Granin Family in Parkinson’s Disease. Sci. Rep. 2020, 10, 2479. [Google Scholar] [CrossRef]
  30. Chen, X.; Li, X.; Hu, X.; Jiang, F.; Shen, Y.; Xu, R.; Wu, L.; Wei, P.; Shen, X. LUM Expression and Its Prognostic Significance in Gastric Cancer. Front. Oncol. 2020, 10, 605. [Google Scholar] [CrossRef]
  31. Huang, M.; Qi, W.; Fang, S.; Jiang, P.; Yang, C.; Mo, Y.; Dong, C.; Li, Y.; Zhong, J.; Cai, W.; et al. Pigment Epithelium-Derived Factor Plays a Role in Alzheimer’s Disease by Negatively Regulating Aβ42. Neurotherapeutics 2018, 15, 728–741. [Google Scholar] [CrossRef]
  32. Wurster, C.D.; Koch, J.C.; Cordts, I.; Dreyhaupt, J.; Otto, M.; Uzelac, Z.; Witzel, S.; Winter, B.; Kocak, T.; Schocke, M.; et al. Routine Cerebrospinal Fluid (CSF) Parameters in Patients with Spinal Muscular Atrophy (SMA) Treated with Nusinersen. Front. Neurol. 2019, 10, 1179. [Google Scholar] [CrossRef] [PubMed]
  33. Musaeus, C.S.; Gleerup, H.S.; Høgh, P.; Waldemar, G.; Hasselbalch, S.G.; Simonsen, A.H. Cerebrospinal Fluid/Plasma Albumin Ratio as a Biomarker for Blood-Brain Barrier Impairment Across Neurodegenerative Dementias. J. Alzheimers. Dis. 2020, 75, 429–436. [Google Scholar] [CrossRef]
  34. Shelton, A.L.; Wang, J.Y.; Fourie, E.; Tassone, F.; Chen, A.; Frizzi, L.; Hagerman, R.J.; Ferrer, E.; Hessl, D.; Rivera, S.M. Middle Cerebellar Peduncle Width-A Novel MRI Biomarker for FXTAS? Front. Neurosci. 2018, 12, 379. [Google Scholar] [CrossRef] [PubMed]
  35. Bacalman, S.; Farzin, F.; Bourgeois, J.A.; Cogswell, J.; Goodlin-Jones, B.L.; Gane, L.W.; Grigsby, J.; Leehey, M.A.; Tassone, F.; Hagerman, R.J. Psychiatric Phenotype of the Fragile X-Associated Tremor/ataxia Syndrome (FXTAS) in Males: Newly Described Fronto-Subcortical Dementia. J. Clin. Psychiatry 2006, 67, 87–94. [Google Scholar] [CrossRef] [PubMed]
  36. Zafarullah, M.; Tassone, F. Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS). Methods Mol. Biol. 2019, 1942, 173–189. [Google Scholar]
  37. Tassone, F.; Pan, R.; Amiri, K.; Taylor, A.K.; Hagerman, P.J. A Rapid Polymerase Chain Reaction-Based Screening Method for Identification of All Expanded Alleles of the Fragile X (FMR1) Gene in Newborn and High-Risk Populations. J. Mol. Diagn. 2008, 10, 43–49. [Google Scholar] [CrossRef]
  38. Filipovic-Sadic, S.; Sah, S.; Chen, L.; Krosting, J.; Sekinger, E.; Zhang, W.; Hagerman, P.J.; Stenzel, T.T.; Hadd, A.G.; Latham, G.J.; et al. A Novel FMR1 PCR Method for the Routine Detection of Low Abundance Expanded Alleles and Full Mutations in Fragile X Syndrome. Clin. Chem. 2010, 56, 399–408. [Google Scholar] [CrossRef]
  39. Rohart, F.; Gautier, B.; Singh, A.; Cao, K.-A.L. mixOmics: An R Package for ‘omics Feature Selection and Multiple Data Integration. PLoS Comput. Biol. 2017, 13, e1005752. [Google Scholar] [CrossRef]
Figure 1. Blood proteome analysis of the present study. The sparse partial least squares discriminant analysis (sparse PLSDA) score plot based on the data of the blood proteome from converters and non-converters (V1 and V2) and healthy controls.
Figure 1. Blood proteome analysis of the present study. The sparse partial least squares discriminant analysis (sparse PLSDA) score plot based on the data of the blood proteome from converters and non-converters (V1 and V2) and healthy controls.
Ijms 24 13477 g001
Figure 2. Differential protein expression levels among the HC and the PM groups. Heatmap of the 79 most significantly altered proteins (adjusted p < 0.05) in the PM group as compared to HC at both V1 and V2. Heatmap was generated using R code; red indicates high and blue indicates low gene expression.
Figure 2. Differential protein expression levels among the HC and the PM groups. Heatmap of the 79 most significantly altered proteins (adjusted p < 0.05) in the PM group as compared to HC at both V1 and V2. Heatmap was generated using R code; red indicates high and blue indicates low gene expression.
Ijms 24 13477 g002
Figure 3. Protein pathways altered from V1 to V2 in CON and NCON groups. Heatmap of the protein pathways that are altered between Visit 2 and Visit 1 in NCON and CON groups. Heatmap was generated using R code; the color from blue to red indicates the increase in statistical significance.
Figure 3. Protein pathways altered from V1 to V2 in CON and NCON groups. Heatmap of the protein pathways that are altered between Visit 2 and Visit 1 in NCON and CON groups. Heatmap was generated using R code; the color from blue to red indicates the increase in statistical significance.
Ijms 24 13477 g003
Figure 4. Protein pathways altered between CON and NCON groups. Heatmap of the protein pathways that are altered (p < 0.05) between CON and NCON at V1 and V2. Heatmap was generated using R code; the color from blue to red indicates the increase in statistical significance.
Figure 4. Protein pathways altered between CON and NCON groups. Heatmap of the protein pathways that are altered (p < 0.05) between CON and NCON at V1 and V2. Heatmap was generated using R code; the color from blue to red indicates the increase in statistical significance.
Ijms 24 13477 g004
Figure 5. Enriched pathways for the proteins that are consistently differentially expressed between CON and NCON from V1 to V2. Protein-protein interactions from STRING database are represented as edges between proteins.
Figure 5. Enriched pathways for the proteins that are consistently differentially expressed between CON and NCON from V1 to V2. Protein-protein interactions from STRING database are represented as edges between proteins.
Ijms 24 13477 g005
Figure 6. Molecular functions altered between CON and NCON at V1 and V2. Gene ontology molecular functions, including the mitochondrial, protein synthesis machinery and transport, and positive regulation of BMP signaling pathways enriched in the proteins (enclosed in orange circle) that are consistently differentially expressed between CON and NCON at V1 and V2. The blue color represents the proteins that are up-regulated in CON. Yellow represents the down-regulated ones. While Grey is representing the non-differential proteins and Red is FMR1.
Figure 6. Molecular functions altered between CON and NCON at V1 and V2. Gene ontology molecular functions, including the mitochondrial, protein synthesis machinery and transport, and positive regulation of BMP signaling pathways enriched in the proteins (enclosed in orange circle) that are consistently differentially expressed between CON and NCON at V1 and V2. The blue color represents the proteins that are up-regulated in CON. Yellow represents the down-regulated ones. While Grey is representing the non-differential proteins and Red is FMR1.
Ijms 24 13477 g006
Figure 7. Significantly altered pathways comparing CON to NCON at V2. Protein-protein interactions from STRING database are represented as edges between proteins.
Figure 7. Significantly altered pathways comparing CON to NCON at V2. Protein-protein interactions from STRING database are represented as edges between proteins.
Ijms 24 13477 g007
Figure 8. Sphingolipid and amino acid metabolism altered in CON. Proteins associated with the sphingolipid and amino-acid pathways, including glycine, serine, and threonine metabolism (enclosed in orange circle), are found to be enriched in the comparison between CON and NCON at V2. The blue color represents the proteins that are up-regulated in CON. Yellow represents the down-regulated ones. The level of the significance is indicated with the intensity of the color.
Figure 8. Sphingolipid and amino acid metabolism altered in CON. Proteins associated with the sphingolipid and amino-acid pathways, including glycine, serine, and threonine metabolism (enclosed in orange circle), are found to be enriched in the comparison between CON and NCON at V2. The blue color represents the proteins that are up-regulated in CON. Yellow represents the down-regulated ones. The level of the significance is indicated with the intensity of the color.
Ijms 24 13477 g008
Figure 9. Comparison of CSF and blood proteomic profile. Cerebrospinal fluid (CSF) proteome of FXTAS patients identified 414 [13]. Blood proteome profile of CON at V2 (in pink) identified a total of 2166 proteins of which 97 were found in common. By looking at significantly altered proteins from blood proteomic profile (n = 110) and CSF proteomic profile (n = 46), 8 proteins were found to be in common (indicated in red ink), including Complement C3, Alpha-2-HS-glycoprotein, Pigment epithelium-derived factor, Inter-alpha-trypsin inhibitor heavy chain H2, Retinol-binding protein 4, Alpha-2-macroglobulin, Prothrombin, and Lumican.
