Next Article in Journal
Extracellular Polymeric Substances Produced by Actinomycetes of the Genus Rhodococcus for Biomedical and Environmental Applications
Previous Article in Journal
Gyrase and Topoisomerase IV as Antibacterial Targets for Gepotidacin and Zoliflodacin: Teaching Old Enzymes New Tricks
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Fingolimod Effects on Motor Function and BDNF-TrkB Signaling in a Huntington’s Mouse Model Are Disease-Stage-Dependent

Cone Laboratory, Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2026, 27(1), 494; https://doi.org/10.3390/ijms27010494
Submission received: 13 January 2025 / Revised: 8 December 2025 / Accepted: 24 December 2025 / Published: 3 January 2026
(This article belongs to the Section Biochemistry)

Abstract

Huntington’s Disease (HD) is characterized by prominent degeneration of the principal neurons of the striatum and by progressive motor and cognitive deterioration. Striatal neurons degenerate in HD due to multiple cell-autonomous and non-autonomous factors. Impaired neurotrophin signaling by brain-derived neurotrophic factor (BDNF) and its cognate receptor Tropomyosin receptor kinase B (TrkB) is an important mechanism underlying neuronal loss in HD. Fingolimod, a clinically approved oral drug for Multiple Sclerosis, was originally developed based on its anti-inflammatory properties. Recent work suggests that fingolimod can also promote BDNF expression and enhance neurotrophic support in the brain. We hypothesized that fingolimod treatment initiated during the presymptomatic phase would increase striatal BDNF levels and protect against motor dysfunction in HD. In wild-type mice, fingolimod treatment increases striatal BDNF levels and enhances BDNF-TrkB signaling. However, chronic fingolimod therapy (0.1 mg/kg, i.p., twice per week, over 7 weeks) initiated at age 4 weeks in the R6/2 mouse model of HD failed to improve behavioral locomotor deficits and exacerbated limb clasping. Furthermore, fingolimod treatment in these presymptomatic R6/2 mice acutely decreased BDNF-TrkB signaling in the striatum in a dose-dependent manner. In contrast, acute administration of fingolimod in symptomatic 7-week-old R6/2 mice increased striatal BDNF-TrkB signaling in a dose-dependent manner, consistent with previous work suggesting that chronic fingolimod can improve motor behavior when given during the symptomatic phase. Thus, the effects of fingolimod striatal BDNF-TrkB signaling and motor behavior in HD are complex and vary with disease stage. Addressing this variability is critical for the design of neuroprotective drug trials in HD, including those utilizing sphingosine-1-phosphate receptor (S1P) modulators.

1. Introduction

The striatum, a central nucleus within the basal ganglia, plays a pivotal role in processing programs that shape motor, cognitive, and emotional behaviors [1,2,3]. GABAergic medium spiny projection neurons (MSPs) constitute the majority (>90%) of striatal neurons, with a smaller population of interneurons. Striatal MSPs express TrkB (Tropomyosin receptor kinase B), the receptor for brain-derived neurotrophic factor (BDNF). MSPs rely on the BDNF-TrkB neurotrophic signaling pathway to promote developmental survival [4,5], maintain MSP phenotype [6,7,8], modulate neurotransmitter activity [9,10], and protect against toxic stressors [8,11,12]. For example, BDNF-TrkB signaling is essential for maintaining the expression of DARPP-32, an MSP-enriched protein that is a central regulator of neurotransmitter signaling in MSPs [13,14,15].
Early dysfunction and prominent degeneration of striatal MSPs are key factors contributing to the hallmark hyperkinetic motor symptoms of Huntington’s disease (HD) [16,17,18]. HD is an autosomal dominant genetic disorder caused by polymorphic expansions of CAG trinucleotide repeats in the HTT gene, which produces mutant huntingtin protein with an elongated N-terminal poly-glutamine domain [19]. The expression of mutant huntingtin protein leads to the disruption of various cellular processes that increase the vulnerability of striatal neurons to degeneration and drive pathological progression [20,21]. Reduced afferent BDNF supply [22,23,24,25] and impaired TrkB signaling [26,27] are implicated in the degeneration of striatal MSPs in HD. Therefore, enhancing BDNF-TrkB signaling may be a promising therapeutic strategy for mitigating striatal degeneration and improving motor and cognitive symptoms in HD [28,29].
Fingolimod, a clinically approved oral drug for Multiple Sclerosis [30], is primarily known for its ability to reduce pathogenic immune responses [31]. However, recent work suggests that it may also offer neuroprotective effects by enhancing neurotrophic support in various animal models of neurodegenerative diseases [32,33,34,35,36]. Fingolimod is lipophilic and readily crosses the blood–brain barrier to exert direct effects on cells in the central nervous system (CNS) [37,38,39]. It is converted to phospho-fingolimod (p-fingolimod), an analogue of sphingosine-1-phosphate (S1P), a naturally occurring signaling molecule that activates various S1P receptors (S1P1-5 subtypes). P-fingolimod and S1P can bind S1P receptors that modulate glutamate release and excitatory activity in neurons [40,41,42,43]. Additionally, p-fingolimod can act as a functional antagonist by inducing S1P receptor internalization [44,45], potentially attenuating endogenous S1P-mediated effects on neuronal activation [46]. Given its complex pharmacodynamics and potential therapeutic role in HD, it is crucial to evaluate fingolimod’s effects, particularly during the prodromal phase prior to neuronal degeneration, before considering broader clinical applications [47,48]. Notably, fingolimod can promote activity-induced expression of BDNF, enhancing neurotrophic support for neurons [32,49]. Moreover, in a mouse model of Rett’s syndrome, early therapy with fingolimod attenuates motor symptoms in association with enhanced BDNF-mediated striatal neuroprotection [32]. We therefore investigated the effectiveness of initiating fingolimod therapy at a presymptomatic stage in a mouse model of HD.
We employed the R6/2 transgenic mouse model of HD, which expresses the N-terminal fragment of mutant huntingtin with approximately 120 glutamine repeats. By 9 weeks of age, this model displays significant motor dysfunction, with progressive decrease in spontaneous locomotion, impaired coordination on a Rotarod test, and limb-clasping dystonia. These motor deficits coincide with the progressive degeneration of striatal MSPs [50,51,52]. The R6/2 model has been extensively used to investigate neurotrophic mechanisms contributing to striatal neuron vulnerability in HD, including impaired BDNF-TrkB signaling [27,53,54,55]. We hypothesized that early intervention with chronic fingolimod therapy could protect R6/2 mice from motor decline by increasing BDNF levels and enhancing TrkB signaling in the striatum. However, our results demonstrate that chronic fingolimod treatment, when initiated in young presymptomatic R6/2 mice, does not improve progressive decline in motor function at later stages. Moreover, we observed a decrease in striatal BDNF-TrkB signaling in these presymptomatic R6/2 mice after a single dose of fingolimod. In contrast, acute fingolimod administration in older symptomatic mice led to a significant increase in striatal BDNF-TrkB signaling. These results indicate that the effects of S1P receptor ligands such as fingolimod depend on the neurodegenerative state. Therapeutic effects may therefore vary depending on whether the drug is administered during presymptomatic or later phases of HD.

2. Results

2.1. Effects of Chronic Fingolimod on Motor Behavior in WT and R6/2 Mice

Motor phenotypes in R6/2 mice are noted mainly after age 7 weeks, with progressive worsening of locomotor activity, incoordination, dystonic phenotype, and associated reduction in weight gain [52,56,57]. Cohorts of WT mice and R6/2 littermates were divided into two treatment groups receiving either saline or fingolimod (0.1 mg/kg, i.p., every 3.5 days- q3.5d) starting at 4 weeks of age, prior to onset of R6/2 motor symptoms. Motor behavior was assessed at regular intervals up to 11 weeks. This dosing regimen was based on previous work in a mouse model of Rett’s syndrome, where fingolimod (0.1 mg/kg i.p.) administration every 4 days for 2–4 weeks improved motor behavior and increased striatal and cortical BDNF levels [32].
To determine the effect of fingolimod on weight gain, WT and R6/2 mice in saline or drug treatment groups were weighed twice per week from 4 to 11 weeks of age (Figure 1a,b). Both WT and R6/2 mice exhibited age-related changes in weight, with comparatively less weight gain in the HD model. Fingolimod treatment mildly impaired weight gain over time in WT mice but did not affect weight in R6/2 mice. WT mice receiving fingolimod showed reduced weight gain compared to saline controls after 8.5 weeks of age. In contrast, both saline- and fingolimod-treated R6/2 mice displayed similar weight gain patterns, peaking at 8.5 weeks before declining. Specifically, statistical analysis (two-factor mixed ANOVA) of weight data for WT mice (Figure 1a) indicates significant effects of Age (F13,182 = 228.726; p < 0.001) and interaction of Age x Treatment (F13,182 = 1.788; p < 0.050) but no effect of Treatment (F1,14 = 0.033; p = 0.858), suggesting that fingolimod-treated WT mice exhibit a different age-related weight gain compared to saline-treated WT mice. Post hoc (Bonferroni) comparisons within the WT saline group (n = 8) show higher week-over-week weights up to 10.5 weeks (e.g., 9.5 vs. 10.5 weeks: 23.1 ± 1.0 g vs. 24.2 ± 1.0 g; p < 0.050). Post hoc comparisons within the WT fingolimod group (n = 8) show higher week-over-week weights only up to 8.5 weeks (e.g., 7.5 vs. 8.5 weeks: 21.8 ± 1.0 g vs. 22.6 ± 1.0 g; p < 0.050). In R6/2 mice (Figure 1b), statistical analysis of weights indicates a significant effect of Age (F13,273 = 75.472; p < 0.001) but no effect of Treatment (F1,21 = 0.820; p = 0.376). Post hoc comparisons within the R6/2 saline group (n = 11) show higher week-over-week weights up to 7 weeks (e.g., 6 vs. 7 weeks: 21.2 ± 0.9 g vs. 22.0 ± 0.8 g; p < 0.001) with a peak at 8.5 weeks (22.5 ± 0.9 g), followed by a lack of weight gain in older mice (e.g., 10.5 weeks: 21.5 ± 0.8 g; p = 0.964 vs. 8.5 weeks). Post hoc comparisons within the R6/2 fingolimod group (n = 12) show weights are higher week-over-week up to age 6.5 weeks (e.g., 5.5 vs. 6.5 weeks: 21.8 ± 0.9 g vs. 22.9 ± 0.8 g; p < 0.050) and then peak at 8.5 weeks (23.7 ± 0.9 g) and become significantly lower at older ages (e.g., 10.5 weeks: 21.8 ± 0.8 g; p < 0.050 vs. 8.5 weeks).
Open field assessments at 9 and 11 weeks revealed no significant effect of fingolimod on spontaneous locomotor activity in either WT or R6/2 mice (Figure 1c,d). While R6/2 mice exhibited a significant age-related decline in locomotor activity (F1,16 = 43.612; p < 0.001), the decline was similar in both saline and fingolimod treatment groups, indicating that fingolimod does not alter the natural progression of locomotor deficits in this HD model.
To determine the effects of fingolimod on motor coordination and balance, WT and R6/2 mice were assessed at age 10 weeks on an Accelerating Rotarod apparatus (Figure 1e,f). WT mice performed better with fingolimod treatment (181 ± 11 s, n = 7; p < 0.050) compared to saline treatment (143 ± 7 s, n = 8). R6/2 mice performed similarly with fingolimod treatment (154 ± 14 s, n = 12) compared to saline treatment (136 ± 25 s, n = 11).
To determine if fingolimod treatment alters dystonic behavior in R6/2 mice, limb clasping during tail suspension was assessed in saline- and fingolimod-treated groups at age 6, 8, and 10 weeks (Figure 1g). ANOVA of clasping scores shows significant effects of Age (F2,42 = 44.082; p < 0.001) and Treatment (F1,21 = 6.028; p < 0.050). Post hoc (Bonferroni) comparisons within the saline group show higher clasping scores at 8 weeks (1.42 ± 0.12, n = 11; p < 0.050) and 10 weeks (1.82 ± 0.10, n = 11; p < 0.001) compared to 6 weeks (0.96 ± 0.18, n = 11). Post hoc comparisons within the fingolimod group show higher clasping scores at 8 weeks (1.90 ± 0.12, n = 12; p < 0.001) and 10 weeks (2.21 ± 0.09, n = 12; p < 0.001) compared to 6 weeks (1.21 ± 0.17, n = 12). Post hoc comparisons between treatment groups show lower clasping scores in the saline vs. fingolimod group (8 weeks: 1.42 + 0.12 vs. 1.90 + 0.12; p < 0.050; 10 weeks: 1.82 ± 0.10 vs. 2.21 ± 0.09; p < 0.050). Thus, chronic fingolimod treatment when initiated in presymptomatic R6/2 mice exacerbates limb clasping dystonia.

2.2. Effects of Chronic Fingolimod on BDNF-TrkB Signaling in Forebrain Motor Regions of R6/2 Mice

Given that increased striatal BDNF-TrkB signaling has been linked to improvements in motor and learning deficits in various neurodegenerative disease models [32,33,58], we investigated whether this protein pathway is altered in striatal tissue samples from the above cohort of R6/2 mice at the end of behavioral testing, following chronic fingolimod treatment (0.1 mg/kg, i.p., q3.5d) from 4 to 11 weeks of age. Motor cortex samples were also analyzed for comparison.
Striatal protein extracts from individual fingolimod- or saline-treated 11-week-old R6/2 mice were run in parallel on Western Blots (Figure 2). Optical density analysis of immunoblot images of BDNF-TrkB pathway proteins [7,27,59,60] was used to calculate relative protein values standardized to the saline-treated R6/2 group. The results show striatal BDNF levels are significantly higher in fingolimod-treated R6/2 mice (relative optical density—rel. OD = 1.49 ± 0.10, n = 9) compared to saline controls (rel. OD = 1.00 ± 0.12, n = 9; p < 0.050) (Figure 2a,b). Total levels of the downstream pathway proteins, full-length TrkB receptors, and DARPP-32 are similar between the two treatment groups (Figure 2a,b). Levels of activated phospho-TrkB receptors are marginally higher in fingolimod-treated R6/2 mice (fingolimod: rel. OD = 1.20 ± 0.09, n = 9; saline: OD = 1.00 ± 0.05, n = 9; p = 0.071). Additional analyses of the ratio of activated phospho-TrkB relative to total TrkB receptors in the striatum show that TrkB signaling activation in chronic fingolimod-treated R6/2 mice (1.03 ± 0.03, n = 9) is similar to saline controls (0.98 ± 0.05, n = 9) (Figure 2e).
BDNF levels are also higher in the motor cortex of fingolimod-treated 11-week-old R6/2 mice (rel. OD = 1.36 ± 0.08, n = 9) compared to saline controls (rel. OD = 1.01 ± 0.06, n = 9; p < 0.050) (Figure 2c,d). Cortical levels of activated phospho-TrkB and total full-length TrkB receptors are not altered by chronic fingolimod treatment (Figure 2c,d). The ratio of activated phospho-TrkB–total TrkB in the cortex of chronic fingolimod-treated R6/2 mice (fingolimod: 1.05 ± 0.06, n = 9) is similar to saline controls (0.99 ± 0.05, n = 9) (Figure 2f).
In summary, chronic treatment with fingolimod (0.1 mg/kg) initiated in 4-week-old R6/2 mice augments BDNF levels in the cortex and striatum by 11 weeks but does not enhance downstream TrkB signaling or DARPP-32 levels in the striatum.

