Next Article in Journal
Fusion Potential of Human Osteoclasts In Vitro Reflects Age, Menopause, and In Vivo Bone Resorption Levels of Their Donors—A Possible Involvement of DC-STAMP
Next Article in Special Issue
Ethionamide Preconditioning Enhances the Proliferation and Migration of Human Wharton’s Jelly-Derived Mesenchymal Stem Cells
Previous Article in Journal
Decoding Susceptibility to Respiratory Viral Infections and Asthma Inception in Children
Previous Article in Special Issue
Activation of Nrf2 by Natural Bioactive Compounds: A Promising Approach for Stroke?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Proteomic Characterization of the Olfactory Molecular Imbalance in Dementia with Lewy Bodies

by
Mercedes Lachén-Montes
1,2,3,
Naroa Mendizuri
1,3,
Domitille Schvartz
4,
Joaquín Fernández-Irigoyen
1,2,3,
Jean Charles Sánchez
4 and
Enrique Santamaría
1,2,3,*
1
Clinical Neuroproteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain
2
Proteored-ISCIII. Proteomics Platform, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain
3
IdiSNA, Navarra Institute for Health Research, Spain Irunlarrea 3, 31008 Pamplona, Spain
4
Translational Biomarker Group, Department of Medicine, University of Geneva, Rue Michel Servet 1, 1211 Geneve, Switzerland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(17), 6371; https://doi.org/10.3390/ijms21176371
Submission received: 29 July 2020 / Revised: 18 August 2020 / Accepted: 1 September 2020 / Published: 2 September 2020
(This article belongs to the Special Issue Molecular Research on Neurodegenerative Diseases 2.0)

Abstract

:
Olfactory dysfunction is one of the prodromal symptoms in dementia with Lewy bodies (DLB). However, the molecular pathogenesis associated with decreased smell function remains largely undeciphered. We generated quantitative proteome maps to detect molecular alterations in olfactory bulbs (OB) derived from DLB subjects compared to neurologically intact controls. A total of 3214 olfactory proteins were quantified, and 99 proteins showed significant alterations in DLB cases. Protein interaction networks disrupted in DLB indicated an imbalance in translation and the synaptic vesicle cycle. These alterations were accompanied by alterations in AKT/MAPK/SEK1/p38 MAPK signaling pathways that showed a distinct expression profile across the OB–olfactory tract (OT) axis. Taken together, our data partially reflect the missing links in the biochemical understanding of olfactory dysfunction in DLB.

1. Introduction

Dementia with Lewy bodies (DLB) constitutes one of the most common causes of dementia only behind Alzheimer’s disease (AD), accounting for approximately 25% of dementia cases [1,2]. Clinically, DLB is manifested by fluctuating cognition with severe variation in attention, recurrent formed and detailed visual hallucinations, and spontaneous parkinsonism motor features. Other common characteristics are temporary consciousness loss, delusions, syncope, repeated falls, systematized hallucinations and neuroleptic sensitivity [2,3,4,5]. In general, all these symptoms are preceded by rapid eye movement sleep disorder, psychiatric symptoms, dysautonomia together with occipital hypo-metabolism, cognitive impairment and smell loss [6,7]. Olfactory impairment has been demonstrated in a plethora of neurological disorders, including AD, Parkinson’s disease (PD) and DLB [8,9]. Although not currently implemented in the clinic, olfactory assessment has been postulated as a promising tool for the differential diagnosis of AD and DLB [10,11,12,13,14]. Previous clinicopathological studies have shown that olfactory dysfunction is associated with Lewy body pathology in limbic and neocortical regions [15], and to date, most of the studies have revealed that olfactory deficits are more severe in DLB than in AD [8,13,14,16]. However, despite the great attention that has caught in the last decade, the sense of smell is often undermine in the clinical diagnosis, and clinicians rarely test this deficit in patients with suspicion of neurological disease.
DLB is pathologically characterized by the presence of Lewy bodies and Lewy neurites in the brainstem, limbic system, and cortical areas [17]. Those are insoluble aggregates of abnormal α-synuclein, which is phosphorylated, nitrated and truncated, producing oligomeric species that afterwards constitute the so-called Lewy bodies and Lewy neurites [18,19,20,21]. Thus, together with Parkinson’s disease (PD) and others, DLB is classified among α-synucleopathies. Importantly, although some controversy still exists, DLB is differentiated from PD dementia (PDD) in the cognitive impairment onset. Following the international guidelines, DLB is diagnosed when cognitive impairment precedes parkinsonian motor symptoms or begins within one year from its onset [22]. On the other hand, cognitive impairments develop after a well-established parkinsonism in PDD patients [23].
Lewy pathology has also been found in the olfactory system, including the olfactory bulb (OB) and the anterior olfactory nucleus (AON) [24,25]. Recent reports have demonstrated the presence of α-synuclein pathology in the OB in 97% of DLB cases [24]. Interestingly, the Braak staging model has suggested that α-synuclein pathology starts at the OB and the AON, progressing afterwards in a caudorostral direction through vulnerable interconnected neurons [26]. This so-called prion hypothesis suggests that the formation of α-synuclein does not occur in a cell-autonomous mechanism. Instead, this process occurs in a determined number of cells and then spreads to the neighboring neuronal cells, reaching distant brain regions [27,28]. Interestingly, this has been demonstrated by the α-synuclein inoculation in the OB and its posterior spreading through among 23 different brain regions [29]. Nevertheless, whether this cell-to-cell spreading mechanisms is the main responsible for the neurodegenerative process remains unknown. In addition, information regarding the OB molecular alterations occurring in consequence is still lacking.
The immense cellular heterogeneity that comprises the central nervous system still represents a big challenge for the study of brain-related disorders. Nowadays, high-throughput techniques such as proteomics have emerged as a valuable approach to deep into the complex molecular mechanisms that underlie the process of neurodegeneration [30,31,32]. Thus, tissue-based quantitative clinical proteomics has already been aimed to provide more insights into DLB pathology [33,34,35,36]. However, although olfactory proteomics has been successfully applied in neurodegenerative phenotypes in mice and humans [37,38,39,40,41,42,43,44,45,46], the olfactory proteostatic imbalance that occurs in DLB remains unknown. To our knowledge, this is the first study aiming to decipher the specific molecular mechanisms that underlie the process of neurodegeneration at the level of primary olfactory areas in DLB.

2. Results and Discussion

Due to the fact that DLB progression is accompanied by an acute olfactory dysfunction [8] and α-synuclein propagation from the OB has been proposed as a potential onset of this pathology [29], we have performed an in-depth quantitative proteomic study in combination with functional interaction data and biochemical approaches to determine the dysregulation of the OB proteome in DLB (Figure 1A). Post-mortem OB specimens were subjected to isobaric chemical tags (tandem mass tags, TMT) coupled to mass spectrometry (four cases/condition). Among 3214 quantified proteins (Supplementary Table S1), differential analysis revealed 99 deregulated proteins in OBs derived from DLB subjects compared to neurological intact controls (16 up-regulated and 83 downregulated proteins) (Table 1). It is important to note that due to the technical workflow used, we failed to accurately quantify many proteins expressed at low levels that might also participate in the DLB phenotype. Moreover, our study does not distinguish between the numerous cell types coexisting in the OB, so in principle, it is not possible to directly assign protein changes to a specific cell type. To partially overcome these drawbacks, and based on prior quantitative thresholding of RNA-seq abundance data, a cell type enrichment analysis across OB quantified proteome and the OB differential dataset was performed using previous characterized cell type-enriched marker genes derived from different OB cell populations [47]. As shown in Figure 1B, cluster-enriched genes corresponding to different neuronal subtypes are represented in the quantified OB proteome. Interestingly, part of the proteostatic alterations are specifically enriched to granular cells (PRUNE2, STXBP1, CAMK2A, PSD3, PENK, RPS27, CAMK2B, GRIA2, TUBB2A, PSIP1, PRKAR1B, SYNPR, NAPB), astrocytes (RPL37, RPL31), external plexiform layer (EPL) interneurons (SCAMP5), mitral/Tufted cells (RAB3B, SYNGR3, ATP1A3, SLC17A7, MYCBP2, NRXN1, SYP, ATP1B1, CD47, GRM1, REEP5, NCEH1, ATP6V0A1, SV2A, CNTNAP2), olfactory sensory neurons (OSN) (RPL22, RPL36, RPL23A, RPS25, RPL38, RPS2, RPL23), and periglomerular cells (LY6H, SLC32A1, RAB3A, ATP6V1A) (Figure 1C). All these data shed new light on the molecular disturbances that accompany the neurodegenerative process across each cellular homeostasis in DLB. With the aim to complement and validate quantitative proteome measurements, subsequent experiments were performed to check the steady-state levels of a subset of differential proteins using downstream assays. As shown in Figure 1D, PI16 and ALDH1B1 are up-regulated in DLB at the level of the OB, confirming the mass spectrometry data. While this is the first report linking PI16 with any neurodegenerative process, mitochondrial ALDH1B1 plays a crucial role concerning the oxidative metabolism of toxic aldehydes in the brain. Thus, the alteration of ALDH1B1 in the OB together with the recent emerging evidence for ALDH2 stands for its implication in the process of neurodegeneration [48].