Figure 9. Comparison of CSF and blood proteomic profile. Cerebrospinal fluid (CSF) proteome of FXTAS patients identified 414 [13]. Blood proteome profile of CON at V2 (in pink) identified a total of 2166 proteins of which 97 were found in common. By looking at significantly altered proteins from blood proteomic profile (n = 110) and CSF proteomic profile (n = 46), 8 proteins were found to be in common (indicated in red ink), including Complement C3, Alpha-2-HS-glycoprotein, Pigment epithelium-derived factor, Inter-alpha-trypsin inhibitor heavy chain H2, Retinol-binding protein 4, Alpha-2-macroglobulin, Prothrombin, and Lumican.
Ijms 24 13477 g009
Table 1. Subjects Baseline Characteristics.
Table 1. Subjects Baseline Characteristics.
Non-Converter (n = 19)Converter (n = 17)Healthy Control (n = 12)p-Value
Age
N191712
Mean (SD)57.2 (8.2)53.2 (6.9)50.2 (5.2)0.0319
Median (Range)59 (44–68)53 (42–65)49 (45–63)
CGG repeat
N191712
Mean (SD)82.9 (22)90.2 (21.4)29.8 (2.4)<0.001
Median (Range)78 (56–135)85 (60–141)30 (23–32)
Table 2. Differential expression statistics (BH adjusted p-values) among converters, non-converters, and healthy controls.
Table 2. Differential expression statistics (BH adjusted p-values) among converters, non-converters, and healthy controls.
Sr #PG Protein AccessionsPG GenesPG Protein DescriptionsConverter V2_v_HealthyControlConverter_v_NonConverter_V1Converter_v_NonConverter_V2Pre_v_Control_BaselineV2_v_V1_ConverterV2_v_V1_NonConverter
1P55957BIDBH3-interacting domain death agonist0.3309610.0001630.01844260.9031130.0001260.2063338
2P00403MT-CO2Cytochrome c oxidase subunit 20.0019020.00000830.25575240.3866888.5 × 10−60.5831517
3O75531BANF1Barrier-to-autointegration factor0.00019680.00001730.14352310.1767370.0032150.9627627
4P20674COX5ACytochrome c oxidase subunit 5A, mitochondrial0.00019680.00000020.73460920.135 × 10−70.9955487
5Q8NFW8CMASN-acylneuraminate cytidylyltransferase0.7071280.0000040.05902330.9581160.0007620.8246921
6Q15370ELOBElongin-B0.97783480.00058510.00040760.8865940.2344480.499658
7Q9Y3B2EXOSC1Exosome complex component CSL40.93985730.00002760.04509910.910060.0097820.8639569
8P62310LSM3U6 snRNA-associated Sm-like protein LSm30.01359750.00003490.41977210.3429449.16 × 10−50.8092159
9Q92769HDAC2Histone deacetylase 20.58177820.00158610.0021990.4211680.0557640.3596959
10P42025ACTR1BBeta-centractin0.74638960.00342870.00001980.9203650.4982850.2063338
11P17676CEBPBCCAAT/enhancer-binding protein beta0.37726530.00069460.04439460.6497230.0354080.7433883
12Q6P1A2LPCAT3Lysophospholipid acyltransferase 50.27812980.00158610.0778950.7831580.0082470.499658
13O00422SAP18Histone deacetylase complex subunit SAP180.27335540.00823360.12015450.9410930.0039190.3596959
14P14406COX7A2Cytochrome c oxidase subunit 7A2, mitochondrial0.07935350.00174150.14934140.4772680.0086430.6176757
15P01909HLA-DQA1HLA class II histocompatibility antigen, DQ alpha 1 chain0.87057040.00167080.04052230.3892860.0145660.4822303
16O95716RAB3DRas-related protein Rab-3D0.49955090.00155290.06394650.8331190.0310630.6751625
17O95182NDUFA7NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 70.88131780.01914250.00306630.6258910.1149890.2063338
18P83881; Q969Q0RPL36A; RPL36AL60S ribosomal protein L36a; 60S ribosomal protein L36a-like0.36133390.00371130.1321790.8904010.0339570.6898521
19Q8N699MYCT1Myc target protein 10.55332070.00433940.01555360.9421970.0177780.2948038
20P51148RAB5CRas-related protein Rab-5C0.46211680.0031530.03867190.6304960.0889830.6539073
21P12829MYL4Myosin light chain 40.88103390.01801410.00026220.9371170.6331670.2948038
22Q8NHV1GIMAP7GTPase IMAP family member 70.50830150.00110110.00416750.5854340.140750.5669873
23Q9UDW1UQCR10Cytochrome b-c1 complex subunit 90.62148310.01856650.14456050.7715410.0108010.4096213
24P62854RPS2640S ribosomal protein S260.76751590.00110110.16908440.8469430.0088950.7317184
25P07919UQCRHCytochrome b-c1 complex subunit 6, mitochondrial0.02765320.00069460.79319930.6887360.0006460.9420505
26Q96CN7ISOC1Isochorismatase domain-containing protein 10.95402230.00069460.22219590.6427020.0044670.7459911
27P30048PRDX3Thioredoxin-dependent peroxide reductase, mitochondrial0.81266830.00248860.00081390.9031130.5830970.633638
28O14980XPO1Exportin-10.4968060.00336450.01578410.2921740.0889830.5036518
29P10155RO6060 kDa SS-A/Ro ribonucleoprotein0.89584940.0039920.09825650.8794250.0418990.6641875
30P14854COX6B1Cytochrome c oxidase subunit 6B10.25232290.00198480.24344410.6921780.0156930.8107049
31P27338MAOBAmine oxidase [flavin-containing] B0.75796990.01142130.07914050.9584010.0776410.5831517
32Q9NWH9SLTMSAFB-like transcription modulator0.657670.00110110.36157140.8220370.006030.8663002
33Q7LBR1CHMP1BCharged multivesicular body protein 1b0.99831770.00805530.0778950.7571830.0487390.5340208
34O14949UQCRQCytochrome b-c1 complex subunit 80.27812980.00255690.36184050.8644670.0044670.7117886
35O76021RSL1D1Ribosomal L1 domain-containing protein 10.95561110.00291010.09617990.9344030.0820460.8256684
36Q9BY77POLDIP3Polymerase delta-interacting protein 30.99397510.00158610.01930430.9719530.182990.7712084
37P84098RPL1960S ribosomal protein L190.4475670.00260240.2081540.642760.0506680.9332484
38Q8NEW0SLC30A7Zinc transporter 70.90474380.00110110.26819980.7855380.0145660.9132308
39O60831PRAF2PRA1 family protein 20.76751590.01872090.04451520.9211530.1914740.5792395
40P25490YY1Transcriptional repressor protein YY10.34258980.00069460.19710430.3401620.1089370.7852614
41P54578USP14Ubiquitin carboxyl-terminal hydrolase 140.70013690.01171660.1551950.9143960.0110720.4529446
42O95299NDUFA10NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 10, mitochondrial0.76751590.023680.01779510.2515170.0272930.2063338
43P50552VASPVasodilator-stimulated phosphoprotein0.00019680.00069460.16453950.2811541.93 × 10−50.7253255
44P62993GRB2Growth factor receptor-bound protein 20.00028780.02068620.95794790.110540.0036850.8107049
45Q9NRX4PHPT114 kDa phosphohistidine phosphatase0.61103570.00473350.24581140.8817670.0750830.9420505
46P51809VAMP7Vesicle-associated membrane protein 70.94873590.10440920.00077220.9802050.3147210.0612594
47Q9Y3A3MOB4MOB-like protein phocein0.50920930.09153210.05230550.7809410.2512710.4529446
48Q9P1F3ABRACLCostars family protein ABRACL0.89538550.03082480.12693680.7730450.0487390.481216
49P30049ATP5F1DATP synthase subunit delta, mitochondrial0.08272370.02421030.18524710.2295570.1784170.7992426
50P42285MTREXExosome RNA helicase MTR40.41531170.00805530.01226750.4218570.1943760.499658
51Q9UK76JPT1Jupiter microtubule associated homolog 10.50637270.01898570.19240870.8312640.048870.6224493
52Q8IV08PLD3Phospholipase D30.86889990.00301720.04304640.7831580.0929230.6898521
53P62306SNRPFSmall nuclear ribonucleoprotein F0.03207780.01171660.72745260.7522060.0103460.8387102
54Q99536VAT1Synaptic vesicle membrane protein VAT-1 homolog0.34258980.00291010.39145260.609270.0196680.9275825
55Q15102PAFAH1B3Platelet-activating factor acetylhydrolase IB subunit gamma0.76026930.00438430.30730040.8331190.0117570.7161634
56Q9Y3B7MRPL1139S ribosomal protein L11, mitochondrial0.49194520.01675990.03330480.308620.0969420.4594807
57Q9Y5Z4HEBP2Heme-binding protein 20.89668220.02421030.04105070.9505720.1061760.4529446
58O95139NDUFB6NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 60.70013690.01099810.26561040.8817670.0304250.7063057
59Q6IAA8LAMTOR1Ragulator complex protein LAMTOR10.88922560.06766550.03330480.5533470.1379190.3596959
60O00483NDUFA4Cytochrome c oxidase subunit NDUFA40.77877890.00438430.00040760.7715410.9689410.6960786
61P49065ALBSerum albumin0.00019680.39588620.00175810.1017520.0506680.9332484
62P04217A1BGAlpha-1B-glycoprotein0.00000050.08826040.00081390.0760830.3104410.8111847
63P02452COL1A1Collagen alpha-1(I) chain0.00019680.90981290.01128940.0120230.0207650.8570423
64P10412HIST1H1EHistone H1.40.00028780.19258620.00308930.0585880.0063620.4096213
65P02760AMBPProtein AMBP0.00028780.22200950.00914120.0585880.4224310.8428062
66P05543SERPINA7Thyroxine-binding globulin0.00561390.34156650.00146460.234070.123320.7834307
67P60660MYL6Myosin light polypeptide 60.00028780.36740640.01996390.0243470.0418990.499658
68P08697SERPINF2Alpha-2-antiplasmin0.00079360.02970950.00040760.2371640.4146010.8286978
69Q15907RAB11BRas-related protein Rab-11B0.00240770.77772950.00928490.382210.0110720.8428062
70Q9Y6W5WASF2Wiskott-Aldrich syndrome protein family member 20.00028780.41132620.00736020.4680220.0044670.7106443
71P12109COL6A1Collagen alpha-1(VI) chain0.0019020.85303940.02005180.2929340.0295980.9623254
72Q9BRA2TXNDC17Thioredoxin domain-containing protein 170.00453390.84190970.0205760.2782140.1155430.8520701
73Q8N386LRRC25Leucine-rich repeat-containing protein 250.03496950.05148340.0002970.4333590.5749020.5565245
74P51884LUMLumican0.01196580.5024470.01844260.308620.279290.8465036
75O95168NDUFB4NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 40.0056010.01994890.00033070.322460.9299630.5115525
76P63027VAMP2Vesicle-associated membrane protein 20.00031530.9561010.34890730.0688550.2913360.9204359
77Q8IZ07ANKRD13AAnkyrin repeat domain-containing protein 13A0.09124380.16936050.00214750.1471050.808390.4980031
78Q8WYJ6SEPTIN1Septin-10.02726540.62666390.08829020.3584340.1527440.876325
79P51570GALK1Galactokinase0.01719240.56360750.00081390.1835220.6583350.2746342
Table 3. Differentially expressed proteins between converter and non-converter at Visit 1.