2.3. Acute Effects of Fingolimod on BDNF-TrkB Signaling in Presymptomatic and Symptomatic R6/2 Mice

Previous work indicates that initiating chronic fingolimod therapy in symptomatic R6/2 mice (i.e., from 7- or 10-week-old) improves some motor deficits and correlates with increases in striatal BDNF protein levels [35]. In contrast, we find that initiating chronic fingolimod therapy in presymptomatic 4-week-old R6/2 mice does not improve spontaneous locomotion in an open field and worsens limb clasping symptoms. We therefore determined if fingolimod has distinct acute effects on striatal BDNF-TrkB signaling in presymptomatic and symptomatic R6/2 mice. Striatal tissue was extracted 48 h after a single injection of fingolimod at the 0.1 mg/kg dose used for chronic therapy. BDNF-TrkB signaling protein levels were compared between fingolimod- vs. saline-treated WT and R6/2 mice (Figure 3).
BDNF levels are higher in the striatum of 4-week-old WT mice 48 h after a single dose of 0.1 mg/kg fingolimod (rel. OD = 1.26 ± 0.12, n = 10) compared to saline controls (rel. OD = 0.99 ± 0.08, n = 12) (Figure 3a,b), but the difference is only marginally significant (p = 0.067). However, striatal levels of downstream activated phospho-TrkB receptors are significantly higher in the fingolimod group (rel. OD = 1.54 ± 0.21, n = 10; p < 0.050) compared to saline controls (rel. OD = 1.00 ± 0.07, n = 12). Levels of total full-length TrkB receptors are also significantly higher in the fingolimod group (rel. OD = 1.36 ± 0.15, n = 10; p < 0.050) compared to saline controls (rel. OD = 1.00 ± 0.02, n = 12). Striatal levels of DARPP-32 remain unchanged with 0.1 mg/kg fingolimod treatment in 4-week-old WT mice (Figure 3a,b). The ratio of activated phospho-TrkB–total TrkB in the striatum is similar between fingolimod- compared to saline-treated 4-week-old WT mice (fingolimod: 1.03 ± 0.05, n = 10; saline: 0.93 ± 0.07, n = 12) (Figure 3e).
In contrast to WT mice, striatal BDNF levels are significantly lower in 4-week-old R6/2 mice after acute treatment with 0.1 mg/kg fingolimod (rel. OD = 0.72 ± 0.05, n = 8; p < 0.050) compared to saline controls (rel. OD = 0.98 ± 0.06, n = 7) (Figure 3c,d). Striatal phospho-TrkB levels are also lower in the fingolimod group (rel. OD = 0.77 ± 0.10, n = 8) compared to saline controls (rel. OD = 0.98 ± 0.06, n = 7), but the difference was not significant (p = 0.108). Striatal levels of total full-length TrkB and DARPP-32 remain unchanged with 0.1 mg/kg fingolimod treatment in 4-week-old R6/2 mice (Figure 3c,d). The ratio of activated phospho-TrkB–total TrkB in the striatum is significantly lower in fingolimod-treated 4-week-old R6/2 mice (fingolimod: 0.88 ± 0.09, n = 8; saline: 1.10 ± 0.04, n = 7; p < 0.050) (Figure 3f).
To determine if the acute effects of 0.1 mg/kg fingolimod are distinct at later stages of HD, levels of BDNF and downstream signaling proteins were examined in 7-week-old R6/2 mice, a timepoint when overt motor symptoms are apparent in this HD model [52,61]. Striatal levels of BDNF and downstream proteins are similar between fingolimod-treated and saline-treated WT mice at 7 weeks old (Figure 4a,b). The ratio of phospho-TrkB–total TrkB in the striatum is similar between fingolimod- and saline-treated WT mice (Figure 4e). Moreover, 7-week-old R6/2 mice show no differences in striatal levels of BDNF and downstream proteins after acute fingolimod treatment compared to saline treatment (Figure 4c,d). The ratio of phospho-TrkB–total TrkB is similar in fingolimod- and saline-treated R6/2 mice (Figure 4f). Thus, in contrast to 4-week-old R6/2 mice that show a reduction in striatal BDNF levels following acute treatment with 0.1 mg/kg fingolimod, 7-week-old R6/2 mice show no change in striatal BDNF levels with this dose of fingolimod.

2.4. Fingolimod Dose Effect on BDNF-TrkB Signaling Proteins in Striatum of WT and R6/2 Mice

Since acute administration of 0.1 mg/kg fingolimod (above) revealed different trends in the BDNF-TrkB signaling pathway at early and later disease stages, further dose escalation was tested on additional cohorts of WT and R6/2 mice at age 4 or 7 weeks. Treatment groups received a single injection of either saline, 1.0 or 3.0 mg/kg of fingolimod, then 48 h later striatal tissue samples were extracted and assayed by Western blot. Motor cortex samples were also assayed, as this region is a major source of afferent BDNF for the striatum.
In 4-week-old WT mice (Figure 5a,b), there is a significant main effect of fingolimod treatment on striatal levels of BDNF (F2,12 = 5.145; p < 0.050) and phospho-TrkB (F2,12 = 4.216; p < 0.050). Post hoc (LSD) comparisons show striatal BDNF levels are significantly higher in the 1.0 mg/kg dose group (rel. OD = 1.40 ± 0.09, n = 5; p < 0.050) compared to saline controls (rel. OD = 0.99 ± 0.08, n = 5) (Figure 5b). There is no further increase in BDNF levels in the 3.0 mg/kg group (rel. OD = 1.26 ± 0.14, n = 3; p = 0.354) compared to the 1.0 mg/kg group. Striatal phospho-TrkB levels are not significantly higher in the 1.0 mg/kg group (rel. OD = 1.10 ± 0.02, n = 5; p = 0.159) compared to saline controls (rel. OD = 1.00 ± 0.07, n = 5), but they are significantly higher in the 3.0 mg/kg group (rel. OD = 1.23 ± 0.06, n = 3; p < 0.050). Striatal levels of DARPP-32 and total full-length TrkB are unchanged by these higher doses of fingolimod. There is a significant main effect of fingolimod (F2,12 = 4.793; p < 0.050) on the ratio of activated phospho-TrkB–total TrkB in the striatum of 4-week-old WT mice (Figure 5e). This ratio is significantly higher than the 3.0 mg/kg group (1.40 ± 0.04, n = 3) compared to saline controls (0.98 ± 0.03, n = 5; p < 0.050) and marginally higher compared to the 1.0 mg/kg group (1.05 ± 0.02, n = 5; p = 0.080) (Figure 5e). Thus, in 4-week-old WT mice, acute administration of fingolimod increases striatal levels of BDNF and downstream activated phospho-TrkB receptors.
In cortical tissue from 4-week-old WT mice (Supplementary Figure S1a,b), there is a significant main effect of fingolimod (F2,16 = 3.766; p < 0.050) on BDNF levels. Post hoc comparisons show that cortical BDNF levels in the 3.0 mg/kg dose group (rel. OD = 3.31 ± 1.00, n = 6) are significantly higher compared to saline controls (rel. OD = 0.90 ± 0.22, n = 6; p < 0.050) and marginally higher compared to the 1.0 mg/kg dose group (rel. OD = 1.42 ± 1.00, n = 5; p = 0.070) (Supplementary Figure S1b).
In 4-week-old R6/2 mice (Figure 5c,d), there is a significant main effect of fingolimod on striatal levels of BDNF (F2,8 = 22.595; p < 0.050), phospho-TrkB (F2,8 = 6.979; p < 0.050), total full-length TrkB (F2,8 = 8.427; p < 0.050), and DARPP-32 (F2,8 = 10.378; p < 0.050). Post hoc (LSD) comparisons show that striatal BDNF levels are significantly lower in both 1.0 mg/kg (rel. OD = 0.75 ± 0.01, n = 3; p < 0.050) and 3.0 mg/kg (rel. OD = 0.62 ± 0.03, n = 3; p < 0.001) dose groups compared to saline controls (rel. OD = 1.00 ± 0.06; n = 3) (Figure 5d). phospho-TrkB levels are similar between saline controls (rel. OD = 1.00 ± 0.03, n = 3) and the 1.0 mg/kg group (rel. OD = 0.95 ± 0.03, n = 3), but they are significantly lower in the 3.0 mg/kg group (rel. OD = 0.84 ± 0.03, n = 3; p < 0.050). Full-length TrkB levels are significantly lower in the 3.0 mg/kg group (rel. OD = 0.76 ± 0.04, n = 3; p < 0.050) compared to saline controls (rel. OD = 1.00 ± 0.05, n = 3). DARPP-32 levels are similar between saline controls (rel. OD = 1.00 ± 0.08, n = 3) and the 1.0 mg/kg group (rel. OD = 1.21 ± 0.04, n = 3), but they are significantly lower in the 3.0 mg/kg group (rel. OD = 0.77 ± 0.08, n = 3; p < 0.050) compared to the 1.0 mg/kg group. There is a significant main effect of fingolimod (F2,6 = 8.263; p < 0.050) on the ratio of activated phospho-TrkB–total TrkB in the striatum of 4-week-old R6/2 mice (Figure 5f). This ratio is significantly higher in the 3.0 mg/kg dose group (1.08 ± 0.06, n = 3) compared to saline controls (0.95 ± 0.03, n = 3; p < 0.050). Thus, in contrast to WT mice, administration of fingolimod in 4-week-old R6/2 mice decreases striatal levels of BDNF and its receptor TrkB, ultimately diminishing downstream phospho-TrkB and DARPP-32 levels in the striatum.
In cortical tissue from 4-week-old R6/2 mice (Supplementary Figure S1c,d), there is a significant main effect of fingolimod treatment on BDNF levels (F2,13 = 8.272; p < 0.050). Post hoc comparisons show that cortical BDNF levels are significantly lower in the 3.0 mg/kg dose group (rel. OD = 0.62 ± 0.12, n = 5; p < 0.050) compared to the 1.0 mg/kg group (0.95 ± 0.06, n = 4; p < 0.050) and compared to saline controls (rel. OD = 1.03 ± 0.03, n = 5; p < 0.050) (Supplementary Figure S1d).
In 7-week-old WT mice, there is a significant main effect of fingolimod treatment on striatal levels of BDNF (F2,30 = 4.074; p < 0.050), phospho-TrkB (F2,35 = 4.575; p < 0.050), DARPP-32 (F2,35 = 7.962; p < 0.050), and full-length TrkB (F2,35 = 8.680; p < 0.001) (Figure 6a,b). Post hoc (LSD) comparisons show that striatal BDNF levels are significantly higher in both 1.0 mg/kg (rel. OD = 1.30 ± 0.11, n = 9; p < 0.050) and 3.0 mg/kg (rel. OD = 1.35 ± 0.12, n = 11; p < 0.050) groups compared to saline controls (rel. OD = 1.00 ± 0.05, n = 11) (Figure 6b). Phospho-TrkB levels are significantly higher in both 1.0 mg/kg (rel. OD = 1.37 ± 0.12, n = 12; p < 0.050) and 3.0 mg/kg (rel. OD = 1.39 ± 0.12, n = 12; p < 0.050) groups compared to saline controls (rel. OD = 0.98 ± 0.08, n = 12). DARPP-32 levels are significantly higher in both 1.0 mg/kg (rel. OD = 1.30 ± 0.07, n = 12; p < 0.050) and 3.0 mg/kg (rel. OD = 1.26 ± 0.04, n = 12; p < 0.050) groups compared to saline (rel. OD = 1.01 ± 0.04, n = 12). Full-length TrkB levels are significantly higher in both 1.0 mg/kg (rel. OD = 1.25 ± 0.06, n = 12; p < 0.050) and 3.0 mg/kg (rel. OD = 1.30 ± 0.06, n = 12; p < 0.050) groups compared to saline controls (rel. OD = 0.99 ± 0.05, n = 12). There is no main effect of fingolimod (F2,35 = 0.798) on the ratio of phospho-TrkB–total TrkB in the striatum of 7-week-old WT mice. Phospho-TrkB–total TrkB ratios are similar between treatment groups (Figure 6e). Thus, in 7-week-old WT mice, acute fingolimod treatment increases striatal levels of BDNF and its receptor TrkB and also increases downstream activated phospho-TrkB and DARPP-32 levels.
In cortical tissue of 7-week-old WT mice (Supplementary Figure S2a,b), there is a marginally significant main effect of fingolimod (F2,17 = 2.944; p = 0.083) on BDNF levels. Post hoc comparisons show that cortical BDNF levels are significantly higher in the 3.0 mg/kg group (rel. OD = 1.83 ± 0.27, n = 5; p < 0.050) compared to saline controls (rel. OD = 0.99 ± 0.11, n = 6) (Supplementary Figure S2b).
In 7-week-old R6/2 mice, there is a significant main effect of fingolimod treatment on striatal levels of BDNF (F2,20 = 13.537; p < 0.001) (Figure 6c,d). Post hoc (LSD) comparisons show that striatal BDNF levels are significantly higher in the 3.0 mg/kg dose group (rel. OD = 1.41 ± 0.08, n = 7; p < 0.001) compared to saline controls (rel. OD = 1.00 ± 0.04, n = 7). The main effect of fingolimod treatment on striatal levels of phospho-TrkB (F2,20 = 2.413; p = 0.118) and DARPP-32 (F2,20 = 2.418; p = 0.117) did not reach statistical significance in 7-week-old R6/2 mice. However, post hoc comparisons show that phospho-TrkB levels are significantly higher in the 3.0 mg/kg group (rel. OD = 1.45 ± 0.20, n = 7; p < 0.050) compared to saline controls (rel. OD = 1.00 ± 0.10, n = 7), while DARPP-32 levels are marginally higher in the 3.0 mg/kg group (rel. OD = 1.18 ± 0.05, n = 7; p = 0.062) compared to saline controls (rel. OD = 1.01 ± 0.06, n = 7). There is a marginally significant main effect of fingolimod (F2,20 = 3.227; p = 0.063) on the ratio of phospho-TrkB–total TrkB in the striatum of 7-week-old R6/2 mice. This ratio is significantly higher in the 3.0 mg/kg group (1.10 ± 0.08, n = 7; p < 0.050) compared to saline controls (0.86 ± 0.06, n = 7) (Figure 6f). Thus, similarly to WT mice, acute administration of fingolimod in 7-week-old R6/2 mice increases striatal levels of BDNF and downstream activated phospho-TrkB receptors.
In cortical tissue of 7-week-old R6/2 mice (Supplementary Figure S2c,d), the main effect of fingolimod (F2,11 = 1.312) on BDNF levels is not statistically significant. Cortical BDNF levels tend to be higher in both 1.0 mg/kg (rel. OD = 1.18 ± 0.20, n = 5; p = 0.580) and the 3.0 mg/kg (rel. OD = 1.51 ± 0.28, n = 5; p = 0.143) dose groups compared to saline controls (rel. OD = 1.00 ± 0.15, n = 4), but the differences are not statistically significant in 7-week-old R6/2 mice (Supplementary Figure S2d).

