Hostname: page-component-848d4c4894-v5vhk Total loading time: 0 Render date: 2024-06-20T05:57:09.826Z Has data issue: false hasContentIssue false

Sirtuins and neuropeptide y downregulation in Flinders Sensitive Line rat model of depression

Published online by Cambridge University Press:  20 October 2021

Miranda Stiernborg
Affiliation:
Department of Molecular Medicine and Surgery, Neurogenetics Unit, Karolinska Institutet, Stockholm, Sweden Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
Paschalis Efstathopoulos
Affiliation:
Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
Andreas Lennartsson
Affiliation:
Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
Catharina Lavebratt
Affiliation:
Department of Molecular Medicine and Surgery, Neurogenetics Unit, Karolinska Institutet, Stockholm, Sweden Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
Aleksander A. Mathé*
Affiliation:
Department of Clinical Neuroscience, Center for Psychiatry Research (CPF), Karolinska Institutet, Stockholm, Sweden
*
Author for correspondence: Aleksander A. Mathé, Email: aleksander.mathe@ki.se
Rights & Permissions [Opens in a new window]

Abstract

Since the NAD+-dependent histone deacetylases sirtuin-1 (SIRT1) and sirtuin-2 (SIRT2) are critically involved in epigenetics, endocrinology and immunology and affect the longevity in model organisms, we investigated their expression in brains of 3-month-old and 14–15 months old rat model of depression Flinders Sensitive Line (FSL) and control Flinders Resistant Line (FRL) rats. In view of the dysregulated NPY system in depression, we also studied NPY in young and old FSL to explore the temporal trajectory of depressive-like–ageing interaction. Sirt1, Sirt2 and Npy mRNA were determined using qRT-PCR in prefrontal cortex (PFC) from young and old FSL and FRL, and in hippocampi from young FSL and FRL. PFC: Sirt1 expression was decreased in FSL (p = 0.001). An interaction between age and genotype was found (p = 0.032); young FSL had lower Sirt1 with respect to both age (p = 0.026) and genotype (p = 0.001). Sirt2 was lower in FSL (p = 0.003). Npy mRNA was downregulated in FSL (p = 0.001) but did not differ between the young and old rat groups. Hippocampus: Sirt1 was reduced in young FSL compared to young FRL (p = 0.005). There was no difference in Sirt2 between FSL and FRL. Npy levels were decreased in hippocampus of young FSL compared to young FRL (p = 0.003). Effects of ageing could not be investigated due to loss of samples. To conclude, i this is the first demonstration that SIRT1 and SIRT2 are changed in brain of FSL, a rat model of depression; ii the changes are age-dependent; iii sirtuins are potential targets for treatment of age-related neurodegenerative diseases.

Type
Original Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
© The Author(s), 2021. Published by Cambridge University Press on behalf of Scandinavian College of Neuropsychopharmacology

Significant outcomes

Major depressive disorder is the predominant cause of ‘Years of life lived with disability’ and ‘Years of life lost because of premature death’. Severity and prevalence of depression increase with age, but the molecular underpinnings of the disorder have only partially been elucidated. Since the NAD+-dependent histone deacetylases sirtuin-1 and sirtuin-2 affect the longevity in model organisms, we assessed Sirt1 and Sirt2 mRNA in the prefrontal cortex and hippocampus of young and old FSL rats, a well-characterised model of depression, and control FRL rats with the aim to explore the depressive-like ageing trajectory. Sirt1 levels were decreased in the young FSL rats compared to the aged FSL as well as to the age-matched young FRL rats (cf Table 1); the former finding is of interest for depression-like–ageing interaction and the latter for elucidating depression pathology. Concluding, this is the first demonstration that SIRT1 and SIRT2 are changed in brain of the FSL rats and that the changes are age-dependent. Thus, sirtuins are potential targets for treatment of age-related neurodegenerative diseases.

Limitations

(i) The small number of animals, (ii) due to lack of samples we could not determine the protein, and (iii) due to the loss of samples we could not compare hippocampal tissue from ‘young’ and ‘aged’ rats (cf Table 1).

Introduction

Major depressive disorder (MDD) is a devastating, life-threatening disorder with a major and increasing burden on the public health (Greenberg et al., Reference Greenberg, Fournier, Sisitsky, Pike and Kessler2015). The understanding of MDD aetiology and pathophysiology is limited (McEwen et al., Reference McEwen, Bowles, Gray, Hill, Hunter, Karatsoreos and Nasca2015) and optimal treatments are lacking. Using the currently available drugs, 30–40% of the patients fail to respond adequately (Tundo et al., Reference Tundo, de Filippis and Proietti2015). However, ketamine and esketamine (the latter approved by FDA and EMA in 2020 for treatment-resistant depression) have shown acute antidepressant effect (Henter et al., Reference Henter, Park and Zarate2021). Moreover, recent papers translationally based on rat studies (Cohen et al., Reference Cohen, Liu, Kozlovsky, Kaplan, Zohar and Mathé2012) have shown that insufflated NPY alleviated MDD symptoms (Mathé et al., Reference Mathé, Michaneck, Berg, Charney and Murrough2020) and PTSD symptoms (Sayed et al., Reference Sayed, Van Dam, Horn, Kautz, Parides, Costi, Collins, Iacoviello, Iosifescu, Mathé, Southwick, Feder, Charney and Murrough2018).