Table 3. Differentially expressed proteins between converter and non-converter at Visit 1.
Sr #Protein AccessionsGeneslogFCAve Exprp-Valueadj.P ValProtein Descriptions
1P20674COX5A1.1113.738.45 × 10−111.83 × 10−7Cytochrome c oxidase subunit 5A, mitochondrial
2Q8NFW8CMAS1.8213.43.73 × 10−94.04 × 10−6N-acylneuraminate cytidylyltransferase
3P00403MT-CO21.2214.451.15 × 10−88.27 × 10−6Cytochrome c oxidase subunit 2
4O75531BANF13.2313.923.19 × 10−81.73 × 10−5Barrier-to-autointegration factor
5Q9Y3B2EXOSC10.8512.986.37 × 10−82.76 × 10−5Exosome complex component CSL4
6P62310LSM31.0913.499.67 × 10−83.49 × 10−5U6 snRNA-associated Sm-like protein LSm3
7P55957BID1.5913.875.27 × 10−70.00016297BH3-interacting domain death agonist
8Q15370ELOB113.112.16 × 10−60.00058509Elongin-B
9P25490YY10.6913.273.26 × 10−60.0006946Transcriptional repressor protein YY1
10Q96N66MBOAT71.4913.453.48 × 10−60.0006946Lysophospholipid acyltransferase 7
11P17676CEBPB2.1613.573.67 × 10−60.0006946CCAAT/enhancer-binding protein beta
12P50552VASP−0.4515.43.90 × 10−60.0006946Vasodilator-stimulated phosphoprotein
13Q96CN7ISOC11.2913.184.39 × 10−60.0006946Isochorismatase domain-containing protein 1
14P07919UQCRH0.9713.24.49 × 10−60.0006946Cytochrome b-c1 complex subunit 6, mitochondrial
15P62854RPS262.2814.528.22 × 10−60.0011010940S ribosomal protein S26
16Q8NHV1GIMAP70.9138.71 × 10−60.00110109GTPase IMAP family member 7
17Q8NEW0SLC30A72.2913.658.81 × 10−60.00110109Zinc transporter 7
18Q9NWH9SLTM3.0813.689.48 × 10−60.00110109SAFB-like transcription modulator
19O15347HMGB30.8415.949.66 × 10−60.00110109High mobility group protein B3
20O95716RAB3D2.0614.751.43 × 10−50.00155291Ras-related protein Rab-3D
21Q92769HDAC20.5212.951.58 × 10−50.00158606Histone deacetylase 2
22P31949S100A110.7714.521.66 × 10−50.00158606Protein S100-A11
23Q9BY77POLDIP30.8712.951.75 × 10−50.00158606Polymerase delta-interacting protein 3
24Q6P1A2LPCAT31.5213.451.76 × 10−50.00158606Lysophospholipid acyltransferase 5
25P01909HLA-DQA10.4612.91.93 × 10−50.00167082HLA class II histocompatibility antigen, DQ alpha 1 chain
26P14406COX7A21.5613.672.09 × 10−50.00174149Cytochrome c oxidase subunit 7A2, mitochondrial
27P14854COX6B11.1313.722.47 × 10−50.00198483Cytochrome c oxidase subunit 6B1
28Q9UBW5BIN2−0.3314.522.93 × 10−50.00226447Bridging integrator 2
29P02656APOC31.5814.743.27 × 10−50.00244275Apolipoprotein C-III
30P30048PRDX30.7514.713.45 × 10−50.00248864Thioredoxin-dependent peroxide reductase, mitochondrial
31P62857RPS280.9915.393.69 × 10−50.0025569340S ribosomal protein S28
32O14949UQCRQ1.1813.373.78 × 10−50.00255693Cytochrome b-c1 complex subunit 8
33P84098RPL191.2114.213.96 × 10−50.0026024260S ribosomal protein L19
34O43760SYNGR20.813.784.20 × 10−50.00267673Synaptogyrin-2
35Q02750MAP2K12.5415.394.77 × 10−50.00291015Dual specificity mitogen-activated protein kinase kinase 1
36Q99536VAT10.5313.634.91 × 10−50.00291015Synaptic vesicle membrane protein VAT-1 homolog
37O76021RSL1D11.0713.435.09 × 10−50.00291015Ribosomal L1 domain-containing protein 1
38P62995TRA2B1.2613.665.11 × 10−50.00291015Transformer-2 protein homolog beta
39Q8IV08PLD30.6312.925.48 × 10−50.00301717Phospholipase D3
40P98179RBM30.6813.435.57 × 10−50.00301717RNA-binding protein 3
41P51148RAB5C0.8613.945.97 × 10−50.00315297Ras-related protein Rab-5C
42O14980XPO1−0.2612.956.52 × 10−50.00336451Exportin-1
43Q02108GUCY1A10.4912.96.71 × 10−50.00338228Guanylate cyclase soluble subunit alpha-1
44O75439PMPCB1.3813.857.10 × 10−50.00342866Mitochondrial-processing peptidase subunit beta
45Q7Z4Q2HEATR30.4412.817.36 × 10−50.00342866HEAT repeat-containing protein 3
46Q13884SNTB1−0.3113.217.37 × 10−50.00342866Beta-1-syntrophin
47Q9Y266NUDC1.413.077.57 × 10−50.00342866Nuclear migration protein nudC
48P42025ACTR1B0.5212.997.60 × 10−50.00342866Beta-centractin
49Q04323UBXN12.3613.618.12 × 10−50.00359083UBX domain-containing protein 1
50P83881; Q969Q0RPL36A; RPL36AL1.2413.668.57 × 10−50.0037112760S ribosomal protein L36a; 60S ribosomal protein L36a-like
51Q86WV1SKAP11.5713.519.38 × 10−50.00398485Src kinase-associated phosphoprotein 1
52P10155RO600.5113.529.58 × 10−50.0039920360 kDa SS-A/Ro ribonucleoprotein
53P62877RBX13.2714.680.000105250.00428208E3 ubiquitin-protein ligase RBX1
54P53041PPP5C0.9313.060.000106760.00428208Serine/threonine-protein phosphatase 5
55Q8N699MYCT11.3113.320.000110190.00433936Myc target protein 1
56Q15102PAFAH1B31.6913.310.000114440.00438431Platelet-activating factor acetylhydrolase IB subunit gamma
57O00483NDUFA41.9514.820.000115380.00438431Cytochrome c oxidase subunit NDUFA4
58Q86YP4GATAD2A2.4514.080.0001180.00440654Transcriptional repressor p66-alpha
59Q9NRX4PHPT10.6312.90.00012940.0047335214 kDa phosphohistidine phosphatase
60O75116ROCK2−0.2713.280.000131120.00473352Rho-associated protein kinase 2
61P55265ADAR−0.3613.040.000134910.00479042Double-stranded RNA-specific adenosine deaminase
62P16333NCK11.0513.170.000138310.00480724Cytoplasmic protein NCK1
63P31645SLC6A41.5713.260.000139820.00480724Sodium-dependent serotonin transporter
64Q9UK45LSM74.3915.020.000144120.00486792U6 snRNA-associated Sm-like protein LSm7
65P0DP23; P0DP24; P0DP25CALM1; CALM2; CALM30.4913.840.000146080.00486792Calmodulin-1; Calmodulin-2; Calmodulin-3
66P78406RAE1−0.3613.070.000149530.00490735mRNA export factor
67O95433AHSA10.9913.170.00016340.00528231Activator of 90 kDa heat shock protein ATPase homolog 1
68Q9BQ61TRIR0.7812.890.000165980.00528682Telomerase RNA component interacting RNase
69P04350TUBB4A0.5612.940.000171330.0053782Tubulin beta-4A chain
70P02751FN1−0.313.70.00021190.00655692Fibronectin
71Q13363CTBP10.4213.340.000256160.00781477C-terminal-binding protein 1
72Q7LBR1CHMP1B2.3213.740.000270020.00805526Charged multivesicular body protein 1b
73P42285MTREX0.5212.850.000271480.00805526Exosome RNA helicase MTR4
74O00422SAP182.514.610.000281820.00823363Histone deacetylase complex subunit SAP18
75O00193SMAP0.4312.870.00028510.00823363Small acidic protein
76Q9Y5X3SNX5−0.5612.780.000367230.01046605Sorting nexin-5
77P20339RAB5A0.7114.80.000373750.01051357Ras-related protein Rab-5A
78P62273RPS290.5213.310.000380650.0105704440S ribosomal protein S29
79P46379BAG60.9113.180.000400040.01096815Large proline-rich protein BAG6
80O95139NDUFB60.9913.250.000406210.01099807NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 6
81P63165SUMO1213.750.000423950.01133665Small ubiquitin-related modifier 1
82P27338MAOB−0.313.330.000432390.01142134Amine oxidase [flavin-containing] B
83Q9H4G4GLIPR20.5415.520.000457830.0117166Golgi-associated plant pathogenesis-related protein 1
84Q96EY8MMAB0.4712.960.000458940.0117166Corrinoid adenosyltransferase