3. Discussion

In HD patients, mutant huntingtin protein is expressed from fetal stages; however, clinical locomotor symptoms and pathological neuronal degeneration manifest mainly in adulthood [17,29,62]. This suggests a significant prodromal period when neuroprotective strategies may be applied. A variety of cell autonomous and non-autonomous mechanisms have been proposed for the prominent striatal neuron degeneration in HD [63,64], including excitotoxicity [62,65,66] and loss of afferent neurotrophic support [67,68,69]. We determined whether fingolimod, a drug known to modulate neurotrophin levels in the CNS, can augment BDNF levels and exert a protective effect in the R6/2 mouse model of HD, particularly when treatment is initiated at presymptomatic stages prior to the onset of motor symptoms and striatal neuronal degeneration.
Fingolimod, an analogue of the endogenous lipid signaling molecule sphingosine, is a drug with pleiotropic effects that modulates sphingosine-1-phosphate (S1P) receptor signaling in diverse cell populations [48,70,71,72]. It is used for therapy in Multiple Sclerosis, where its main mechanism of action is attributed to S1P receptor-mediated suppression of inflammatory activity in the periphery and in the CNS [37]. It also acts on neuronal S1P receptors, which can modulate glutamate release and excitatory activity [40,73,74,75,76], ultimately regulating activity-induced BDNF expression [49,77]. Impaired BDNF-TrkB neurotrophic signaling is an important mechanism underlying striatal degeneration in the R6/2 mouse model of HD [27,52,53,55]. We investigated whether fingolimod treatment in R6/2 mice, initiated at early stages before the onset of striatal neurodegeneration and motor symptoms, could upregulate BDNF expression and protect against progressive locomotor dysfunction. Our results indicate that initiating chronic fingolimod treatment in presymptomatic R6/2 mice fails to rescue them from deteriorating motor function. Moreover, exposing presymptomatic 4-week-old R6/2 mice to a single dose of fingolimod decreases cortical and striatal BDNF levels, attenuates downstream TrkB signaling, and reduces DARPP-32 expression, a phenotypic marker of mature striatal neurons [6,27,78]. In contrast, acute administration of fingolimod in symptomatic 7-week-old R6/2 mice increases cortical and striatal BDNF levels, an effect also seen in WT mice. Therefore, fingolimod’s effects on forebrain neurotrophin levels depend on disease state in this animal model of HD.
Previous work in different models of neurodegenerative disease suggests that fingolimod can improve motor and behavioral symptoms [72,79]. For example, the MeCP2 mutant mouse model of Rett’s syndrome shows patterns of progressive motor dysfunction similar to R6/2 mice [52], including progressive deterioration of spontaneous locomotion and motor coordination, dystonic limb clasping, and striatal degeneration over a 12-week lifespan [80]. Chronic treatment with fingolimod (0.1 mg/kg, i.p., every 4 days over 4 weeks) improves motor symptoms in MeCP2 mutant mice and increases forebrain BDNF levels [32]. Using a similar dosing regimen initiated in R6/2 mice during the presymptomatic phase (0.1 mg/kg, i.p., every 3.5 days from age 4 to 11 weeks), we show a lack of therapeutic effect of fingolimod on the severity of major motor symptoms, including spontaneous locomotor activity and motor coordination, and no effect on weight loss. Moreover, chronic treatment with fingolimod initiated in young presymptomatic R6/2 mice worsens limb clasping, a dystonic motor symptom that is reliably noted as R6/2 mice progress through their 13- to 15-week lifespan [52,61,81]. These results suggest that young R6/2 mice have distinct responses to fingolimod compared to MeCP2 mutant mice. MeCP2 mutant mice show pathologically decreased cortical neuron excitability from a young age [82,83], and enhanced excitatory activity by fingolimod provides a salutary effect [32]. In contrast, although young presymptomatic R6/2 mice exhibit relatively normal cortical neuron activity, aberrant receptor properties in cortical and striatal neurons render them vulnerable to adverse excitatory signaling [66,84,85,86,87]. This vulnerability may be exacerbated by fingolimod’s enhancement of neuronal glutamate release and excitatory activation. At symptomatic R6/2 stages, striatal neurons exhibit deficits in afferent glutamatergic signaling [86,88,89,90]. Therefore, at this later stage, fingolimod may exert therapeutic effects via enhanced excitatory transmission and neurotrophin signaling. Indeed, a previous study using R6/2 mice demonstrated that chronic fingolimod treatment initiated at symptomatic stages improves motor symptoms, elevates striatal BDNF levels, and promotes neurotrophic support for striatal neurons [35]. While the present study also finds that BDNF levels are elevated after chronic fingolimod treatment from presymptomatic through to symptomatic stages, initiation of treatment at presymptomatic stages ultimately fails to improve R6/2 motor symptoms.
Deficiencies in BDNF supply [22,53] or impaired TrkB receptor-mediated signaling [26,27] are important mechanisms that contribute to progressive striatal neuron degeneration and declining motor function in HD [23,25,91]. Increasing BDNF supply to the striatum may therefore be neuroprotective in HD. Previous studies show fingolimod can promote excitatory activity-associated gene transcription, including the expression of BDNF [32,76,92]. In wild-type cortical cultures, fingolimod can induce BDNF mRNA and protein expression in a manner dependent on excitatory activity via glutamate receptors [32,49]. Moreover, acute administration of fingolimod in WT mice promotes Erk1/2-CREB signaling and elevates cortical BDNF expression [32]. CREB signaling is a well-established pathway that underlies glutamatergic activity-induced BDNF expression [93,94]. The present results further demonstrate that fingolimod treatment increases cerebral cortical BDNF protein levels and promotes BDNF-TrkB signaling at the striatal target in both 4 and 7-week-old WT mice. In contrast, similar treatment in presymptomatic 4-week-old R6/2 mice decreases cortical BDNF levels, attenuates striatal BDNF-TrkB signaling, and decreases striatal DARPP-32 levels. We propose that this early impairment of striatal neurotrophic support occurs with chronic fingolimod in presymptomatic R6/2 mice, ultimately contributing to striatal pathology and exacerbating locomotor decline. Indeed, we found that initiating chronic fingolimod therapy in presymptomatic R6/2 mice fails to improve motor deficits and even worsens limb-clasping motor behavior.
Previous work suggests that acute administration of fingolimod (0.1 mg/kg) augments BDNF levels in both the cortex and striatum of WT mice when assayed by ELISA [32], a method that does not distinguish between different protein forms of BDNF, including pre-proBDNF (32 kDa), proBDNF (28 kDa), and mature BDNF (14 kDa) [95]. Other ELISA-based studies report no effect of sub-chronic or chronic fingolimod treatment on cortical or striatal BDNF levels in WT mice [33,58,96,97]. Using Western blot, we demonstrate that fingolimod treatment in 4- and 7-week-old WT mice mediates a positive drug-effect that elevates striatal levels of mature BDNF and increases downstream activated phospho-TrkB receptors. In contrast, in presymptomatic 4-week-old R6/2 mice, fingolimod (0.1 to 3.0 mg/kg) significantly decreases striatal BDNF levels. This reduction is accompanied by a parallel decline in downstream activated phospho-TrkB receptors and striatal DARPP-32 protein levels. Conversely, in 7-week-old symptomatic R6/2 mice, fingolimod increases striatal BDNF levels. This finding is consistent with prior reports of neurotrophic elevation following chronic treatment in symptomatic R6/2 mice [35] and other neurodegenerative models [32,33,58].
Accumulating evidence indicates that fingolimod enhances neuronal glutamate signaling and BDNF expression in several neurodegenerative models via S1P receptor-dependent mechanisms [32,33,58,76,92,98]. However, S1P receptor-independent pathways—such as epigenetic modulation via HDACs [99,100]—may also drive neurotrophic signaling and contribute to our observations. Despite the critical role of S1P receptors in neuronal development [101], their specific contribution to cellular dysfunction in HD remains unclear. While S1P receptor expression appears normal in symptomatic HD mice [102,103,104], our data suggest that S1P receptor function warrants further investigation across all stages of disease progression.
Altered forebrain excitability may explain the differential responses to fingolimod across the R6/2 lifespan. Studies in R6/2 and other HD models indicate that prodromal changes, specifically a decreased ratio of synaptic to extrasynaptic NMDA receptors (NMDARs) [87,105,106] and impaired synaptic glutamate clearance [84,107,108], could increase neuronal vulnerability to excitotoxicity and disrupt neurotrophic signaling [106,109]. While synaptic NMDARs couple glutamate signaling to the Erk1/2-CREB pathway to drive BDNF expression [93], extrasynaptic NMDARs antagonize this pathway [110,111,112,113,114]. Consequently, in the cerebral cortex of presymptomatic R6/2 mice, fingolimod-induced glutamate release may cause spillover, preferentially activating extrasynaptic NMDARs and suppressing cortical and striatal BDNF levels. Other prodromal deficits, including reduced synaptic NMDAR and AMPAR currents and increased GABAergic inhibition [106,115], may further uncouple fingolimod-induced glutamate release from BDNF production [32,40,41,58]. Importantly, these cortical abnormalities reverse during the symptomatic phase, where synaptic NMDAR and AMPAR currents increase, and GABAergic inhibition decreases [106,115,116]. This shift likely facilitates the coupling of fingolimod-induced glutamate release to BDNF expression, allowing for salutary effects when the drug is initiated during the symptomatic phase.
Beyond its direct effects on neurons, fingolimod may modulate forebrain neurotrophins by altering inflammatory pathways. Pro-inflammatory cytokines (e.g., TNFα, IL-1β, IL-6) are activated in R6/2 mice in a complex, stage-dependent manner, paralleling the progression of inflammatory pathology in human HD [117,118,119,120,121]. Presymptomatic HD patients and animal models of HD, including R6/2 mice, are characterized by comparatively low levels of pro-inflammatory cytokines and activated inflammatory cell populations [120,121,122]. In contrast, the symptomatic phase is marked by heightened peripheral inflammation, elevated CNS cytokine levels, and widespread astrocyte and microglial activation [120,121,122,123]. Consequently, fingolimod’s impact may depend on the prevailing inflammatory milieu. In pro-inflammatory states, fingolimod can dampen CNS inflammation by inhibiting NF-κB signaling and glial activation [124], thereby enhancing BDNF expression [38,103]. However, in the quiescent presymptomatic state, fingolimod may promote gliosis and cytokine production, leading to BDNF downregulation [125,126,127]. This effect may be amplified by the progressive decline of S1P, the endogenous ligand for S1P receptors [128,129]. The relative absence of endogenous S1P reduces competition for the receptor, potentially enhancing fingolimod’s agonist activity and its modulation of neuronal excitability and inflammation. Finally, given that HD alters liver metabolism [56,130] and blood–brain barrier permeability [131,132], fingolimod bioavailability likely shifts as the disease advances. Therefore, a comparative analysis of fingolimod pharmacokinetics and pharmacodynamics across presymptomatic and symptomatic R6/2 stages represents a critical avenue for future research.

4. Materials and Methods

4.1. Animals

Animal procedures were in accordance with the Canadian Council on Animal Care guidelines for ethical use and welfare of animals in research, as administered by the McGill University Animal Care Committee. Female mice with ovarian transplants from R6/2 mice (C57Bl6/J background; Jackson Laboratory, Bar Harbor, MA, USA) carrying a mutant HTT exon-1 transgene with 120 ± 5 CAG repeats, were mated with wild-type (WT) males from the same genetic background to obtain WT and R6/2 offspring. Only F1 generation offspring were used to mitigate generational instability of CAG-repeat length, which could prolong symptom onset in R6/2 strains with >160 CAG-repeats [61,122]. Tail-tip samples from newborn pups were tested for the R6/2 transgene, and CAG repeat size (125 ± 2.18 repeats) was confirmed by PCR genotyping (Laragen Inc, Culver City, CA, USA). WT and R6/2 littermates of both genders were used for chronic treatment experiments assessing behavioral performance and forebrain protein levels and for acute treatment experiments assessing forebrain protein levels.