2.1. Olfactory Protein Translation and Synaptic Function are Impaired in DLB

To characterize in detail the olfactory proteotype in DLB phenotypes, we performed proteome-scale interaction networks merging the 99 differential proteins detected at the level of the OB. As shown in Figure 2A, the protein interactome was mainly composed by specific protein clusters related to translation, unblocking of NMDA receptors, synaptic vesicle cycle and innate immune system. Principally, OB proteostatic imbalance affects translation-related processes, defined by the significant downregulation of a great number of ribosomal proteins (Figure 2A). In this context, alteration in mRNA expression of ribosomal proteins has been previously observed in frontal cortex from DLB patients [49], suggesting a common pathological mechanism in these two brain regions. Moreover, alterations in protein synthesis have also been related to the presence of α-synuclein oligomers in PD-related brain areas [50]. Accompanying the reduction in EEF1A2 levels, the disruption of OB protein synthesis was confirmed by the drop in the P70 S6K beta (ribosomal protein S6 kinase beta) (Figure 2B), a serine/threonine kinase involved in the selective translation of unique family of mRNAs coding the ribosomal proteins and elongation factors [51]. In accordance with our data, reduced protein levels of the highly homologous EEF1A1 isoform have been observed in the synaptic fraction of DLB brain specimens [52], suggesting a dysregulation of the synaptic protein translation in DLB. Accordingly, several types of dementia, including DLB, are characterized by an important loss of synaptic transmission [53,54]. Additional functional analyses have commonly pointed out potential alterations in olfactory synapsis-related processes (Figure 2A,C). These data are in agreement with alterations found at prefrontal cortical level across DLB and PDD pathologies, where a significant loss of multiple synaptic proteins has been reported [33]. In addition, among those synapsis-related deregulated proteins, Rab3b significantly downregulated in the OB of DLB subjects has been recently proposed as a predictive marker for cognitive decline in DLB patients [33].

2.2. Specific Olfactory Derangements in Survival Pathways in DLB

We performed additional proteome-scale interactive networks using the deregulated OB proteome. Using the Ingenuity Pathway Analysis (IPA) (QIAGEN Redwood City, www.qiagen.com/ingenuity), both AKT (protein kinase B) and ERK (extracellular signal-regulated kinase 1/2) appeared as main nodes in protein interactome maps (Figure 3A). Although we did not detected changes in the expression of these survival kinases in the proteomic approach, alterations in some of their targets may be in accordance with their potential alteration at the level of the OB in DLB cases. In fact, an activation of AKT is observed in DLB cases, in parallel with an increment in the phosphorylation of BAD (Bcl2-associated agonist of cell death) at Ser112 (Figure 3B). This serine is a canonical target of AKT, modulating apoptosis [55]. This phosphorylation phenomenon has been related with BAD binding to 14-3-3t protein, which sequesters BAD from BclxL [56]. Thus, the inhibition of the proapoptotic factor pBAD, together with the tendency to increase in BclxL protein suggests readjustments in the survival potential of the OB from DLB subjects. Previous reports have demonstrated a differential deregulation of multiple survival kinases in AD and PD at the level of the OB [39,40,42]. Subsequent experiments were performed to monitor upstream and downstream effectors of the following specific routes: (i) MAPK pathway: ERK1/2 (extracellular signal-regulated kinase 1/2) and MEK1/2 (mitogen-activated protein kinase kinase 1/2), (ii) SEK1 (mitogen-activated protein kinase kinase 4)-SAPK/JNK (stress-activated protein kinase/Jun-amino terminal kinase) axis, and (iii) the MKK3/6 (dual specificity mitogen-activated protein kinase kinase 3/6)-p38 MAPK axis. Although still under controversy, AKT and ERK have been previously related to α-synuclein deposits [57,58]. However, the expression profiling detected in DLB (Figure 3C) differs from the observed during AD or PD progression. No alterations in AKT signaling have been previously observed during both AD and PD stage-dependent OB analysis [39,42]. On the other hand, previous work has demonstrated the activation of olfactory ERK survival factor during AD [42]. Interestingly, although both PD and DLB are characterized by the presence of α-synuclein deposits, an opposite pattern concerning ERK protein levels was observed in PD at the level of the OB [39]. Moreover, while no alterations are found in upstream MEK in DLB OBs (Figure S1A), a significant increase was observed in advanced stages of PD [39]. Altogether, these results point out that olfactory AKT/MAPK pathways are differential molecular features between both alpha-synucleinopathies. Moreover, stress-responsive kinases targeted by activated AKT [59] were evaluated in DLB OBs. Only a clear increase in OB SEK1 levels was noticed in DLB (Figure 3C), while no appreciable changes were detected in its activated status (data not shown). On the contrary, an activation of olfactory SEK1 has been observed in AD subjects [42]. Otherwise, downstream targets such as SAPK/JNK (stress-activated protein kinase/Jun-amino terminal kinase) or PKAc (protein kinase A) were unchanged (Figure S1B,C). Although the p38 MAPK cascade has been previously related to PD pathogenesis [60], no specific information is available concerning its involvement in DLB. The p38 MAPK signaling route mediates numerous cell effects in response to external signals and interestingly, an early activation has been observed in AD OBs [42]. However, no such alterations have been observed for PD [39]. In our analysis, a significantly increment in p38 MAPK levels was observed in OB specimens derived from DLB cases (Figure 3C). These alterations were not observed in two of its main upstream kinases, MKK3 and MKK6 (dual specificity mitogen-activated protein kinase kinase 3/6) (Figure S1D).

2.3. Survival Kinome Differs across the OB–OT Axis in DLB

The olfactory tract (OT) is constituted by the axons coming from the mitral and tufted neurons located in the OB. A significant degeneration of axons has been detected in the OT from AD subjects [61]. In pre-AD mild cognitive impairment subjects, the loss of fiber OT integrity corresponds to a loss of gray matter density in parallel with a reduced glucose metabolism in central olfactory structures [62,63]. Moreover, OTs undergo early and sequential morphological alterations that correlate with the development of dementia [64]. On the other hand, atrophy and changes in the structural integrity of OT have been also observed in PD subjects compared to controls [65]. The deposition of pathological α-synuclein protein has been observed not only in the OB but also in the olfactory tract (OT) [9]. Considering the fact that the analysis of the OT area may provide clues regarding the molecular alterations along the olfactory system during the neurodegenerative process, subsequent experiments were performed to characterize the survival kinome in this region. Our analysis revealed the following: (a) OT-specific alterations in MEK, AKT and PKA (Figure 4); (b) opposite alterations between OB and OT protein levels for SEK1 and p38 MAPK (Figure 4); and (c) unchanged OB/OT protein levels for SAPK1 and MKK3/MKK6 (Figure S2). Altogether, these results point out different protein expression profiles across the OB–OT axis, laying the foundation for future exhaustive molecular profiling of OT areas in order to increase our understanding about the smell deficits that accompany the neurodegenerative process in alpha-synucleinopathies.

3. Materials and Methods

3.1. Materials

The following reagents and materials were used: anti-ALDH1B1 (ref. sc-374090) was purchased from Santa Cruz; anti-PI16 (ref. PAS 31882) from Thermo; anti-P70 S6K beta (ref: ab184551) from Abcam; and anti-Bcl-xL (ref. 2764), anti-pAkt (Ser473) (ref. 4060), anti-Akt (ref. 4685), anti-pMEK1/2 (Ser217/221) (ref. 9154), anti-MEK1/2 (ref. 9126), anti-pERK1/2 (Thr202/Tyr204) (ref. 4370), anti-ERK1/2 (ref. 9102), anti-pPKA (Thr197) (ref. 5661), anti-PKA (ref. 4782), anti-pSEK1/MKK4 (Ser257/Thr261) (ref. 9156), anti-SEK1/MKK4 (ref. 9152), anti pSAPK/JNK (Thr183/Tyr185) (ref. 9255), anti pSAPK/JNK (ref. 9252S), anti pMKK3-6 (Thr183/Tyr185) (ref. 9231), anti MKK3 (ref. 5674), anti p-p38 MAPK (Thr180/Tyr182) (ref. 4511), anti p38 MAPK (ref. 9212) were purchased from Cell Signaling. Electrophoresis reagents were purchased from Biorad and trypsin from Promega.