85O00487PSMD141.1213.340.000467040.011716626S proteasome non-ATPase regulatory subunit 14
86P04424ASL0.4513.020.000469850.0117166Argininosuccinate lyase
87Q96K37SLC35E1−0.4412.860.000475210.0117166Solute carrier family 35 member E1
88A5YKK6CNOT10.4612.80.000478140.0117166CCR4-NOT transcription complex subunit 1
89P62306SNRPF1.1414.310.000484710.0117166Small nuclear ribonucleoprotein F
90P54578USP140.5113.490.000486840.0117166Ubiquitin carboxyl-terminal hydrolase 14
91Q9UIA9XPO72.1414.440.000503460.01188018Exportin-7
92P49959MRE11−0.312.90.000504610.01188018Double-strand break repair protein MRE11
93P18206VCL−0.3214.870.00052650.01226224Vinculin
94O43290SART11.3213.350.000545980.01258082U4/U6.U5 tri-snRNP-associated protein 1
95O95819MAP4K40.5714.630.000592750.01351476Mitogen-activated protein kinase kinase kinase kinase 4
96Q5T1M5FKBP150.4813.350.000601150.01356334FK506-binding protein 15
97P02765AHSG−0.7418.910.000618440.0137706Alpha-2-HS-glycoprotein
98O00170AIP−0.3513.190.000623050.0137706AH receptor-interacting protein
99Q12907LMAN20.5814.570.00062960.01377496Vesicular integral-membrane protein VIP36
100Q8NFV4ABHD110.3912.850.000656160.01421252Protein ABHD11
101P63000RAC1−0.2615.490.000666360.01429044Ras-related C3 botulinum toxin substrate 1
102Q8NCG7DAGLB0.5212.870.0007270.01543803Sn1-specific diacylglycerol lipase beta
103Q9BQE9BCL7B2.0613.520.000785040.0165087B-cell CLL/lymphoma 7 protein family member B
104P61964WDR51.5813.480.000807430.01675991WD repeat-containing protein 5
105Q9Y3B7MRPL110.8313.360.000812460.0167599139S ribosomal protein L11, mitochondrial
106Q8IX12CCAR10.8713.450.000846940.01730627Cell division cycle and apoptosis regulator protein 1
107Q01658DR11.613.340.00087220.01764242Protein Dr1
108Q8IVB4SLC9A90.7213.010.000879680.01764242Sodium/hydrogen exchanger 9
109P12829MYL41.0113.410.000906530.01801412Myosin light chain 4
110Q99961SH3GL1−0.3413.410.000923810.01819059Endophilin-A2
111Q9UDW1UQCR100.4612.950.000951470.01856648Cytochrome b-c1 complex subunit 9
112O60831PRAF20.7913.170.000968030.01872093PRA1 family protein 2
113Q9UK76JPT10.3613.010.000990480.01898573Jupiter microtubule associated homolog 1
114P41226UBA7−0.2813.150.001010870.01910907Ubiquitin-like modifier-activating enzyme 7
115O15173PGRMC20.313.430.001014560.01910907Membrane-associated progesterone receptor component 2
116O95182NDUFA71.0913.780.001025170.01914249NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 7
117P62633CNBP0.8713.430.001047340.01928702Cellular nucleic acid-binding protein
118Q9UIG0BAZ1B1.413.310.001050720.01928702Tyrosine-protein kinase BAZ1B
119O95168NDUFB41.414.470.001095990.01994894NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 4
120Q93008USP9X−0.3713.010.0011150.02012574Probable ubiquitin carboxyl-terminal hydrolase FAF-X
121Q8WXF1PSPC10.4313.620.00114140.02040483Paraspeckle component 1
122Q96JB5CDK5RAP31.4913.690.00114930.02040483CDK5 regulatory subunit-associated protein 3
123Q6DD87ZNF7870.4512.840.001160820.02044178Zinc finger protein 787
124Q7Z6Z7HUWE1−0.2412.950.001177460.02056758E3 ubiquitin-protein ligase HUWE1
125P62993GRB20.3514.040.00119380.02068617Growth factor receptor-bound protein 2
126P68402PAFAH1B20.4412.910.001227820.0210104Platelet-activating factor acetylhydrolase IB subunit beta
127Q7RTV0PHF5A1.9514.440.001231910.0210104PHD finger-like domain-containing protein 5A
128O75368SH3BGRL0.5814.020.001253090.02120469SH3 domain-binding glutamic acid-rich-like protein
129Q9Y4L1HYOU1−0.2513.330.001272330.02136329Hypoxia up-regulated protein 1
130Q9P035HACD3−0.2913.020.001287890.02145828Very-long-chain (3R)-3-hydroxyacyl-CoA dehydratase 3
131Q9Y333LSM2−0.3312.850.00131310.0215621U6 snRNA-associated Sm-like protein LSm2
132Q8TBC4UBA31.9713.450.001323730.0215621NEDD8-activating enzyme E1 catalytic subunit
133Q9C0C9UBE2O−0.2713.080.001323990.0215621(E3-independent) E2 ubiquitin-conjugating enzyme
134Q92542NCSTN1.0313.260.001470970.02366738Nicastrin
135Q15056EIF4H0.6213.810.001475110.02366738Eukaryotic translation initiation factor 4H
136O95299NDUFA10−0.2212.90.001486830.02368002NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 10, mitochondrial
137P30049ATP5F1D1.9615.010.00154320.02421028ATP synthase subunit delta, mitochondrial
138Q9H3P7ACBD30.9413.220.001547520.02421028Golgi resident protein GCP60
139Q96EP5DAZAP10.7814.450.001555650.02421028DAZ-associated protein 1
140O00299CLIC1−0.2616.010.001571570.02421028Chloride intracellular channel protein 1
141Q9Y5Z4HEBP20.2313.050.001576020.02421028Heme-binding protein 2
142P49585PCYT1A0.6213.120.001588220.02422601Choline-phosphate cytidylyltransferase A
143P19784CSNK2A21.4413.10.00164450.024909Casein kinase II subunit alpha’
144A0AVT1UBA6−0.2612.990.001687550.02533419Ubiquitin-like modifier-activating enzyme 6
145P23141CES10.3813.730.001695960.02533419Liver carboxylesterase 1
146P21333FLNA−0.2815.690.00172930.02565525Filamin-A
147P04075ALDOA−0.2515.310.001869560.02749417Fructose-bisphosphate aldolase A
148Q8IZP0ABI10.2812.970.001878640.02749417Abl interactor 1
149Q9C0E8LNPK0.6713.080.001943980.02818302Endoplasmic reticulum junction formation protein lunapark
150P00747PLG−0.3513.730.001952930.02818302Plasminogen
151P06239LCK0.4213.250.001966210.02818302Tyrosine-protein kinase Lck
152O14735CDIPT1.1113.530.001978830.02818302CDP-diacylglycerol-inositol 3-phosphatidyltransferase
153O00154ACOT7−0.3512.910.001990770.02818302Cytosolic acyl coenzyme A thioester hydrolase
154Q13435SF3B2−0.2713.780.002007780.02823925Splicing factor 3B subunit 2
155Q14642INPP5A0.7913.030.002027160.02828249Inositol polyphosphate-5-phosphatase A
156P62328TMSB4X−0.5114.40.002036970.02828249Thymosin beta-4
157Q9Y3Y2CHTOP0.6613.880.002062870.02830664Chromatin target of PRMT1 protein
158Q8NBQ5HSD17B110.5813.120.002064840.02830664Estradiol 17-beta-dehydrogenase 11
159Q15833STXBP2−0.2113.630.002090620.0284634Syntaxin-binding protein 2
160Q32P28P3H10.4612.870.002102560.0284634Prolyl 3-hydroxylase 1
161Q9Y5S9RBM8A0.4614.020.002146390.02887632RNA-binding protein 8A
162Q8N392ARHGAP18−0.3613.810.002171550.02903448Rho GTPase-activating protein 18
163P04234CD3D1.0713.670.002219570.02942847T-cell surface glycoprotein CD3 delta chain
164P61803DAD10.9714.320.002232530.02942847Dolichyl-diphosphooligosaccharide-protein glycosyltransferase subunit DAD1
165Q01518CAP1−0.2115.060.002241780.02942847Adenylyl cyclase-associated protein 1
166P08697SERPINF21.2615.880.00227690.02970948Alpha-2-antiplasmin
167Q8WXF7ATL10.3512.870.002337050.03031163Atlastin-1
168Q6P1M0SLC27A40.6112.780.002358430.03040693Long-chain fatty acid transport protein 4
