Next Article in Journal
Comprehensive Insight into Tapetum-Mediated Pollen Development in Arabidopsis thaliana
Next Article in Special Issue
Age-Dependent Surface Receptor Expression Patterns in Immature Versus Mature Platelets in Mouse Models of Regenerative Thrombocytopenia
Previous Article in Journal
Tight Junction Protein Signaling and Cancer Biology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Multi-Trait Association Analysis of Brain Disorders and Platelet Traits Identifies Novel Susceptibility Loci for Major Depression, Alzheimer’s and Parkinson’s Disease

by
Alfonsina Tirozzi
1,
Miriam Shasa Quiccione
1,
Chiara Cerletti
1,
Maria Benedetta Donati
1,
Giovanni de Gaetano
1,
Licia Iacoviello
1,2,* and
Alessandro Gialluisi
1,2
1
Department of Epidemiology and Prevention, IRCCS Neuromed, Via dell’Elettronica, 86077 Pozzilli, Italy
2
EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Via Ravasi 2, 21100 Varese, Italy
*
Author to whom correspondence should be addressed.
Cells 2023, 12(2), 245; https://doi.org/10.3390/cells12020245
Submission received: 24 November 2022 / Revised: 24 December 2022 / Accepted: 31 December 2022 / Published: 6 January 2023
(This article belongs to the Special Issue Platelet Function and Platelet Proteomics in Ageing-Related Diseases)

Abstract

:
Among candidate neurodegenerative/neuropsychiatric risk-predictive biomarkers, platelet count, mean platelet volume and platelet distribution width have been associated with the risk of major depressive disorder (MDD), Alzheimer’s disease (AD) and Parkinson’s disease (PD) through epidemiological and genomic studies, suggesting partial co-heritability. We exploited these relationships for a multi-trait association analysis, using publicly available summary statistics of genome-wide association studies (GWASs) of all traits reported above. Gene-based enrichment tests were carried out, as well as a network analysis of significantly enriched genes. We analyzed 4,540,326 single nucleotide polymorphisms shared among the analyzed GWASs, observing 149 genome-wide significant multi-trait LD-independent associations (p < 5 × 10−8) for AD, 70 for PD and 139 for MDD. Among these, 27 novel associations were detected for AD, 34 for PD and 40 for MDD. Out of 18,781 genes with annotated variants within ±10 kb, 62 genes were enriched for associations with AD, 70 with PD and 125 with MDD (p < 2.7 × 10−6). Of these, seven genes were novel susceptibility loci for AD (EPPK1, TTLL1, PACSIN2, TPM4, PIF1, ZNF689, AZGP1P1), two for PD (SLC26A1, EFNA3) and two for MDD (HSPH1, TRMT61A). The resulting network showed a significant excess of interactions (enrichment p = 1.0 × 10−16). The novel genes that were identified are involved in the organization of cytoskeletal architecture (EPPK1, TTLL1, PACSIN2, TPM4), telomere shortening (PIF1), the regulation of cellular aging (ZNF689, AZGP1P1) and neurodevelopment (EFNA3), thus, providing novel insights into the shared underlying biology of brain disorders and platelet parameters.

1. Introduction

Platelets have represented, for decades, an interesting setting to investigate the biological underpinnings of neuropsychiatric and neurodegenerative disorders since they are considered “circulating mirrors of neurons” [1]. Indeed, despite their different embryonic origin, platelets and neurons share common characteristics in subcellular organization and in protein composition. There are proteins typically expressed in both neurons and circulating platelets, and they were found to regulate processes such as platelet activation, hemostasis and thrombosis [2]. For example, Reelin- neuronal protein that regulates cell migration, synaptic plasticity and memory formation- is also expressed in blood and is actively released following platelet activation [3,4]. Amyloid Aβ peptides, which accumulate in senile plaques in dementia, and the amyloid precursor protein (APP), are expressed in megakaryocytes, stored in platelet α-granules and released upon platelet activation [1]. Released APP is able to participate in hemostasis and can trigger platelet activation, adhesion and aggregation through a number of different pathways [5]. Soluble APP inhibits the activity of the blood coagulation factors IXa, XIa and Xa, and, to a lesser extent, of factor VIIa–tissue factor complex [6]. It plays a role in the coagulation cascade, modulating hemostasis following vascular injury. Brain derived neurotrophic factor (BDNF)-a secretory protein regulating the development and function of neural circuits-is expressed in both central and peripheral nervous systems but also in human megakaryocyte α-granules together with platelet factor 4 (PF4), where they are stored and released by platelets at the site of injury during platelet aggregation [5,7]. Similarly, serotonin—a neurotransmitter with important roles in controlling behavior and sociality—is stored in platelet-dense granules, where it is released upon activation to act as a weak agonist [3,4].
Epidemiological and genetic evidence also supports the existence of a bridge between platelet traits and neurodegenerative/neuropsychiatric disorders, such as Alzheimer’s disease (AD), Parkinson’s disease (PD) and major depressive disorder (MDD), which often suffer from a lack of risk-predictive biomarkers [5]. Among candidate circulating biomarkers, platelet parameters such as platelet count (Plt), mean platelet volume (MPV) and platelet distribution width (PDW) have been associated with the risk of MDD, AD and PD [7]. Some epidemiological studies consistently reported a positive association between MPV and MDD [5,7,8,9,10], as well as between depressive symptoms and PDW, while the evidence of an association between Plt and MDD status is less consistent [7]. Similarly, increased MPV was also reported for PD-although other studies observed an inverse correlation with the staging of the disease [7,11]-and with AD and cognitive performance, although not always consistently across studies and types of dementia [7]. PDW has been instead more consistently (inversely) associated not only with AD risk [5] but also with mild cognitive impairment and vascular dementia [12].
More recently, genomic studies investigated the genetic correlation-or single nucleotide polymorphism (SNP)-based co-heritability-of platelet traits and brain disorders, as well as potential causality links through Mendelian randomization (MR) approaches. In a large genome-wide association study (GWAS) of blood cell measures, Astle et al. performed a multivariable MR analysis on platelet parameters and MDD risk, which revealed no significant causal effect of the formers on the latter, although MPV and PDW showed marginally significant effects [5]. Later on, Wray et al. investigated genetic correlations between MDD risk and Plt and MPV, reporting no significant genetic correlations between depression and platelet parameters [13], which we instead observed for PDW and MDD risk [14]. Our group later detected a significant genomic overlap between PDW and PD risk through linkage disequilibrium (LD) score regression, which was confirmed also by a polygenic score analysis, as well as a trend of significance for genetic correlations between PDW and AD risk [5]. Despite these promising genetic findings, the variants and genes at the basis of the genomic overlap between platelet parameters and brain disorders remain largely under-investigated. A first attempt was made to carry out a multi-trait genetic association analysis between PD age at onset and MPV, which revealed novel associations in interesting candidate genes such as KALRN (Kalirin RhoGEF Kinase), encoding a PINK1 interactor previously implicated in schizophrenia, AD and PD [5].
Here, we aimed to identify novel genetic associations with AD, PD and MDD using a multi-trait association analysis (MTAG) approach [15], exploiting their reciprocal genetic correlations and evidence of genomic overlap previously identified among themselves and with three common platelet parameters, namely, Plt, MPV and PDW. Such an approach provided higher power to detect novel susceptibility variants for neuropsychiatric and neurodegenerative disorders, as well as overlapping genes and pathways enriched for these associations. This allowed us to untangle the resulting molecular networks at a more fine-grained resolution, revealing potential molecular targets for future treatments of these disorders.