3.2. Human Samples

According to the Spanish Law 14/2007 of Biomedical Research, written consent forms of the Neurological Tissue Bank of Navarra Health Service and Neurological Tissue Bank of IDIBAPS-Hospital Clinic (Barcelona, Spain) were obtained for research purposes. All assessments, post-mortem evaluations, and procedures were approved by the Clinical Ethics Committee of Navarra (code 2016/36) and were conducted in accordance with the Declaration of Helsinki. For the proteomic phase, four DLB cases were analyzed, while four cases from elderly subjects with no history or histological findings of any neurological disease were used as controls. All cases had a post-mortem interval (PMI) lower than 16 h. For the validation phase, OB tissue from additional subjects was included (n = 12) (Table 2). Neuropathological evaluation was performed according to standardized guidelines [2].

3.3. Sample Preparation for Proteomic Analysis

OB specimens derived from DLB cases and non-neurological subjects were homogenized using “mini potters’’ in lysis buffer containing 8 M urea and 0.1 M TEAB as previously described [39]. The homogenates were spun down at 14,000× g for 15 min at 4 °C. Protein quantitation was performed with the Bradford assay kit (Bio-Rad, Hercules, CA, USA).

3.4. Reduction, Alkylation, Digestion and TMT Labeling

For the proteomic analysis, 25 µg of protein from each sample was used. Proteins were first reduced with 10 mM TCEP for 1 h at 30 °C. Then, alkylation was performed for 30 min at room temperature using 40 mM IAA. Samples were diluted 5 times to reduce the urea concentration <1 M. Trypsinization and peptide labeling was performed as previously described [39]. Control cases were labeled with 127N, 127C, 128N, and 128C and disease cases were labeled with 129N, 129C, 130N and 130C (Labels 126 and 131 were used as intern quality control).

3.5. Off-Gel Electrophoresis and LC-MS/MS Analysis

Samples previously purified with C18 Macro Spin Columns (Harvard Apparatus) were fractionated using off-gel electrophoresis (OGE) separation using an Agilent 3100 off-gel fractionator as previously described [66]. Samples were then desalted and purified using MicroSpins C18 columns, dried in a speed-vacuum and stored at −20 °C until analysis. An LTQ Orbitrap Q-exactive Plus mass spectrometer (Thermo Fisher, Waltham, MA, USA) coupled with nano-HPLC was used to analyzed the OGE fractions [42]. Peptides were reconstituted using 5% ACN and 0.1% FA; were trapped on a 2 cm × 75 µm ID, 3 µm pre-column; and separated on a 50 cm × 75 µm ID, PepMap C18, 2 µm easy-spray column (Thermo Scientific, Waltham, MA, USA). The analytical separation was run for 60 min using a gradient of H2O/FA (99.9%/0.1%; solvent A) and CH3CN/FA (99.9%/0.1%; solvent B) at a flow rate of 300 nL/min. For MS survey scans, the OT resolution, the ion population, the number of precursor ions and the collision energy used have been previously described [39].

3.6. Protein Identification and Quantification

MS data were processed using EasyProt. Peak list was obtained using OGE fractions, and the combination of HCD-CID raw data peak list was generated. These data were submitted to the EasyProt software platform (version 2.3, build 718) that uses Phenyx software (GeneBio, Geneva, Switzerland) for protein identification [67]. The search was performed using the Uniprot/Swiss-Prot database (2014-10, 66903) using Homo Sapiens taxonomy; oxidized methionine (variable modification); and TMT-10 amino-terminus, TMT-10 lysine and cysteine carbamethylation (fixed modifications). One missed cleavage was selected, and parent-ion tolerance was set to 10 ppm and the accuracy of fragment ions to 0.6 Da. Only proteins with a false discovery rate (FDR) lower than 1% and at least 2 unique peptides were considered (peptide length of 6 amino acids). Isobaric quantification was performed using the Isobar R package [68]. To correct the isotopic impurities of TMT-10 reporter ion intensities, the manufacturer’s isotopic distribution data were applied. To normalize the reporter intensities, the equal median intensity method was considered. Biological and technical variability were calculated for each protein ratio in order to test the ratio’s accuracy and biological significance. For each variable, a ratio and sample p-values were calculated. Proteins with a cut-off threshold value higher than 1.33 or lower than 0.77 were considered. All MS data files have been deposited to the ProteomeXchange Consortium [69] (http://proteomecentral.proteomexchange.org) via the PRIDE partner repository with the dataset identifier PXD016069 (username: [email protected]; password: O8KNli2b).

3.7. Bioinformatics

The proteomic information was analyzed using STRING (Search Tool for the Retrieval of Interacting Genes) software (v.11) (http://stringdb.org/) [70] to infer protein interactomes and detect differentially activated/deactivated pathways as a result of DLB phenotypes. The identification of specifically dysregulated regulatory/metabolic networks in DLB was analyzed through the use of QIAGEN’s Ingenuity Pathway Analysis (QIAGEN Redwood City, www.qiagen.com/ingenuity).

3.8. Western-Blotting

Equal amounts of protein (10 µg) were resolved in 4–15% stain free SDS-PAGE gels (Bio-rad). OB/OT proteins derived from control and DLB subjects were electrophoretically transferred onto nitrocellulose membranes as previously described [39]. Membranes were probed with primary antibodies at 1:1000 dilution in 5% nonfat milk or BSA and using a horseradish peroxidase-conjugated secondary antibody at a 1:5000 dilution. Enhanced chemiluminescence (Perkin Elmer, Madrid, Spain) was used to visualize the immunoreactivity using a Chemidoc MP Imaging System (Bio-Rad). Equal loading in the gels was evaluated using stain-free imaging technology [71]. Thus, protein normalization was performed by measuring the total protein directly on the gels used for Western blotting. Image Lab Software (v.5.2; Bio-Rad) was used for densitometric analyses. Optical density values were normalized to total protein levels and considered as arbitrary units.

4. Conclusions

Overall, the present study provides new insights regarding the molecular mechanisms governing the olfactory dysfunction in DLB. Besides the pathological alpha-synuclein depositions previously observed in olfactory areas, we have demonstrated a disarrangement in the OB proteostasis, affecting translation and the synaptic vesicle cycle and showing disturbances in the protein expression profile of cell survival routes across the OB–OT axis. Thus, our findings provide basic information for understanding the implication of olfactory structures in the pathophysiology of DLB, identifying protein mediators that may be used as potential therapeutic agents or even explored in biofluids as candidate biomarkers for DLB diagnosis and evolution.

Supplementary Materials

The following are available online at https://www.mdpi.com/1422-0067/21/17/6371/s1, Table S1: Quantified OB proteins across DLB phenotypes. Figure S1: Unchanged protein levels and activation state of MEK (A), SAPK (B), PKA (C) and MKK3-6 (D). Data are presented as mean ± SEM. n.s: non-significant. Figure S2: Unchanged protein levels for SAPK and MKK3-6 in the OT of DLB subjects. Data are presented as mean ± SEM. n.s: non-significant. Figure S3: Main features used by IPA software including molecule shapes, relationship labels and types. Figures S4 and S5: High-resolution images corresponding to protein networks shown in Figure 3A.

Author Contributions

Conceptualization, E.S.; data curation, E.S., M.L.-M.; formal analysis, M.L.-M., N.M.; funding acquisition, E.S., J.F.-I.; investigation, M.L.-M., N.M., D.S., J.F.-I., J.C.S. and E.S.; methodology, M.L.-M., N.M., D.S., J.F.-I. and J.C.S.; resources, J.C.S.; software, M.L.-M., D.S. and E.S.; supervision, J.F.-I. and E.S.; validation, M.L.-M. and N.M.; writing—original draft, M.L.-M., E.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by grants from the Spanish Ministry of Economy and Competitiveness (Ref. SAF2014-59340-R to E.S) and the Spanish Ministry of Science Innovation and Universities (Ref. PID2019-110356RB-I00 to J.F.I and E.S). M.L-M was supported by a postdoctoral fellowship from the Public University of Navarra (UPNA) and mobility fellowships from UPNA, La Caixa (Res. Nº 810/2016) and the Navarra Health Department (Res. Nº 993/2016).