169Q9P1F3ABRACL−0.212.790.002409180.03082475Costars family protein ABRACL
170P01911HLA-DRB10.6414.510.00242110.03082475HLA class II histocompatibility antigen, DRB1-15 beta chain
171Q16643DBN1−0.313.490.002433530.03082475Drebrin
172P11908PRPS21.4313.640.002450040.03085338Ribose-phosphate pyrophosphokinase 2
173Q99439CNN2−0.2914.250.002501220.03131584Calponin-2
174Q86UT6NLRX1−0.3512.840.002519820.03134018NLR family member X1
175Q9NRL3STRN41.4314.380.00253210.03134018Striatin-4
176Q9NZ45CISD1−0.4212.920.002579350.03174358CDGSH iron-sulfur domain-containing protein 1
177P84103SRSF30.5413.920.002701480.03305881Serine/arginine-rich splicing factor 3
178Q8TF42UBASH3B−0.3113.220.002845780.03451842Ubiquitin-associated and SH3 domain-containing protein B
179P98194ATP2C1−0.3513.110.002852630.03451842Calcium-transporting ATPase type 2C member 1
180O95870ABHD16A−0.2213.10.002915590.03496139Phosphatidylserine lipase ABHD16A
181Q06187BTK−0.3213.210.002921520.03496139Tyrosine-protein kinase BTK
182Q86V81ALYREF0.3313.650.003010630.03582983THO complex subunit 4
183Q96HC4PDLIM5−0.2413.110.003115110.03674076PDZ and LIM domain protein 5
184P50502ST13−0.3914.490.003136020.03674076Hsc70-interacting protein
185O75874IDH1−0.1813.260.003138060.03674076Isocitrate dehydrogenase [NADP] cytoplasmic
186Q9UIQ6LNPEP−0.2613.040.003164210.03684776Leucyl-cystinyl aminopeptidase
187P05204HMGN20.5313.650.00319310.03698535Non-histone chromosomal protein HMG-17
188Q9UBE0SAE11.7713.380.003254040.03749071SUMO-activating enzyme subunit 1
189Q27J81INF2−0.2513.290.003294490.03764605Inverted formin-2
190P17568NDUFB70.913.50.003302290.03764605NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 7
191P42226STAT60.4712.820.00339130.03818853Signal transducer and activator of transcription 6
192Q96A72MAGOHB0.5113.650.003391340.03818853Protein mago nashi homolog 2
193Q969T9WBP2−0.4415.050.003402760.03818853WW domain-binding protein 2
194P25098GRK2−0.2413.10.003454880.03857356Beta-adrenergic receptor kinase 1
195Q9UHA4LAMTOR30.3812.830.003523140.03898614Ragulator complex protein LAMTOR3
196O75558STX11−0.2613.120.003527830.03898614Syntaxin-11
197Q9NS28RGS180.6613.350.003570690.03925947Regulator of G-protein signaling 18
198P07737PFN1−0.3816.530.00361220.03951523Profilin-1
199P13807GYS10.913.130.003632190.03953429Glycogen [starch] synthase, muscle
200P28838LAP3−0.2813.870.003822860.04140152Cytosol aminopeptidase
201Q8NBS9TXNDC5−0.2713.770.003907720.0421101Thioredoxin domain-containing protein 5
202P04114APOB−0.3713.830.003962570.04248971Apolipoprotein B-100
203Q92597NDRG10.6913.960.004019890.04289198Protein NDRG1
204Q10472GALNT11.113.60.004064430.04315433Polypeptide N-acetylgalactosaminyltransferase 1
205P16930FAH−0.3613.130.004084320.04315433Fumarylacetoacetase
206Q9Y2T2AP3M1−0.3513.230.004108910.04320338AP-3 complex subunit mu-1
207Q01813PFKP−0.213.410.004160370.04353312ATP-dependent 6-phosphofructokinase, platelet type
208Q00577PURA0.6913.20.004192250.04363857Transcriptional activator protein Pur-alpha
209O15143ARPC1B−0.2214.670.004210740.04363857Actin-related protein 2/3 complex subunit 1B
210Q7KZF4SND1−0.1613.840.004344830.04458141Staphylococcal nuclease domain-containing protein 1
211Q7L576CYFIP1−0.2313.530.004363860.04458141Cytoplasmic FMR1-interacting protein 1
212Q3ZCW2LGALSL−0.2913.070.004369390.04458141Galectin-related protein
213Q8N8A2ANKRD440.7113.040.004384040.04458141Serine/threonine-protein phosphatase 6 regulatory ankyrin repeat subunit B
214Q5RKV6EXOSC60.8813.390.004642160.04698563Exosome complex component MTR3
215P61952GNG111.6914.010.004717230.04752336Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-11
216P16157ANK1−0.4313.090.00484620.04859658Ankyrin-1
217Q9NZ01TECR0.9413.350.004889120.04880109Very-long-chain enoyl-CoA reductase
218Q14644RASA3−0.1813.250.004939510.04886889Ras GTPase-activating protein 3
219Q13813SPTAN1−0.213.120.004954710.04886889Spectrin alpha chain, non-erythrocytic 1
220Q9Y262EIF3L0.2513.390.00496360.04886889Eukaryotic translation initiation factor 3 subunit L
221P62701RPS4X0.1913.690.004995050.0489560440S ribosomal protein S4, X isoform
222Q8N4P3HDDC30.3512.820.0050640.04901974Guanosine-3′,5′-bis(diphosphate) 3′-pyrophosphohydrolase MESH1
223O75165DNAJC13−0.2512.880.005069320.04901974DnaJ homolog subfamily C member 13
224Q9P2X0DPM30.813.050.005069450.04901974Dolichol-phosphate mannosyltransferase subunit 3
225Q13057COASY−0.2512.880.005164730.04971913Bifunctional coenzyme A synthase
226Q9Y3L3SH3BP1−0.3213.550.005231760.04993232SH3 domain-binding protein 1
227O75083WDR1−0.1714.370.005232980.04993232WD repeat-containing protein 1
Table 4. Differentially expressed proteins between converter and non-converter at Visit 2.
Table 4. Differentially expressed proteins between converter and non-converter at Visit 2.
Sr #Protein AccessionsGeneslogFCAveExprp-Valueadj.P.ValProtein Descriptions
1P42025ACTR1B0.7912.999.15 × 10−91.98 × 10−5Beta-centractin
2P02656APOC32.0314.741.86 × 10−70.000202Apolipoprotein C-III
3P12829MYL41.6313.413.63 × 10−70.000262Myosin light chain 4
4Q08380LGALS3BP1.1613.25.15 × 10−70.000279Galectin-3-binding protein
5Q8N386LRRC251.1913.336.85 × 10−70.000297Leucine-rich repeat-containing protein 25
6O95168NDUFB42.214.479.16 × 10−70.000331NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 4
7P08697SERPINF22.0515.881.69 × 10−60.000408Alpha-2-antiplasmin
8Q02108GUCY1A10.5912.91.82 × 10−60.000408Guanylate cyclase soluble subunit alpha-1
9Q15370ELOB0.9913.111.85 × 10−60.000408Elongin-B
10O00483NDUFA42.4514.821.88 × 10−60.000408Cytochrome c oxidase subunit NDUFA4
11Q92597NDRG11.1513.963.48 × 10−60.000685Protein NDRG1
12P51809VAMP70.5612.944.36 × 10−60.000772Vesicle-associated membrane protein 7
13P04424ASL0.613.024.63 × 10−60.000772Argininosuccinate lyase
14P04217A1BG2.3616.035.69 × 10−60.000814Alpha-1B-glycoprotein
15P30048PRDX30.8214.715.84 × 10−60.000814Thioredoxin-dependent peroxide reductase, mitochondrial
16P04350TUBB4A0.6812.946.23 × 10−60.000814Tubulin beta-4A chain
17P51570GALK10.9713.26.39 × 10−60.000814Galactokinase
18A6NHR9SMCHD1−0.3213.097.35 × 10−60.000885Structural maintenance of chromosomes flexible hinge domain-containing protein 1
19A8MWD9; P62308SNRPGP15; SNRPG0.9614.179.53 × 10−60.001087Putative small nuclear ribonucleoprotein G-like protein 15; Small nuclear ribonucleoprotein G
20P18669PGAM10.7914.771.05 × 10−50.001132Phosphoglycerate mutase 1
21P05543SERPINA71.3513.381.42 × 10−50.001465Thyroxine-binding globulin
22P49065ALB2.0115.741.79 × 10−50.001758Serum albumin
23Q02750MAP2K12.6415.391.89 × 10−50.001758Dual specificity mitogen-activated protein kinase kinase 1