2. Materials and Methods

We used MTAG [15] to perform a multi-trait association analysis of three neuropsychiatric/neurodegenerative disorders-AD, PD and MDD-and three common platelet parameters that have been associated with their risk, namely, Plt, MPV and PDW [7]. MTAG is a generalization of standard, inverse-variance-weighted meta-analysis between two or more genetically correlated traits, which generates trait-specific associations for each genetic variant shared among the source GWAS studies, using linkage disequilibrium (LD) score regression to account for potential sample overlap [15]. The summary statistics of AD (71,880 cases, 383,378 controls and 13,309,438 SNPs) [16], PD (37,688 cases, 18,618 UK Biobank proxy-cases, 1,417,791 controls and 10,081,487 SNPs) [17], MDD (246,363 cases, 561,190 controls and 7,880,531 SNPs) [18], Plt (38,561,936 SNPs), MPV (41,254,093 SNPs) and PDW (41,253,708 SNPs; all with N = 408,112 participants of European ancestry) [19] were obtained from published GWAS summary statistics data (see URLs). To carry out MTAG analysis, we first pre-processed and quality-controlled summary statistics from each study involved: SNP (rs) ids were retrieved, and Z-score was computed by log (OR)/SE when these were not available; MAF threshold was set at 0.01; indels were removed and SNPs mapping to the same position of other variants and/or showing conflicting alleles among different studies were dropped. After variant filtering, the number of variants left for analysis was 8,632,257 for AD; 6,582,074 for PD; 7,183,400 for MDD; 8,933,201 for Plt; 8,933,763 for MPV; and 8,934,170 for PDW. Of these, 4,540,326 variants were in common among the different studies and, therefore, underwent MTAG analysis. Using Functional Mapping and Annotation (FUMA) platform [20], we first identified LD-independent genome-wide significant SNPs within each study (p < 5.0 × 10−8; pairwise r2 < 0.6 in a 1 Mb window). Then, we selected novel genome-wide significant SNP associations with the analyzed disorders, defined as associations not detected as genome-wide significant in the original study, nor in previous GWASs of the same disorder, based on the GWAS catalog and literature search until 30 April 2022 (Table 1).
To have further biological insights into the underlying biology of common variance in different neuropsychiatric/neurodegenerative disorders and platelet parameters, associations underwent gene, gene ontology and pathway enrichment analysis through MAGMA v1.08 [21], within the FUMA platform [20]. This was carried out for all the protein-coding genes to which at least one SNP was annotated within a ±10 kb interval, namely, 18,781 genes. A Bonferroni correction for multiple testing was applied accordingly, based on the number of genes tested (α = 2.7 × 10−6) (Table 2).
To estimate protein–protein interactions (PPIs) among the genes enriched for associations, we used the search tool for the retrieval of interacting genes/proteins in db-STRING v11.5 [22]. We analyzed genes significantly enriched for associations with AD, PD and MDD, first separately and then merged into a single list, to compute “global” interactions among all the genes significantly enriched for any of the three disorders. The obtained network included both direct (physical) and indirect (functional) associations, specifically evidence of interaction from curated databases; evidence experimentally determined; gene neighborhood; gene fusions; gene co-occurrence; joint gene mentioning based on text mining in published articles; gene co-expression; and protein homology. We set a minimum interaction score of >0.7 so that only high-confidence interactions between proteins were included in the analysis. The observed excess of interactions compared to the expected number of edges among nodes, average node degree (i.e., the average number of edges per node in the graph) and local clustering coefficient (i.e., a measure of the extent to which nodes in the graph tend to cluster together) were taken as measures of network density and clustering levels.

3. Results

In the multi-trait association analysis of three different brain disorders (AD, PD, MDD) and three different platelet parameters (Plt, MPV and PDW), we analyzed 4,540,326 variants shared across the different source studies that passed QC (see Methods section). Given the main aim of the manuscript, we report in detail below the results of the associations with AD, PD and MDD, along with the overlap with platelet parameters in terms of single variant and the gene enrichment of associations. The full results of the MTAG analysis of platelet traits are reported in Supplementary Materials (Figures S1 and S2) and online (see Data Availability Statement). MTAG revealed 358 genome-wide significant multi-trait associations (p < 5 × 10−8; pairwise r2 < 0.6; Figure 1): 149 for AD (top hit at rs1081105, within the APOE gene, p = 2.36 × 10−180), 70 for PD (top hit at rs356219, within the SNCA gene, p = 6.01 × 10−41) and 139 for MDD (top hit at rs2232429, within the ZSCAN12 gene, p = 6.41 × 10−19). Among these, we observed 27 novel associations with AD (top hit at rs2232429, within the ITGB5 gene, p = 6.41 × 10−19), 34 with PD (top hit at rs1372518 within the SNCA gene, p = 3.33 × 10−28) and 40 with MDD (top hit at rs200965 in a transcription factor binding site on 6p22.1, p = 4.07 × 10−15). Of these SNPs, seven were mapped in novel susceptibility genes for AD (EPPK1, TTLL1, PACSIN2, TPM4, PIF1, ZNF689 and AZGP1P1), two for PD (SLC26A1 and EFNA3) and two for MDD (HSPH1 and TRMT61A). The analysis of 18,781 genes tested in the gene-based enrichment analysis revealed 62, 70 and 125 genes with a significant enrichment of associations with AD, PD and MDD, surviving correction for multiple gene testing (p < 2.7 × 10−6, Figure 2). Of these, eight genes represented novel associations with AD (PVRIG, FAM57B, NDFUS2, C16orf92, KLC3, B4GALT3, ZNF688 and DEDD), 14 with PD (DPM3, SLC26A1, FBXL19, SMIM15, ERCC8, FAM200B, CTF1, ADAM15, PRSS8, NCOR1, VKORC1, ZNF668, PRSS36 and SRCAP) and 38 with MDD (ZNF165, BTN2A2, OR2W1, OR12D3, TRMT61A, OR2J1, HMGN4, OR2B3, BTN3A3, ZNF197, ZNF35, TRIM27, OR2B6, DLST, RHOBTB1, ZNF660, RBM4B, ITGB6, RBM14-RBM4, HIST1H4K, HIST1H2AK, HIST1H2BL, HIST1H3H, HIST1H2AL, HIST1H1B, HIST1H3I, HIST1H4J, HIST1H2BM, HIST1H2AI, HIST1H2AJ, HIST1H2AM, HIST1H3J, HIST1H2BN, HIST1H2AG, HIST1H2BO, HIST1H4L, HIST1H2BJ and HIST1H4I). None of these genes were overlapping across the three disorders. However, when we checked the overlap between each neurodegenerative/neuropsychiatric disorder and platelet parameters in terms of single variant associations, we found 24 SNPs associated with AD and 3 associated with PD, which were also associated with 1 or more platelet parameters. Of these, 12 AD- and 3 PD-associated SNPs represented novel associations never detected before (Table 3a,b). As for overlapping gene enrichments with any of the platelet parameters analyzed, we identified 29 genes matching with AD, 14 with PD and 15 with MDD. Of these, 6 were novel susceptibility loci for AD, 4 for PD and 12 for MDD (Table 4a–c).
The molecular network resulting from the gene enrichment test, as produced by STRING v11.5 analysis, showed more significant interactions than expected for AD (enrichment p = 1.0 × 10−16; 57 nodes, 57 edges vs. 2 expected, average node degree 2.00 and average local clustering coefficient 0.36), PD (enrichment p = 8.46 × 10−9; 63 nodes, 18 edges vs. 3 expected, average node degree 0.57 and average local clustering coefficient 0.23) and MDD (enrichment p = 1.0 × 10−16; 120 nodes, 264 edges vs. 49 expected, average node degree 4.4 and average local clustering coefficient 0.47) (Figure S3a–c; Table S1). Similarly, we observed evidence of an interaction also when the genes enriched for the three disorders were analyzed together (enrichment p = 1.0 × 10−16; 240 nodes, 281 edges vs. 61 expected, average node degree 2.34 and average local clustering coefficient 0.3) (Figure 3; Table S1).
Gene-set analysis also revealed significant enrichment for the three disorders, the most associated gene ontology (GO) term was negative regulation of amyloid precursor protein catabolic process (13 genes, β(SE) = 2.23(0.26); enrichment p after Bonferroni correction = 2.7 × 10−13) for AD, IgG binding (9 genes, β(SE) = 0.03(0.31); Pbonf = 0.0056) for PD and GABAergic synapse (64 genes, β(SE) = 0.74(0.13); Pbonf = 9.5 × 10−5) for MDD (Table S2; see URLs to access the full list of pathways tested and the genes driving these enrichments).

4. Discussion

We report the first multi-trait association analysis of structural platelet parameters routinely assessed in blood tests and three of the most common neurodegenerative/neuropsychiatric disorders, identifying novel candidate susceptibility genes for AD, PD and MDD. The most significant associations were detected in some of the most implicated genes in neurodegenerative/neuropsychiatric disorders, namely, APOE (apolipoprotein E, with AD) [39], SNCA (alpha synuclein, with PD) [40] and ZSCAN12 (zinc finger and SCAN domain-containing 12, with MDD) [41]. APOE is a protein associated with lipid particles that mainly functions in lipoprotein-mediated lipid transport between organs via plasma and interstitial fluids [42]. Alpha synuclein is involved in synaptic activities such as the regulation of synaptic vesicle trafficking and subsequent neurotransmitter release [43,44]; moreover, it modulates DNA repair processes, including the repair of double-strand breaks [45]. ZSCAN12 encodes a Zinc finger and SCAN domain-containing protein involved in transcriptional regulation.
Still, MTAG analysis also revealed novel genes showing significant multi-trait associations, which, to our knowledge, were never associated with these disorders before. Among genes associated with AD, EPPK1, TTLL1, PACSIN2 and TPM4 play a role in the organization of cytoskeletal architecture, which has been identified as an important component in the development of neurodegenerative disorders [46,47,48,49]. PIF1 prevents telomere elongation by inhibiting the action of telomerase, while ZNF689 and AZGP1P1 are transcription factors involved in cell viability and apoptosis, and both molecular functions can affect cellular aging and the development of age-related disorders such as AD and PD [24]. Moreover, TTLL1 [19], PACSIN2 [26,40], TPM4 [27] and PIF1 [19] were previously associated with platelet parameters, suggesting a possible pleiotropic effect of these genes. EFNA3, a novel gene resulting in an association with PD, encodes a member of the ephrin family, previously implicated in mediating developmental events, especially in the central nervous system [50].
A gene-based enrichment analysis also revealed novel genes associated with AD, PD and MDD. Interestingly, among these genes are several encode transcription factors that may be involved in development, maintenance and survival of neurons and olfactory receptors [51]. Indeed, olfactory dysfunction, which is thought to be due to the loss of synaptic function, has been linked with most neurodegenerative, neuropsychiatric and communication disorders [52]. Moreover, among these genes, there are also some histone complex proteins, in line with some recent studies revealing associations between histone methylation/acetylation and AD [53] and implicating several histone deacetylases in the pathogenesis of PD [54]. These findings suggest the pleiotropic influence of several genes on the risk of neurodegenerative and neuropsychiatric disorders, which were not previously detected through classical univariate GWAS analyses.
Of note, we found several overlaps between genes and SNPs multi-trait associations with the brain disorders and platelet parameters analyzed. Among genes enriched for association, we identified clusters of genes encoding products involved in mitochondrial function (e.g., NDUFS2, NDUFAF2 and TOMM40L), cytoskeleton remodeling (CD2AP and KLC3, as discussed above) and histone proteins (HIST1H2BK, HIST1H4K, HIST1H2AK and HIST1H3B, as explained below). Indeed, NDUFS2 and NDUFAF2 encode for a subunit and for a chaperone involved in the assembly of complex I, located on the inner mitochondrial membrane, while TOMM40L is involved in mitochondrial transmembrane translocation. Several studies suggest that platelet mitochondrial dysfunction may be involved in neurodegenerative diseases such as AD and PD [55,56,57]. Still, further studies are needed to clarify the variant association overlap between platelet parameters and MDD, which we were not able to identify here, possibly due to the genetic and phenotypic heterogeneity of depression.
Protein–protein interaction analysis revealed a significant excess of interactions among enriched genes for the brain disorders tested both separately and jointly, suggesting that their gene products are highly likely to be linked in a global molecular network.
In particular, this highlighted some local networks of interests, such as the one among the histone proteins complex-HIST1H2BI, HIST1H2BF and HIST1H2BJ-which may play a role in the onset of neurodegenerative diseases due to the alteration of methylation patterns [58]. Similarly, the apolipoproteins APOE, APOA2, APOC1 and APOC4 have been repeatedly implicated in triglyceride and cholesterol transport and metabolism [59,60], as well as in neurodegenerative [61] and cardiovascular risk [62], while the local network among EPHA1, EPHB2, EFNA1, EFNA3 and EFNA4, highlights the importance of the interactions between ephrins and ephrin receptors in the etiology of several neurodegenerative and neuroinflammatory disorders, suggesting potential links with (cellular) immunity [63].
Gene-set analysis revealed significant enrichments of GO terms involved in the regulation, formation and catabolic processes of amyloid beta and in the negative regulation of metalloendopeptidase activity for AD, supporting the hypothesis that metallopeptidases are implicated in the pathogenesis of several central nervous system diseases such as multiple sclerosis and AD [64]. For PD, significant enrichments of IgG binding is interesting in light of a higher fraction of IgG, a different IgG glycosylation profile [65], and of increased IgG (but not IgM) binding in dopaminergic neurons of PD cases vs. controls [66]. Moreover, serum IgG levels in PD patients are negatively associated with mood/cognition scores [67], in line with a potential pleiotropic role of humoral immunity at the interface among the mood, cognitive and motor control domains. Similarly, the significant enrichment of GABAergic synapse GO term in MDD analysis corroborates the hypothesis that the alteration in GABAergic receptors may play a role in long-term depression [68].
Overall, we provide insights into the shared underlying biology of these disorders and related platelet parameters, proposing novel molecular targets for the risk prediction and treatment of these disorders.

Strengths and Limitations

The strengths of this study include the novelty of the analysis performed; indeed, to our knowledge, this represents the first attempt to identify the shared genomic underpinnings of platelet parameters and three of the most common neurodegenerative/neuropsychiatric disorders through a comprehensive approach, including not only multi-trait association analysis but also gene-/gene-set enrichment and molecular network analyses. Moreover, our analyses are based on large GWASs, with an important amount of genetic data, which confers robustness to our observations. Last, our focus on novel associations allowed identifying proteins and biological pathways, which should be functionally validated in the future.
This study presents some limitations. First, currently only partial evidence of genetic correlation among the disorders and platelet parameters tested exists, which may have hampered the power of the analyses. Second, in multi-trait association approaches, association significance is often driven by the largest source GWAS involved in the MTAG analysis, which may have biased the analyses towards the largest studies. Still, this represents a useful approach to identifying the pleiotropic variants and genes influencing multiple traits and/or disorders, which were already proven successful with multiple correlated phenotypes [15]. Third, functional studies are warranted to explain the role of the novel susceptibility genes identified here, both in neurodegenerative/neuropsychiatric risk and platelet variability.
However, although there are some limitations, these studies may reveal potential molecular targets for future treatments of three of the most common neurodegenerative/neuropsychiatric disorders.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cells12020245/s1, Figure S1: Manhattan plots of multi-trait associations with (a) MPV, (b) PDW and (c) Plt; Figure S2: Manhattan plots of gene-based enrichments of multi-trait associations with (a) MPV, (b) PDW and (c) Plt; Figure S3: Protein–protein interaction network of genes significantly enriched for associations with (a) AD, (b) PD and (c) MDD; Table S1: Network statistics of protein–protein interaction networks of genes significantly enriched for associations with AD, PD and MDD; Table S2: Significant gene-set enrichments of associations with (a) AD, (b) PD and (c) MDD.

Author Contributions

A.G., L.I. and G.d.G. conceived this study. A.T. and M.S.Q. performed analyses and data curation under the supervision of A.G., L.I., G.d.G., M.B.D. and C.C. provided critical feedback on the study. All co-authors contributed to manuscript writing and critical review. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by Ricerca Corrente 2022-24 (Italian Ministry of Health) and partly supported by Progetto Platone-Platform for Integrated Health Life (Italian Ministry of Economic Development; PON I&C 2014–2020 call, project code: F/080032/01/X35) and by INAIL-Bric 2019 ID 47 project (Italian National Institute for Insurance against Accidents at Work; 2019 call, project CUP code: F24I19000630008).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

AD summary statistics: https://ctg.cncr.nl/software/summary_statistics (accessed on 10 January 2019); PD summary statistics: https://drive.google.com/file/d/1FZ9UL99LAqyWnyNBxxlx6qOUlfAnublN/view?usp=sharing (accessed on 26 March 2020); MDD Summary statistics: https://datashare.ed.ac.uk/handle/10283/3203 (accessed on 30 January 2019); Plt summary statistics: http://ftp.ebi.ac.uk/pub/databases/gwas/summary_statistics/GCST90002001-GCST90003000/GCST90002402/ (accessed on 30 January 2022); MPV summary statistics: http://ftp.ebi.ac.uk/pub/databases/gwas/summary_statistics/GCST90002001-GCST90003000/GCST90002395/ (accessed on 30 January 2022); PDW summary statistics: http://ftp.ebi.ac.uk/pub/databases/gwas/summary_statistics/GCST90002001-GCST90003000/GCST90002401/ (accessed on 30 January 2022).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Canobbio, I. Blood platelets: Circulating mirrors of neurons? Res. Pract. Thromb. Haemost. 2019, 3, 564–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Canobbio, I.; Barbieri, S.S. Are platelets more than a model of brain neurons? Bleeding Thromb. Vasc. Biol. 2022, 1, 1. [Google Scholar] [CrossRef]
  3. Tseng, W.-L.; Huang, C.-L.; Chong, K.-Y.; Liao, C.-H.; Stern, A.; Cheng, J.-C.; Tseng, C.-P. Reelin is a platelet protein and functions as a positive regulator of platelet spreading on fibrinogen. Cell. Mol. Life Sci. 2010, 67, 641–653. [Google Scholar] [CrossRef] [PubMed]
  4. Krueger, I.; Gremer, L.; Mangels, L.; Klier, M.; Jurk, K.; Willbold, D.; Bock, H.H.; Elvers, M. Reelin Amplifies Glycoprotein VI Activation and AlphaIIb Beta3 Integrin Outside-In Signaling via PLC Gamma 2 and Rho GTPases. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 2391–2403. [Google Scholar] [CrossRef]
  5. Tirozzi, A.; Izzi, B.; Noro, F.; Marotta, A.; Gianfagna, F.; Hoylaerts, M.F.; Cerletti, C.; Donati, M.B.; De Gaetano, G.; Iacoviello, L.; et al. Assessing genetic overlap between platelet parameters and neurodegenerative disorders. Front. Immunol. 2020, 11, 2127. [Google Scholar] [CrossRef]
  6. Canobbio, I.; Abubaker, A.A.; Visconte, C.; Torti, M.; Pula, G. Role of amyloid peptides in vascular dysfunction and platelet dysregulation in Alzheimer’s disease. Front. Cell. Neurosci. 2015, 9, 65. [Google Scholar] [CrossRef] [Green Version]
  7. Izzi, B.; Tirozzi, A.; Cerletti, C.; Donati, M.B.; de Gaetano, G.; Hoylaerts, M.F.; Iacoviello, L.; Gialluisi, A. Beyond Haemostasis and Thrombosis: Platelets in Depression and Its Co-Morbidities. Int. J. Mol. Sci. 2020, 21, 817. [Google Scholar] [CrossRef]
  8. Bondade, S.; Supriya; Seema, H.S.; Shivakumar, B.K. Mean Platelet Volume in Depression and Anxiety Disorder- a Hospital Based Case-control Study. Int. Neuropsychiatr. Dis. J. 2018, 11, 1–8. [Google Scholar] [CrossRef]
  9. Cai, L.; Xu, L.; Wei, L.; Chen, W. Relationship of mean platelet volume to MDD: A retrospective study. Shanghai Arch. Psychiatry 2017, 29, 21–29. [Google Scholar]
  10. Tirozzi, A.; Parisi, R.; Cerletti, C.; Donati, M.B.; de Gaetano, G.; Iacoviello, L.; Gialluisi, A. Genomic Overlap between Platelet Parameters Variability and Age at Onset of Parkinson Disease. Appl. Sci. 2021, 11, 6927. [Google Scholar] [CrossRef]
  11. Nalls, M.A.; Blauwendraat, C.; Vallerga, C.L.; Heilbron, K.; Bandres-Ciga, S.; Chang, D.; Tan, M.; Kia, D.A.; Noyce, A.J.; Xue, A.; et al. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: A meta-analysis of genome-wide association studies. Lancet Neurol. 2019, 18, 1091–1102. [Google Scholar] [CrossRef] [PubMed]
  12. Liang, Q.C.; Jin, D.; Li, Y.; Wang, R.T. Mean platelet volume and platelet distribution width in vascular dementia and Alzheimer’s disease. Platelets 2014, 25, 433–438. [Google Scholar] [CrossRef] [PubMed]
  13. Wray, N.R.; Ripke, S.; Mattheisen, M.; Trzaskowski, M.; Byrne, E.M.; Abdellaoui, A.; Adams, M.J.; Agerbo, E.; Air, T.M.; Andlauer, T.M.F.; et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat. Genet. 2018, 50, 668–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gialluisi, A.; Izzi, B.; Di Castelnuovo, A.; Cerletti, C.; Donati, M.B.; de Gaetano, G.; Iacoviello, L. Revisiting the link between platelets and depression through genetic epidemiology: New insights from platelet distribution width. Haematologica 2019, 105, e246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Turley, P.; Walters, R.K.; Maghzian, O.; Okbay, A.; Lee, J.J.; Fontana, M.A.; Nguyen-Viet, T.A.; Wedow, R.; Zacher, M.; Furlotte, N.A.; et al. Multi-trait analysis of genome-wide association summary statistics using MTAG. Nat. Genet. 2018, 50, 229–237. [Google Scholar] [CrossRef]
  16. Jansen, I.E.; Savage, J.E.; Watanabe, K.; Bryois, J.; Williams, D.M.; Steinberg, S.; Sealock, J.; Karlsson, I.K.; Hägg, S.; Athanasiu, L.; et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat. Genet. 2019, 51, 404–413. [Google Scholar] [CrossRef] [PubMed]
  17. Blauwendraat, C.; Heilbron, K.; Vallerga, C.L.; Bandres-Ciga, S.; von Coelln, R.; Pihlstrøm, L.; Simón-Sánchez, J.; Schulte, C.; Sharma, M.; Krohn, L.; et al. Parkinson’s disease age at onset genome-wide association study: Defining heritability, genetic loci, and α-synuclein mechanisms. Mov. Disord. 2019, 34, 866–875. [Google Scholar] [CrossRef] [Green Version]
  18. Howard, D.M.; Adams, M.J.; Clarke, T.K.; Hafferty, J.D.; Gibson, J.; Shirali, M.; Coleman, J.R.I.; Hagenaars, S.P.; Ward, J.; Wigmore, E.M.; et al. Genome-wide meta-analysis of depression identifies 102 independent variants and highlights the importance of the prefrontal brain regions. Nat. Neurosci. 2019, 22, 343–352. [Google Scholar] [CrossRef] [Green Version]
  19. Vuckovic, D.; Bao, E.L.; Akbari, P.; Lareau, C.A.; Mousas, A.; Jiang, T.; Chen, M.H.; Raffield, L.M.; Tardaguila, M.; Huffman, J.E.; et al. The Polygenic and Monogenic Basis of Blood Traits and Diseases. Cell 2020, 182, 1214–1231.e11. [Google Scholar] [CrossRef]
  20. Watanabe, K.; Taskesen, E.; Van Bochoven, A.; Posthuma, D. Functional mapping and annotation of genetic associations with FUMA. Nat. Commun. 2017, 8, 1826. [Google Scholar] [CrossRef] [Green Version]
  21. de Leeuw, C.A.; Mooij, J.M.; Heskes, T.; Posthuma, D. MAGMA: Generalized gene-set analysis of GWAS data. PLoS Comput. Biol. 2015, 11, e1004219. [Google Scholar] [CrossRef]
  22. Szklarczyk, D.; Gable, A.L.; Nastou, K.C.; Lyon, D.; Kirsch, R.; Pyysalo, S.; Doncheva, N.T.; Legeay, M.; Fang, T.; Bork, P.; et al. The STRING database in 2021: Customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021, 49, D605–D612. [Google Scholar] [CrossRef] [PubMed]
  23. Schwartzentruber, J.; Cooper, S.; Liu, J.Z.; Barrio-Hernandez, I.; Bello, E.; Kumasaka, N.; Young, A.M.H.; Franklin, R.J.M.; Johnson, T.; Estrada, K.; et al. Genome-wide meta-analysis, fine-mapping and integrative prioritization implicate new Alzheimer’s disease risk genes. Nat. Genet. 2021, 53, 392–402. [Google Scholar] [CrossRef]
  24. Bone, W.P.; Siewert, K.M.; Jha, A.; Klarin, D.; Damrauer, S.M.; Chang, K.-M.; Tsao, P.S.; Assimes, T.L.; Ritchie, M.D.; Voight, B.F. Multi-trait association studies discover pleiotropic loci between Alzheimer’s disease and cardiometabolic traits. Alzheimers. Res. Ther. 2021, 13, 34. [Google Scholar] [CrossRef]
  25. Chen, M.-H.; Raffield, L.M.; Mousas, A.; Sakaue, S.; Huffman, J.E.; Moscati, A.; Trivedi, B.; Jiang, T.; Akbari, P.; Vuckovic, D.; et al. Trans-ethnic and Ancestry-Specific Blood-Cell Genetics in 746,667 Individuals from 5 Global Populations. Cell 2020, 182, 1198–1213.e14. [Google Scholar] [CrossRef]
  26. Li, Q.S.; Tian, C.; Hinds, D.; Seabrook, G.R. The association of clinical phenotypes to known AD/FTD genetic risk loci and their inter-relationship. PLoS ONE 2020, 15, e0241552. [Google Scholar] [CrossRef]
  27. Astle, W.J.; Elding, H.; Jiang, T.; Allen, D.; Ruklisa, D.; Mann, A.L.; Mead, D.; Bouman, H.; Riveros-Mckay, F.; Kostadima, M.A.; et al. The Allelic Landscape of Human Blood Cell Trait Variation and Links to Common Complex Disease. Cell 2016, 167, 1415–1429.e19. [Google Scholar] [CrossRef] [Green Version]
  28. Cavalcanti, A.B.; Zampieri, F.G.; Rosa, R.G.; Azevedo, L.C.P.; Veiga, V.C.; Avezum, A.; Damiani, L.P.; Marcadenti, A.; Kawano-Dourado, L.; Lisboa, T.; et al. Hydroxychloroquine with or without Azithromycin in Mild-to-Moderate Covid-19. N. Engl. J. Med. 2020, 383, 2041–2052. [Google Scholar] [CrossRef]
  29. Park, Y.H.; Pyun, J.-M.; Hodges, A.; Jang, J.-W.; Bice, P.J.; Kim, S.; Saykin, A.J.; Nho, K. Dysregulated expression levels of APH1B in peripheral blood are associated with brain atrophy and amyloid-β deposition in Alzheimer’s disease. Alzheimer’s Res. Ther. 2021, 13, 183. [Google Scholar] [CrossRef] [PubMed]
  30. Zhang, X.; Yu, W.; Cao, X.; Wang, Y.; Zhu, C.; Guan, J. Identification of Serum Biomarkers in Patients with Alzheimer’s Disease by 2D-DIGE Proteomics. Gerontology 2022, 68, 686–698. [Google Scholar] [CrossRef] [PubMed]
  31. Xiao, Q.; Xi, J.; Wang, R.; Zhao, Q.; Liang, X.; Wu, W.; Zheng, L.; Guo, Q.; Hong, Z.; Fu, H.; et al. The Relationship Between Low-Density Lipoprotein Cholesterol and Progression of Mild Cognitive Impairment: The Influence of rs6859 in PVRL2. Front. Genet. 2022, 13, 823406. [Google Scholar] [CrossRef] [PubMed]
  32. Sierksma, A.; Lu, A.; Mancuso, R.; Fattorelli, N.; Thrupp, N.; Salta, E.; Zoco, J.; Blum, D.; Buée, L.; De Strooper, B.; et al. Novel Alzheimer risk genes determine the microglia response to amyloid-β but not to TAU pathology. EMBO Mol. Med. 2020, 12, e10606. [Google Scholar] [CrossRef] [PubMed]
  33. Wightman, D.P.; Jansen, I.E.; Savage, J.E.; Shadrin, A.A.; Bahrami, S.; Holland, D.; Rongve, A.; Børte, S.; Winsvold, B.S.; Drange, O.K.; et al. A genome-wide association study with 1,126,563 individuals identifies new risk loci for Alzheimer’s disease. Nat. Genet. 2021, 53, 1276–1282. [Google Scholar] [CrossRef] [PubMed]
  34. Sakaue, S.; Kanai, M.; Tanigawa, Y.; Karjalainen, J.; Kurki, M.; Koshiba, S.; Narita, A.; Konuma, T.; Yamamoto, K.; Akiyama, M.; et al. A cross-population atlas of genetic associations for 220 human phenotypes. Nat. Genet. 2021, 53, 1415–1424. [Google Scholar] [CrossRef]
  35. Lu, S.-S.; Gong, F.-F.; Feng, F.; Hu, C.-Y.; Qian, Z.-Z.; Wu, Y.-L.; Yang, H.-Y.; Sun, Y.-H. Association of microtubule associated protein tau/Saitohin (MAPT/STH) MAPT_238bp/STH Q7R polymorphisms and Parkinson’s disease: A meta-analysis. Biochem. Biophys. Res. Commun. 2014, 453, 653–661. [Google Scholar] [CrossRef]
  36. Lutz, M.W.; Sprague, D.; Barrera, J.; Chiba-Falek, O. Shared genetic etiology underlying Alzheimer’s disease and major depressive disorder. Transl. Psychiatry 2020, 10, 88. [Google Scholar] [CrossRef] [Green Version]
  37. Zhang, F.; Cao, H.; Baranova, A. Shared Genetic Liability and Causal Associations Between Major Depressive Disorder and Cardiovascular Diseases. Front. Cardiovasc. Med. 2021, 8, 735136. [Google Scholar] [CrossRef]
  38. Loh, P.-R.; Kichaev, G.; Gazal, S.; Schoech, A.P.; Price, A.L. Mixed-model association for biobank-scale datasets. Nat. Genet. 2018, 50, 906–908. [Google Scholar] [CrossRef]
  39. Yin, Y.; Wang, Z. ApoE and Neurodegenerative Diseases in Aging. Adv. Exp. Med. Biol. 2018, 1086, 77–92. [Google Scholar] [CrossRef]
  40. Cherian, A.; Divya, K.P. Genetics of Parkinson’s disease. Acta Neurol. Belg. 2020, 120, 1297–1305. [Google Scholar] [CrossRef] [PubMed]
  41. Al-Naama, N.; Mackeh, R.; Kino, T. C(2)H(2)-Type Zinc Finger Proteins in Brain Development, Neurodevelopmental, and Other Neuropsychiatric Disorders: Systematic Literature-Based Analysis. Front. Neurol. 2020, 11, 32. [Google Scholar] [CrossRef] [PubMed]
  42. Krimbou, L.; Denis, M.; Haidar, B.; Carrier, M.; Marcil, M.; Genest, J.J. Molecular interactions between apoE and ABCA1: Impact on apoE lipidation. J. Lipid Res. 2004, 45, 839–848. [Google Scholar] [CrossRef] [Green Version]
  43. Logan, T.; Bendor, J.; Toupin, C.; Thorn, K.; Edwards, R.H. α-Synuclein promotes dilation of the exocytotic fusion pore. Nat. Neurosci. 2017, 20, 681–689. [Google Scholar] [CrossRef] [PubMed]
  44. Huang, C.-C.; Chiu, T.-Y.; Lee, T.-Y.; Hsieh, H.-J.; Lin, C.-C.; Kao, L.-S. Soluble α-synuclein facilitates priming and fusion by releasing Ca2+ from the thapsigargin-sensitive Ca2+ pool in PC12 cells. J. Cell Sci. 2018, 131, jcs213017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Schaser, A.J.; Osterberg, V.R.; Dent, S.E.; Stackhouse, T.L.; Wakeham, C.M.; Boutros, S.W.; Weston, L.J.; Owen, N.; Weissman, T.A.; Luna, E.; et al. Alpha-synuclein is a DNA binding protein that modulates DNA repair with implications for Lewy body disorders. Sci. Rep. 2019, 9, 10919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Shimada, H.; Nambu-Niibori, A.; Wilson-Morifuji, M.; Mizuguchi, S.; Araki, N.; Sumiyoshi, H.; Sato, M.; Mezaki, Y.; Senoo, H.; Ishikawa, K.; et al. Epiplakin modifies the motility of the HeLa cells and accumulates at the outer surfaces of 3-D cell clusters. J. Dermatol. 2013, 40, 249–258. [Google Scholar] [CrossRef]
  47. Yao, G.; Su, X.; Nguyen, V.; Roberts, K.; Li, X.; Takakura, A.; Plomann, M.; Zhou, J. Polycystin-1 regulates actin cytoskeleton organization and directional cell migration through a novel PC1-Pacsin 2-N-Wasp complex. Hum. Mol. Genet. 2014, 23, 2769–2779. [Google Scholar] [CrossRef] [Green Version]
  48. Marchenko, M.; Nefedova, V.; Artemova, N.; Kleymenov, S.; Levitsky, D.; Matyushenko, A. Structural and Functional Peculiarities of Cytoplasmic Tropomyosin Isoforms, the Products of TPM1 and TPM4 Genes. Int. J. Mol. Sci. 2021, 22, 5141. [Google Scholar] [CrossRef]
  49. Bodakuntla, S.; Yuan, X.; Genova, M.; Gadadhar, S.; Leboucher, S.; Birling, M.-C.; Klein, D.; Martini, R.; Janke, C.; Magiera, M.M. Distinct roles of α- and β-tubulin polyglutamylation in controlling axonal transport and in neurodegeneration. EMBO J. 2021, 40, e108498. [Google Scholar] [CrossRef]
  50. Duffy, S.L.; Steiner, K.A.; Tam, P.P.L.; Boyd, A.W. Expression analysis of the Epha1 receptor tyrosine kinase and its high-affinity ligands Efna1 and Efna3 during early mouse development. Gene Expr. Patterns 2006, 6, 719–723. [Google Scholar] [CrossRef]
  51. Wang, R.; Yang, S.; Nie, T.; Zhu, G.; Feng, D.; Yang, Q. Transcription Factors: Potential Cell Death Markers in Parkinson’s Disease. Neurosci. Bull. 2017, 33, 552–560. [Google Scholar] [CrossRef] [PubMed]
  52. Bhatia-Dey, N.; Heinbockel, T. The Olfactory System as Marker of Neurodegeneration in Aging, Neurological and Neuropsychiatric Disorders. Int. J. Environ. Res. Public Health 2021, 18, 6976. [Google Scholar] [CrossRef] [PubMed]
  53. Zima, L.; West, R.; Smolen, P.; Kobori, N.; Hergenroeder, G.W.; Choi, H.A.; Moore, A.N.; Redell, J.B.; Dash, P.K. Epigenetic modifications and their potential contributions to traumatic brain injury pathobiology and outcome. J. Neurotrauma 2022, 39, 1279–1288. [Google Scholar] [CrossRef] [PubMed]
  54. Li, Y.; Gu, Z.; Lin, S.; Chen, L.; Dzreyan, V.; Eid, M.; Demyanenko, S.; He, B. Histone Deacetylases as Epigenetic Targets for Treating Parkinson’s Disease. Brain Sci. 2022, 12, 672. [Google Scholar] [CrossRef] [PubMed]
  55. Koçer, A.; Yaman, A.; Niftaliyev, E.; Dürüyen, H.; Eryilmaz, M.; Koçer, E. Assessment of platelet indices in patients with neurodegenerative diseases: Mean platelet volume was increased in patients with Parkinson’s disease. Curr. Gerontol. Geriatr. Res. 2013, 2013, 986254. [Google Scholar] [CrossRef]
  56. Subrahmanian, N.; LaVoie, M.J. Is there a special relationship between complex I activity and nigral neuronal loss in Parkinson’s disease? A critical reappraisal. Brain Res. 2021, 1767, 147434. [Google Scholar] [CrossRef] [PubMed]
  57. Melchinger, H.; Jain, K.; Tyagi, T.; Hwa, J. Role of Platelet Mitochondria: Life in a Nucleus-Free Zone. Front. Cardiovasc. Med. 2019, 6, 153. [Google Scholar] [CrossRef] [Green Version]
  58. Kaur, G.; Rathod, S.S.S.; Ghoneim, M.M.; Alshehri, S.; Ahmad, J.; Mishra, A.; Alhakamy, N.A. DNA Methylation: A Promising Approach in Management of Alzheimer’s Disease and Other Neurodegenerative Disorders. Biology 2022, 11, 90. [Google Scholar] [CrossRef]
  59. Fuior, E.V.; Gafencu, A.V. Apolipoprotein C1: Its Pleiotropic Effects in Lipid Metabolism and Beyond. Int. J. Mol. Sci. 2019, 20, 5939. [Google Scholar] [CrossRef] [Green Version]
  60. Dominiczak, M.H.; Caslake, M.J. Apolipoproteins: Metabolic role and clinical biochemistry applications. Ann. Clin. Biochem. 2011, 48, 498–515. [Google Scholar] [CrossRef]
  61. Van Giau, V.; Bagyinszky, E.; An, S.S.A.; Kim, S.Y. Role of apolipoprotein E in neurodegenerative diseases. Neuropsychiatr. Dis. Treat. 2015, 11, 1723–1737. [Google Scholar] [CrossRef]
  62. Zhou, Y.; Mägi, R.; Milani, L.; Lauschke, V.M. Global genetic diversity of human apolipoproteins and effects on cardiovascular disease risk. J. Lipid Res. 2018, 59, 1987–2000. [Google Scholar] [CrossRef] [Green Version]
  63. Darling, T.K.; Lamb, T.J. Emerging roles for Eph receptors and ephrin ligands in immunity. Front. Immunol. 2019, 10, 1473. [Google Scholar] [CrossRef] [Green Version]
  64. Wee Yong, V.; Forsyth, P.A.; Bell, R.; Krekoski, C.A.; Edwards, D.R. Matrix metalloproteinases and diseases of the CNS. Trends Neurosci. 1998, 21, 75–80. [Google Scholar] [CrossRef] [PubMed]
  65. Russell, A.C.; Šimurina, M.; Garcia, M.T.; Novokmet, M.; Wang, Y.; Rudan, I.; Campbell, H.; Lauc, G.; Thomas, M.G.; Wang, W. The N-glycosylation of immunoglobulin G as a novel biomarker of Parkinson’s disease. Glycobiology 2017, 27, 501–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Orr, C.F.; Rowe, D.B.; Mizuno, Y.; Mori, H.; Halliday, G.M. A possible role for humoral immunity in the pathogenesis of Parkinson’s disease. Brain 2005, 128, 2665–2674. [Google Scholar] [CrossRef] [Green Version]
  67. Sun, C.; Yu, W.; Zhao, Z.; Song, C.; Liu, Y.; Jia, G.; Wang, X.; Liu, Y. Peripheral Humoral Immune Response Is Associated With the Non-motor Symptoms of Parkinson’s Disease. Front. Neurosci. 2019, 13, 1057. [Google Scholar] [CrossRef] [Green Version]
  68. Sastry, B.R.; Morishita, W.; Yip, S.; Shew, T. Gaba-ergic transmission in deep cerebellar nuclei. Prog. Neurobiol. 1997, 53, 259–271. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Manhattan plots of multi-trait associations with (a) AD, (b) PD and (c) MDD. The x-axis shows chromosomal position, and the y-axis shows association p-values on a −log10 scale. Red dashed line represents the statistical significance thresholds (α = 5 × 10−8).
Figure 1. Manhattan plots of multi-trait associations with (a) AD, (b) PD and (c) MDD. The x-axis shows chromosomal position, and the y-axis shows association p-values on a −log10 scale. Red dashed line represents the statistical significance thresholds (α = 5 × 10−8).
Cells 12 00245 g001aCells 12 00245 g001b
Figure 2. Manhattan plots of gene-based enrichments of multi-trait associations with (a) AD, (b) PD and (c) MDD. The x-axis shows chromosomal position, and the y-axis shows association p-values on a −log10 scale. Red dashed line represents the statistical significance thresholds (α = 2.7 × 10−6).
Figure 2. Manhattan plots of gene-based enrichments of multi-trait associations with (a) AD, (b) PD and (c) MDD. The x-axis shows chromosomal position, and the y-axis shows association p-values on a −log10 scale. Red dashed line represents the statistical significance thresholds (α = 2.7 × 10−6).
Cells 12 00245 g002aCells 12 00245 g002b
Figure 3. Protein–protein interaction network of genes significantly enriched for associations with AD, PD and MDD. The reported network, including both direct (physical) and indirect (functional) associations, was based on the STRING v11.5 database [22]. Only high-confidence interactions between proteins are reported (interaction score > 0.7), while disconnected nodes in the network are hidden. Each node represents all the proteins produced by a single protein-coding gene locus, while edges represent protein–protein associations. Line color indicates the type of interaction evidence: light blue— from curated databases; purple—experimentally determined; green—gene neighborhood; red—gene fusions; blue—gene co-occurrence; yellow—text mining; black—co-expression; and violet—protein homology.
Figure 3. Protein–protein interaction network of genes significantly enriched for associations with AD, PD and MDD. The reported network, including both direct (physical) and indirect (functional) associations, was based on the STRING v11.5 database [22]. Only high-confidence interactions between proteins are reported (interaction score > 0.7), while disconnected nodes in the network are hidden. Each node represents all the proteins produced by a single protein-coding gene locus, while edges represent protein–protein associations. Line color indicates the type of interaction evidence: light blue— from curated databases; purple—experimentally determined; green—gene neighborhood; red—gene fusions; blue—gene co-occurrence; yellow—text mining; black—co-expression; and violet—protein homology.
Cells 12 00245 g003
Table 1. Novel genes associated with neuropsychiatric/neurodegenerative disorders based on single variant associations.
Table 1. Novel genes associated with neuropsychiatric/neurodegenerative disorders based on single variant associations.
CHRPosREFALTrsIDpnSNPsGeneDisorder
8144,992,361CTrs78225118.82 × 10−11139EPPK1AD
2243,414,330AGrs30913643.82 × 10−1016TTLL1AD
2243,279,611AGrs48222183.91 × 10−109PACSIN2AD
1916,211,630AGrs595084941.49 × 10−93TPM4AD
1565,170,949CTrs20135556.37 × 10−972PIF1AD
1630,902,353AGrs800956803.62 × 10−889ZNF689AD
799,581,469CTrs117618824.64 × 10−834AZGP1P1AD
4975,238CTrs732118132.99 × 10−1045SLC26A1PD
1155,053,719CTrs14628554.17 × 10−836EFNA3PD
14104,000,183CTrs27561278.20 × 10−11108TRMT61AMDD
1331,733,057AGrs412921515.34 × 10−923HSPH1MDD
Table 2. Novel genes associated with (a) AD, (b) PD and (c) MDD based on gene-based enrichment analysis.
Table 2. Novel genes associated with (a) AD, (b) PD and (c) MDD based on gene-based enrichment analysis.
(a)
SYMBOLCHRSTARTSTOPNSNPSZSTATp
PVRIG799,805,86499,829,113435.57521.24 × 10−8
FAM57B1630,025,74830074299425.11351.58 × 10−7
NDUFS211.61 × 1081.61 × 108435.06892.00 × 10−7
C16orf921630,024,65530,049,057274.85556.01 × 10−7
KLC31945,826,69245,864,778464.69011.37 × 10−6
B4GALT311.61 × 1081.61 × 108184.65141.65 × 10−6
ZNF6881630,570,66730,594,05554.64521.70 × 10−6
DEDD11.61 × 1081.61 × 108224.62931.83 × 10−6
(b)
SYMBOLCHRSTARTSTOPNSNPSZSTATp
DPM311.55 × 1081.55 × 108177.38047.89 × 10−14
SLC26A14962,861997,228606.34911.08 × 10−10
FBXL191630,924,37630,970,104266.27871.71 × 10−10
SMIM15560,443,53660,468,301386.14593.98 × 10−10
ERCC8560,159,65860,250,9001335.7275.11 × 10−9
FAM200B415,673,28515,717,188665.22958.50 × 10−8
CTF11630,897,92830,924,881135.21239.32 × 10−8
ADAM1511.55 × 1081.55 × 108245.12361.50 × 10−7
PRSS81631,132,75631,157,083175.07041.99 × 10−7
NCOR11715,922,47116,131,4992065.02382.53 × 10−7
VKORC11631,092,16331,117,301104.90644.64 × 10−7
ZNF6681631,062,16431,095,641194.80857.61 × 10−7
PRSS361631,140,24631,171,415194.77149.15 × 10−7
ZNF6681631,062,81331,083,451144.73051.12 × 10−6
SRCAP1630,699,53030,765,602184.65671.61 × 10−6
(c)
SYMBOLCHRSTARTSTOPNSNPSZSTATp
ZNF165628,038,75328,067,341457.3141.30 × 10−13
BTN2A2626,373,32426,405,102886.20642.71 × 10−10
OR2W1629,001,99029023,01755.95451.30 × 10−9
OR12D3629,331,20029,353,06865.93921.43 × 10−9
TRMT61A141.04 × 1081.04 × 108445.93121.50 × 10−9
OR2J1629,058,38629,079,65855.63468.77 × 10−9
HMGN4626,528,63326,556,482305.58891.14 × 10−8
OR2B3629,043,98529,065,09045.22668.63 × 10−8
BTN3A3626,430,70026,463,643544.98933.03 × 10−7
ZNF197344,616,38044,699,963704.85845.92 × 10−7
ZNF35344,680,21944,712,283184.83616.62 × 10−7
TRIM27628,860,77928,901,766114.78268.65 × 10−7
OR2B6627,915,01927,935,960144.72881.13 × 10−6
DLST1475,338,59475,380,448624.69851.31 × 10−6
RHOBTB11062,619,19662,771,1982384.68241.42 × 10−6
ZNF660344,609,71544,651,186464.67311.48 × 10−6
RBM4B1166,422,46966,455,392194.67171.49 × 10−6
ITGB621.61 × 1081.61 × 1083124.66361.55 × 10−6
RBM14-RBM41166,374,09766,423,940324.58342.29 × 10−6
Legend: CHR—chromosome; NSNPS—number of SNPs in the gene; START/STOP—start/stop position (bp) of the variant in the genome (GRCh37/hg19 coordinates); ZSTAT—enrichment Z-score statistics; and P—p-value.
Table 3. Single variant association overlap of (a) AD and (b) PD with platelet parameters.
Table 3. Single variant association overlap of (a) AD and (b) PD with platelet parameters.
(a)
SNPs Overlap betweenSNPGeneChr:positionFunctionPreviously Associated with ADPreviously Associated with Platelet Parameters
AD and all platelet parametersrs6727023EHD3 2:31475960Upstream
rs62118504EXOC3L2 19:45734751Intronic[23]
rs2143926 22:43185180
rs4822218PACSIN2 22:43279611Intronic
rs2267487PACSIN222:43411389Upstream
AD, MPV and PDWrs4575098B4GALT31:161155392Upstream[23]
rs585021ITGB53:124482869Intronic
rs11620465LRCH113:47250344Intronic
rs3091364 22:43414330
AD, MPV and Pltrs12461065PPP1R3719:45605308Intronic[23]
rs12461144EXOC3L219:45723706Intronic[23]
rs123187 19:45830947 [23]
AD, PDW and Pltrs9357551 6:47606029 [24]Plt [25]
AD and MPVrs10934680KALRN3:124440780Downstream
rs11669338NECTIN219:45382984Downstream[24]
rs138235833 19:45415285
rs620807 19:45706952 [24]
rs4803806 19:45708947
AD and PDWrs858502CASTOR37:99843353Intronic[24]
rs7113976 11:85869737 [24]
rs283810NECTIN219:45388241Downstream[24]
rs1160983TOMM4019:45397229Synonymous variant
rs584007APOC119:45416478Upstream[24]
rs117648021EIF3L22:38274632Intronic
(b)
SNPs Overlap betweenSNPGeneChr:positionFunctionPreviously Associated with PDPreviously Associated with Platelet Parameters
PD, MPV and PDWrs10847839HIP1R12:122838013Intronic
PD and MPVrs17689966CRHR117:45833089Intronic
rs9899833MAPT17:45915577Intronic
No overlaps were found between SNPs associated with MDD and those associated with platelet parameters. Legend: AD—Alzheimer’s disease; PD—Parkinson’s disease; Plt—platelet count; MPV—mean platelet volume; and PDW—platelet distribution width.
Table 4. Gene enrichment overlap of (a) AD, (b) PD and (c) MDD with platelet parameters.
Table 4. Gene enrichment overlap of (a) AD, (b) PD and (c) MDD with platelet parameters.
(a)
Genes Overlap betweenGenePreviously Associated with ADPreviously Associated with Platelet Parameters
AD and all platelet parametersAC005779.2
AC006126.3
CD2AP[26]MPV [26] and Plt [26]
CKM[16]
EHD3 MPV [27], PDW [19] and Plt [27]
EXOC3L2[24]
KLC3
L47234.1
MARK4[24]MPV [25], PDW [27] and Plt [25]
PVR[24]MPV [27]
AD, MPV and PDWADAMTS4[24]
AL590714.1
APOA2
B4GALT3
DEDD[24]
NDUFS2
TOMM40L[28]
AD, MPV and PltGEMIN7[24]
AD, PDW and PltGATS[29]
AZGP1[30]
PILRA[24]
AD and MPVBLOC1S3[24]MPV [25], PDW [27] and Plt [27]
PPP1R37[24]
PVRL2[31]
AD and PDWAPOC1[23]PDW [19]
APOE[24]PDW [19] and Plt [27]
GPC2[32]
PVRIG
SLC24A4[24]PDW [19]
STAG3[33]
TOMM40[24]
AD and PltIGSF23[24]
PICALM[24]
(b)
Genes Overlap betweenGenePreviously Associated with PDPreviously Associated with Platelet Parameters
PD and MPVWNT3[34]
AC008498.1
SPPL2C[34]
ARHGAP27[34]
CRHR1[34]
MAPT[34]Plt [25]
ELOVL7[34]MPV [19]
ERCC8
SMIM15
PLEKHM1[34]
KANSL1[34]
NSF[34]
NDUFAF2[34]
STH[35]
PD and PDWSRCAP MPV [19]
(c)
Genes Overlap betweenGenePreviously Associated with ADPreviously Associated with Platelet Parameters
MDD and MPVOR2J3[36]
TRIM27
OR5V1[37]
C11orf31
OR2B3
NRD1
OR12D3
HIST1H2BK
OR2J1
OR2W1
HIST1H4K
HIST1H2AK
MDD and PDWMARK3[37]PDW [38]
TRMT61A
MDD, PDW and PltHIST1H3B
Legend: AD—Alzheimer’s disease; PD—Parkinson’s disease; MDD—major depressive disorder; Plt—platelet count; MPV—mean platelet volume; and PDW—platelet distribution width.
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

Tirozzi, A.; Quiccione, M.S.; Cerletti, C.; Donati, M.B.; de Gaetano, G.; Iacoviello, L.; Gialluisi, A. A Multi-Trait Association Analysis of Brain Disorders and Platelet Traits Identifies Novel Susceptibility Loci for Major Depression, Alzheimer’s and Parkinson’s Disease. Cells 2023, 12, 245. https://doi.org/10.3390/cells12020245

AMA Style

Tirozzi A, Quiccione MS, Cerletti C, Donati MB, de Gaetano G, Iacoviello L, Gialluisi A. A Multi-Trait Association Analysis of Brain Disorders and Platelet Traits Identifies Novel Susceptibility Loci for Major Depression, Alzheimer’s and Parkinson’s Disease. Cells. 2023; 12(2):245. https://doi.org/10.3390/cells12020245

Chicago/Turabian Style

Tirozzi, Alfonsina, Miriam Shasa Quiccione, Chiara Cerletti, Maria Benedetta Donati, Giovanni de Gaetano, Licia Iacoviello, and Alessandro Gialluisi. 2023. "A Multi-Trait Association Analysis of Brain Disorders and Platelet Traits Identifies Novel Susceptibility Loci for Major Depression, Alzheimer’s and Parkinson’s Disease" Cells 12, no. 2: 245. https://doi.org/10.3390/cells12020245

APA Style

Tirozzi, A., Quiccione, M. S., Cerletti, C., Donati, M. B., de Gaetano, G., Iacoviello, L., & Gialluisi, A. (2023). A Multi-Trait Association Analysis of Brain Disorders and Platelet Traits Identifies Novel Susceptibility Loci for Major Depression, Alzheimer’s and Parkinson’s Disease. Cells, 12(2), 245. https://doi.org/10.3390/cells12020245

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