Individuals with depression have an enhanced risk of developing age-related diseases. Moreover, persons diagnosed with repeated episodes of MDD have decreased life expectancy, often due to cardiovascular disease and diabetes (Mezuk et al., Reference Mezuk, Eaton, Albrecht and Golden2008; Walker et al., Reference Walker, McGee and Druss2015). In a longitudinal study of ageing (ELSA), an association between the duration of depressive symptoms and mortality risk has been demonstrated (White et al., Reference White, Zaninotto, Walters, Kivimäki, Demakakos, Biddulph, Kumari, De Oliveira, Gallacher and Batty2016). However, the underlying biological mechanisms have only in part been elucidated. Of particular interest for our work that focuses on depression and ageing are sirtuins, a family of 7 NAD+-dependent histone deacetylases (HDACs) that alter the chromatin structure through deacetylation of histones resulting in altered gene expression (Sun et al., Reference Sun, Kennedy and Nestler2013; Lu et al., Reference Lu, Li, Zhang, Zhao, Yan and Yang2018). Sirtuins have several targets including the histone modifications H4K16ac and H3K18ac as well as H3K9m3 (Vaquero et al., Reference Vaquero, Scher, Lee, Erdjument-Bromage, Tempst and Reinberg2004, Reference Vaquero, Scher, Hwei-Ling, Alt, Serrano, Sterglanz and Reinberg2006; Vaquero et al., Reference Vaquero, Scher, Erdjument-Bromage, Tempst, Serrano and Reinberg2007; Eskandarian et al., Reference Eskandarian, Impens, Nahori, Soubigou, Coppée, Cossart and Hamon2013). Thus, as HDACs, sirtuins regulate several essential cellular processes, such as mitochondrial activity, synaptic plasticity, apoptosis, cellular stress resistance, inflammation and memory processes through epigenetic marking (Brunet et al., Reference Brunet, Sweeney, Sturgill, Chua, Greer, Lin, Tran, Ross, Mostoslavsky, Cohen, Hu, Cheng, Jedrychowski, Gygi, Sinclair, Alt and Greenberg2004; Gao et al., Reference Gao, Wang, Mao, Graff, Guan, Pan, Mak, Kim, Su and Tsai2010; Michan et al., Reference Michan, Li, Chou, Parrella, Ge, Long, Allard, Lewis, Miller, Xu, Mervis, Chen, Guerin, Smith, McBurney, Sinclair, Baudry, de Cabo and Longo2010; Yoshizaki et al., Reference Yoshizaki, Schenk, Imamura, Babendure, Sonoda, Bae, Oh, Lu, Milne, Westphal, Bandyopadhyay and Olefsky2010; Wątroba and Szukiewicz, Reference Wątroba and Szukiewicz2016). With regard to ageing, sirtuins also alter the longevity in model organisms, where an extra copy of Sirt2, an orthologue to the mammalian sirtuin genes, extended the replicative lifespan in yeast by 50%, while a deletion of the gene shortened the lifespan (Haigis and Guarente, Reference Haigis and Guarente2006). Thus, sirtuins may play an important role in ageing. Indeed, several studies suggest their role in the pathogenesis of many age-related diseases, including neurodegenerative diseases (reviewed in Wątroba and Szukiewicz, Reference Wątroba and Szukiewicz2016).

Furthermore, several studies have shown that alterations of sirtuin levels have been associated with depression-like behaviour in animal models and depression in humans. For example, a sparse whole genome sequencing of a large population of depressed patients identified alterations of two single nucleotide polymorphisms (SNPs) that contributed to risk of MDD, with one of them located near the SIRT1 gene (CONVERGE consortium, 2015). Moreover, a decrease in SIRT1 and SIRT2 expression in leukocytes was suggested to be a state marker in MDD and bipolar disorder patients (Abe et al., Reference Abe, Uchida, Otsuki, Hobara, Yamagata, Higuchi, Shibata and Watanabe2011). In accordance with this, another study reported decreased levels of SIRT1 in peripheral blood from MDD patients (McGrory et al., Reference McGrory, Ryan, Kolshus, Finnegan and McLoughlin2018). This was further supported by finding of reduced Sirt1 activity in the dentate gyrus in a mouse model of chronic stress. Results from rodent models regarding the role of Sirt1 in depression have been conflicting. A pharmacologic inhibition of hippocampal Sirt1 activity increased depression-like behaviour, while Sirt1 upregulation led to stress resilience, with corresponding effects on dendrite length and spine plasticity for dentate gyrus granule neurons (Abe-Higuchi et al., Reference Abe-Higuchi, Uchida, Yamagata, Higuchi, Hobara, Hara, Kobayashi and Watanabe2016). Ferland et al. (Reference Ferland, Hawley, Puckett, Wineberg, Lubin, Dohanich and Schrader2013), in contrast to Abe-Higuchi, demonstrated that mice subjected to chronic stress had increased Sirt1 activity in dentate gyrus. In addition, mice with a Sirt1 knockout in brain showed decreased anxiety and mice globally overexpressing Sirt1 showed increased anxiety (Libert et al., Reference Libert, Pointer, Bell, Das, Cohen, Asara, Bergmann, Preisig, Otowa, Kendler, Chen, Hettema, vandenOord, Rubio and Guarente2011), and the Sirt1 activator Resveratrol had an antidepressant-like effect in rat models of depression. However, resveratrol is hypothesised to also act through other pathways leading to antidepressant-like effect (Howitz et al., Reference Howitz, Bitterman, Cohen, Lamming, Lavu, Wood, Zipkin, Chung, Kisielewski, Zhang, Scherer and Sinclair2003; Hurley et al., Reference Hurley, Akinfiresoye, Kalejaiye and Tizabi2014; Liu et al., Reference Liu, Xie, Yang, Gu, Ge, Wang and Wang2014). With regard to Sirt2, mice exposed to chronic social defeat stress had increased Sirt2 mRNA and protein levels in the prefrontal cortex (PFC), whereas repeated injections of imipramine reduced the Sirt2 levels (Erburu et al., Reference Erburu, Muñoz-Cobo, Domínguez-Andrés, Beltran, Suzuki, Mai, Valente, Puerta and Tordera2015). Administration of a Sirt2 inhibitor into the mouse PFC reversed the chronic mild stress-induced elevated PFC Sirt2 levels and depression-like behaviour (Erburu et al., Reference Erburu, Muñoz-Cobo, Diaz-Perdigon, Mellini, Suzuki, Puerta and Tordera2017).