Acknowledgments

We are very grateful to the patients and relatives who generously donated the brain tissue for research purposes. We are indebted to the Neurological Tissue Bank of the Biobank from the Hospital Clinic-Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS, Barcelona), the Neurological Tissue Bank of HUB-ICO-IDIBELL (Barcelona, Spain) and the Neurological Tissue Bank of Navarrabiomed (Pamplona, Spain) for providing us the OB/OT specimens as well as the associated clinical and neuropathological data. Authors thank the PRIDE team for helping with the mass spectrometric data deposit in ProteomeXChange/PRIDE. The Proteomics Unit of Navarrabiomed is a member of Proteored, PRB3-ISCIII, and is supported by grant PT17/0019/009 to JFI, of the PE I+D+I 2013-2016 funded by ISCIII and FEDER. The Clinical Neuroproteomics Unit of Navarrabiomed is member of the Global Consortium for Chemosensory Research (GCCR) and the Spanish Olfactory Network (ROE) (supported by grant RED2018-102662-T funded by Spanish Ministry of Science and Innovation). This project is part of the HUPO Brain Proteome Project, and these results are lined up with the Spanish Initiative on the Human Proteome Project.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAlzheimer’s disease
BADBcl2-associated agonist of cell death
CAMK IICalmodulin-dependent protein kinase II
DLBDementia with lewy bodies
ERKExtracellular signal-regulated kinase
FDRFalse discovery rate
MEKMitogen-activated protein kinase kinase
MKK3Dual specificity mitogen-activated protein kinase kinase 3
MKK6Dual specificity mitogen-activated protein kinase kinase 6
OB/OTOlfactory bulb/olfactory tract
P70 S6KP70 S6K beta (Ribosomal protein S6 kinase beta
PDParkinson’s disease
PDDParkinson’s disease dementia
PSPProgressive supranuclear palsy
PKAProtein kinase A
PKCProtein kinase C
p38 MAPKp38 mitogen-activated protein kinase
PMIPost-mortem interval
SAPK/JNKStress-activated protein kinase/Jun-amino terminal kinase
SEK1Mitogen-activated protein kinase Kinase 4