24Q00577PURA1.0513.21.95 × 10−50.001758Transcriptional activator protein Pur-alpha
25Q9H8H3METTL7A1.3614.822.27 × 10−50.001967Methyltransferase-like protein 7A
26Q8IZ07ANKRD13A0.9212.92.58 × 10−50.002148Ankyrin repeat domain-containing protein 13A
27Q92769HDAC20.512.952.74 × 10−50.002199Histone deacetylase 2
28Q92530PSMF10.5712.93.55 × 10−50.002683Proteasome inhibitor PI31 subunit
29Q86X76NIT11.3213.353.59 × 10−50.002683Deaminated glutathione amidase
30P55795HNRNPH20.7214.13.79 × 10−50.002735Heterogeneous nuclear ribonucleoprotein H2
31C4AMC7; Q6VEQ5WASH3P; WASH2P0.8813.213.94 × 10−50.002751Putative WAS protein family homolog 3; WAS protein family homolog 2
32O95182NDUFA71.3713.784.53 × 10−50.003066NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 7
33P10412HIST1H1E−0.5417.514.71 × 10−50.003089Histone H1.4
34P05387RPLP20.8114.334.88 × 10−50.00310860S acidic ribosomal protein P2
35Q06587RING10.7612.946.84 × 10−50.004089E3 ubiquitin-protein ligase RING1
36Q8NFV4ABHD110.4612.856.90 × 10−50.004089Protein ABHD11
37P16401HIST1H1B−0.6916.826.98 × 10−50.004089Histone H1.5
38P17568NDUFB71.2313.57.45 × 10−50.004168NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 7
39Q8NHV1GIMAP70.78137.50 × 10−50.004168GTPase IMAP family member 7
40P15153RAC20.8614.688.65 × 10−50.004687Ras-related C3 botulinum toxin substrate 2
41P31483TIA10.6312.940.0001070.005643Nucleolysin TIA-1 isoform p40
42P27986PIK3R10.3312.910.0001140.005884Phosphatidylinositol 3-kinase regulatory subunit alpha
43Q13363CTBP10.4313.340.0001380.006932C-terminal-binding protein 1
44Q6DD87ZNF7870.5212.840.0001490.007156Zinc finger protein 787
45O95379TNFAIP80.613.020.0001560.007156Tumor necrosis factor alpha-induced protein 8
46P56279TCL1A1.2713.060.0001580.007156T-cell leukemia/lymphoma protein 1A
47Q9H9G7; Q9HCK5; Q9UL18AGO3; AGO4; AGO11.5813.520.0001590.007156Protein argonaute-3; Protein argonaute-4; Protein argonaute-1
48O00505KPNA31.0213.40.0001590.007156Importin subunit alpha-4
49P30043BLVRB0.6713.490.0001680.00736Flavin reductase (NADPH)
50Q9P2R7SUCLA2−0.4714.110.000170.00736Succinate-CoA ligase [ADP-forming] subunit beta, mitochondrial
51Q9NUQ9FAM49B0.3613.560.0001750.00736Protein FAM49B
52Q7Z4Q2HEATR30.4112.810.0001790.00736HEAT repeat-containing protein 3
53Q9Y6W5WASF20.6614.350.000180.00736Wiskott-Aldrich syndrome protein family member 2
54Q93050ATP6V0A10.5512.820.0001840.007376V-type proton ATPase 116 kDa subunit a isoform 1
55P43304GPD2−0.2913.50.0001870.007376Glycerol-3-phosphate dehydrogenase, mitochondrial
56Q16630CPSF60.413.580.0002010.007785Cleavage and polyadenylation specificity factor subunit 6
57Q96NY7; Q9NZA1CLIC6; CLIC51.9213.950.0002130.008057Chloride intracellular channel protein 6; Chloride intracellular channel protein 5
58P46379BAG60.9413.180.0002180.008057Large proline-rich protein BAG6
59B2RUZ4SMIM1−0.5413.360.0002190.008057Small integral membrane protein 1
60Q06323PSME10.514.20.0002480.008953Proteasome activator complex subunit 1
61P02760AMBP1.1116.260.0002570.009141Protein AMBP
62Q15907RAB11B−0.3914.990.0002660.009285Ras-related protein Rab-11B
63Q00059TFAM−0.3513.40.0002980.010133Transcription factor A, mitochondrial
64Q15287RNPS11.8314.510.0002990.010133RNA-binding protein with serine-rich domain 1
65Q13813SPTAN1−0.2613.120.0003250.01077Spectrin alpha chain, non-erythrocytic 1
66P14678; P63162SNRPB; SNRPN0.7914.220.0003280.01077Small nuclear ribonucleoprotein-associated proteins B and B’; Small nuclear ribonucleoprotein-associated protein N
67P02452COL1A11.2914.380.0003540.011289Collagen alpha-1(I) chain
68P05452CLEC3B1.2314.080.0003540.011289Tetranectin
69Q8NG11TSPAN14−0.4413.540.0003760.011791Tetraspanin-14
70P26641EEF1G−0.3814.60.0003990.012224Elongation factor 1-gamma
71O75746SLC25A12−0.313.140.0004010.012224Calcium-binding mitochondrial carrier protein Aralar1
72Q9Y333LSM2−0.3712.850.0004090.012267U6 snRNA-associated Sm-like protein LSm2
73P42285MTREX0.512.850.0004130.012267Exosome RNA helicase MTR4
74P62318SNRPD31.0215.60.000470.013767Small nuclear ribonucleoprotein Sm D3
75Q9BUJ2HNRNPUL10.4213.810.0004920.014049Heterogeneous nuclear ribonucleoprotein U-like protein 1
76Q8WXF7ATL10.412.870.0004990.014049Atlastin-1
77P20933AGA1.1614.150.0004990.014049N(4)-(beta-N-acetylglucosaminyl)-L-asparaginase
78P28838LAP3−0.3413.870.0005120.014231Cytosol aminopeptidase
79P32942ICAM3−0.5714.850.000520.014251Intercellular adhesion molecule 3
80Q96BM9ARL8A−0.4214.460.000530.014361ADP-ribosylation factor-like protein 8A
81Q9H098FAM107B1.8813.650.000580.015505Protein FAM107B
82Q8N699MYCT11.1313.320.0005890.015554Myc target protein 1
83O75116ROCK2−0.2413.280.00060.01557Rho-associated protein kinase 2
84Q9UIA9XPO72.0714.440.0006070.01557Exportin-7
85P34910EVI2B0.9413.60.0006110.01557Protein EVI2B
86O14980XPO1−0.2112.950.0006270.015784Exportin-1
87P23743DGKA−0.3113.180.0006380.015889Diacylglycerol kinase alpha
88P14543NID1−0.3513.680.0006820.016787Nidogen-1
89Q9UIQ6LNPEP−0.313.040.000720.017408Leucyl-cystinyl aminopeptidase
90Q32P28P3H10.512.870.000730.017408Prolyl 3-hydroxylase 1
91P06730EIF4E0.7713.270.0007380.017408Eukaryotic translation initiation factor 4E
92Q15008PSMD6−0.2212.960.0007470.01740826S proteasome non-ATPase regulatory subunit 6
93P10599TXN0.4513.660.0007470.017408Thioredoxin
94O95299NDUFA10−0.2312.90.0007780.017795NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 10, mitochondrial
95Q52LJ0FAM98B0.713.590.000780.017795Protein FAM98B
96O95819MAP4K40.5514.630.0007910.01784Mitogen-activated protein kinase kinase kinase kinase 4
97Q8IV53DENND1C0.6613.190.0008350.018443DENN domain-containing protein 1C
98P51884LUM2.0315.330.000850.018443Lumican
99P09917ALOX5−0.1912.90.0008510.018443Arachidonate 5-lipoxygenase
100P55957BID0.9813.870.0008510.018443BH3-interacting domain death agonist
101Q9BPX5ARPC5L0.713.690.0008840.018961Actin-related protein 2/3 complex subunit 5-like protein
102Q9NS28RGS180.7513.350.0009070.019263Regulator of G-protein signaling 18
103O00193SMAP0.3812.870.0009470.019304Small acidic protein
104O95544NADK0.8413.340.0009480.019304NAD kinase
105P11908PRPS21.5413.640.0009480.019304Ribose-phosphate pyrophosphokinase 2
106Q13057COASY−0.312.880.0009550.019304Bifunctional coenzyme A synthase
107Q9BY77POLDIP30.6412.950.0009570.019304Polymerase delta-interacting protein 3
108Q86YP4GATAD2A2.0314.080.0009630.019304Transcriptional repressor p66-alpha
109P17900GM2A0.9513.050.000980.019473Ganglioside GM2 activator
110Q8NI27THOC2−0.4212.970.001020.019964THO complex subunit 2
111P60660MYL6−0.3515.440.0010390.019964Myosin light polypeptide 6
112Q13045FLII−0.213.480.0010440.019964Protein flightless-1 homolog