Neuropeptide Y (NPY) is the most abundant peptide in mammal brain and decreased NPY levels have been demonstrated in the cerebrospinal fluid of MDD and posttraumatic stress disorder (PTSD) patients as well as bipolar patients at risk for suicide. These findings have been replicated preclinically in models such as the Flinders Sensitive Line (FSL) an extensively characterised model of depression, displaying depressive-like behaviour, and Flinders Resistant Line (FRL) rats (Heilig et al., Reference Heilig, Zachrisson, Thorsell, Ehnvall, Mottagui-Tabar, Sjögren, Asberg, Ekman, Wahlestedt and Agren2004; Overstreet et al., Reference Overstreet, Friedman, Mathé and Yadid2005; Wu et al., Reference Wu, Feder, Wegener, Bailey, Saxena, Charney and Mathé2011; Cohen et al., Reference Cohen, Liu, Kozlovsky, Kaplan, Zohar and Mathé2012; Overstreet and Wegener, Reference Overstreet and Wegener2013; Sandberg et al., Reference Sandberg, Jakobsson, Pålsson, Landén and Mathé2014; Thorsell and Mathé, Reference Thorsell and Mathé2017). FSL rats display several features that resemble the human depression, including elevated REM sleep, psychomotor retardation and increased immobility in the forced swim test (Overstreet et al., Reference Overstreet, Friedman, Mathé and Yadid2005; Overstreet and Wegener, Reference Overstreet and Wegener2013). Additionally, several molecular features are shared with human depression, such as elevated proinflammatory cytokine IL-6, lower central expression of the glial-specific protein S100B and complement factor C3 in several brain regions; shortened telomere length, dysregulated NPY system as well as abnormalities in the glutamatergic system (Jimenez-Vasquez et al., Reference Jimenez-Vasquez, Overstreet and Mathé2000, Reference Jiménez-Vasquez, Diaz-Cabiale, Caberlotto, Bellido, Overstreet, Fuxe and Mathé2007; Hascup et al., Reference Hascup, Hascup, Stephens, Glaser, Yoshitake, Mathé, Gerhardt and Kehr2011; Melas et al., Reference Melas, Mannervik, Mathé and Lavebratt2012; Wei et al., Reference W, ei, Melas, Wegener, Mathé and Lavebratt2014, Reference Wei, Backlund, Wegener, Mathé and Lavebratt2015; Strenn et al., Reference Strenn, Suchankova, Nilsson, Fischer, Wegener, Mathé and Ekman2015; Du Jardin et al., Reference Du Jardin, Muller, Sanchez, Wegener and Elfving2017). Sirt2 has been shown to modulate the glutamate system (Erburu et al., Reference Erburu, Muñoz-Cobo, Diaz-Perdigon, Mellini, Suzuki, Puerta and Tordera2017). Thus, in the PFC of the FSL rat, no differences in glutamate dynamics were observed in brains of young (3–6 months old) FRL and FSL rats, but a significant increase in resting glutamate levels was found in the aged (12–15 months old) FSL compared with the young FSL and age-matched FRL rats. In FSL, percent change in glutamate release during stress was also higher in the aged rat compared to younger rat (Hascup et al., Reference Hascup, Hascup, Stephens, Glaser, Yoshitake, Mathé, Gerhardt and Kehr2011). These findings confirmed and extended interactions between depression and age in animal models. In view of these findings, we investigated the mRNA expression levels of Sirt1, Sirt2 as well as Npy in the FSL and the control FRL rat strains. The behavioural and associated brain differences between FSL and FRL, inter alia in NPY expression, are well documented and we measured NPY as a positive control. In addition, we have previously found NPY changes with age (Husum et al., Reference Husum, Aznar, Hoyer-Hansen, Larsen, Mikkelsen, Moller, Mathé and Wörtwein2006). The analyses were performed in the PFC and hippocampus, brain regions implicated in MDD and rodent depression models (Overstreet et al., Reference Overstreet, Friedman, Mathé and Yadid2005; Neumann et al., Reference Neumann, Wegener, Homberg, Cohen, Slattery, Zohar, Olivier and Mathé2011; Overstreet and Wegener, Reference Overstreet and Wegener2013).

Materials and methods

Animals and tissue samples

FSL and FRL rats were maintained at the Animal Facility, Karolinska Institutet, Huddinge, under controlled conditions of temperature (22 ± 1°C), relative humidity (45–55%) and daylight cycle (12:12 h, lights on at 6:00 am). Standard rat chow and tap water were available ad libitum. The PFC and hippocampi were dissected according to Glowinski and Iversen (Reference Glowinski and Iversen1966) and stored at −80°C until subsequent analyses. Expression levels of Sirt1, Sirt2 and Npy were determined in the PFC of male (’young’ = 3-month-old and ‘aged’ = 14–15 months old) FSL young n = 4, FSL aged n = 7, FRL young n = 5 and FRL aged n = 9. Since hippocampal tissue from the aged rats was lost, we could only analyse tissue from the 3-month-old rats (FSL n = 8; FRL n = 7). All experimental work was approved by the Ethical Committee for protection of animals at the Karolinska Institutet.

RNA extraction and reverse transcription

Tissue-Tearor (Biospec Products Inc., Bartlesville, OK, USA) was used to homogenise the brain samples. Total RNA was extracted using AllPrep DNA/RNA mini kit according to the manufactures protocol (Qiagen, Hilden, Germany) and was treated with DNase I to eliminate the contamination of DNA (Qiagen). The total RNA concentrations were measured spectrophotometrically using NanoDrop 2000 (Thermo Fisher Scientific, Rockford, IL, USA). The RNA quality of 10 samples was examined by Agilent 2100 BioAnalyzer (Agilent Technologies, Germany). Complementary DNA (cDNA) was synthesised by reverse transcription of total RNA using SuperScript III First-Strand Synthesis System for qPCR with random hexamers according to the manufacturer’s protocol (Invitrogen, Carlsbad, CA, USA). cDNA was stored at −20°C and RNA at −80°C.