References

  1. McRitchie, D.A.; Reid, W.G.; Halliday, G.M.; Hely, M.A.; Brooks, W.S.; Morris, J.G. Neuropathology of three clinical cases prospectively diagnosed as dementia with Lewy bodies. J. Clin. Neurosci. 1999, 6, 149–154. [Google Scholar] [CrossRef]
  2. McKeith, I.G.; Dickson, D.W.; Lowe, J.; Emre, M.; O’Brien, J.T.; Feldman, H.; Cummings, J.; Duda, J.E.; Lippa, C.; Perry, E.K.; et al. Diagnosis and management of dementia with Lewy bodies: Third report of the DLB Consortium. Neurology 2005, 65, 1863–1872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. McKeith, I.G.; Galasko, D.; Kosaka, K.; Perry, E.K.; Dickson, D.W.; Hansen, L.A.; Salmon, D.P.; Lowe, J.; Mirra, S.S.; Byrne, E.J.; et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): Report of the consortium on DLB international workshop. Neurology 1996, 47, 1113–1124. [Google Scholar] [CrossRef] [PubMed]
  4. McKeith, I.; Mintzer, J.; Aarsland, D.; Burn, D.; Chiu, H.; Cohen-Mansfield, J.; Dickson, D.; Dubois, B.; Duda, J.E.; Feldman, H.; et al. Dementia with Lewy bodies. Lancet Neurol. 2004, 3, 19–28. [Google Scholar] [CrossRef]
  5. Mayo, M.C.; Bordelon, Y. Dementia with Lewy bodies. Semin. Neurol. 2014, 34, 182–188. [Google Scholar] [CrossRef] [Green Version]
  6. Fujishiro, H.; Nakamura, S.; Sato, K.; Iseki, E. Prodromal dementia with Lewy bodies. Geriatr. Gerontol. Int. 2015, 15, 817–826. [Google Scholar] [CrossRef]
  7. Donaghy, P.C.; O’Brien, J.T.; Thomas, A.J. Prodromal dementia with Lewy bodies. Psychol. Med. 2015, 45, 259–268. [Google Scholar] [CrossRef]
  8. Doty, R.L. Olfactory dysfunction in neurodegenerative diseases: Is there a common pathological substrate? Lancet Neurol. 2017, 16, 478–488. [Google Scholar] [CrossRef]
  9. Attems, J.; Walker, L.; Jellinger, K.A. Olfactory bulb involvement in neurodegenerative diseases. Acta Neuropathol. 2014, 127, 459–475. [Google Scholar] [CrossRef]
  10. Westervelt, H.J.; Bruce, J.M.; Faust, M.A. Distinguishing Alzheimer’s disease and dementia with Lewy bodies using cognitive and olfactory measures. Neuropsychology 2016, 30, 304–311. [Google Scholar] [CrossRef]
  11. Westervelt, H.J.; Stern, R.A.; Tremont, G. Odor identification deficits in diffuse lewy body disease. Cogn. Behav. Neurol. 2003, 16, 93–99. [Google Scholar] [CrossRef] [PubMed]
  12. Yoo, H.S.; Jeon, S.; Chung, S.J.; Yun, M.; Lee, P.H.; Sohn, Y.H.; Evans, A.C.; Ye, B.S. Olfactory dysfunction in Alzheimer’s disease- and Lewy body-related cognitive impairment. Alzheimers Dement 2018, 14, 1243–1252. [Google Scholar] [CrossRef]
  13. Yoon, J.H.; Kim, M.; Moon, S.Y.; Yong, S.W.; Hong, J.M. Olfactory function and neuropsychological profile to differentiate dementia with Lewy bodies from Alzheimer’s disease in patients with mild cognitive impairment: A 5-year follow-up study. J. Neurol. Sci. 2015, 355, 174–179. [Google Scholar] [CrossRef] [PubMed]
  14. Beach, T.G.; Adler, C.H.; Zhang, N.; Serrano, G.E.; Sue, L.I.; Driver-Dunckley, E.; Mehta, S.H.; Zamrini, E.E.; Sabbagh, M.N.; Shill, H.A.; et al. Severe hyposmia distinguishes neuropathologically confirmed dementia with Lewy bodies from Alzheimer’s disease dementia. PLoS ONE 2020, 15, e0231720. [Google Scholar] [CrossRef] [Green Version]
  15. Wilson, R.S.; Yu, L.; Schneider, J.A.; Arnold, S.E.; Buchman, A.S.; Bennett, D.A. Lewy bodies and olfactory dysfunction in old age. Chem. Senses 2011, 36, 367–373. [Google Scholar] [CrossRef] [Green Version]
  16. McShane, R.H.; Nagy, Z.; Esiri, M.M.; King, E.; Joachim, C.; Sullivan, N.; Smith, A.D. Anosmia in dementia is associated with Lewy bodies rather than Alzheimer’s pathology. J. Neurol. Neurosurg. Psychiatry 2001, 70, 739–743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Fujishiro, H.; Ferman, T.J.; Boeve, B.F.; Smith, G.E.; Graff-Radford, N.R.; Uitti, R.J.; Wszolek, Z.K.; Knopman, D.S.; Petersen, R.C.; Parisi, J.E.; et al. Validation of the neuropathologic criteria of the third consortium for dementia with Lewy bodies for prospectively diagnosed cases. J. Neuropathol. Exp. Neurol. 2008, 67, 649–656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Goedert, M. Alpha-synuclein and neurodegenerative diseases. Nat. Rev. Neurosci. 2001, 2, 492–501. [Google Scholar] [CrossRef]
  19. Hasegawa, M.; Fujiwara, H.; Nonaka, T.; Wakabayashi, K.; Takahashi, H.; Lee, V.M.; Trojanowski, J.Q.; Mann, D.; Iwatsubo, T. Phosphorylated alpha-synuclein is ubiquitinated in alpha-synucleinopathy lesions. J. Biol. Chem. 2002, 277, 49071–49076. [Google Scholar] [CrossRef] [Green Version]
  20. Mukaetova-Ladinska, E.B.; McKeith, I.G. Pathophysiology of synuclein aggregation in Lewy body disease. Mech. Ageing Dev. 2006, 127, 188–202. [Google Scholar] [CrossRef]
  21. Spillantini, M.G.; Crowther, R.A.; Jakes, R.; Hasegawa, M.; Goedert, M. Alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with lewy bodies. Proc. Natl. Acad. Sci. USA 1998, 95, 6469–6473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. McKeith, I.G.; Boeve, B.F.; Dickson, D.W.; Halliday, G.; Taylor, J.P.; Weintraub, D.; Aarsland, D.; Galvin, J.; Attems, J.; Ballard, C.G.; et al. Diagnosis and management of dementia with Lewy bodies: Fourth consensus report of the DLB Consortium. Neurology 2017, 89, 88–100. [Google Scholar] [CrossRef] [Green Version]
  23. Emre, M.; Aarsland, D.; Brown, R.; Burn, D.J.; Duyckaerts, C.; Mizuno, Y.; Broe, G.A.; Cummings, J.; Dickson, D.W.; Gauthier, S.; et al. Clinical diagnostic criteria for dementia associated with Parkinson’s disease. Mov. Disord. 2007, 22, 1689–1707. [Google Scholar] [CrossRef] [PubMed]
  24. Beach, T.G.; White, C.L., 3rd; Hladik, C.L.; Sabbagh, M.N.; Connor, D.J.; Shill, H.A.; Sue, L.I.; Sasse, J.; Bachalakuri, J.; Henry-Watson, J.; et al. Olfactory bulb alpha-synucleinopathy has high specificity and sensitivity for Lewy body disorders. Acta Neuropathol. 2009, 117, 169–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Silveira-Moriyama, L.; Holton, J.L.; Kingsbury, A.; Ayling, H.; Petrie, A.; Sterlacci, W.; Poewe, W.; Maier, H.; Lees, A.J.; Revesz, T. Regional differences in the severity of Lewy body pathology across the olfactory cortex. Neurosci. Lett. 2009, 453, 77–80. [Google Scholar] [CrossRef] [PubMed]
  26. Braak, H.; Del Tredici, K.; Rub, U.; de Vos, R.A.; Jansen Steur, E.N.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef]
  27. Goedert, M. Alzheimer’s and Parkinson’s diseases: The prion concept in relation to assembled Abeta, tau, and alpha-synuclein. Science 2015, 349, 1255555. [Google Scholar] [CrossRef]
  28. Brundin, P.; Melki, R.; Kopito, R. Prion-like transmission of protein aggregates in neurodegenerative diseases. Nat. Rev. Mol. Cell Biol. 2010, 11, 301–307. [Google Scholar] [CrossRef] [Green Version]
  29. Rey, N.L.; Steiner, J.A.; Maroof, N.; Luk, K.C.; Madaj, Z.; Trojanowski, J.Q.; Lee, V.M.; Brundin, P. Widespread transneuronal propagation of alpha-synucleinopathy triggered in olfactory bulb mimics prodromal Parkinson’s disease. J. Exp. Med. 2016, 213, 1759–1778. [Google Scholar] [CrossRef]
  30. Hosp, F.; Mann, M. A Primer on Concepts and Applications of Proteomics in Neuroscience. Neuron 2017, 96, 558–571. [Google Scholar] [CrossRef]
  31. Medland, S.E.; Jahanshad, N.; Neale, B.M.; Thompson, P.M. Whole-genome analyses of whole-brain data: Working within an expanded search space. Nat. Neurosci. 2014, 17, 791–800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Pollock, J.D.; Wu, D.Y.; Satterlee, J.S. Molecular neuroanatomy: A generation of progress. Trends Neurosci. 2014, 37, 106–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bereczki, E.; Branca, R.M.; Francis, P.T.; Pereira, J.B.; Baek, J.H.; Hortobagyi, T.; Winblad, B.; Ballard, C.; Lehtio, J.; Aarsland, D. Synaptic markers of cognitive decline in neurodegenerative diseases: A proteomic approach. Brain 2018, 141, 582–595. [Google Scholar] [CrossRef]
  34. Datta, A.; Chai, Y.L.; Tan, J.M.; Lee, J.H.; Francis, P.T.; Chen, C.P.; Sze, S.K.; Lai, M.K.P. An iTRAQ-based proteomic analysis reveals dysregulation of neocortical synaptopodin in Lewy body dementias. Mol. Brain 2017, 10, 36. [Google Scholar] [CrossRef]
  35. Lundstrom, S.L.; Zhang, B.; Rutishauser, D.; Aarsland, D.; Zubarev, R.A. SpotLight Proteomics: Uncovering the hidden blood proteome improves diagnostic power of proteomics. Sci. Rep. 2017, 7, 41929. [Google Scholar] [CrossRef] [Green Version]