113P14174MIF1.8415.480.0010490.019964Macrophage migration inhibitory factor
114Q86Y39NDUFA110.4813.480.0010510.019964NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 11
115P12109COL6A11.6614.750.0010740.020052Collagen alpha-1(VI) chain
116Q08722CD47−1.2115.130.0010770.020052Leukocyte surface antigen CD47
117Q9NVZ3NECAP20.613.390.0010830.020052Adaptin ear-binding coat-associated protein 2
118Q9BRA2TXNDC170.6513.950.0011210.020576Thioredoxin domain-containing protein 17
119Q16401PSMD5−0.5512.830.0012080.02199426S proteasome non-ATPase regulatory subunit 5
120P14625HSP90B1−0.2213.990.0012980.023421Endoplasmin
121Q8NCG7DAGLB0.4812.870.0013530.024214Sn1-specific diacylglycerol lipase beta
122Q15796SMAD20.8413.290.001380.024501Mothers against decapentaplegic homolog 2
123P01911HLA-DRB10.6614.510.0015030.02647HLA class II histocompatibility antigen, DRB1-15 beta chain
124P49773HINT10.3413.970.0016030.027994Histidine triad nucleotide-binding protein 1
125P84243H3-3A0.7112.90.0016530.028452Histone H3.3
126Q8IYM9TRIM22−0.3112.820.0016550.028452E3 ubiquitin-protein ligase TRIM22
127Q15056EIF4H0.613.810.0017340.029567Eukaryotic translation initiation factor 4H
128P04259KRT6B1.7913.90.001830.030966Keratin, type II cytoskeletal 6B
129Q13576IQGAP2−0.2213.260.0018650.031298Ras GTPase-activating-like protein IQGAP2
130Q93009USP7−0.2713.320.0018920.031298Ubiquitin carboxyl-terminal hydrolase 7
131P09543CNP−0.3412.780.0018930.0312982′,3′-cyclic-nucleotide 3′-phosphodiesterase
132P54709ATP1B30.3613.190.0019270.031614Sodium/potassium-transporting ATPase subunit beta-3
133Q8N4P3HDDC30.3812.820.0019610.031932Guanosine-3′,5′-bis(diphosphate) 3′-pyrophosphohydrolase MESH1
134P68402PAFAH1B20.4112.910.0020190.0324Platelet-activating factor acetylhydrolase IB subunit beta
135P22694PRKACB0.6213.10.0020190.0324cAMP-dependent protein kinase catalytic subunit beta
136P39687ANP32A0.3614.590.0020710.032871Acidic leucine-rich nuclear phosphoprotein 32 family member A
137Q9H3G5CPVL0.3913.610.0020790.032871Probable serine carboxypeptidase CPVL
138P62304SNRPE0.4312.950.0021220.033305Small nuclear ribonucleoprotein E
139P02749APOH0.7814.820.0021540.033305Beta-2-glycoprotein 1
140Q8N5M9JAGN10.4413.090.0021710.033305Protein jagunal homolog 1
141Q6IAA8LAMTOR1−0.312.720.002180.033305Ragulator complex protein LAMTOR1
142Q9Y3B7MRPL110.7413.360.0021830.03330539S ribosomal protein L11, mitochondrial
143Q96JB5CDK5RAP31.3713.690.0022880.034652CDK5 regulatory subunit-associated protein 3
144P18583SON1.113.470.002310.034748Protein SON
145Q9Y2T2AP3M1−0.3613.230.0023780.035172AP-3 complex subunit mu-1
146P49327FASN−0.2213.120.002380.035172Fatty acid synthase
147O14735CDIPT1.0713.530.0023950.035172CDP-diacylglycerol-inositol 3-phosphatidyltransferase
148P50851LRBA−0.2313.290.0024030.035172Lipopolysaccharide-responsive and beige-like anchor protein
149Q86VM9ZC3H18−0.3612.910.0024550.035476Zinc finger CCCH domain-containing protein 18
150Q9NZK5ADA2−0.1913.150.0024660.035476Adenosine deaminase 2
151P01042KNG10.7413.860.0024730.035476Kininogen-1
152P46926GNPDA1−0.3112.920.0024960.035565Glucosamine-6-phosphate isomerase 1
153Q9BUQ8DDX23−0.3912.960.002530.03569Probable ATP-dependent RNA helicase DDX23
154Q92506HSD17B8−0.5613.610.0025380.03569Estradiol 17-beta-dehydrogenase 8
155Q9P035HACD3−0.2713.020.0025850.036072Very-long-chain (3R)-3-hydroxyacyl-CoA dehydratase 3
156O43670ZNF2070.7113.660.0025980.036072BUB3-interacting and GLEBS motif-containing protein ZNF207
157P16109SELP−0.7114.990.0026920.037136P-selectin
158P07996THBS1−0.4415.630.0027590.037818Thrombospondin-1
159P51148RAB5C0.6113.940.0028390.038672Ras-related protein Rab-5C
160P30046DDT0.3213.570.0029670.040172D-dopachrome decarboxylase
161Q9BTT0ANP32E0.4413.670.0029970.040324Acidic leucine-rich nuclear phosphoprotein 32 family member E
162P01909HLA-DQA10.312.90.0030310.040522HLA class II histocompatibility antigen, DQ alpha 1 chain
163Q9H4I9SMDT10.6813.480.0030920.041051Essential MCU regulator, mitochondrial
164Q9Y5Z4HEBP20.2113.050.0031080.041051Heme-binding protein 2
165P42126ECI10.613.030.0031310.041095Enoyl-CoA delta isomerase 1, mitochondrial
166Q9C0C9UBE2O−0.2413.080.0031920.041121(E3-independent) E2 ubiquitin-conjugating enzyme
167P62857RPS280.6715.390.0031940.04112140S ribosomal protein S28
168Q02338BDH11.5214.410.0031950.041121D-beta-hydroxybutyrate dehydrogenase, mitochondrial
169Q86WV1SKAP11.1413.510.003210.041121Src kinase-associated phosphoprotein 1
170P28072PSMB60.3413.130.0032270.041121Proteasome subunit beta type-6
171O75947ATP5PD−0.3114.520.0032940.04173ATP synthase subunit d, mitochondrial
172P01344IGF21.2614.880.0033530.042227Insulin-like growth factor II
173Q96CX2KCTD120.6114.270.0034150.042752BTB/POZ domain-containing protein KCTD12
174P16150SPN−0.6615.380.0034440.042806Leukosialin
175P07477PRSS12.0617.290.0034580.042806Trypsin-1
176Q8IV08PLD30.4312.920.003510.043046Phospholipase D3
177P62277RPS130.3414.070.0035180.04304640S ribosomal protein S13
178P62195PSMC5−0.2812.860.0035490.04314626S proteasome regulatory subunit 8
179O95866MPIG6B1.0716.40.0035660.043146Megakaryocyte and platelet inhibitory receptor G6b
180Q13761RUNX30.4112.980.0036040.043363Runt-related transcription factor 3
181P08708RPS170.313.250.0036640.04352740S ribosomal protein S17
182P62330ARF60.8713.460.0036880.043527ADP-ribosylation factor 6
183P25789PSMA40.4213.690.0037010.043527Proteasome subunit alpha type-4
184P12236SLC25A6−0.2915.30.0037030.043527ADP/ATP translocase 3
185Q9NQG5RPRD1B−0.3713.010.0037180.043527Regulation of nuclear pre-mRNA domain-containing protein 1B
186P20340RAB6A−0.3514.330.0038160.044395Ras-related protein Rab-6A
187P17676CEBPB1.2613.570.0038330.044395CCAAT/enhancer-binding protein beta
188O60831PRAF20.6813.170.0038790.044515PRA1 family protein 2
189P62140PPP1CB0.9115.630.0038840.044515Serine/threonine-protein phosphatase PP1-beta catalytic subunit
190Q86UT6NLRX1−0.3312.840.0039260.044761NLR family member X1
191Q9Y3B2EXOSC10.4112.980.0039770.045099Exosome complex component CSL4
192O76074PDE5A−0.2713.360.0040520.045708cGMP-specific 3′,5′-cyclic phosphodiesterase
193Q9UII2ATP5IF1−0.3913.870.004170.046799ATPase inhibitor, mitochondrial
194Q99961SH3GL1−0.2913.410.0042310.047238Endophilin-A2
195O00487PSMD140.8813.340.0044190.04908226S proteasome non-ATPase regulatory subunit 14
196Q96K37SLC35E1−0.3512.860.0044640.049327Solute carrier family 35 member E1
Table 5. Differentially expressed proteins between converter and non-converter at Visit 1 and Visit 2.
Table 5. Differentially expressed proteins between converter and non-converter at Visit 1 and Visit 2.