4.2. Drug Treatment

A cohort of WT and R6/2 littermates was divided into two chronic treatment groups receiving either fingolimod (0.1 mg/kg, i.p., Cayman Chemicals, Ann Arbor, MI, USA, #10006292; WT-Fing, n = 8; R6/2-Fing, n = 12) or saline (WT-sal, n = 8; R6/2-sal, n = 11) every 3.5 days (q3.5d) from postnatal day (P) 28 to 73 (10.5 weeks). Mice were placed in an open field apparatus at ages 9 and 11 to evaluate spontaneous locomotion. Limb clasping was evaluated at 6, 8, and 10 weeks. Rotorod performance was evaluated at 9 weeks. Finally, animals were sacrificed at 11 weeks for Western blot assays of forebrain protein levels.
A second cohort of 4-week-old WT and R6/2 littermates was divided into two acute treatment groups receiving a single injection (i.p.) of either saline (WT-sal, n = 12; R6/2-sal, n = 7) or fingolimod (0.1 mg/kg; WT-Fing 0.1 mg/kg, n = 10; R6/2-Fing 0.1 mg/kg, n = 8) and then sacrificed 48 h later for Western blot assays. A third cohort of 4-week-old WT and R6/2 littermates was divided into three acute treatment groups receiving a single injection of either saline (WT-sal, n = 5; R6/2-sal, n = 3) or fingolimod at 1.0 mg/kg (WT-Fing 1 mg/kg, n = 5; R6/2-Fing 1 mg/kg, n = 3) or 3.0 mg/kg (WT-Fing 3 mg/kg, n = 3; R6/2-Fing 3 mg/kg, n = 3) and then sacrificed 48 h later for Western blot assays.
A fourth cohort of 7-week-old WT and R6/2 littermates received a single injection of either saline (WT-sal, n = 12; R6/2-sal, n = 7) or fingolimod (0.1 mg/kg; WT-Fing 0.1 mg/kg, n = 10; R6/2-Fing 0.1 mg/kg, n = 8), and then, they were sacrificed 48 h later for Western blot assays. Finally, a fifth cohort of 7-week-old WT and R6/2 littermates was divided into three acute treatment groups receiving a single injection of either saline (WT-sal, n = 12; R6/2-sal, n = 7) or fingolimod at 1.0 mg/kg (WT-Fing 1 mg/kg, n = 12; R6/2-Fing 1 mg/kg, n = 7) or 3.0 mg/kg (WT-Fing 3 mg/kg, n = 12; R6/2-Fing 3 mg/kg, n = 7), and then, they were sacrificed 48 h later for Western blot assays.

4.3. Tissue Preparation and Western Blot Assays

Mice were sacrificed by guillotine decapitation, and whole brains were dissected, cooled in isopentane (−80 °C), and stored in a −80 °C freezer. Next, whole brains were mounted on a freezing microtome with Tissue Tek gel (Sakura Finetek, Torrance, CA, USA). Tissue samples were obtained from the primary motor cortex and the dorsolateral motor striatum using a 1.0 × 1.5 mm cylindrical micro-punch (Stoelting, Wood Dale, IL, USA). Tissue samples were lysed in 60 µL of RIPA buffer (containing protease and phosphatase inhibitors) and then centrifuged to obtain soluble protein fractions that were individually stored at −80 °C.
Tissue samples from either WT or R6/2 mice were processed on different Western Blots. Equal volumes of tissue lysates from individual mice were loaded in parallel lanes on 4–15% gradient gels, along with a lane of molecular weight protein markers (Kaleidoscope™ Prestained Protein Standards, BioRad, Hercules, CA, USA). Proteins were separated through SDS-PAGE and then transferred to Nitrosecellulose membranes. Protein blots were cut into horizontal sections delimited by molecular weight markers and blocked with 2.5% BSA-TBST or 5% milk-TBST. Blot sections were separately immuno-labeled with antibodies to detect mature BDNF at 14 kDa (#sc546, 1:500; Santa Cruz, Santa Cruz, CA, USA), phospho-TrkB at 145 kDa (#sc135645, 1:500; Santa Cruz), phospho-Erk1/2 at 42–43 kDa (#9106, 1:2000; Cell Signaling Tech., Danvers, MA, USA), DARPP-32 at 32 kDa (#ab1656, 1:2000; Millipore, Burlington, MA, USA), total full-length TrkB at 145 kDa (#5374, 1:2000; Millipore, USA), total Erk1/2 at 42–43 kDa (#9102, 1:2000; Cell Signaling Tech., USA), and β-III-Tubulin at 50 kDa (#PRB435, 1:10,000; Covance, Princeton, NJ, USA). Immuno-labeled blots were incubated in secondary HRP-conjugated antibodies (goat anti-rabbit or -mouse; 1:10,000; Millipore) and then developed using either Super Signal West Femto (Pierce, Appleton, WI, USA) or Clarity (Bio-Rad, USA; for full-length TrkB and Erk1/2) chemiluminescence substrate and digitally imaged using a high-resolution (3-megapixel) and high-sensitivity (16-bit) camera system (Chemocam Imager, Intas Science Imaging, Göttingen, Germany).
Labimage 1D software (version L360, Kapelan Bio-Imaging Solutions, Leipzig, Germany) was used to quantify the optical density (OD) of specific protein bands. The OD value of each protein band was standardized to that of β-III-Tubulin (a neuronal-specific protein) in the same sample lane, thereby controlling for total neuronal protein loading for each individual tissue sample. The final relative OD value for each protein band was calculated relative to the average OD value of the saline-treated samples in each blot [133]. Each blot always included all treatment groups in order to calculate the relative OD value of each sample. Samples were run multiple times in each mouse to obtain at least 3 blots, and relative OD values were averaged for individual animals. Subsequently, individual values were averaged within each treatment group.

4.4. Evaluation of Motor Behavior

WT and R6/2 mice were weighed prior to receiving chronic drug treatments every 3.5 days from age P28 to P74. Locomotor activity was assessed between ages P63 and P77 by testing spontaneous ambulatory movement in an open field apparatus with infrared backlighting [134]. Spontaneous movement was recorded during a one-hour period using the Videotrack system equipped with motion analysis software (VideoTrack 3.1, Viewpoint Life Sciences, Montreal, Canada), and the total distance traveled was calculated.
Limb dystonia was assessed at ages P42, P56, and P70 by testing limb clasping during 20 s tail suspensions repeated three times with a 30 min rest in between each trial. Limb movements were videotaped and later scored by an observer blinded to genotype. Clasping movements were observed as a retraction of a limb toward the body. Clasping score was based on an index of clasps towards midline for each limb: none = 0, no clasping; mild = 0.25, limb retraction towards midline but contraction not sustained (<5 s); moderate = 0.5, high-amplitude limb retraction to or beyond the midline that was not sustained or partial limb retraction that was sustained (>5 consecutive seconds); severe = 0.75, high-amplitude limb retraction beyond the midline that was sustained (>5 s). Scores for each limb are added for a maximum total limb clasping score of 3. The highest clasping score from the 3 sessions was used for analysis [81].
Motor coordination was assessed at age P67 by testing performance on a Rotorod apparatus (Columbus Instruments, Columbus, OH, USA) during an accelerating rotation protocol [135,136]. Rotorod baseline training occurred over 3 consecutive days with four 5 min trials on each day. The maximum speed at the end of the training period was 24 r.p.m. Testing on day 4 consisted of five trials. At each trial, the speed was gradually increased from 5 to 44 r.p.m. over 5 min, with a 1-hour rest period between trials. The latency to fall at each trial was recorded, and the mean of the 3 best performances was used for analysis.

4.5. Statistical Analysis

Motor behavior scores and weight for the fingolimod and saline groups were compared by two-way ANOVA for main effects and interaction of age and treatment. Post hoc comparisons were carried out to identify simple effects between age or treatment groups, as detailed in the results. Non-parametric statistics were used to analyze the results of clasping behavior. Protein levels are reported as mean relative optical density (rel. OD ± SEM) of groups based on genotype and drug treatment (saline, 0.1, 1.0, and 3.0 mg/kg). Relative protein levels were compared by one-way ANOVA with appropriate post hoc tests to identify differences between treatment groups.

5. Conclusions

Fingolimod, an approved immunomodulator for relapsing-remitting Multiple Sclerosis, exhibits complex and stage-dependent effects on neurotrophin pathways and motor behavior in Huntington’s disease mouse models. When initiated in the presymptomatic phase, fingolimod decreases BDNF levels and TrkB signaling in the forebrain and fails to improve motor behavior. However, during the symptomatic phase, acute fingolimod administration enhances striatal BDNF levels and TrkB receptor signaling, consistent with previous findings indicating its potential to improve motor behavior when administered during this later stage. Further research is warranted to elucidate the mechanisms underlying these stage-dependent effects, particularly the influence of the evolving excitability and inflammatory environments as Huntington’s disease progresses. These findings emphasize the importance of tailoring treatment approaches in HD and other neurodegenerative diseases, recognizing the dynamic nature of disease progression and the complex interactions between immunomodulatory drugs and neurotrophin signaling pathways.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms27010494/s1.

Author Contributions

Conceptualization, A.F.S. and K.Q.N.; methodology, K.Q.N. and V.V.R.; writing—original draft preparation, K.Q.N.; writing—review and editing, K.Q.N., A.F.S. and V.V.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by grants from the Canadian Institutes of Health Research (CIHR), the National Science and Engineering Research Council (NSERC), Healthy Brains for Healthy Lives (HBHL) and Canadian Foundation for Innovation (CFI) to AFS. KQN was supported by the Fonds de Recherche du Québec-Santé (FRSQ; #21667).

Institutional Review Board Statement

The animal study protocol was approved by the Centre for Neurological Disease Models of the Montreal Neurological Institute (AUP: MNI-4532, 25 April 2022).

Informed Consent Statement

Not applicable.

Data Availability Statement

The raw data supporting the conclusions of this article will be made available by the authors on request.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

BDNFBrain-derived neurotrophic factor
NMDAN-methyl-D-aspartate
AMPAα-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
HDHuntington’s disease

References

  1. Albin, R.L.; Young, A.B.; Penney, J.B. The functional anatomy of basal ganglia disorders. Trends Neurosci. 1989, 12, 366–375. [Google Scholar] [CrossRef]
  2. Parent, A.; Hazrati, L.-N. Functional anatomy of the basal ganglia. I. The cortico-basal ganglia-thalamo-cortical loop. Brain Res. Rev. 1995, 20, 91–127. [Google Scholar] [CrossRef]
  3. Bolam, J.P.; Hanley, J.J.; Booth, P.A.; Bevan, M.D. Synaptic organisation of the basal ganglia. J. Anat. 2000, 196, 527–542. [Google Scholar] [CrossRef] [PubMed]
  4. Baydyuk, M.; Russell, T.; Liao, G.-Y.; Zang, K.; An, J.J.; Reichardt, L.F.; Xu, B. TrkB receptor controls striatal formation by regulating the number of newborn striatal neurons. Proc. Natl. Acad. Sci. USA 2011, 108, 1669–1674. [Google Scholar] [CrossRef] [PubMed]
  5. Baydyuk, M.; Xie, Y.; Tessarollo, L.; Xu, B. Midbrain-derived neurotrophins support survival of immature striatal projection neurons. J. Neurosci. 2013, 33, 3363–3369. [Google Scholar] [CrossRef] [PubMed]
  6. Ivkovic, S.; Polonskaia, O.; Fariñas, I.; Ehrlich, M.E. Brain-derived neurotrophic factor regulates maturation of the DARPP-32 phenotype in striatal medium spiny neurons: Studies in vivo and in vitro. Neuroscience 1997, 79, 509–516. [Google Scholar] [CrossRef]
  7. Ivkovic, S.; Ehrlich, M.E. Expression of the Striatal DARPP-32/ARPP-21 Phenotype in GABAergic Neurons Requires Neurotrophins In Vivo and In Vitro. J. Neurosci. 1999, 19, 5409–5419. [Google Scholar] [CrossRef] [PubMed]
  8. Nakao, N.; Brundin, P.; Funa, K.; Lindvall, O.; Odin, P. Trophic and protective actions of brain-derived neurotrophic factor on striatal DARPP-32-containing neurons in vitro. Dev. Brain Res. 1995, 90, 92–101. [Google Scholar] [CrossRef]
  9. Cheng, P.-L.; Song, A.-H.; Wong, Y.-H.; Wang, S.; Zhang, X.; Poo, M.-M. Self-amplifying autocrine actions of BDNF in axon development. Proc. Natl. Acad. Sci. USA 2011, 108, 18430–18435. [Google Scholar] [CrossRef]
  10. Park, H.; Poo, M.-M. Neurotrophin regulation of neural circuit development and function. Nat. Rev. Neurosci. 2013, 14, 7–23. [Google Scholar] [CrossRef]
  11. Gomes, J.R.; Costa, J.T.; Melo, C.V.; Felizzi, F.; Monteiro, P.; Pinto, M.J.; Inácio, A.R.; Wieloch, T.; Almeida, R.D.; Grãos, M.; et al. Excitotoxicity Downregulates TrkB.FL Signaling and Upregulates the Neuroprotective Truncated TrkB Receptors in Cultured Hippocampal and Striatal Neurons. J. Neurosci. 2012, 32, 4610. [Google Scholar] [CrossRef]
  12. Bemelmans, A.P.; Horellou, P.; Pradier, L.; Brunet, I.; Colin, P.; Mallet, J. Brain-derived neurotrophic factor-mediated protection of striatal neurons in an excitotoxic rat model of Huntington’s disease, as demonstrated by adenoviral gene transfer. Hum. Gene Ther. 1999, 10, 2987–2997. [Google Scholar] [CrossRef]
  13. Bibb, J.A.; Yan, Z.; Svenningsson, P.; Snyder, G.L.; Pieribone, V.A.; Horiuchi, A.; Nairn, A.C.; Messer, A.; Greengard, P. Severe deficiencies in dopamine signaling in presymptomatic Huntington’s disease mice. Proc. Natl. Acad. Sci. USA 2000, 97, 6809–6814. [Google Scholar] [CrossRef]
  14. Greengard, P.; Allen, P.B.; Nairn, A.C. Beyond the dopamine receptor: The DARPP-32/protein phosphatase-1 cascade. Neuron 1999, 23, 435–447. [Google Scholar] [CrossRef]
  15. Svenningsson, P.; Nishi, A.; Fisone, G.; Girault, J.A.; Nairn, A.C.; Greengard, P. DARPP-32: An integrator of neurotransmission. Annu. Rev. Pharmacol. Toxicol. 2004, 44, 269–296. [Google Scholar] [CrossRef]
  16. Biglan, K.M.; Ross, C.A.; Langbehn, D.R.; Aylward, E.H.; Stout, J.C.; Queller, S.; Carlozzi, N.E.; Duff, K.; Beglinger, L.J.; Paulsen, J.S. Motor abnormalities in premanifest persons with Huntington’s disease: The PREDICT-HD study. Mov. Disord. 2009, 24, 1763–1772. [Google Scholar] [CrossRef]
  17. Paulsen, J.; Long, J.; Johnson, H.; Aylward, E.; Ross, C.; Williams, J.; Nance, M.; Erwin, C.; Westervelt, H.; Harrington, D.; et al. Clinical and biomarker changes in premanifest Huntington disease show trial feasibility: A decade of the PREDICT-HD study. Front. Aging Neurosci. 2014, 6, 78. [Google Scholar] [CrossRef]
  18. Rüb, U.; Seidel, K.; Heinsen, H.; Vonsattel, J.P.; den Dunnen, W.F.; Korf, H.W. Huntington’s disease (HD): The neuropathology of a multisystem neurodegenerative disorder of the human brain. Brain Pathol. 2016, 26, 726–740. [Google Scholar] [CrossRef] [PubMed]
  19. The Huntington’s Disease Collaborative Research Group; MacDonald, M.E.; Ambrose, C.M.; Duyao, M.P.; Myers, R.H.; Lin, C.; Srinidhi, L.; Barnes, G.; Taylor, S.A.; James, M.; et al. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell 1993, 72, 971–983. [Google Scholar] [CrossRef] [PubMed]
  20. Harjes, P.; Wanker, E.E. The hunt for huntingtin function: Interaction partners tell many different stories. Trends Biochem. Sci. 2003, 28, 425–433. [Google Scholar] [CrossRef] [PubMed]
  21. Borrell-Pages, M.; Zala, D.; Humbert, S.; Saudou, F. Huntington’s disease: From huntingtin function and dysfunction to therapeutic strategies. Cell. Mol. Life Sci. 2006, 63, 2642–2660. [Google Scholar] [CrossRef]
  22. Gauthier, L.R.; Charrin, B.C.; Borrell-Pagès, M.; Dompierre, J.P.; Rangone, H.; Cordelières, F.P.; De Mey, J.; MacDonald, M.E.; Leßmann, V.; Humbert, S.; et al. Huntingtin Controls Neurotrophic Support and Survival of Neurons by Enhancing BDNF Vesicular Transport along Microtubules. Cell 2004, 118, 127–138. [Google Scholar] [CrossRef]
  23. Zuccato, C.; Cattaneo, E. Role of brain-derived neurotrophic factor in Huntington’s disease. Prog. Neurobiol. 2007, 81, 294–330. [Google Scholar] [CrossRef]
  24. Yu, C.; Li, C.H.; Chen, S.; Yoo, H.; Qin, X.; Park, H. Decreased BDNF Release in Cortical Neurons of a Knock-in Mouse Model of Huntington’s Disease. Sci. Rep. 2018, 8, 16976. [Google Scholar] [CrossRef]
  25. Zhou, Z.; Zhong, S.; Zhang, R.; Kang, K.; Zhang, X.; Xu, Y.; Zhao, C.; Zhao, M. Functional analysis of brain derived neurotrophic factor (BDNF) in Huntington’s disease. Aging 2021, 13, 6103–6114. [Google Scholar] [CrossRef] [PubMed]
  26. Liot, G.; Zala, D.; Pla, P.; Mottet, G.; Piel, M.; Saudou, F. Mutant Huntingtin alters retrograde transport of TrkB receptors in striatal dendrites. J. Neurosci. 2013, 33, 6298–6309. [Google Scholar] [CrossRef]
  27. Nguyen, K.Q.; Rymar, V.V.; Sadikot, A.F. Impaired TrkB signaling underlies reduced BDNF-mediated trophic support of striatal neurons in the R6/2 mouse model of Huntington’s Disease. Front. Cell. Neurosci. 2016, 10, 37. [Google Scholar] [CrossRef] [PubMed]
  28. Tabrizi, S.J.; Estevez-Fraga, C.; van Roon-Mom, W.M.C.; Flower, M.D.; Scahill, R.I.; Wild, E.J.; Muñoz-Sanjuan, I.; Sampaio, C.; Rosser, A.E.; Leavitt, B.R. Potential disease-modifying therapies for Huntington’s disease: Lessons learned and future opportunities. Lancet Neurol. 2022, 21, 645–658. [Google Scholar] [CrossRef]
  29. Bates, G.P.; Dorsey, R.; Gusella, J.F.; Hayden, M.R.; Kay, C.; Leavitt, B.R.; Nance, M.; Ross, C.A.; Scahill, R.I.; Wetzel, R.; et al. Huntington disease. Nat. Rev. Dis. Primers 2015, 1, 15005. [Google Scholar] [CrossRef] [PubMed]
  30. Sharma, S.; Mathur, A.G.; Pradhan, S.; Singh, D.B.; Gupta, S. Fingolimod (FTY720): First approved oral therapy for multiple sclerosis. J. Pharmacol. Pharmacother. 2011, 2, 49–51. [Google Scholar] [CrossRef]
  31. Brinkmann, V.; Billich, A.; Baumruker, T.; Heining, P.; Schmouder, R.; Francis, G.; Aradhye, S.; Burtin, P. Fingolimod (FTY720): Discovery and development of an oral drug to treat multiple sclerosis. Nat. Rev. Drug Discov. 2010, 9, 883–897, Erratum in Nat. Rev. Drug Discov. 2010, 9, 955. [Google Scholar] [CrossRef]
  32. Deogracias, R.; Yazdani, M.; Dekkers, M.P.J.; Guy, J.; Ionescu, M.C.S.; Vogt, K.E.; Barde, Y.-A. Fingolimod, a sphingosine-1 phosphate receptor modulator, increases BDNF levels and improves symptoms of a mouse model of Rett syndrome. Proc. Natl. Acad. Sci. USA 2012, 109, 14230–14235. [Google Scholar] [CrossRef] [PubMed]
  33. Fukumoto, K.; Mizoguchi, H.; Takeuchi, H.; Horiuchi, H.; Kawanokuchi, J.; Jin, S.; Mizuno, T.; Suzumura, A. Fingolimod increases brain-derived neurotrophic factor levels and ameliorates amyloid β-induced memory impairment. Behav. Brain Res. 2014, 268, 88–93. [Google Scholar] [CrossRef] [PubMed]
  34. Vargas-Medrano, J.; Krishnamachari, S.; Villanueva, E.; Godfrey, W.H.; Lou, H.; Chinnasamy, R.; Arterburn, J.B.; Perez, R.G. Novel FTY720-Based Compounds Stimulate Neurotrophin Expression and Phosphatase Activity in Dopaminergic Cells. ACS Med. Chem. Lett. 2014, 5, 782–786. [Google Scholar] [CrossRef]
  35. Di Pardo, A.; Amico, E.; Favellato, M.; Castrataro, R.; Fucile, S.; Squitieri, F.; Maglione, V. FTY720 (fingolimod) is a neuroprotective and disease-modifying agent in cellular and mouse models of Huntington disease. Hum. Mol. Genet. 2014, 23, 2251–2265. [Google Scholar] [CrossRef]
  36. Ren, M.; Han, M.; Wei, X.; Guo, Y.; Shi, H.; Zhang, X.; Perez, R.G.; Lou, H. FTY720 Attenuates 6-OHDA-Associated Dopaminergic Degeneration in Cellular and Mouse Parkinsonian Models. Neurochem. Res. 2017, 42, 686–696. [Google Scholar] [CrossRef]
  37. Groves, A.; Kihara, Y.; Chun, J. Fingolimod: Direct CNS effects of sphingosine 1-phosphate (S1P) receptor modulation and implications in multiple sclerosis therapy. J. Neurol. Sci. 2013, 328, 9–18. [Google Scholar] [CrossRef]
  38. Noda, H.; Takeuchi, H.; Mizuno, T.; Suzumura, A. Fingolimod phosphate promotes the neuroprotective effects of microglia. J. Neuroimmunol. 2013, 256, 13–18. [Google Scholar] [CrossRef]
  39. Hunter, S.F.; Bowen, J.D.; Reder, A.T. The Direct Effects of Fingolimod in the Central Nervous System: Implications for Relapsing Multiple Sclerosis. CNS Drugs 2016, 30, 135–147. [Google Scholar] [CrossRef]
  40. Kajimoto, T.; Okada, T.; Yu, H.; Goparaju, S.K.; Jahangeer, S.; Nakamura, S.-i. Involvement of Sphingosine-1-Phosphate in Glutamate Secretion in Hippocampal Neurons. Mol. Cell. Biol. 2007, 27, 3429–3440. [Google Scholar] [CrossRef] [PubMed]
  41. Kanno, T.; Nishizaki, T.; Proia, R.L.; Kajimoto, T.; Jahangeer, S.; Okada, T.; Nakamura, S. Regulation of synaptic strength by sphingosine 1-phosphate in the hippocampus. Neuroscience 2010, 171, 973–980. [Google Scholar] [CrossRef]
  42. Norman, E.; Cutler, R.G.; Flannery, R.; Wang, Y.; Mattson, M.P. Plasma membrane sphingomyelin hydrolysis increases hippocampal neuron excitability by sphingosine-1-phosphate mediated mechanisms. J. Neurochem. 2010, 114, 430–439. [Google Scholar] [CrossRef] [PubMed]
  43. Darios, F.D.; Jorgacevski, J.; Flašker, A.; Zorec, R.; García-Martinez, V.; Villanueva, J.; Gutiérrez, L.M.; Leese, C.; Bal, M.; Nosyreva, E.; et al. Sphingomimetic multiple sclerosis drug FTY720 activates vesicular synaptobrevin and augments neuroendocrine secretion. Sci. Rep. 2017, 7, 5958. [Google Scholar] [CrossRef]
  44. Oo, M.L.; Thangada, S.; Wu, M.-T.; Liu, C.H.; Macdonald, T.L.; Lynch, K.R.; Lin, C.-Y.; Hla, T. Immunosuppressive and Anti-angiogenic Sphingosine 1-Phosphate Receptor-1 Agonists Induce Ubiquitinylation and Proteasomal Degradation of the Receptor. J. Biol. Chem. 2007, 282, 9082–9089. [Google Scholar] [CrossRef] [PubMed]
  45. Sykes, D.A.; Riddy, D.M.; Stamp, C.; Bradley, M.E.; McGuiness, N.; Sattikar, A.; Guerini, D.; Rodrigues, I.; Glaenzel, A.; Dowling, M.R.; et al. Investigating the molecular mechanisms through which FTY720-P causes persistent S1P1 receptor internalization. Br. J. Pharmacol. 2014, 171, 4797–4807. [Google Scholar] [CrossRef] [PubMed]
  46. Li, C.; Li, J.-N.; Kays, J.; Guerrero, M.; Nicol, G.D. Sphingosine 1-phosphate enhances the excitability of rat sensory neurons through activation of sphingosine 1-phosphate receptors 1 and/or 3. J. Neuroinflamm. 2015, 12, 70. [Google Scholar] [CrossRef]
  47. Wood, H. Could fingolimod provide cognitive benefits in patients with Huntington disease? Nat. Rev. Neurol. 2015, 11, 426. [Google Scholar] [CrossRef]
  48. Bascuñana, P.; Möhle, L.; Brackhan, M.; Pahnke, J. Fingolimod as a Treatment in Neurologic Disorders Beyond Multiple Sclerosis. Drugs RD 2020, 20, 197–207. [Google Scholar] [CrossRef]
  49. Deogracias, R.; Klein, C.; Matsumoto, T.; Yazdani, M.; Bibel, M.; Barde, Y.; Schubart, A. Expression of brain-derived neurotrophic factor is regulated by fingolimod (FTY720) in cultured neurons. Mult. Scler. J. 2008, 14, 29–293. [Google Scholar] [CrossRef]
  50. Mangiarini, L.; Sathasivam, K.; Seller, M.; Cozens, B.; Harper, A.; Hetherington, C.; Lawton, M.; Trottier, Y.; Lehrach, H.; Davies, S.W.; et al. Exon 1 of the HD Gene with an Expanded CAG Repeat Is Sufficient to Cause a Progressive Neurological Phenotype in Transgenic Mice. Cell 1996, 87, 493–506. [Google Scholar] [CrossRef]
  51. Stack, E.C.; Kubilus, J.K.; Smith, K.; Cormier, K.; Signore, S.J.D.; Guelin, E.; Ryu, H.; Hersch, S.M.; Ferrante, R.J. Chronology of behavioral symptoms and neuropathological sequela in R6/2 Huntington’s disease transgenic mice. J. Comp. Neurol. 2005, 490, 354–370. [Google Scholar] [CrossRef]
  52. Samadi, P.; Boutet, A.; Rymar, V.V.; Rawal, K.; Maheux, J.; Kvann, J.C.; Tomaszewski, M.; Beaubien, F.; Cloutier, J.F.; Levesque, D.; et al. Relationship between BDNF expression in major striatal afferents, striatum morphology and motor behavior in the R6/2 mouse model of Huntington’s disease. Genes Brain Behav. 2013, 12, 108–124. [Google Scholar] [CrossRef]
  53. Zuccato, C.; Liber, D.; Ramos, C.; Tarditi, A.; Rigamonti, D.; Tartari, M.; Valenza, M.; Cattaneo, E. Progressive loss of BDNF in a mouse model of Huntington’s disease and rescue by BDNF delivery. Pharmacol. Res. 2005, 52, 133–139. [Google Scholar] [CrossRef]
  54. Ginés, S.; Paoletti, P.; Alberch, J. Impaired TrkB-mediated ERK1/2 Activation in Huntington Disease Knock-in Striatal Cells Involves Reduced p52/p46 Shc Expression. J. Biol. Chem. 2010, 285, 21537–21548. [Google Scholar] [CrossRef]
  55. Simmons, D.A.; Belichenko, N.P.; Yang, T.; Condon, C.; Monbureau, M.; Shamloo, M.; Jing, D.; Massa, S.M.; Longo, F.M. A small molecule TrkB ligand reduces motor impairment and neuropathology in R6/2 and BACHD mouse models of Huntington’s disease. J. Neurosci. 2013, 33, 18712–18727, Erratum in: J. Neurosci. 2014, 34, 2012. [Google Scholar] [CrossRef]
  56. van der Burg, J.M.M.; Bacos, K.; Wood, N.I.; Lindqvist, A.; Wierup, N.; Woodman, B.; Wamsteeker, J.I.; Smith, R.; Deierborg, T.; Kuhar, M.J.; et al. Increased metabolism in the R6/2 mouse model of Huntington’s disease. Neurobiol. Dis. 2008, 29, 41–51. [Google Scholar] [CrossRef] [PubMed]
  57. Cummings, D.M.; Alaghband, Y.; Hickey, M.A.; Joshi, P.R.; Hong, S.C.; Zhu, C.; Ando, T.K.; André, V.M.; Cepeda, C.; Watson, J.B.; et al. A critical window of CAG repeat-length correlates with phenotype severity in the R6/2 mouse model of Huntington’s disease. J. Neurophysiol. 2012, 107, 677–691. [Google Scholar] [CrossRef] [PubMed]
  58. Becker-Krail, D.; Farrand, A.Q.; Boger, H.A.; Lavin, A. Effects of fingolimod administration in a genetic model of cognitive deficits. J. Neurosci. Res. 2016, 95, 1174–1181. [Google Scholar] [CrossRef]
  59. Sandhya, V.K.; Raju, R.; Verma, R.; Advani, J.; Sharma, R.; Radhakrishnan, A.; Nanjappa, V.; Narayana, J.; Somani, B.L.; Mukherjee, K.K.; et al. A network map of BDNF/TRKB and BDNF/p75NTR signaling system. J. Cell Commun. Signal. 2013, 7, 301–307. [Google Scholar] [CrossRef] [PubMed]
  60. Li, Y.; Yui, D.; Luikart, B.W.; McKay, R.M.; Li, Y.; Rubenstein, J.L.; Parada, L.F. Conditional ablation of brain-derived neurotrophic factor-TrkB signaling impairs striatal neuron development. Proc. Natl. Acad. Sci. USA 2012, 109, 15491–15496. [Google Scholar] [CrossRef]
  61. Menalled, L.; El-Khodor, B.F.; Patry, M.; Suárez-Fariñas, M.; Orenstein, S.J.; Zahasky, B.; Leahy, C.; Wheeler, V.; Yang, X.W.; MacDonald, M.; et al. Systematic behavioral evaluation of Huntington’s disease transgenic and knock-in mouse models. Neurobiol. Dis. 2009, 35, 319–336. [Google Scholar] [CrossRef]
  62. Parsons, M.P.; Raymond, L.A. Chapter 17—Huntington disease. In Neurobiology of Brain Disorders, 2nd ed.; Zigmond, M.J., Wiley, C.A., Chesselet, M.-F., Eds.; Academic Press: Cambridge, MA, USA, 2023; pp. 275–292. [Google Scholar]
  63. Ehrlich, M.E. Huntington’s disease and the striatal medium spiny neuron: Cell-autonomous and non-cell-autonomous mechanisms of disease. Neurotherapeutics 2012, 9, 270–284. [Google Scholar] [CrossRef]
  64. Ross, C.A.; Aylward, E.H.; Wild, E.J.; Langbehn, D.R.; Long, J.D.; Warner, J.H.; Scahill, R.I.; Leavitt, B.R.; Stout, J.C.; Paulsen, J.S.; et al. Huntington disease: Natural history, biomarkers and prospects for therapeutics. Nat. Rev. Neurol. 2014, 10, 204–216. [Google Scholar] [CrossRef]
  65. Levine, M.S.; Klapstein, G.J.; Koppel, A.; Gruen, E.; Cepeda, C.; Vargas, M.E.; Jokel, E.S.; Carpenter, E.M.; Zanjani, H.; Hurst, R.S.; et al. Enhanced sensitivity to N-methyl-D-aspartate receptor activation in transgenic and knockin mouse models of Huntington’s disease. J. Neurosci. Res. 1999, 58, 515–532. [Google Scholar] [CrossRef]
  66. Cepeda, C.; Ariano, M.A.; Calvert, C.R.; Flores-Hernández, J.; Chandler, S.H.; Leavitt, B.R.; Hayden, M.R.; Levine, M.S. NMDA receptor function in mouse models of Huntington disease. J. Neurosci. Res. 2001, 66, 525–539. [Google Scholar] [CrossRef]
  67. Jaaro-Peled, H.; Kumar, S.; Hughes, D.; Sumitomo, A.; Kim, S.-H.; Zoubovsky, S.; Hirota-Tsuyada, Y.; Zala, D.; Bruyere, J.; Katz, B.M.; et al. Regulation of sensorimotor gating via Disc1/Huntingtin-mediated Bdnf transport in the cortico-striatal circuit. Mol. Psychiatry 2022, 27, 1805–1815. [Google Scholar] [CrossRef] [PubMed]
  68. Zuccato, C.; Marullo, M.; Conforti, P.; MacDonald, M.E.; Tartari, M.; Cattaneo, E. Systematic Assessment of BDNF and Its Receptor Levels in Human Cortices Affected by Huntington’s Disease. Brain Pathol. 2008, 18, 225–238. [Google Scholar] [CrossRef] [PubMed]
  69. Speidell, A.; Bin Abid, N.; Yano, H. Brain-Derived Neurotrophic Factor Dysregulation as an Essential Pathological Feature in Huntington’s Disease: Mechanisms and Potential Therapeutics. Biomedicines 2023, 11, 2275. [Google Scholar] [CrossRef] [PubMed]
  70. Huwiler, A.; Zangemeister-Wittke, U. The sphingosine 1-phosphate receptor modulator fingolimod as a therapeutic agent: Recent findings and new perspectives. Pharmacol. Ther. 2018, 185, 34–49. [Google Scholar] [CrossRef]
  71. Hla, T.; Brinkmann, V. Sphingosine 1-phosphate (S1P). Neurology 2011, 76, S3–S8. [Google Scholar] [CrossRef]
  72. Grassi, S.; Mauri, L.; Prioni, S.; Cabitta, L.; Sonnino, S.; Prinetti, A.; Giussani, P. Sphingosine 1-Phosphate Receptors and Metabolic Enzymes as Druggable Targets for Brain Diseases. Front. Pharmacol. 2019, 10, 807. [Google Scholar] [CrossRef]
  73. Zhang, Y.H.; Fehrenbacher, J.C.; Vasko, M.R.; Nicol, G.D. Sphingosine-1-Phosphate Via Activation of a G-Protein-Coupled Receptor(s) Enhances the Excitability of Rat Sensory Neurons. J. Neurophysiol. 2006, 96, 1042–1052. [Google Scholar] [CrossRef]
  74. Okada, T.; Kajimoto, T.; Jahangeer, S.; Nakamura, S.-i. Sphingosine kinase/sphingosine 1-phosphate signalling in central nervous system. Cell. Signal. 2009, 21, 7–13. [Google Scholar] [CrossRef]
  75. Riganti, L.; Antonucci, F.; Gabrielli, M.; Prada, I.; Giussani, P.; Viani, P.; Valtorta, F.; Menna, E.; Matteoli, M.; Verderio, C. Sphingosine-1-Phosphate (S1P) Impacts Presynaptic Functions by Regulating Synapsin I Localization in the Presynaptic Compartment. J. Neurosci. 2016, 36, 4624. [Google Scholar] [CrossRef]
  76. Anastasiadou, S.; Knöll, B. The multiple sclerosis drug fingolimod (FTY720) stimulates neuronal gene expression, axonal growth and regeneration. Exp. Neurol. 2016, 279, 243–260. [Google Scholar] [CrossRef]
  77. Doi, Y.; Takeuchi, H.; Horiuchi, H.; Hanyu, T.; Kawanokuchi, J.; Jin, S.; Parajuli, B.; Sonobe, Y.; Mizuno, T.; Suzumura, A. Fingolimod Phosphate Attenuates Oligomeric Amyloid β–Induced Neurotoxicity via Increased Brain-Derived Neurotrophic Factor Expression in Neurons. PLoS ONE 2013, 8, e61988. [Google Scholar] [CrossRef] [PubMed]
  78. Gustafson, E.L.; Ehrlich, M.E.; Trivedi, P.; Greengard, P. Developmental regulation of phosphoprotein gene expression in the caudate-putamen of rat: An in situ hybridization study. Neuroscience 1992, 51, 65–75. [Google Scholar] [CrossRef] [PubMed]
  79. Brunkhorst, R.; Vutukuri, R.; Pfeilschifter, W. Fingolimod for the treatment of neurological diseases—State of play and future perspectives. Front. Cell. Neurosci. 2014, 8, 283. [Google Scholar] [CrossRef]
  80. Stearns, N.A.; Schaevitz, L.R.; Bowling, H.; Nag, N.; Berger, U.V.; Berger-Sweeney, J. Behavioral and anatomical abnormalities in Mecp2 mutant mice: A model for Rett syndrome. Neuroscience 2007, 146, 907–921. [Google Scholar] [CrossRef] [PubMed]
  81. Crevier-Sorbo, G.; Rymar, V.V.; Crevier-Sorbo, R.; Sadikot, A.F. Thalamostriatal degeneration contributes to dystonia and cholinergic interneuron dysfunction in a mouse model of Huntington’s disease. Acta Neuropathol. Commun. 2020, 8, 14. [Google Scholar] [CrossRef] [PubMed]
  82. Chang, Q.; Khare, G.; Dani, V.; Nelson, S.; Jaenisch, R. The Disease Progression of Mecp2 Mutant Mice Is Affected by the Level of BDNF Expression. Neuron 2006, 49, 341–348. [Google Scholar] [CrossRef] [PubMed]
  83. Dani, V.S.; Chang, Q.; Maffei, A.; Turrigiano, G.G.; Jaenisch, R.; Nelson, S.B. Reduced cortical activity due to a shift in the balance between excitation and inhibition in a mouse model of Rett Syndrome. Proc. Natl. Acad. Sci. USA 2005, 102, 12560–12565. [Google Scholar] [CrossRef] [PubMed]
  84. Behrens, P.F.; Franz, P.; Woodman, B.; Lindenberg, K.S.; Landwehrmeyer, G.B. Impaired glutamate transport and glutamate–glutamine cycling: Downstream effects of the Huntington mutation. Brain 2002, 125, 1908–1922. [Google Scholar] [CrossRef] [PubMed]
  85. Liévens, J.-C.; Woodman, B.; Mahal, A.; Bates, G.P. Abnormal Phosphorylation of Synapsin I Predicts a Neuronal Transmission Impairment in the R6/2 Huntington’s Disease Transgenic Mice. Mol. Cell. Neurosci. 2002, 20, 638–648. [Google Scholar] [CrossRef]
  86. Cepeda, C.; Hurst, R.S.; Calvert, C.R.; Hernandez-Echeagaray, E.; Nguyen, O.K.; Jocoy, E.; Christian, L.J.; Ariano, M.A.; Levine, M.S. Transient and Progressive Electrophysiological Alterations in the Corticostriatal Pathway in a Mouse Model of Huntington’s Disease. J. Neurosci. 2003, 23, 961–969. [Google Scholar] [CrossRef]
  87. Ali, N.J.; Levine, M.S. Changes in expression of N-methyl-D-aspartate receptor subunits occur early in the R6/2 mouse model of Huntington’s disease. Dev. Neurosci. 2006, 28, 230–238. [Google Scholar] [CrossRef]
  88. Traficante, A.; Riozzi, B.; Cannella, M.; Rampello, L.; Squitieri, F.; Battaglia, G. Reduced activity of cortico-striatal fibres in the R6/2 mouse model of Huntington’s disease. Neuroreport 2007, 18, 1997–2000. [Google Scholar] [CrossRef]
  89. Cepeda, C.; Starling, A.J.; Wu, N.; Nguyen, O.K.; Uzgil, B.; Soda, T.; André, V.M.; Ariano, M.A.; Levine, M.S. Increased GABAergic function in mouse models of Huntington’s disease: Reversal by BDNF. J. Neurosci. Res. 2004, 78, 855–867. [Google Scholar] [CrossRef]
  90. Milnerwood, A.J.; Raymond, L.A. Early synaptic pathophysiology in neurodegeneration: Insights from Huntington’s disease. Trends Neurosci. 2010, 33, 513–523. [Google Scholar] [CrossRef]
  91. Besusso, D.; Geibel, M.; Kramer, D.; Schneider, T.; Pendolino, V.; Picconi, B.; Calabresi, P.; Bannerman, D.M.; Minichiello, L. BDNF–TrkB signaling in striatopallidal neurons controls inhibition of locomotor behavior. Nat. Commun. 2013, 4, 2031. [Google Scholar] [CrossRef]
  92. Patnaik, A.; Spiombi, E.; Frasca, A.; Landsberger, N.; Zagrebelsky, M.; Korte, M. Fingolimod Modulates Dendritic Architecture in a BDNF-Dependent Manner. Int. J. Mol. Sci. 2020, 21, 3079. [Google Scholar] [CrossRef] [PubMed]
  93. West, A.E.; Griffith, E.C.; Greenberg, M.E. Regulation of transcription factors by neuronal activity. Nat. Rev. Neurosci. 2002, 3, 921–931. [Google Scholar] [CrossRef]
  94. West, A.E.; Pruunsild, P.; Timmusk, T. Neurotrophins: Transcription and translation. Handb. Exp. Pharmacol. 2014, 220, 67–100. [Google Scholar] [CrossRef] [PubMed]
  95. Donegà, S.; Tongiorgi, E. Detecting BDNF Protein Forms by ELISA, Western Blot, and Immunofluorescence. In Neuromethods; Humana Press: Totowa, NJ, USA, 2018. [Google Scholar]
  96. Komnig, D.; Dagli, T.C.; Habib, P.; Zeyen, T.; Schulz, J.B.; Falkenburger, B.H. Fingolimod (FTY720) is not protective in the subacute MPTP mouse model of Parkinson’s disease and does not lead to a sustainable increase of brain-derived neurotrophic factor. J. Neurochem. 2018, 147, 678–691. [Google Scholar] [CrossRef] [PubMed]
  97. Kartalou, G.-I.; Pereira, A.R.S.; Endres, T.; Lesnikova, A.; Casarotto, P.; Pousinha, P.; Delanoe, K.; Edelmann, E.; Castrén, E.; Gottmann, K.; et al. Anti-inflammatory treatment with FTY720 starting after onset of symptoms reverses synaptic and memory deficits in an AD mouse model. Int. J. Mol. Sci. 2020, 21, 8957. [Google Scholar] [CrossRef]
  98. Smith, P.A.; Schmid, C.; Zurbruegg, S.; Jivkov, M.; Doelemeyer, A.; Theil, D.; Dubost, V.; Beckmann, N. Fingolimod inhibits brain atrophy and promotes brain-derived neurotrophic factor in an animal model of multiple sclerosis. J. Neuroimmunol. 2018, 318, 103–113. [Google Scholar] [CrossRef]
  99. Hait, N.C.; Wise, L.E.; Allegood, J.C.; O’Brien, M.; Avni, D.; Reeves, T.M.; Knapp, P.E.; Lu, J.; Luo, C.; Miles, M.F.; et al. Active, phosphorylated fingolimod inhibits histone deacetylases and facilitates fear extinction memory. Nat. Neurosci. 2014, 17, 971–980. [Google Scholar] [CrossRef]
  100. Segura-Ulate, I.; Yang, B.; Vargas-Medrano, J.; Perez, R.G. FTY720 (Fingolimod) reverses alpha-synuclein-induced downregulation of brain-derived neurotrophic factor mRNA in OLN-93 oligodendroglial cells. Neuropharmacology 2017, 117, 149–157. [Google Scholar] [CrossRef]
  101. Mizugishi, K.; Yamashita, T.; Olivera, A.; Miller, G.F.; Spiegel, S.; Proia, R.L. Essential role for sphingosine kinases in neural and vascular development. Mol. Cell. Biol. 2005, 25, 11113–11121. [Google Scholar] [CrossRef]
  102. Wu, J.; Santos-Garcia, I.; Eiriz, I.; Bruning, T.; Kvasnicka, A.; Friedecky, D.; Nyman, T.A.; Pahnke, J. Sex-dependent efficacy of sphingosine-1-phosphate receptor agonist FTY720 in mitigating Huntington’s disease. Pharmacol. Res. 2025, 211, 107557. [Google Scholar] [CrossRef]
  103. Miguez, A.; García-Díaz Barriga, G.; Brito, V.; Straccia, M.; Giralt, A.; Ginés, S.; Canals, J.M.; Alberch, J. Fingolimod (FTY720) enhances hippocampal synaptic plasticity and memory in Huntington’s disease by preventing p75NTR up-regulation and astrocyte-mediated inflammation. Hum. Mol. Genet. 2015, 24, 4958–4970. [Google Scholar] [CrossRef]
  104. Di Pardo, A.; Pepe, G.; Castaldo, S.; Marracino, F.; Capocci, L.; Amico, E.; Madonna, M.; Giova, S.; Jeong, S.K.; Park, B.-M.; et al. Stimulation of Sphingosine Kinase 1 (SPHK1) Is Beneficial in a Huntington’s Disease Pre-clinical Model. Front. Mol. Neurosci. 2019, 12, 100. [Google Scholar] [CrossRef]
  105. Luthi-Carter, R.; Apostol, B.L.; Dunah, A.W.; DeJohn, M.M.; Farrell, L.A.; Bates, G.P.; Young, A.B.; Standaert, D.G.; Thompson, L.M.; Cha, J.-H.J. Complex alteration of NMDA receptors in transgenic Huntington’s disease mouse brain: Analysis of mRNA and protein expression, plasma membrane association, interacting proteins, and phosphorylation. Neurobiol. Dis. 2003, 14, 624–636. [Google Scholar] [CrossRef] [PubMed]
  106. André, V.M.; Cepeda, C.; Venegas, A.; Gomez, Y.; Levine, M.S. Altered Cortical Glutamate Receptor Function in the R6/2 Model of Huntington’s Disease. J. Neurophysiol. 2006, 95, 2108. [Google Scholar] [CrossRef]
  107. Zabel, C.; Mao, L.; Woodman, B.; Rohe, M.; Wacker, M.A.; Klare, Y.; Koppelstatter, A.; Nebrich, G.; Klein, O.; Grams, S.; et al. A large number of protein expression changes occur early in life and precede phenotype onset in a mouse model for huntington disease. Mol. Cell. Proteom. 2009, 8, 720–734. [Google Scholar] [CrossRef]
  108. Cha, J.H.; Kosinski, C.M.; Kerner, J.A.; Alsdorf, S.A.; Mangiarini, L.; Davies, S.W.; Penney, J.B.; Bates, G.P.; Young, A.B. Altered brain neurotransmitter receptors in transgenic mice expressing a portion of an abnormal human huntington disease gene. Proc. Natl. Acad. Sci. USA 1998, 95, 6480–6485. [Google Scholar] [CrossRef] [PubMed]
  109. Milnerwood, A.J.; Gladding, C.M.; Pouladi, M.A.; Kaufman, A.M.; Hines, R.M.; Boyd, J.D.; Ko, R.W.Y.; Vasuta, O.C.; Graham, R.K.; Hayden, M.R.; et al. Early Increase in Extrasynaptic NMDA Receptor Signaling and Expression Contributes to Phenotype Onset in Huntington’s Disease Mice. Neuron 2010, 65, 178–190, Correction in: Neuron 2010, 65, 436. [Google Scholar] [CrossRef] [PubMed]
  110. Parsons, M.P.; Raymond, L.A. Extrasynaptic NMDA Receptor Involvement in Central Nervous System Disorders. Neuron 2014, 82, 279–293. [Google Scholar] [CrossRef]
  111. Hardingham, G.E.; Bading, H. Synaptic versus extrasynaptic NMDA receptor signalling: Implications for neurodegenerative disorders. Nat. Rev. Neurosci. 2010, 11, 682–696. [Google Scholar] [CrossRef]
  112. Hardingham, G.E.; Fukunaga, Y.; Bading, H. Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. Nat. Neurosci. 2002, 5, 405–414. [Google Scholar] [CrossRef]
  113. Léveillé, F.; El Gaamouch, F.; Gouix, E.; Lecocq, M.; Lobner, D.; Nicole, O.; Buisson, A. Neuronal viability is controlled by a functional relation between synaptic and extrasynaptic NMDA receptors. FASEB J. 2008, 22, 4258–4271. [Google Scholar] [CrossRef]
  114. Thomas, G.M.; Huganir, R.L. MAPK cascade signalling and synaptic plasticity. Nat. Rev. Neurosci. 2004, 5, 173–183. [Google Scholar] [CrossRef]
  115. Cummings, D.M.; André, V.M.; Uzgil, B.O.; Gee, S.M.; Fisher, Y.E.; Cepeda, C.; Levine, M.S. Alterations in Cortical Excitation and Inhibition in Genetic Mouse Models of Huntington’s Disease. J. Neurosci. 2009, 29, 10371. [Google Scholar] [CrossRef]
  116. Raymond, L.A.; André, V.M.; Cepeda, C.; Gladding, C.M.; Milnerwood, A.J.; Levine, M.S. Pathophysiology of Huntington’s disease: Time-dependent alterations in synaptic and receptor function. Neuroscience 2011, 198, 252–273. [Google Scholar] [CrossRef] [PubMed]
  117. Wild, E.; Björkqvist, M.; Tabrizi, S.J. Immune markers for Huntington’s disease? Expert Rev. Neurother. 2008, 8, 1779–1781. [Google Scholar] [CrossRef]
  118. Jia, Q.; Li, S.; Li, X.-J.; Yin, D. Neuroinflammation in Huntington’s disease: From animal models to clinical therapeutics. Front. Immunol. 2022, 13, 1088124. [Google Scholar] [CrossRef] [PubMed]
  119. Hsiao, H.-Y.; Chen, Y.-C.; Chen, H.-M.; Tu, P.-H.; Chern, Y. A critical role of astrocyte-mediated nuclear factor-κB-dependent inflammation in Huntington’s disease. Hum. Mol. Genet. 2013, 22, 1826–1842. [Google Scholar] [CrossRef] [PubMed]
  120. Pido-Lopez, J.; Andre, R.; Benjamin, A.C.; Ali, N.; Farag, S.; Tabrizi, S.J.; Bates, G.P. In vivo neutralization of the protagonist role of macrophages during the chronic inflammatory stage of Huntington’s disease. Sci. Rep. 2018, 8, 11447. [Google Scholar] [CrossRef]
  121. Gasser, J.; Gillet, G.; Valadas, J.S.; Rouvière, L.; Kotian, A.; Fan, W.; Keaney, J.; Kadiu, I. Innate immune activation and aberrant function in the R6/2 mouse model and Huntington’s disease iPSC-derived microglia. Front. Mol. Neurosci. 2023, 16, 1191324. [Google Scholar] [CrossRef] [PubMed]
  122. Bjorkqvist, M.; Wild, E.J.; Thiele, J.; Silvestroni, A.; Andre, R.; Lahiri, N.; Raibon, E.; Lee, R.V.; Benn, C.L.; Soulet, D.; et al. A novel pathogenic pathway of immune activation detectable before clinical onset in Huntington’s disease. J. Exp. Med. 2008, 205, 1869–1877. [Google Scholar] [CrossRef]
  123. Etxeberria-Rekalde, E.; Alzola-Aldamizetxebarria, S.; Flunkert, S.; Hable, I.; Daurer, M.; Neddens, J.; Hutter-Paier, B. Quantification of Huntington’s Disease Related Markers in the R6/2 Mouse Model. Front. Mol. Neurosci. 2021, 13, 617229. [Google Scholar] [CrossRef]
  124. Rothhammer, V.; Kenison, J.E.; Tjon, E.; Takenaka, M.C.; de Lima, K.A.; Borucki, D.M.; Chao, C.-C.; Wilz, A.; Blain, M.; Healy, L.; et al. Sphingosine 1-phosphate receptor modulation suppresses pathogenic astrocyte activation and chronic progressive CNS inflammation. Proc. Natl. Acad. Sci. USA 2017, 114, 2012–2017. [Google Scholar] [CrossRef]
  125. Sorensen, S.D.; Nicole, O.; Peavy, R.D.; Montoya, L.M.; Lee, C.J.; Murphy, T.J.; Traynelis, S.F.; Hepler, J.R. Common Signaling Pathways Link Activation of Murine PAR-1, LPA, and S1P Receptors to Proliferation of Astrocytes. Mol. Pharmacol. 2003, 64, 1199. [Google Scholar] [CrossRef]
  126. Mullershausen, F.; Craveiro, L.M.; Shin, Y.; Cortes-Cros, M.; Bassilana, F.; Osinde, M.; Wishart, W.L.; Guerini, D.; Thallmair, M.; Schwab, M.E.; et al. Phosphorylated FTY720 promotes astrocyte migration through sphingosine-1-phosphate receptors. J. Neurochem. 2007, 102, 1151–1161. [Google Scholar] [CrossRef]
  127. Reick, C.; Ellrichmann, G.; Tsai, T.; Lee, D.-H.; Wiese, S.; Gold, R.; Saft, C.; Linker, R.A. Expression of brain-derived neurotrophic factor in astrocytes—Beneficial effects of glatiramer acetate in the R6/2 and YAC128 mouse models of Huntington’s disease. Exp. Neurol. 2016, 285, 12–23. [Google Scholar] [CrossRef]
  128. Di Pardo, A.; Amico, E.; Basit, A.; Armirotti, A.; Joshi, P.; Neely, D.M.; Vuono, R.; Castaldo, S.; Digilio, A.F.; Scalabrì, F.; et al. Defective Sphingosine-1-phosphate metabolism is a druggable target in Huntington’s disease. Sci. Rep. 2017, 7, 5280, Erratum in Sci. Rep. 2018, 8, 8266. [Google Scholar] [CrossRef] [PubMed]
  129. Pirhaji, L.; Milani, P.; Dalin, S.; Wassie, B.T.; Dunn, D.E.; Fenster, R.J.; Avila-Pacheco, J.; Greengard, P.; Clish, C.B.; Heiman, M.; et al. Identifying therapeutic targets by combining transcriptional data with ordinal clinical measurements. Nat. Commun. 2017, 8, 623. [Google Scholar] [CrossRef] [PubMed]
  130. García-González, X.; Cubo, E.; Simón-Vicente, L.; Mariscal, N.; Alcaraz, R.; Aguado, L.; Rivadeneyra-Posadas, J.; Sanz-Solas, A.; Saiz-Rodríguez, M. Pharmacogenetics in the Treatment of Huntington’s Disease: Review and Future Perspectives. J. Pers. Med. 2023, 13, 385. [Google Scholar] [CrossRef]
  131. Kao, Y.-H.; Chern, Y.; Yang, H.-T.; Chen, H.-M.; Lin, C.-J. Regulation of P-glycoprotein expression in brain capillaries in Huntington’s disease and its impact on brain availability of antipsychotic agents risperidone and paliperidone. J. Cereb. Blood Flow Metab. 2016, 36, 1412–1423. [Google Scholar] [CrossRef] [PubMed]
  132. Wu, K.-C.; Lin, C.-J. The regulation of drug-metabolizing enzymes and membrane transporters by inflammation: Evidences in inflammatory diseases and age-related disorders. J. Food Drug Anal. 2019, 27, 48–59. [Google Scholar] [CrossRef]
  133. Miller, L. Analyzing Gels and Western Blots with Imagej. Available online: https://lukemiller.org/index.php/2010/11/analyzing-gels-and-western-blots-with-image-j/ (accessed on 23 December 2025).
  134. Bailoo, J.D.; Bohlen, M.O.; Wahlsten, D. The precision of video and photocell tracking systems and the elimination of tracking errors with infrared backlighting. J. Neurosci. Methods 2010, 188, 45–52. [Google Scholar] [CrossRef] [PubMed]
  135. Carter, R.J.; Lione, L.A.; Humby, T.; Mangiarini, L.; Mahal, A.; Bates, G.P.; Dunnett, S.B.; Morton, A.J. Characterization of Progressive Motor Deficits in Mice Transgenic for the Human Huntington’s Disease Mutation. J. Neurosci. 1999, 19, 3248–3257. [Google Scholar] [CrossRef] [PubMed]
  136. Monville, C.; Torres, E.M.; Dunnett, S.B. Comparison of incremental and accelerating protocols of the rotarod test for the assessment of motor deficits in the 6-OHDA model. J. Neurosci. Methods 2006, 158, 219–223. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effects of chronic fingolimod treatment on locomotor behavior. WT (wildtype) and R6/2 mice received saline (i.p., q3.5d; Sal) or fingolimod (0.1 mg/kg, i.p., q3.5d; Fing) treatments starting at age 4 weeks. (a) WT mice show progressive weight gain in both saline and fingolimod treatment groups. (b) R6/2 mice show impaired weight gain after age 7 weeks in both treatment groups. (c) Open field assay shows fingolimod does not alter spontaneous locomotor activity in either WT or (d) R6/2 mice, and R6/2 mice exhibited age-related decline in locomotor activity regardless of treatment, reflecting progression of the degenerative process (* p < 0.050). (e) Rotarod assay shows that fingolimod improves motor coordination in 10-week-old WT mice, as indicated by the increased latency to fall (* p < 0.050). (f) However, no such effect was observed in R6/2 mice with similar latency to fall for both treatment groups. (g) Limb clasping scores increase with age in R6/2 mice, indicating progressive motor dystonia in both treatment groups (* p < 0.050; ** p < 0.001). Fingolimod treatment exacerbates limb dystonia in R6/2 mice at both 8 and 10 weeks of age (# p < 0.050 vs. saline).
Figure 1. Effects of chronic fingolimod treatment on locomotor behavior. WT (wildtype) and R6/2 mice received saline (i.p., q3.5d; Sal) or fingolimod (0.1 mg/kg, i.p., q3.5d; Fing) treatments starting at age 4 weeks. (a) WT mice show progressive weight gain in both saline and fingolimod treatment groups. (b) R6/2 mice show impaired weight gain after age 7 weeks in both treatment groups. (c) Open field assay shows fingolimod does not alter spontaneous locomotor activity in either WT or (d) R6/2 mice, and R6/2 mice exhibited age-related decline in locomotor activity regardless of treatment, reflecting progression of the degenerative process (* p < 0.050). (e) Rotarod assay shows that fingolimod improves motor coordination in 10-week-old WT mice, as indicated by the increased latency to fall (* p < 0.050). (f) However, no such effect was observed in R6/2 mice with similar latency to fall for both treatment groups. (g) Limb clasping scores increase with age in R6/2 mice, indicating progressive motor dystonia in both treatment groups (* p < 0.050; ** p < 0.001). Fingolimod treatment exacerbates limb dystonia in R6/2 mice at both 8 and 10 weeks of age (# p < 0.050 vs. saline).
Ijms 27 00494 g001
Figure 2. Effects of chronic fingolimod treatment on BDNF-TrkB signaling proteins in forebrain of 11-week-old R6/2 mice. (a) Representative Western blot of striatal proteins (rows): BDNF (14 kDa), phospho-(p)TrkB (145 kDa), DARPP-32 (32 kDa), full-length (fl)TrkB (145 kDa), and β-III-Tubulin (50 kDa) from individual (columns) R6/2 mice after chronic treatment with saline (sal, −) or fingolimod (0.1 mg/kg, i.p.; Fing, +). Molecular weight standards are indicated on the right. (b) Graph of relative levels of striatal BDNF and downstream signaling proteins in R6/2 mice normalized to saline controls. Striatal BDNF levels are higher after chronic fingolimod treatments vs. saline controls (* p < 0.050). (c) Representative blots of motor cortex proteins from individual R6/2 mice after chronic treatment with saline (−) or fingolimod (+). (d) Graph of relative cortical protein levels in R6/2 mice shows that BDNF levels are higher after chronic fingolimod treatments vs. saline controls (* p < 0.050). (e) Ratios of pTrkB/total TrkB levels in the striatum and (f) motor cortex in 11-week-old R6/2 mice show that the activation of TrkB signaling is similar after chronic fingolimod treatments vs. saline controls.
Figure 2. Effects of chronic fingolimod treatment on BDNF-TrkB signaling proteins in forebrain of 11-week-old R6/2 mice. (a) Representative Western blot of striatal proteins (rows): BDNF (14 kDa), phospho-(p)TrkB (145 kDa), DARPP-32 (32 kDa), full-length (fl)TrkB (145 kDa), and β-III-Tubulin (50 kDa) from individual (columns) R6/2 mice after chronic treatment with saline (sal, −) or fingolimod (0.1 mg/kg, i.p.; Fing, +). Molecular weight standards are indicated on the right. (b) Graph of relative levels of striatal BDNF and downstream signaling proteins in R6/2 mice normalized to saline controls. Striatal BDNF levels are higher after chronic fingolimod treatments vs. saline controls (* p < 0.050). (c) Representative blots of motor cortex proteins from individual R6/2 mice after chronic treatment with saline (−) or fingolimod (+). (d) Graph of relative cortical protein levels in R6/2 mice shows that BDNF levels are higher after chronic fingolimod treatments vs. saline controls (* p < 0.050). (e) Ratios of pTrkB/total TrkB levels in the striatum and (f) motor cortex in 11-week-old R6/2 mice show that the activation of TrkB signaling is similar after chronic fingolimod treatments vs. saline controls.
Ijms 27 00494 g002
Figure 3. Effects of acute fingolimod treatment on BDNF-TrkB signaling proteins in 4-week-old WT and R6/2 mice. At 48 h after a single injection of saline (sal) or 0.1 mg/kg fingolimod (Fing), striatal tissues were extracted and analyzed by Western blot. (a) Representative blots of striatal proteins (rows) from individual (columns) WT mice treated at 4 weeks with saline (−) or fingolimod (+). (b) Graph of relative levels of striatal BDNF and downstream signaling proteins in WT mice normalized to saline controls. pTrkB and fl-TrkB levels are increased by this low dose of fingolimod vs. saline controls at 4 weeks (* p < 0.050). (c) Representative blots of striatal proteins from individual R6/2 mice treated at 4 weeks with saline (−) or fingolimod (+). (d) Graph of relative levels of striatal proteins in R6/2 mice shows that BDNF, pTrkB, and fl-TrkB are decreased by this low dose of fingolimod vs. saline controls at 4 weeks (* p < 0.050 ). (e) Ratios of activated pTrkB/total TrkB levels in the striatum of WT mice and (f) R6/2 mice show that activation of TrkB signaling is attenuated after fingolimod treatment in R6/2 mice (* p < 0.050 vs. saline).
Figure 3. Effects of acute fingolimod treatment on BDNF-TrkB signaling proteins in 4-week-old WT and R6/2 mice. At 48 h after a single injection of saline (sal) or 0.1 mg/kg fingolimod (Fing), striatal tissues were extracted and analyzed by Western blot. (a) Representative blots of striatal proteins (rows) from individual (columns) WT mice treated at 4 weeks with saline (−) or fingolimod (+). (b) Graph of relative levels of striatal BDNF and downstream signaling proteins in WT mice normalized to saline controls. pTrkB and fl-TrkB levels are increased by this low dose of fingolimod vs. saline controls at 4 weeks (* p < 0.050). (c) Representative blots of striatal proteins from individual R6/2 mice treated at 4 weeks with saline (−) or fingolimod (+). (d) Graph of relative levels of striatal proteins in R6/2 mice shows that BDNF, pTrkB, and fl-TrkB are decreased by this low dose of fingolimod vs. saline controls at 4 weeks (* p < 0.050 ). (e) Ratios of activated pTrkB/total TrkB levels in the striatum of WT mice and (f) R6/2 mice show that activation of TrkB signaling is attenuated after fingolimod treatment in R6/2 mice (* p < 0.050 vs. saline).
Ijms 27 00494 g003aIjms 27 00494 g003b
Figure 4. Effects of acute fingolimod treatment on BDNF-TrkB signaling proteins in 7-week-old WT and R6/2 mice. At 48 h after a single injection of saline (sal) or 0.1 mg/kg fingolimod (Fing), striatal tissues were extracted and analyzed by Western blot. (a) Representative blots of striatal proteins (rows) from individual (columns) WT mice treated at 7 weeks with saline (−) or fingolimod (+). (b) Graph of relative levels of striatal BDNF and downstream signaling proteins in WT mice normalized to saline controls. There are no significant differences between this low dose of fingolimod vs. saline treatment at 7 weeks. (c) Representative blots of striatal proteins from individual R6/2 mice treated at 7 weeks with saline (−) or fingolimod (+). (d) Graph of relative levels of striatal proteins in R6/2 mice shows no significant differences between this low dose of fingolimod vs. saline treatment at 7 weeks old. (e) Ratios of pTrkB/total TrkB levels in the striatum of 7-week-old WT mice and (f) R6/2 mice show that activation of TrkB signaling is not significantly different after fingolimod vs. saline treatment in WT mice and R6/2 mice.
Figure 4. Effects of acute fingolimod treatment on BDNF-TrkB signaling proteins in 7-week-old WT and R6/2 mice. At 48 h after a single injection of saline (sal) or 0.1 mg/kg fingolimod (Fing), striatal tissues were extracted and analyzed by Western blot. (a) Representative blots of striatal proteins (rows) from individual (columns) WT mice treated at 7 weeks with saline (−) or fingolimod (+). (b) Graph of relative levels of striatal BDNF and downstream signaling proteins in WT mice normalized to saline controls. There are no significant differences between this low dose of fingolimod vs. saline treatment at 7 weeks. (c) Representative blots of striatal proteins from individual R6/2 mice treated at 7 weeks with saline (−) or fingolimod (+). (d) Graph of relative levels of striatal proteins in R6/2 mice shows no significant differences between this low dose of fingolimod vs. saline treatment at 7 weeks old. (e) Ratios of pTrkB/total TrkB levels in the striatum of 7-week-old WT mice and (f) R6/2 mice show that activation of TrkB signaling is not significantly different after fingolimod vs. saline treatment in WT mice and R6/2 mice.
Ijms 27 00494 g004
Figure 5. Acute effects of higher doses of fingolimod on BDNF-TrkB signaling proteins in 4-week-old WT and R6/2 mice. At 48 h after a single injection of either saline (Sal) or 1 mg/kg or 3 mg/kg fingolimod (Fing), striatal tissues were extracted and analyzed by Western blot. (a) Representative blots of striatal proteins (rows) from individual (columns) 4-week-old WT mice. (b) Graph of relative levels of striatal BDNF and downstream signaling proteins in WT mice normalized to saline controls. BDNF and pTrkB levels are higher after fingolimod vs. saline treatment at 4 weeks (* p < 0.050). (c) Representative blots of striatal proteins from individual 4-week-old R6/2 mice. (d) Graph of relative levels of striatal proteins in R6/2 mice shows that BDNF, pTrkB, and DARPP-32 are lower after fingolimod vs. saline treatment at 4 weeks old (* p < 0.050; ** p < 0.001). (e) Ratios of pTrkB/total TrkB levels in the striatum of 4-week-old WT mice and (f) R6/2 mice show that activation of TrkB signaling is elevated by fingolimod treatment in both WT mice and R6/2 mice (* p < 0.050 vs. saline).
Figure 5. Acute effects of higher doses of fingolimod on BDNF-TrkB signaling proteins in 4-week-old WT and R6/2 mice. At 48 h after a single injection of either saline (Sal) or 1 mg/kg or 3 mg/kg fingolimod (Fing), striatal tissues were extracted and analyzed by Western blot. (a) Representative blots of striatal proteins (rows) from individual (columns) 4-week-old WT mice. (b) Graph of relative levels of striatal BDNF and downstream signaling proteins in WT mice normalized to saline controls. BDNF and pTrkB levels are higher after fingolimod vs. saline treatment at 4 weeks (* p < 0.050). (c) Representative blots of striatal proteins from individual 4-week-old R6/2 mice. (d) Graph of relative levels of striatal proteins in R6/2 mice shows that BDNF, pTrkB, and DARPP-32 are lower after fingolimod vs. saline treatment at 4 weeks old (* p < 0.050; ** p < 0.001). (e) Ratios of pTrkB/total TrkB levels in the striatum of 4-week-old WT mice and (f) R6/2 mice show that activation of TrkB signaling is elevated by fingolimod treatment in both WT mice and R6/2 mice (* p < 0.050 vs. saline).
Ijms 27 00494 g005
Figure 6. Acute effects of higher doses of fingolimod on BDNF-TrkB signaling proteins in 7-week-old WT or R6/2 mice. At 48 h after a single injection of either saline (Sal) or 1 mg/kg or 3 mg/kg fingolimod (Fing), striatal tissues were extracted and analyzed by Western blot. (a) Representative blots of striatal proteins (rows) from individual (columns) 7-week-old WT mice. (b) Graph of relative levels of striatal BDNF and downstream signaling proteins in WT mice normalized to saline controls. BDNF, pTrkB, DARPP-32, and fl-TrkB levels are higher after fingolimod vs. saline treatment at 7 weeks old (* p < 0.050, ** p < 0.001). (c) Representative blots of striatal proteins from individual 7-week-old R6/2 mice. (d) Graph of relative levels of striatal proteins in R6/2 mice shows that BDNF and DARPP-32 are higher after fingolimod vs. saline treatment at 7 weeks old (* p < 0.050 ). (e) Ratios of pTrkB/total TrkB levels in the striatum of 7-week-old WT mice and (f) R6/2 mice show activation of TrkB signaling is elevated by fingolimod treatment in R6/2 mice (* p < 0.050 vs. saline).
Figure 6. Acute effects of higher doses of fingolimod on BDNF-TrkB signaling proteins in 7-week-old WT or R6/2 mice. At 48 h after a single injection of either saline (Sal) or 1 mg/kg or 3 mg/kg fingolimod (Fing), striatal tissues were extracted and analyzed by Western blot. (a) Representative blots of striatal proteins (rows) from individual (columns) 7-week-old WT mice. (b) Graph of relative levels of striatal BDNF and downstream signaling proteins in WT mice normalized to saline controls. BDNF, pTrkB, DARPP-32, and fl-TrkB levels are higher after fingolimod vs. saline treatment at 7 weeks old (* p < 0.050, ** p < 0.001). (c) Representative blots of striatal proteins from individual 7-week-old R6/2 mice. (d) Graph of relative levels of striatal proteins in R6/2 mice shows that BDNF and DARPP-32 are higher after fingolimod vs. saline treatment at 7 weeks old (* p < 0.050 ). (e) Ratios of pTrkB/total TrkB levels in the striatum of 7-week-old WT mice and (f) R6/2 mice show activation of TrkB signaling is elevated by fingolimod treatment in R6/2 mice (* p < 0.050 vs. saline).
Ijms 27 00494 g006aIjms 27 00494 g006b
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

Nguyen, K.Q.; Rymar, V.V.; Sadikot, A.F. Fingolimod Effects on Motor Function and BDNF-TrkB Signaling in a Huntington’s Mouse Model Are Disease-Stage-Dependent. Int. J. Mol. Sci. 2026, 27, 494. https://doi.org/10.3390/ijms27010494

AMA Style

Nguyen KQ, Rymar VV, Sadikot AF. Fingolimod Effects on Motor Function and BDNF-TrkB Signaling in a Huntington’s Mouse Model Are Disease-Stage-Dependent. International Journal of Molecular Sciences. 2026; 27(1):494. https://doi.org/10.3390/ijms27010494

Chicago/Turabian Style

Nguyen, Khanh Q., Vladimir V. Rymar, and Abbas F. Sadikot. 2026. "Fingolimod Effects on Motor Function and BDNF-TrkB Signaling in a Huntington’s Mouse Model Are Disease-Stage-Dependent" International Journal of Molecular Sciences 27, no. 1: 494. https://doi.org/10.3390/ijms27010494

APA Style

Nguyen, K. Q., Rymar, V. V., & Sadikot, A. F. (2026). Fingolimod Effects on Motor Function and BDNF-TrkB Signaling in a Huntington’s Mouse Model Are Disease-Stage-Dependent. International Journal of Molecular Sciences, 27(1), 494. https://doi.org/10.3390/ijms27010494

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