  36. Remnestal, J.; Just, D.; Mitsios, N.; Fredolini, C.; Mulder, J.; Schwenk, J.M.; Uhlen, M.; Kultima, K.; Ingelsson, M.; Kilander, L.; et al. CSF profiling of the human brain enriched proteome reveals associations of neuromodulin and neurogranin to Alzheimer’s disease. Proteom. Clin. Appl. 2016, 10, 1242–1253. [Google Scholar] [CrossRef] [PubMed]
  37. Lachen-Montes, M.; Fernandez-Irigoyen, J.; Santamaria, E. Deconstructing the molecular architecture of olfactory areas using proteomics. Proteom. Clin. Appl. 2016, 10, 1178–1190. [Google Scholar] [CrossRef] [PubMed]
  38. Lachen-Montes, M.; Gonzalez-Morales, A.; de Morentin, X.M.; Perez-Valderrama, E.; Ausin, K.; Zelaya, M.V.; Serna, A.; Aso, E.; Ferrer, I.; Fernandez-Irigoyen, J.; et al. An early dysregulation of FAK and MEK/ERK signaling pathways precedes the beta-amyloid deposition in the olfactory bulb of APP/PS1 mouse model of Alzheimer’s disease. J. Proteom. 2016, 148, 149–158. [Google Scholar] [CrossRef]
  39. Lachen-Montes, M.; Gonzalez-Morales, A.; Iloro, I.; Elortza, F.; Ferrer, I.; Gveric, D.; Fernandez-Irigoyen, J.; Santamaria, E. Unveiling the olfactory proteostatic disarrangement in Parkinson’s disease by proteome-wide profiling. Neurobiol. Aging 2019, 73, 123–134. [Google Scholar] [CrossRef]
  40. Lachen-Montes, M.; Gonzalez-Morales, A.; Palomino, M.; Ausin, K.; Gomez-Ochoa, M.; Zelaya, M.V.; Ferrer, I.; Perez-Mediavilla, A.; Fernandez-Irigoyen, J.; Santamaria, E. Early-Onset Molecular Derangements in the Olfactory Bulb of Tg2576 Mice: Novel Insights Into the Stress-Responsive Olfactory Kinase Dynamics in Alzheimer’s Disease. Front. Aging Neurosci. 2019, 11, 141. [Google Scholar] [CrossRef] [Green Version]
  41. Lachen-Montes, M.; Gonzalez-Morales, A.; Schvartz, D.; Zelaya, M.V.; Ausin, K.; Fernandez-Irigoyen, J.; Sanchez, J.C.; Santamaria, E. The olfactory bulb proteotype differs across frontotemporal dementia spectrum. J. Proteom. 2019, 201, 37–47. [Google Scholar] [CrossRef] [PubMed]
  42. Lachen-Montes, M.; Gonzalez-Morales, A.; Zelaya, M.V.; Perez-Valderrama, E.; Ausin, K.; Ferrer, I.; Fernandez-Irigoyen, J.; Santamaria, E. Olfactory bulb neuroproteomics reveals a chronological perturbation of survival routes and a disruption of prohibitin complex during Alzheimer’s disease progression. Sci. Rep. 2017, 7, 9115. [Google Scholar] [CrossRef] [PubMed]
  43. Lachen-Montes, M.; Zelaya, M.V.; Segura, V.; Fernandez-Irigoyen, J.; Santamaria, E. Progressive modulation of the human olfactory bulb transcriptome during Alzheimer s disease evolution: Novel insights into the olfactory signaling across proteinopathies. Oncotarget 2017, 8, 69663–69679. [Google Scholar] [CrossRef] [Green Version]
  44. Zelaya, M.V.; Perez-Valderrama, E.; de Morentin, X.M.; Tunon, T.; Ferrer, I.; Luquin, M.R.; Fernandez-Irigoyen, J.; Santamaria, E. Olfactory bulb proteome dynamics during the progression of sporadic Alzheimer’s disease: Identification of common and distinct olfactory targets across Alzheimer-related co-pathologies. Oncotarget 2015, 6, 39437–39456. [Google Scholar] [CrossRef]
  45. Palomino-Alonso, M.; Lachen-Montes, M.; Gonzalez-Morales, A.; Ausin, K.; Perez-Mediavilla, A.; Fernandez-Irigoyen, J.; Santamaria, E. Network-Driven Proteogenomics Unveils an Aging-Related Imbalance in the Olfactory IkappaBalpha-NFkappaB p65 Complex Functionality in Tg2576 Alzheimer’s Disease Mouse Model. Int. J. Mol. Sci. 2017, 18, 2260. [Google Scholar] [CrossRef] [PubMed]
  46. Fernandez-Irigoyen, J.; Santamaria, E. Olfactory proteotyping: Towards the enlightenment of the neurodegeneration. Neural Regen. Res. 2019, 14, 979–981. [Google Scholar] [CrossRef]
  47. Tepe, B.; Hill, M.C.; Pekarek, B.T.; Hunt, P.J.; Martin, T.J.; Martin, J.F.; Arenkiel, B.R. Single-Cell RNA-Seq of Mouse Olfactory Bulb Reveals Cellular Heterogeneity and Activity-Dependent Molecular Census of Adult-Born Neurons. Cell Rep. 2018, 25, 2689–2703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Deza-Ponzio, R.; Herrera, M.L.; Bellini, M.J.; Virgolini, M.B.; Herenu, C.B. Aldehyde dehydrogenase 2 in the spotlight: The link between mitochondria and neurodegeneration. Neurotoxicology 2018, 68, 19–24. [Google Scholar] [CrossRef] [Green Version]
  49. Garcia-Esparcia, P.; Lopez-Gonzalez, I.; Grau-Rivera, O.; Garcia-Garrido, M.F.; Konetti, A.; Llorens, F.; Zafar, S.; Carmona, M.; Del Rio, J.A.; Zerr, I.; et al. Dementia with Lewy Bodies: Molecular Pathology in the Frontal Cortex in Typical and Rapidly Progressive Forms. Front. Neurol. 2017, 8, 89. [Google Scholar] [CrossRef] [Green Version]
  50. Garcia-Esparcia, P.; Hernandez-Ortega, K.; Koneti, A.; Gil, L.; Delgado-Morales, R.; Castano, E.; Carmona, M.; Ferrer, I. Altered machinery of protein synthesis is region- and stage-dependent and is associated with alpha-synuclein oligomers in Parkinson’s disease. Acta Neuropathol. Commun. 2015, 3, 76. [Google Scholar] [CrossRef] [Green Version]
  51. Bahrami, B.F.; Ataie-Kachoie, P.; Pourgholami, M.H.; Morris, D.L. p70 Ribosomal protein S6 kinase (Rps6kb1): An update. J. Clin. Pathol. 2014, 67, 1019–1025. [Google Scholar] [CrossRef] [PubMed]
  52. Blumenstock, S.; Angelo, M.F.; Peters, F.; Dorostkar, M.M.; Ruf, V.C.; Luckner, M.; Crux, S.; Slapakova, L.; Arzberger, T.; Claverol, S.; et al. Early defects in translation elongation factor 1alpha levels at excitatory synapses in alpha-synucleinopathy. Acta Neuropathol. 2019, 138, 971–986. [Google Scholar] [CrossRef]
  53. Revuelta, G.J.; Rosso, A.; Lippa, C.F. Neuritic pathology as a correlate of synaptic loss in dementia with lewy bodies. Am. J. Alzheimers Dis. Demen. 2008, 23, 97–102. [Google Scholar] [CrossRef] [PubMed]
  54. Schulz-Schaeffer, W.J. The synaptic pathology of alpha-synuclein aggregation in dementia with Lewy bodies, Parkinson’s disease and Parkinson’s disease dementia. Acta Neuropathol. 2010, 120, 131–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Del Peso, L.; Gonzalez-Garcia, M.; Page, C.; Herrera, R.; Nunez, G. Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt. Science 1997, 278, 687–689. [Google Scholar] [CrossRef] [PubMed]
  56. Datta, S.R.; Dudek, H.; Tao, X.; Masters, S.; Fu, H.; Gotoh, Y.; Greenberg, M.E. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell 1997, 91, 231–241. [Google Scholar] [CrossRef] [Green Version]
  57. Gomez-Santos, C.; Ferrer, I.; Reiriz, J.; Vinals, F.; Barrachina, M.; Ambrosio, S. MPP+ increases alpha-synuclein expression and ERK/MAP-kinase phosphorylation in human neuroblastoma SH-SY5Y cells. Brain Res. 2002, 935, 32–39. [Google Scholar] [CrossRef]
  58. Tolo, J.; Taschenberger, G.; Leite, K.; Stahlberg, M.A.; Spehlbrink, G.; Kues, J.; Munari, F.; Capaldi, S.; Becker, S.; Zweckstetter, M.; et al. Pathophysiological Consequences of Neuronal alpha-Synuclein Overexpression: Impacts on Ion Homeostasis, Stress Signaling, Mitochondrial Integrity, and Electrical Activity. Front. Mol. Neurosci. 2018, 11, 49. [Google Scholar] [CrossRef] [Green Version]
  59. Song, G.; Ouyang, G.; Bao, S. The activation of Akt/PKB signaling pathway and cell survival. J. Cell Mol. Med. 2005, 9, 59–71. [Google Scholar] [CrossRef]
  60. Jha, S.K.; Jha, N.K.; Kar, R.; Ambasta, R.K.; Kumar, P. p38 MAPK and PI3K/AKT Signalling Cascades inParkinson’s Disease. Int. J. Mol. Cell Med. 2015, 4, 67–86. [Google Scholar]
  61. Davies, D.C.; Brooks, J.W.; Lewis, D.A. Axonal loss from the olfactory tracts in Alzheimer’s disease. Neurobiol. Aging 1993, 14, 353–357. [Google Scholar] [CrossRef]
  62. Thomann, P.A.; Dos Santos, V.; Seidl, U.; Toro, P.; Essig, M.; Schroder, J. MRI-derived atrophy of the olfactory bulb and tract in mild cognitive impairment and Alzheimer’s disease. J. Alzheimers Dis. 2009, 17, 213–221. [Google Scholar] [CrossRef] [PubMed]
  63. Cross, D.J.; Anzai, Y.; Petrie, E.C.; Martin, N.; Richards, T.L.; Maravilla, K.R.; Peskind, E.R.; Minoshima, S. Loss of olfactory tract integrity affects cortical metabolism in the brain and olfactory regions in aging and mild cognitive impairment. J. Nucl. Med. 2013, 54, 1278–1284. [Google Scholar] [CrossRef] [Green Version]
  64. Bathini, P.; Mottas, A.; Jaquet, M.; Brai, E.; Alberi, L. Progressive signaling changes in the olfactory nerve of patients with Alzheimer’s disease. Neurobiol. Aging 2019, 76, 80–95. [Google Scholar] [CrossRef] [PubMed]
  65. Scherfler, C.; Schocke, M.F.; Seppi, K.; Esterhammer, R.; Brenneis, C.; Jaschke, W.; Wenning, G.K.; Poewe, W. Voxel-wise analysis of diffusion weighted imaging reveals disruption of the olfactory tract in Parkinson’s disease. Brain 2006, 129, 538–542. [Google Scholar] [CrossRef] [PubMed]
  66. Dayon, L.; Pasquarello, C.; Hoogland, C.; Sanchez, J.C.; Scherl, A. Combining low- and high-energy tandem mass spectra for optimized peptide quantification with isobaric tags. J. Proteom. 2010, 73, 769–777. [Google Scholar] [CrossRef] [PubMed]
  67. Gluck, F.; Hoogland, C.; Antinori, P.; Robin, X.; Nikitin, F.; Zufferey, A.; Pasquarello, C.; Fetaud, V.; Dayon, L.; Muller, M.; et al. EasyProt—An easy-to-use graphical platform for proteomics data analysis. J. Proteom. 2013, 79, 146–160. [Google Scholar] [CrossRef]
  68. Breitwieser, F.P.; Muller, A.; Dayon, L.; Kocher, T.; Hainard, A.; Pichler, P.; Schmidt-Erfurth, U.; Superti-Furga, G.; Sanchez, J.C.; Mechtler, K.; et al. General statistical modeling of data from protein relative expression isobaric tags. J Proteom. Res. 2011, 10, 2758–2766. [Google Scholar] [CrossRef]
  69. Vizcaino, J.A.; Deutsch, E.W.; Wang, R.; Csordas, A.; Reisinger, F.; Rios, D.; Dianes, J.A.; Sun, Z.; Farrah, T.; Bandeira, N.; et al. ProteomeXchange provides globally coordinated proteomics data submission and dissemination. Nat. Biotechnol. 2014, 32, 223–226. [Google Scholar] [CrossRef]
  70. Szklarczyk, D.; Gable, A.L.; Lyon, D.; Junge, A.; Wyder, S.; Huerta-Cepas, J.; Simonovic, M.; Doncheva, N.T.; Morris, J.H.; Bork, P.; et al. STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019, 47, D607–D613. [Google Scholar] [CrossRef] [Green Version]
  71. Moritz, C.P. Tubulin or Not Tubulin: Heading Toward Total Protein Staining as Loading Control in Western Blots. Proteomics 2017, 17, 1600189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. (A) An overview of the workflow used for the molecular characterization of the olfactory bulbs (OB) derived from dementia with Lewy bodies (DLB) subjects. (B) Quantified proteome distribution across OB cell layers. (C) Cluster-enriched genes in specific OB cell layers that are differentially expressed at the level of the OB in DLB subjects. (D) OB protein expression changes of ALDH1B1 and PI16 in DLB subjects by Western blotting. Data are presented as mean ± SEM. * p < 0.05 vs. control group; *** p < 0.001 vs. control group (a.u: arbitrary units; DEPs: differential expressed proteins; EPL: external plexiform layer; OSN: olfactory sensory neuron).
Figure 1. (A) An overview of the workflow used for the molecular characterization of the olfactory bulbs (OB) derived from dementia with Lewy bodies (DLB) subjects. (B) Quantified proteome distribution across OB cell layers. (C) Cluster-enriched genes in specific OB cell layers that are differentially expressed at the level of the OB in DLB subjects. (D) OB protein expression changes of ALDH1B1 and PI16 in DLB subjects by Western blotting. Data are presented as mean ± SEM. * p < 0.05 vs. control group; *** p < 0.001 vs. control group (a.u: arbitrary units; DEPs: differential expressed proteins; EPL: external plexiform layer; OSN: olfactory sensory neuron).
Ijms 21 06371 g001
Figure 2. Impairment of OB protein translation and synaptic function in DLB. (A) Interactome network for OB deregulated proteome using STRING (Search Tool for the Retrieval of Interacting Genes). Proteins are represented with nodes and the physical/functional interactions with continuous lines. Interactions tagged as “high confidence” (>0.7) in the STRING database were exclusively considered. K means clustering was used. (B) Western-blotting of P70 S6K protein levels across controls and DLB subjects. Data are presented as mean ± SEM. * p < 0.05 vs. control group. In this case, equal loading of the gel was assessed using stain-free imaging technology, and protein normalization was performed by measuring total protein directly on the gels (a.u; arbitrary units). (C) Significantly enriched synapsis-related pathways in the OB differential proteome using the STRING tool.
Figure 2. Impairment of OB protein translation and synaptic function in DLB. (A) Interactome network for OB deregulated proteome using STRING (Search Tool for the Retrieval of Interacting Genes). Proteins are represented with nodes and the physical/functional interactions with continuous lines. Interactions tagged as “high confidence” (>0.7) in the STRING database were exclusively considered. K means clustering was used. (B) Western-blotting of P70 S6K protein levels across controls and DLB subjects. Data are presented as mean ± SEM. * p < 0.05 vs. control group. In this case, equal loading of the gel was assessed using stain-free imaging technology, and protein normalization was performed by measuring total protein directly on the gels (a.u; arbitrary units). (C) Significantly enriched synapsis-related pathways in the OB differential proteome using the STRING tool.
Ijms 21 06371 g002
Figure 3. Disruption of OB signaling routes in DLB. (A) Visual representation of protein interactome maps for OB differentially expressed proteins in DLB. Upregulated proteins in red and downregulated proteins in green. Continuous lines represent direct interactions, while discontinuous lines correspond to indirect functional interactions. See complete legend and high resolution images in Figures S3–S5. (B) Activation state and protein measurements of AKT and Bcl2-associated agonist of cell death (BAD) across controls and DLB subjects by Western-blotting. (C) OB ERK1/2, SEK1 and p38 MAPK levels at the level of OB. Data are presented as mean ± SEM. * p < 0.05 vs. control group; *** p < 0.001 vs. control group. Equal loading of the gels was assessed using stain-free imaging technology, and protein normalization was performed by measuring total protein directly on the gels (a.u; arbitrary units).
Figure 3. Disruption of OB signaling routes in DLB. (A) Visual representation of protein interactome maps for OB differentially expressed proteins in DLB. Upregulated proteins in red and downregulated proteins in green. Continuous lines represent direct interactions, while discontinuous lines correspond to indirect functional interactions. See complete legend and high resolution images in Figures S3–S5. (B) Activation state and protein measurements of AKT and Bcl2-associated agonist of cell death (BAD) across controls and DLB subjects by Western-blotting. (C) OB ERK1/2, SEK1 and p38 MAPK levels at the level of OB. Data are presented as mean ± SEM. * p < 0.05 vs. control group; *** p < 0.001 vs. control group. Equal loading of the gels was assessed using stain-free imaging technology, and protein normalization was performed by measuring total protein directly on the gels (a.u; arbitrary units).
Ijms 21 06371 g003
Figure 4. Disruption of olfactory tract (OT) signaling routes in DLB. Steady-state levels of AKT, MEK, PKAc, SEK1 and p38 MAPK in the OTs derived from controls and DLB subjects. Data are presented as mean ± SEM. * p < 0.05 vs. control group; *** p < 0.001 vs. control group. Equal loading of the gels was assessed using stain-free imaging technology, and protein normalization was performed by measuring total protein directly on the gels (a.u; arbitrary units).
Figure 4. Disruption of olfactory tract (OT) signaling routes in DLB. Steady-state levels of AKT, MEK, PKAc, SEK1 and p38 MAPK in the OTs derived from controls and DLB subjects. Data are presented as mean ± SEM. * p < 0.05 vs. control group; *** p < 0.001 vs. control group. Equal loading of the gels was assessed using stain-free imaging technology, and protein normalization was performed by measuring total protein directly on the gels (a.u; arbitrary units).
Ijms 21 06371 g004
Table 1. Significantly deregulated proteins in DLB OB specimens.
Table 1. Significantly deregulated proteins in DLB OB specimens.
DescriptionGene NameUniprotPeptide CountRatiop Value Sample
Down-regulated proteins across DLB
60S ribosomal protein L34RPL34P4920720.444.6181 × 10−13
60S ribosomal protein L37RPL37P6192720.4657.3563 × 10−14
Protein prune homolog 2PRUNE2Q8WUY330.54544.0993 × 10−10
60S ribosomal protein L22RPL22P3526820.5741.4869 × 10−12
Ras-related protein Rab-3BRAB3BP2033720.61256.995 × 10−9
60S ribosomal protein L36RPL36Q9Y3U820.61392.0299 × 10−10
V-type proton ATPase subunit d 1ATP6V0D1P61421100.61446.1715 × 10−11
Heterogeneous nuclear ribonucleoprotein UHNRNPUQ00839200.61811.9382 × 10−11
DNA-dependent protein kinase catalytic subunitPRKDCP78527190.62641.5042 × 10−11
FACT complex subunit SPT16SUPT16HQ9Y5B920.63288.6739 × 10−11
Synaptogyrin-3SYNGR3O4376130.65065.5741 × 10−11
Liprin-alpha-4PPFIA4O7533510.6544.3343 × 10−10
60S ribosomal protein L10RPL10P2763520.66722.1869 × 10−10
60S ribosomal protein L23aRPL23AP6275030.66754.0283 × 10−10
40S ribosomal protein S11RPS11P6228030.66794.1716 × 10−9
HMG nucleosome × 10−binding domain-containing protein 3HMGN3Q1565120.66921.4543 × 10−9
Sodium/potassium-transporting ATPase subunit alpha-3ATP1A3P13637260.66935.6496 × 10−11
Lymphocyte antigen 6HLY6HO9477250.67342.4723 × 10−9
40S ribosomal protein S25RPS25P6285130.67411.1199 × 10−9
Neurofilament light polypeptideNEFLP07196180.67437.1158 × 10−10
40S ribosomal protein S3aRPS3AP6124740.67823.4891 × 10−9
Vesicular inhibitory amino acid transporterSLC32A1Q9H59860.68162.3798 × 10−10
Vesicular glutamate transporter 1SLC17A7Q9P2U720.68182.7575 × 10−11
Plasma membrane calcium-transporting ATPase 3ATP2B3Q1672020.6864.3062 × 10−9
E3 ubiquitin-protein ligase MYCBP2MYCBP2O7559220.68623.7771 × 10−10
Syntaxin-1ASTX1AQ1662370.68794.8927 × 10−9
Syntaxin-binding protein 1STXBP1P61764290.69041.4785 × 10−10
Neurofilament medium polypeptideNEFMP07197270.69321.5816 × 10−9
Secretory carrier-associated membrane protein 5SCAMP5Q8TAC940.69462.3559 × 10−10
Calcium/calmodulin-dependent protein kinase type II subunit alphaCAMK2AQ9UQM780.70125.1862 × 10−11
Neurexin-1NRXN1Q9ULB110.70147.3383 × 10−10
PH and SEC7 domain-containing protein 3PSD3Q9NYI0110.70371.2708 × 10−9
Gamma-aminobutyric acid receptor subunit beta-2GABRB2P4787020.70421.5422 × 10−9
Proenkephalin-A cleaved into synenkephalin; Met-enkephalinPENKP0121050.71343.9046 × 10−8
SynaptophysinSYPP0824750.71565.4513 × 10−11
40S ribosomal protein S27, 40S ribosomal protein S27-likeRPS27Q71UM520.71651.5132 × 10−9
Syntaxin-1BSTX1BP61266130.71653.4018 × 10−10
DnaJ homolog subfamily C member 5DNAJC5Q9H3Z450.71722.3736 × 10−10
Calcium/calmodulin-dependent protein kinase type II subunit betaCAMK2BQ13554110.71843.952 × 10−10
Mitochondrial glutamate carrier 1SLC25A22Q9H93620.72049.6273 × 10−7
Sodium/potassium-transporting ATPase subunit beta-1ATP1B1P05026130.72167.7815 × 10−10
Glutamate receptor 2GRIA2P4226250.72382.3865 × 10−9
Tubulin beta-2A chainTUBB2AQ1388520.72461.4984 × 10−9
PC4 and SFRS1-interacting proteinPSIP1O75475110.72581.7917 × 10−9
60S ribosomal protein L38RPL38P6317320.72723.6241 × 10−9
Clathrin heavy chain 1CLTCQ00610580.73051.1969 × 10−9
Ras-related protein Rab-3ARAB3AP2033640.73122.1299 × 10−9
Leukocyte surface antigen CD47CD47Q0872230.73193.2189 × 10−8
Metabotropic glutamate receptor 1GRM1Q1325530.73654.3436 × 10−9
Receptor expression-enhancing protein 5REEP5Q0076550.73948.049 × 10−9
Probable leucine--tRNA ligase, mitochondrialLARS2Q1503120.73957.1964 × 10−9
Neutral cholesterol ester hydrolase 1NCEH1Q6PIU240.74071.4583 × 10−9
40S ribosomal protein S2RPS2P1588070.74111.147 × 10−8
Glucose 1,6-bisphosphate synthasePGM2L1Q6PCE380.74383.5479 × 10−9
60S ribosomal protein L15RPL15P6131330.74464.3186 × 10−8
Transmembrane protein 35ATMEM35AQ53FP220.74463.4577 × 10−9
60S ribosomal protein L23RPL23P6282920.74481.9037 × 10−7
Disintegrin and metalloproteinase domain-containing protein 22ADAM22Q9P0K120.7462.1643 × 10−8
60S ribosomal protein L31RPL31P6289930.74692.7904 × 10−9
Pre-mRNA-processing-splicing factor 8PRPF8Q6P2Q950.74843.9116 × 10−10
V-type proton ATPase 116 kDa subunit a isoform 1ATP6V0A1Q93050160.75142.8591 × 10−9
Actin-related protein 3BACTR3BQ9P1U150.7526.086 × 10−9
ATP-dependent RNA helicase ADHX9Q08211200.75216.8201 × 10−9
cAMP-dependent protein kinase type I-beta regulatory subunitPRKAR1BP3132110.75222.2148 × 10−7
Ras/Rap GTPase-activating protein SynGAPSYNGAP1Q96PV070.75272.5633 × 10−8
Stearoyl-CoA desaturase 5SCD5Q86SK920.75521.0027 × 10−6
Elongation factor 1-alpha 2EEF1A2Q0563970.75573.9797 × 10−9
Synaptic vesicle glycoprotein 2ASV2AQ7L0J360.75583.2828 × 10−9
Protein rogdi homologROGDIQ9GZN730.75695.4595 × 10−9
X-ray repair cross-complementing protein 6XRCC6P12956150.75732.6216 × 10−10
Protein DEKDEKP3565920.75981.2282 × 10−8
Vesicle-fusing ATPaseNSFP46459320.76082.2491 × 10−9
Cytochrome c oxidase subunit 7A-related protein, mitochondrialCOX7A2LO1454820.76132.1107 × 10−8
Ras-related protein Rab-33BRAB33BQ9H08210.76145.1009 × 10−7
Kelch-like protein 22KLHL22Q53GT120.76332.2287 × 10−8
Dynamin-1DNM1Q05193220.76411.0564 × 10−8
Probable G-protein coupled receptor 158GPR158Q5T84820.76531.1577 × 10−6
Poly [ADP-ribose] polymerase 1PARP1P09874100.76544.795 × 10−7
SynaptoporinSYNPRQ8TBG920.76575.4958 × 10−9
Beta-soluble NSF attachment proteinNAPBQ9H115140.76662.9345 × 10−8
V-type proton ATPase catalytic subunit AATP6V1AP38606230.76741.3052 × 10−8
Contactin-associated protein-like 2CNTNAP2Q9UHC670.76741.2516 × 10−8
ATPase inhibitor, mitochondrialATPIF1Q9UII260.7695.9459 × 10−8
Up-regulated proteins across DLB
Glutathione S-transferase theta-1GSTT1P3071141.346.8584 × 10−8
Phosphoglycerate mutase 2PGAM2P1525941.34052.2404 × 10−9
Cysteine and glycine-rich protein 1CSRP1P21291121.34125.701 × 10−9
Phosphotriesterase-related proteinPTERQ96BW541.34472.9204 × 10−10
Peroxiredoxin-6PRDX6P30041151.3572.3469 × 10−9
Phenazine biosynthesis-like domain-containing proteinPBLDP3003921.36238.8245 × 10−6
Gamma-butyrobetaine dioxygenaseBBOX1O7593651.40038.8403 × 10−10
Heat shock protein beta-6HSPB6O1455821.40744.9675 × 10−10
GMP reductase 1GMPRP3695941.41691.2597 × 10−11
PirinPIRO0062531.42543.3049 × 10−9
Protein S100-A4S100A4P2644731.45042.1294 × 10−9
Aldehyde dehydrogenase X, mitochondrialALDH1B1P3083751.51591.3337 × 10−10
Nicotinate-nucleotide pyrophosphorylase [carboxylating]QPRTQ1527441.5165.6656 × 10−8
Metallothionein-1EMT1EP0473211.58811.6841 × 10−10
Collagen alpha-3(VI) chainCOL6A3P12111281.99921.5501 × 10−14
Peptidase inhibitor 16PI16Q6UXB832.09551.2432 × 10−10
Down-regulated and up-regulated proteins in DLB OB are indicated in green and red respectively.
Table 2. Clinicopathological data of subjects included in this study.
Table 2. Clinicopathological data of subjects included in this study.
GroupsCase (Code)Age (Years)SexPMIOBOTDefinitive DXProteomic PhaseWestern Blot Analysis
ControlBK-030075F-YesYesARP I-IIxx
BK-137878M6 hYesYesmulti-infarctxx
BK-107884F6 hYesYesVascular encephalopathyxx
BK-119582F8 hYesYesAcute stroke left cerebral arteryxx
BK-156379M5 hYesYesAcute stroke left cerebral artery x
DLBCS-062278M8 h 30 minYesYesDLB-AD neocorticalxx
CS-087074M15 hYesYesDLB-AD neocorticalxx
CS-093880M9 h 40 minYesYesDLB-AD neocorticalxx
CS-094772M11 hYesYesDLB-AD neocorticalxx
CS-109673F15 h 30 minYesYesDLB-AD neocortical x
CS-114082F4 h 30 minYesYesDLB-AD neocortical x
CS-115874M10 hYesYesDLB-AD neocortical x
CS-121567F13 h 30 minYesYesDLB-AD neocortical x
CS-128278F5 hYesYesDLB-AD neocortical x
CS-119277F14 h 20 minYesYesDLB-AD neocortical x
43271F9 hYesNoDLB-AD neocortical x
39790F5 hYesNoDLB-AD neocortical x
39470M4 hYesNoDLB-AD neocortical x
33970M4 h 15 minYesNoDLB-AD neocortical x
32591M12 hYesNoDLB-AD neocortical x
PMI: post-mortem interval; DX: diagnosis. “x” indicates that the sample has been used in the proteomic phase, Western-blot analysis or in both approaches.

Share and Cite

MDPI and ACS Style

Lachén-Montes, M.; Mendizuri, N.; Schvartz, D.; Fernández-Irigoyen, J.; Sánchez, J.C.; Santamaría, E. Proteomic Characterization of the Olfactory Molecular Imbalance in Dementia with Lewy Bodies. Int. J. Mol. Sci. 2020, 21, 6371. https://doi.org/10.3390/ijms21176371

AMA Style

Lachén-Montes M, Mendizuri N, Schvartz D, Fernández-Irigoyen J, Sánchez JC, Santamaría E. Proteomic Characterization of the Olfactory Molecular Imbalance in Dementia with Lewy Bodies. International Journal of Molecular Sciences. 2020; 21(17):6371. https://doi.org/10.3390/ijms21176371

Chicago/Turabian Style

Lachén-Montes, Mercedes, Naroa Mendizuri, Domitille Schvartz, Joaquín Fernández-Irigoyen, Jean Charles Sánchez, and Enrique Santamaría. 2020. "Proteomic Characterization of the Olfactory Molecular Imbalance in Dementia with Lewy Bodies" International Journal of Molecular Sciences 21, no. 17: 6371. https://doi.org/10.3390/ijms21176371

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