Sr #Protein
Accessions
GeneslogFC.V1AveExpr.V1logFC.V2AveExpr.V2Protein Descriptions
1Q9Y3B2EXOSC10.84860112.980930.40650412.9809302Exosome complex component CSL4
2P55957BID1.59455313.865450.9824113.8654541BH3-interacting domain death agonist
3Q15370ELOB1.00145113.107070.99303413.1070734Elongin-B
4P17676CEBPB2.15990313.573771.25592713.5737735CCAAT/enhancer-binding protein beta
5Q8NHV1GIMAP70.90498112.995370.7790212.9953678GTPase IMAP family member 7
6Q92769HDAC20.52382112.946680.49836812.9466806Histone deacetylase 2
7Q9BY77POLDIP30.87411212.949390.64052712.949388Polymerase delta-interacting protein 3
8P01909HLA-DQA10.45570512.897880.29886112.8978826HLA class II histocompatibility antigen, DQ alpha 1 chain
9P02656APOC31.5786414.739722.03402514.7397213Apolipoprotein C-III
10P30048PRDX30.75172814.710940.82102114.7109441Thioredoxin-dependent peroxide reductase, mitochondrial
11P62857RPS280.98808915.386770.67067615.386765840S ribosomal protein S28
12Q02750MAP2K12.53559215.394212.64329815.394209Dual specificity mitogen-activated protein kinase kinase 1
13Q8IV08PLD30.6277112.920780.4325212.9207843Phospholipase D3
14P51148RAB5C0.85708613.936790.6082413.9367931Ras-related protein Rab-5C
15O14980XPO1−0.25590812.95095−0.21178312.9509484Exportin-1
16Q02108GUCY1A10.48511212.903770.5885212.9037683Guanylate cyclase soluble subunit alpha-1
17Q7Z4Q2HEATR30.4429412.807010.40896612.8070147HEAT repeat-containing protein 3
18P42025ACTR1B0.51513612.992520.7934412.9925248Beta-centractin
19Q86WV1SKAP11.57396913.509381.13638813.5093818Src kinase-associated phosphoprotein 1
20Q8N699MYCT11.305513.316341.12639513.3163412Myc target protein 1
21O00483NDUFA41.94631314.824272.4473514.8242724Cytochrome c oxidase subunit NDUFA4
22Q86YP4GATAD2A2.44676614.079852.03002414.0798512Transcriptional repressor p66-alpha
23O75116ROCK2−0.26960213.27577−0.23516113.275773Rho-associated protein kinase 2
24P04350TUBB4A0.55669912.936620.67636312.936615Tubulin beta-4A chain
25Q13363CTBP10.41821213.336990.4311513.3369899C-terminal-binding protein 1
26P42285MTREX0.52309112.850320.49744412.8503171Exosome RNA helicase MTR4
27O00193SMAP0.43286712.867670.38434212.8676676Small acidic protein
28P46379BAG60.91091313.181820.94054813.1818226Large proline-rich protein BAG6
29O00487PSMD141.11768113.34010.87919813.340100526S proteasome non-ATPase regulatory subunit 14
30P04424ASL0.44744813.024120.59838713.0241187Argininosuccinate lyase
31Q96K37SLC35E1−0.44273312.86068−0.34835412.860683Solute carrier family 35 member E1
32Q9UIA9XPO72.1424414.441992.07422514.4419886Exportin-7
33O95819MAP4K40.5749314.634260.55137614.6342571Mitogen-activated protein kinase kinase kinase kinase 4
34Q8NFV4ABHD110.39420912.854930.46112912.8549305Protein ABHD11
35Q8NCG7DAGLB0.51579112.869690.47889112.8696866Sn1-specific diacylglycerol lipase beta
36Q9Y3B7MRPL110.83326713.357510.74441813.357508139S ribosomal protein L11, mitochondrial
37P12829MYL41.01123113.407681.62533613.40768Myosin light chain 4
38Q99961SH3GL1−0.34108813.4084−0.28637913.4084045Endophilin-A2
39O60831PRAF20.79331113.169340.6758213.1693404PRA1 family protein 2
40O95182NDUFA71.09282413.775691.36882813.7756949NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 7
41O95168NDUFB41.40330714.47172.19814114.4716973NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 4
42Q96JB5CDK5RAP31.48740813.686591.36552913.6865904CDK5 regulatory subunit-associated protein 3
43Q6DD87ZNF7870.44966212.836110.52482912.8361067Zinc finger protein 787
44P68402PAFAH1B20.44269612.912730.41440112.9127301Platelet-activating factor acetylhydrolase IB subunit beta
45Q9P035HACD3−0.29397713.0163−0.269313.0163005Very-long-chain (3R)-3-hydroxyacyl-CoA dehydratase 3
46Q9Y333LSM2−0.33476612.85472−0.36553712.8547206U6 snRNA-associated Sm-like protein LSm2
47Q9C0C9UBE2O−0.27094913.07538−0.24315313.075381(E3-independent) E2 ubiquitin-conjugating enzyme
48Q15056EIF4H0.62360513.812990.60365213.8129855Eukaryotic translation initiation factor 4H
49O95299NDUFA10−0.21876612.90348−0.22877312.9034786NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 10, mitochondrial
50Q9Y5Z4HEBP20.23125113.050450.2117513.0504482Heme-binding protein 2
51O14735CDIPT1.11177413.534971.07198513.5349659CDP-diacylglycerol-inositol 3-phosphatidyltransferase
52Q32P28P3H10.45867812.867890.49969612.8678948Prolyl 3-hydroxylase 1
53P08697SERPINF21.25598815.878482.05346415.8784812Alpha-2-antiplasmin
54Q8WXF7ATL10.34822712.866940.3965512.8669358Atlastin-1
55P01911HLA-DRB10.64190114.507140.66327414.507142HLA class II histocompatibility antigen, DRB1-15 beta chain
56P11908PRPS21.42542913.637231.54126513.6372317Ribose-phosphate pyrophosphokinase 2
57Q86UT6NLRX1−0.35188412.83793−0.32937412.8379265NLR family member X1
58Q9UIQ6LNPEP−0.2649413.0362−0.30207213.0361981Leucyl-cystinyl aminopeptidase
59P17568NDUFB70.9010613.501541.23117613.5015444NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 7
60Q9NS28RGS180.65970113.352560.7468113.352561Regulator of G-protein signaling 18
61P28838LAP3−0.2795513.87283−0.33548313.8728298Cytosol aminopeptidase
62Q92597NDRG10.68634313.958791.15206913.9587948Protein NDRG1
63Q9Y2T2AP3M1−0.34723913.2346−0.36320713.2345966AP-3 complex subunit mu-1
64Q00577PURA0.68603413.199651.05015613.1996452Transcriptional activator protein Pur-alpha
65Q13813SPTAN1−0.19862813.121−0.25525513.1210042Spectrin alpha chain, non-erythrocytic 1
66Q8N4P3HDDC30.35161312.820650.38485412.8206512Guanosine-3′,5′-bis(diphosphate) 3′-pyrophosphohydrolase MESH1
67Q13057COASY−0.2548212.88284−0.29996412.8828366Bifunctional coenzyme A synthase
Table 6. Differentially expressed proteins between Visit 1 and Visit 2 in converter and non-converter.
Table 6. Differentially expressed proteins between Visit 1 and Visit 2 in converter and non-converter.
Sr #GroupDifferentially Expressed Proteins
1ConverterCOX5A,MT-CO2,VASP,C3,LSM3,EIF5A,GC,BID,ALB,ZNF207,AHSG,RAB32,UQCRH,F2,CMAS,A2M,SH3BGRL,AFP,SERPINF1,SERPINC1,BANF1,CALM1;CALM2;CALM3,GRB2,SAP18,UQCRQ,WASF2,ISOC1,AHNAK,C4A,ADD3,CNN2,SLTM,HIST1H1E,SF3B2,GLIPR2,FN1,LPCAT3,MTPN,COX7A2,SKP1,ABCC4,RPS26,PRPS1,ITIH2,HBA2,NONO,RAB6A,OGDH,EXOSC1,SNRPF,UQCR10,RAB11B,USP14,PAFAH1B3,ITIH3,RAE1,SLC30A7,U2AF2,RBM8A,COX6B1,GP1BA,WARS,GIMAP4,DDT,DNAJC13,MYCT1,ARPC3,SMARCC2,ENO2,HCLS1,APOB,PPP1CA,VAT1,RPL31,FBLN1,BLVRA,COL1A1,CAB39,AK2,OSBPL8,CTSB,CNDP2,TPD52L2,LTA4H,TUFM,ARF3;ARF1,ACTG1,PCYT1A,SUCLA2,SNX2,ST13,LAMTOR4,LMAN2,CLEC1B,SYNGR2,RAB18,NDUFA10,PCMT1,PDXK,COL6A1,SARS,ANXA11,NDUFB6,TRAF3IP3,WAS,RAB3D,ZYX,SLC9A3R1,DAD1,UBXN1,TFAM,SASH3,PGK1,TMPO,G3BP1,ALDOA,HM13,RNH1,BIN2,RPL36A;RPL36AL,PSMC2,ACO2,APOH,CEBPB,RPL9P8,TCP1,HNRNPA3,RBX1,PSIP1,GATD3B;GATD3A,PAK2,HSD17B11,HIST1H2BJ,EEF1A1;EEF1A1P5,SCP2,MRE11,COX5B,CHCHD2,IGFBP2,MYL6,NUDC,RO60,PNKD,RAB6B,SART1,PLPBP,DTD1,SRP9,MAGOHB,GART,INPP5A,BAZ1B,COL1A2,MAT2A,ABRACL,CHMP1B,PRDX1,JPT1,HLA-DQA1
2NonConverterOSBPL8
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zafarullah, M.; Li, J.; Salemi, M.R.; Phinney, B.S.; Durbin-Johnson, B.P.; Hagerman, R.; Hessl, D.; Rivera, S.M.; Tassone, F. Blood Proteome Profiling Reveals Biomarkers and Pathway Alterations in Fragile X PM at Risk for Developing FXTAS. Int. J. Mol. Sci. 2023, 24, 13477. https://doi.org/10.3390/ijms241713477

AMA Style

Zafarullah M, Li J, Salemi MR, Phinney BS, Durbin-Johnson BP, Hagerman R, Hessl D, Rivera SM, Tassone F. Blood Proteome Profiling Reveals Biomarkers and Pathway Alterations in Fragile X PM at Risk for Developing FXTAS. International Journal of Molecular Sciences. 2023; 24(17):13477. https://doi.org/10.3390/ijms241713477

Chicago/Turabian Style

Zafarullah, Marwa, Jie Li, Michelle R. Salemi, Brett S. Phinney, Blythe P. Durbin-Johnson, Randi Hagerman, David Hessl, Susan M. Rivera, and Flora Tassone. 2023. "Blood Proteome Profiling Reveals Biomarkers and Pathway Alterations in Fragile X PM at Risk for Developing FXTAS" International Journal of Molecular Sciences 24, no. 17: 13477. https://doi.org/10.3390/ijms241713477

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop