Abstract

The serotonin (5-hydroxytryptamine, 5-HT) transporter (5-HTT) gene-linked polymorphic region (5-HTTLPR) is thought to alter 5-HT signaling and contribute to behavioral and cognitive phenotypes in depression as well as Alzheimer disease (AD). We explored how well the short (S) and long (L) alleles of the 5-HTTLPR align with serotoninergic indices in 60 autopsied cortical samples from early-onset AD/EOAD and late-onset AD/LOAD donors, and age- and sex-matched controls. Stratifying data by either diagnosis-by-genotype or by sex-by-genotype revealed that the donor's 5-HTTLPR genotype, i.e., L/L, S/L, or S/S, did not affect 5-HTT mRNA or protein expression. However, the glycosylation of 5-HTT was significantly higher in control female (vs. male) samples and tended to decrease in female EOAD/LOAD samples, but remained unaltered in male LOAD samples. Glycosylated forms of the vesicular monoamine transporter (VMAT2) were lower in both male and female AD samples, while a sex-by-genotype stratification revealed a loss of VMAT2 glycosylation specifically in females with an L/L genotype. VMAT2 and 5-HTT glycosylation were correlated in male samples and inversely correlated in female samples in both stratification models. The S/S genotype aligned with lower levels of 5-HT turnover in females (but not males) and with an increased glycosylation of the post-synaptic 5-HT2C receptor. Interestingly, the changes in presynaptic glycosylation were evident primarily in female carriers of the APOE ε4 risk factor for AD. Our data do not support an association between 5-HTTLPR genotype and 5-HTT expression, but they do reveal a non-canonical association of 5-HTTLPR genotype with sex-dependent glycosylation changes in pre- and post-synaptic markers of serotoninergic neurons. These patterns of change suggest adaptive responses in 5-HT signaling and could certainly be contributing to the female prevalence in risk for either depression or AD.

Highlights

  • We examined our samples for 5-HTT mRNA transcript levels based on primer-pairs spanning the acceptor-donor splice sites for Ex3–4, Ex8–9, and Ex12–13. 5-HT transporter (5HTT) mRNA transcript levels [Ex3–4 (P = 0.7020); Ex8–9 (P = 0.1631); Ex12–13 (P = 0.0706)] were not significantly different between controls and a diagnosis of early-onset AD (EOAD) or late-onset/sporadic AD (LOAD). 5-HTT mRNA transcript levels [Ex3–4 (P = 0.7513); Ex8–9 (P = 0.2390); Ex12–13 (P = 0.5263)] were not significantly different between S/S, S/L, and L/L genotypes

  • Levels of the 80 kDa glycosylated form of 5-HTT were diminished in carriers of the ε4 allele in pooled data [i.e., male + female: P = 0.0404] and this was reflected by a tendency for a loss of glycosylated 5-HTT in female carriers of the ε4 allele [P = 0.0625], but not male carriers [P = 0.3947] (Figure 13)

  • There was no influence of ε4 status on the glycosylated form of VMAT2 using pooled data [P = 0.3678], but a significant loss of glycosylated VMAT2 was observed in female carriers of the ε4 allele [P = 0.0196], but not male carriers of the allele [P = 0.6029]

Read more

Summary

INTRODUCTION

The degeneration of serotoninergic cell bodies in the dorsal raphé nucleus and noradrenergic cell bodies in the locus coeruleus, and their respective ascending projections (Marcyniuk et al, 1986; Zweig et al, 1988; Rub et al, 2000; Parvizi et al, 2001; Grudzien et al, 2007) are acknowledged as critical events in the earliest stages of Alzheimer disease (AD) and likely predispose to a range of physiological and neuropsychiatric sequelae in presymptomatic AD-dementia that potentially are maintained throughout later stages of the disease process. 5-HTTLPR Genotype and Glycosylation loss of glycosylation of VMAT2 (a marker of monoaminergic neurons) concurrent with a relative increase in the glycosylated 5-HT2C receptor (i.e., active, membrane-associated) support the complexity of glycosylation events observed in AD (FrenkelPinter et al, 2017) Some of these glycosylation changes in our sample set are sex-dependent and could be reflecting adaptive responses designed (Heiming and Schaser, 2010) to overcome signaling deficits during the course of the disease

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call