Gene expression by quantitative RT-PCR

Quantative RT-PCR was applied to measure mRNA levels of the genes Sirt1, Sirt2 and Npy in PFC and hippocampal tissue. Amplifications of these genes and the housekeeping gene Actin were performed in triplicates of the cDNA samples using Power-SYBR Green (Applied Biosystems Inc., Foster City, CA, USA) and the QuantStudioTM flex 6/7 System (Thermo Fisher Scientific). Sample to sample variation in Sirt1, Sirt2 and Npy expression levels were corrected for by normalising to that of Actin, and interplate variation was corrected for by including a positive control/calibrator sample in each plate. The relative gene expression, fold change, was calculated according to the comparative Ct method using the formula: 2−ΔΔCt. The primers sequences, restricting the amplification to mRNA, were (written 5′ to 3′) Sirt1 Fw:TACCCCATGAAGTGCCTCAA, Sirt1 Rv:AAGTTTGGCATACTCGCCAC, Sirt2 Fw: GCAGGAATCCCTGACTTCCG, Sirt2 Rv: TGCCCAGGATAGAGCTCCTTA, Npy Fw:CGCTCTGCGACACTACATCA, Npy Rv:TGGGGGCATTTTCTGTGCTT, beta-actin Fw: TAAGGCCAACCGTGAAAAGAT and beta-actin Rv: GTGGTACGACCAGAGGCATAC.

Statistical analysis

The Levene’s test and Shapiro−Wilk test were used for assessing the homogeneity of variance and the normality of the data, respectively. The statistical significance of differences in PFC gene expression between the four groups (young and aged FRL and FSL rats) were tested applying two way-ANOVA followed by a post hoc analysis with a Bonferroni-corrected independent sample t-test. As the beforehand mentioned assumptions were met for hippocampus, a two-tailed independent test was used. Statistical significance was assigned for p < 0.05. Outliers marked in the figure legends were identified based on the values below Q1-1.5*IQR or above Q3 + 1.5*IQR. The analyses were made in R (Rstudio version 1.2.1335).

Results

Prefrontal cortex

Expression levels of Sirt1, Sirt2 and Npy in PFC of young and old FSL and FRL rats are depicted in Fig. 1. Sirt1 levels were significantly decreased in the FSL compared to the FRL rats (F[1,21]= 13.8; p = 0.001, two-way ANOVA). Sirt1 levels were also significantly different between the age groups (F[1,21]= 6.22; p = 0.022), and a significant interaction between age and genotype was found (F[1,21]= 5.26; p = 0.032); the young FSL rats had a significantly lower Sirt1 expression with respect to both age (p = 0.026, post hoc test) and genotype (p = 0.001, post hoc test). Sirt2 levels were significantly lower in the FSL (F[1,21]= 14,1; p = 0.001) but did not differ between the age groups. With regard to Npy , in similarity to Sirt2, Npy mRNA levels in PFC were downregulated in the FSL compared to the FRL rats (F[1,21]= 31.3; p < 0.001) but did not differ between the age groups.

Fig. 1. Relative mRNA levels of the Sirt1, Sirt2 and Npy genes in the prefrontal cortex (PFC) of young and aged FSL and FRL rats. (A) Sirt1 levels were significantly decreased in FSL compared to FRL. Moreover, the levels were lower in the young FSL rats compared to the aged FSL. (B) Sirt2 levels were lower in FSL compared to FRL. No difference with regard to age was found. (C) Npy levels were reduced in FSL compared to FRL. No difference with regard to age was found. The relative quantification (R.Q) bars represent mean values and the error bars represent the standard error of the mean (SEM). n = 4 young FSL; n = 7 old FSL, n = 5 young FRL; n = 9 old FRL; * p < 0.05, ** p < 0.01.

Hippocampus

Expression levels of Sirt1, Sirt2 and Npy in hippocampus of young FSL and FRL rats are depicted in Fig. 2. Since samples from the old FSL and FRL rats were lost in processing, effects of ageing could not be investigated. In similarity to findings in the PFC, hippocampal Sirt1 levels were reduced in young FSL compared to young FRL rats (p = 0.005). There was no detectable difference in Sirt2 levels between the FSL and FRL. Lastly, Npy levels were decreased in the hippocampus of the young FSL compared to young FRL rats (p = 0.003).

Fig. 2. Relative mRNA levels of the Sirt1, Sirt2 and Npy genes in the hippocampi of young FSL and FRL rats. (A) Sirt1 levels were significantly decreased in young FSL compared to young FRL. (B) No differences in Sirt2 levels between young FRL and young FSL were detected. n = 8 FSL; n = 7 FRL (one outlier in the FSL group in the Sirt1 gene has been excluded) *p < 0.05, **p < 0.01. (C) Npy levels were significantly decreased in young FSL compared to young FRL. The relative quantification (R.Q) bars represent mean values and the error bars represent the standard error of the mean (SEM) n = 8 FSL; n = 7 FRL (one outlier in the FSL group in the Sirt1 gene has been excluded) *p < 0.05, ** p < 0.01.

Discussion

Our main findings are lower expressions of Sirt1 and Sirt2 in the PFC of the FSL compared to the FRL strain. Moreover, Sirt1 was also decreased in young FSL rats. Relationships of these changes to pathology and the disease trajectory with age are not clear but point to new targets to elucidate depression pathophysiology and develop novel treatments. Vicissitudes of Sirt1 and Sirt2 expression have been reviewed in the Introduction section. Differences in species used (mouse and rat) as well as a variety of experimental procedures have led to divergent, sometimes conflicting results. Nevertheless, a tentative conclusion is that reduced Sirt1 and Sirt2 expressions are associated with depression and anxiety-like behaviours. The downregulated Sirt1 levels in PFC and hippocampus of young FSL rats were partly in agreement with reports showing a reduction of Sirt1 mRNA levels in hippocampus but not in medial PFC using stress-induced behavioural models of depression in 2-month-old mice (Abe-Higuchi et al., Reference Abe-Higuchi, Uchida, Yamagata, Higuchi, Hobara, Hara, Kobayashi and Watanabe2016). In line with this, 2-month-old mice susceptible to chronic social defeat stress had reduced Sirt1 mRNA levels in hippocampus compared to the controls and stress-resilient mice (Kim et al., Reference Kim, Hesterman, Call, Magazu, Keeley, Armenta, Kronman, Neve, Nestler and Ferguson2016). With regard to the downregulated Sirt2 levels, we found in the PFC of the FSL rats, studies in mice focusing on chronic stress and the disrupted vesicular glutamate transporter 1 (VGLUT1+/−) models reported a link between reduction in Sirt2 mRNA and Sirt2 activity in PFC (Erburu et al., Reference Erburu, Muñoz-Cobo, Domínguez-Andrés, Beltran, Suzuki, Mai, Valente, Puerta and Tordera2015, Reference Erburu, Muñoz-Cobo, Diaz-Perdigon, Mellini, Suzuki, Puerta and Tordera2017; Munoz-Cobo et al., Reference Munoz-Cobo, Belloch, Diaz-Perdigon, Puerta and Tordera2017). In those experiments, both antidepressants and a selective Sirt2 inhibitor reduced Sirt2 levels in PFC as well as depression-like behaviour. Probable explanations are that the discrepancies are due to species differences and that FSL rats represent a genetic model of depression clearly different from the stress models and the model of disrupted VGLUT transporter. Furthermore, the differences between our Sirt2 findings and the aforementioned studies in PFC could reflect the heterogeneity in underlying molecular pathways of depression-like behaviour. Another variable that has to be considered in interpreting the results is that of circadian rhythm. For instance, SIRT1 expression also shows circadian rhythm (Chang and Guarente, Reference Chang and Guarente2013) and, in a different context, we have shown that circadian rhythm fluctuations in NPYergic system and HPA) axis underlie differences in vulnerability to stress responses (Cohen et al., Reference Cohen, Vainer, Matar, Kozlovsky, Kaplan, Zohar, Mathé and Cohen2015). However, since we controlled for the time of sacrifice, this factor can be ruled out with regard to results within our study but could have a bearing on comparison with other studies. The Sirt1 mRNA expression in the PFC region was decreased in young FSL rats only, while the levels in the older group did not significantly differ between the FSL and FRL. These findings suggest that during the ageing trajectory, there are compensatory mechanisms that counteract the reduction of Sirt1 levels found in young depressed FSL rats; elucidation of such mechanisms should contribute to understanding the depression–ageing interaction. Finally, our findings are consistent with the reports of reduced SIRT1 and SIRT2 levels in leukocytes from patients with MDD compared to healthy controls (Abe et al., Reference Abe, Uchida, Otsuki, Hobara, Yamagata, Higuchi, Shibata and Watanabe2011; McGrory et al., Reference McGrory, Ryan, Kolshus, Finnegan and McLoughlin2018).

Findings of reduced Npy levels in both PFC and hippocampus of the FSL rats are in agreement with previous results (Jiménez-Vasquez et al., Reference Jimenez-Vasquez, Overstreet and Mathé2000 and Reference Jiménez-Vasquez, Diaz-Cabiale, Caberlotto, Bellido, Overstreet, Fuxe and Mathé2007; Wu et al., Reference Wu, Feder, Wegener, Bailey, Saxena, Charney and Mathé2011; Thorsell and Mathé, Reference Thorsell and Mathé2017) and are thus a quality indicator of procedures used in this study. With regard to the effect of age, in one study (Husum et al., Reference Husum, Aznar, Hoyer-Hansen, Larsen, Mikkelsen, Moller, Mathé and Wörtwein2006), we found that ageing caused an exacerbated loss of NPY immunoreactive cells in the dentate gyrus in the FSL strain compared with FRL. The aged FSL rats also had shortened 5-HT-IR fibres in the dorsal hippocampus, indicative of an impaired 5-HT innervation of this area, compared with FRL. These results are not contradictory due to the basic difference in methodology and call for in depth study of ageing effect on the NPYergic system.

The strengths of the study– the first of its kind – are the findings of significant decrease in Sirt1 and Sirt2 expression in the FSL compared to the FRL strain as well as an increase in Sirt1 with ageing in brain regions of relevance for depression. In addition, we confirmed the decreased Npy expression in FSL versus FRL strains. These findings contribute to mapping the molecular underpinnings of depression and the ageing–depression temporal trajectory and have a potential to contribute to development of novel therapeutic targets.

Table 1. Significant outcomes and limitations

Authors contributions

AAM: concept and initiative, breeding of animals, preparation of brains, and tissue sample contribution; MS and PE: experimental work: all authors: data analysis and interpretation; MS and AAM: draughting the manuscript. MS, PE, CL, AL and AAM: final manuscript and approval.

Financial support

Supported by the Swedish Medical Research Council grant 10414 (AAM, 2016-02955) and the Centre for Psychiatry Research, SLL-KI (AAM).

Conflict of interest

None.

Statement of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Animal welfare ethical statement

All experimental work was approved by the Ethical Committee for protection of animals at the Karolinska Institutet.

Footnotes

*

Shared first authorship.

References

Abe-Higuchi, N, Uchida, S, Yamagata, H, Higuchi, F, Hobara, T, Hara, K, Kobayashi, A and Watanabe, Y (2016) Hippocampal sirtuin 1 signaling mediates depression-like behavior. Biological Psychiatry 80(11), 815826.CrossRefGoogle ScholarPubMed
Abe, N, Uchida, S, Otsuki, K, Hobara, T, Yamagata, H, Higuchi, F, Shibata, T and Watanabe, Y (2011) Altered sirtuin deacetylase gene expression in patients with a mood disorder. Journal of Psychiatric Research 45(8), 11061112.CrossRefGoogle ScholarPubMed
Brunet, A, Sweeney, LB, Sturgill, JF, Chua, KF, Greer, PL, Lin, Y, Tran, H, Ross, SE, Mostoslavsky, R, Cohen, HY, Hu, LS, Cheng, H-L, Jedrychowski, MP, Gygi, SP, Sinclair, DA, Alt, FW and Greenberg, ME (2004) Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303(5666), 20112015.CrossRefGoogle ScholarPubMed
Chang, H-C and Guarente, L (2013) SIRT1 mediates central circadian control in the SCN by a mechanism that decays with aging. Cell 153(7), 14481460.CrossRefGoogle ScholarPubMed
Cohen, H, Liu, T, Kozlovsky, N, Kaplan, Z, Zohar, J and Mathé, AA (2012) The neuropeptide Y (NPY)-ergic system is associated with behavioral resilience to stress exposure in an animal model of post-traumatic stress disorder. Neuropsychopharmacology 37(2), 350363.CrossRefGoogle Scholar
Cohen, S, Vainer, E, Matar, MA, Kozlovsky, N, Kaplan, Z, Zohar, J, Mathé, AA and Cohen, H (2015) Diurnal fluctuations in HPA and neuropeptide Y-ergic systems underlie differences in vulnerability to traumatic stress responses at different zeitgeber times. Neuropsychopharmacology 40(3), 774790.CrossRefGoogle ScholarPubMed
CONVERGE consortium (2015) Sparse whole-genome sequencing identifies two loci for major depressive disorder. Nature 523(7562), 588591.CrossRefGoogle Scholar
Du Jardin, KG, Muller, HK, Sanchez, C, Wegener, G and Elfving, B (2017) Gene expression related to serotonergic and glutamatergic neurotransmission is altered in the flinders sensitive line rat model of depression: effect of ketamine. Synapse 71(1), 3745.CrossRefGoogle ScholarPubMed
Erburu, M, Muñoz-Cobo, I, Diaz-Perdigon, T, Mellini, P, Suzuki, T, Puerta, E, Tordera, RM (2017) SIRT2 inhibition modulate glutamate and serotonin systems in the prefrontal cortex and induces antidepressant-like action. Neuropharmacology 117, 195208.CrossRefGoogle ScholarPubMed
Erburu, M, Muñoz-Cobo, I, Domínguez-Andrés, J, Beltran, E, Suzuki, T, Mai, A, Valente, S, Puerta, E and Tordera, RM (2015) Chronic stress and antidepressant induced changes in Hdac5 and Sirt2 affect synaptic plasticity. European Neuropsychopharmacology 25(11), 20362048.CrossRefGoogle ScholarPubMed
Eskandarian, HA, Impens, F, Nahori, M-A, Soubigou, G, Coppée, J-Y, Cossart, P and Hamon, MA (2013) A role for SIRT2-dependent histone H3K18 deacetylation in bacterial infection. Science 341(6145), 1238858.CrossRefGoogle ScholarPubMed
Ferland, CL, Hawley, WR, Puckett, RE, Wineberg, K, Lubin, FD, Dohanich, GP and Schrader, LA (2013) Sirtuin activity in dentate gyrus contributes to chronic stress-induced behavior and extracellular signal-regulated protein kinases 1 and 2 cascade changes in the hippocampus. Biological Psychiatry 74(12), 927935.CrossRefGoogle ScholarPubMed
Gao, J, Wang, WY, Mao, YW, Graff, J, Guan, JS, Pan, L, Mak, G, Kim, D, Su, SC and Tsai, L-H (2010) A novel pathway regulates memory and plasticity via SIRT1 and miR-134. Nature 466(7310), 11051109.CrossRefGoogle ScholarPubMed
Glowinski, J and Iversen, LL (1966) Regional studies of catecholamines in the rat brain. I. The disposition of [3H]norepinephrine, [3H]dopamine and [3H]dopa in various regions of the brain. Journal of Neurochemistry 13(8), 655669.CrossRefGoogle Scholar
Greenberg, PE, Fournier, AA, Sisitsky, T, Pike, CT and Kessler, RC (2015) The economic burden of adults with major depressive disorder in the United States (2005 and 2010). The Journal of Clinical Psychiatry 76(2), 155162.CrossRefGoogle Scholar
Haigis, MC and Guarente, LP (2006) Mammalian sirtuins--emerging roles in physiology, aging, and calorie restriction. Genes & Development 20(21), 29132921.CrossRefGoogle ScholarPubMed
Hascup, KN, Hascup, ER, Stephens, ML, Glaser, PEA, Yoshitake, T, Mathé, AA, Gerhardt, GA and Kehr, J (2011) Resting glutamate levels and rapid glutamate transients in the prefrontal cortex of the flinders sensitive line rat: a genetic rodent model of depression. Neuropsychopharmacology 36(8), 17691777.CrossRefGoogle ScholarPubMed
Heilig, M, Zachrisson, O, Thorsell, A, Ehnvall, A, Mottagui-Tabar, S, Sjögren, M, Asberg, M, Ekman, R, Wahlestedt, C, Agren, H (2004) Decreased cerebrospinal fluid neuropeptide Y (NPY) in patients with treatment refractory unipolar major depression: preliminary evidence for association with preproNPY gene polymorphism. Journal of Psychiatric Research 38(2), 113121.CrossRefGoogle ScholarPubMed
Henter, ID, Park, LT and Zarate, CA Jr (2021) Novel glutamatergic modulators for the treatment of mood disorders: current status. CNS Drugs 35(5), 527543. doi: 10.1007/s40263-021-00816-x.CrossRefGoogle ScholarPubMed
Howitz, KT, Bitterman, KJ, Cohen, HY, Lamming, DW, Lavu, S, Wood, JG, Zipkin, RE, Chung, P, Kisielewski, A, Zhang, L-L, Scherer, B and Sinclair, DA (2003) Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 425(6954), 191196.CrossRefGoogle ScholarPubMed
Hurley, LL, Akinfiresoye, L, Kalejaiye, O and Tizabi, Y (2014) Antidepressant effects of resveratrol in an animal model of depression. Behavioural Brain Research 268(Suppl. 1), 17.CrossRefGoogle ScholarPubMed
Husum, H, Aznar, S, Hoyer-Hansen, S, Larsen, MH, Mikkelsen, JD, Moller, A, Mathé, AA and Wörtwein, G (2006) Exacerbated loss of cell survival, neuropeptide Y-immunoreactive (IR) cells, and serotonin-IR fiber lengths in the dorsal hippocampus of the aged flinders sensitive line depressed rat: implications for the pathophysiology of depression? Journal of Neuroscience Research 84(6), 12921302.CrossRefGoogle ScholarPubMed
Jimenez-Vasquez, PA, Overstreet, DH and Mathé, AA (2000) Neuropeptide Y in male and female brains of flinders sensitive line, a rat model of depression. Effects of electroconvulsive stimuli. Journal of Psychiatric Research 34(6), 405412.CrossRefGoogle ScholarPubMed
Jiménez-Vasquez, PA, Diaz-Cabiale, Z, Caberlotto, L, Bellido, I, Overstreet, D, Fuxe, K and Mathé, AA (2007) Electroconvulsive stimuli selectively affect behavior and neuropeptide Y (NPY) and NPY Y(1) receptor gene expressions in hippocampus and hypothalamus of flinders sensitive line rat model of depression. European Neuropsychopharmacology 17(4), 298308.CrossRefGoogle Scholar
Kim, HD, Hesterman, J, Call, T, Magazu, S, Keeley, E, Armenta, K, Kronman, H, Neve, RL, Nestler, EJ and Ferguson, D (2016) SIRT1 mediates depression-like behaviors in the nucleus accumbens. Journal of Neuroscience 36(32), 84418452.CrossRefGoogle ScholarPubMed
Libert, S, Pointer, K, Bell, EL, Das, A, Cohen, DE, Asara, JM, Bergmann, S, Preisig, M, Otowa, T, Kendler, KS, Chen, X, Hettema, JM, vandenOord, EJ, Rubio, EJ and Guarente, L (2011) SIRT1 activates MAO-A in the brain to mediate anxiety and exploratory drive. Cell 147(7), 14591472.CrossRefGoogle ScholarPubMed
Liu, D, Xie, K, Yang, X, Gu, J, Ge, L, Wang, X and Wang, Z (2014) Resveratrol reverses the effects of chronic unpredictable mild stress on behavior, serum corticosterone levels and BDNF expression in rats. Behavioural Brain Research 264(4), 916.CrossRefGoogle ScholarPubMed
Lu, G, Li, J, Zhang, H, Zhao, X, Yan, LJ and Yang, X (2018) Role and possible mechanisms of Sirt1 in depression. Oxidative Medicine and Cellular Longevity 2018(2), 16.Google ScholarPubMed
Mathé, AA, Michaneck, M, Berg, E, Charney, DS and Murrough, JW (2020) A randomized controlled trial of intranasal neuropeptide Y in patients with major depressive disorder. International Journal of Neuropsychopharmacology 23(12), 783790. doi: 10.1093/ijnp/pyaa054.CrossRefGoogle ScholarPubMed
McEwen, BS, Bowles, NP, Gray, JD, Hill, MN, Hunter, RG, Karatsoreos, IN and Nasca, C (2015) Mechanisms of stress in the brain. Nature Neuroscience 18(10), 13531363.CrossRefGoogle Scholar
McGrory, CL, Ryan, KM, Kolshus, E, Finnegan, M and McLoughlin, DM (2018) Peripheral blood SIRT1 mRNA levels in depression and treatment with electroconvulsive therapy. European Neuropsychopharmacology 28(9), 10151023.CrossRefGoogle ScholarPubMed
Melas, PA, Mannervik, M, Mathé, AA and Lavebratt, C (2012) Neuropeptide Y: identification of a novel rat mRNA splice-variant that is downregulated in the hippocampus and the prefrontal cortex of a depression-like model. Peptides 35(1), 4955.CrossRefGoogle ScholarPubMed
Mezuk, B, Eaton, WW, Albrecht, S and Golden, SH (2008) Depression and type 2 diabetes over the lifespan: a meta-analysis. Diabetes Care 31(12), 23832390.CrossRefGoogle ScholarPubMed
Michan, S, Li, Y, Chou, MM, Parrella, E, Ge, H, Long, JM, Allard, JS, Lewis, K, Miller, M, Xu, W, Mervis, RF, Chen, J, Guerin, KI, Smith, LEH, McBurney, MW, Sinclair, DA, Baudry, M, de Cabo, R and Longo, VD (2010) SIRT1 is essential for normal cognitive function and synaptic plasticity. Journal of Neuroscience 30(29), 96959707.CrossRefGoogle ScholarPubMed
Munoz-Cobo, I, Belloch, FB, Diaz-Perdigon, T, Puerta, E and Tordera, RM (2017) SIRT2 inhibition reverses anhedonia in the VGLUT1+/- depression model. Behavioural Brain Research 335, 128131.CrossRefGoogle ScholarPubMed
Neumann, ID, Wegener, G, Homberg, JR, Cohen, H, Slattery, DA, Zohar, J, Olivier, JD and Mathé, AA (2011) Animal models of depression and anxiety: what do they tell us about human condition? Progress in Neuro-Psychopharmacology & Biological Psychiatry 35(6):13571375.CrossRefGoogle Scholar
Overstreet, DH and Wegener, G (2013) The flinders sensitive line rat model of depression. 25 years and still producing. Pharmacological Reviews 65(1), 143155.CrossRefGoogle ScholarPubMed
Overstreet, DH, Friedman, E, Mathé, AA and Yadid, G (2005) The flinders sensitive line rat: a selectively bred putative animal model of depression. Neuroscience & Biobehavioral Reviews 29(4-5), 739759.CrossRefGoogle ScholarPubMed
Sandberg, JV, Jakobsson, J, Pålsson, E, Landén, M and Mathé, AA (2014) Low neuropeptide Y in cerebrospinal fluid in bipolar patients is associated with previous and prospective suicide attempts. European Neuropsychopharmacology 24(12):19071915.CrossRefGoogle Scholar
Sayed, S, Van Dam, NT, Horn, SR, Kautz, MM, Parides, M, Costi, S, Collins, KA, Iacoviello, B, Iosifescu, DV, Mathé, AA, Southwick, SM, Feder, A, Charney, DS, Murrough, JW (2018) A randomized dose-ranging study of neuropeptide Y in patients with posttraumatic stress disorder. International Journal of Neuropsychopharmacology 21(1), 311. doi: 10.1093/ijnp/pyx109.CrossRefGoogle ScholarPubMed
Strenn, N, Suchankova, P, Nilsson, S, Fischer, C, Wegener, G, Mathé, AA and Ekman, A (2015) Expression of inflammatory markers in a genetic rodent model of depression. Behavioural Brain Research 281, 348357.CrossRefGoogle Scholar
Sun, H, Kennedy, PJ and Nestler, EJ (2013) Epigenetics of the depressed brain: role of histone acetylation and methylation. Neuropsychopharmacology 38(1), 124137.CrossRefGoogle ScholarPubMed
Thorsell, A and Mathé, AA (2017) Neuropeptide Y in alcohol addiction and affective disorders. Frontiers in Endocrinology 8, 178.CrossRefGoogle ScholarPubMed
Tundo, A, de Filippis, R and Proietti, L (2015) Pharmacologic approaches to treatment resistant depression: evidences and personal experience. World Journal of Psychiatry 5(3), 330341.CrossRefGoogle ScholarPubMed
Vaquero, A, Scher, M, Erdjument-Bromage, H, Tempst, P, Serrano, L and Reinberg, D (2007) SIRT1 regulates the histone methyl-transferase SUV39H1 during heterochromatin formation. Nature 450(7168), 440444.CrossRefGoogle ScholarPubMed
Vaquero, A, Scher, M, Lee, D, Erdjument-Bromage, H, Tempst, P and Reinberg, D (2004) Human SirT1 interacts with histone H1 and promotes formation of facultative heterochromatin. Molecular Cell 16(1), 93105.CrossRefGoogle ScholarPubMed
Vaquero, A, Scher, MB, Hwei-Ling, C, Alt, FW, Serrano, L, Sterglanz, R and Reinberg, D (2006) Sirt2 is a histone deacetylase with preference for histone H4 Lys 16 during mitosis. Genes & Development 20(10), 12561261.CrossRefGoogle ScholarPubMed
Walker, ER, McGee, RE and Druss, BG (2015) Mortality in mental disorders and global disease burden implications: a systematic review and meta-analysis. JAMA Psychiatry 72(4), 334341.CrossRefGoogle ScholarPubMed
Wątroba, M and Szukiewicz, D (2016) The role of sirtuins in aging and age-related diseases. Advances in Medical Sciences 61:5262.CrossRefGoogle ScholarPubMed
W, ei, Y, Melas, PA, Wegener, G, Mathé, AA and Lavebratt, C (2014) Antidepressant-like effect of sodium butyrate is associated with an increase in TET1 and in 5-hydroxymethylation levels in the Bdnf gene. The International Journal of Neuropsychopharmacology 18(2), pyu032.Google Scholar
Wei, YB, Backlund, L, Wegener, G, Mathé, AA and Lavebratt, C (2015) Telomerase dysregulation in the hippocampus of a rat model of depression: normalization by lithium. International Journal of Neuropsychopharmacology 18(7), pyv002.CrossRefGoogle ScholarPubMed
White, J, Zaninotto, P, Walters, K, Kivimäki, M, Demakakos, P, Biddulph, J, Kumari, M, De Oliveira, C, Gallacher, J and Batty, GD (2016) Duration of depressive symptoms and mortality risk: the English Longitudinal Study of Ageing (ELSA). British Journal of Psychiatry 208(4), 337342.CrossRefGoogle Scholar
Wu, G, Feder, A, Wegener, G, Bailey, C, Saxena, S, Charney, D and Mathé, AA (2011) Central functions of neuropeptide Y in mood and anxiety disorders. Expert Opinion on Therapeutic Targets 15(11), 13171331.CrossRefGoogle ScholarPubMed
Yoshizaki, T, Schenk, S, Imamura, T, Babendure, JL, Sonoda, N, Bae, EJ, Oh, DY, Lu, M, Milne, JC, Westphal, C, Bandyopadhyay, G and Olefsky, JM (2010) SIRT1 inhibits inflammatory pathways in macrophages and modulates insulin sensitivity. American Journal of Physiology-Endocrinology and Metabolism 298(3), E419E428.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Relative mRNA levels of the Sirt1, Sirt2 and Npy genes in the prefrontal cortex (PFC) of young and aged FSL and FRL rats. (A) Sirt1 levels were significantly decreased in FSL compared to FRL. Moreover, the levels were lower in the young FSL rats compared to the aged FSL. (B) Sirt2 levels were lower in FSL compared to FRL. No difference with regard to age was found. (C) Npy levels were reduced in FSL compared to FRL. No difference with regard to age was found. The relative quantification (R.Q) bars represent mean values and the error bars represent the standard error of the mean (SEM). n = 4 young FSL; n = 7 old FSL, n = 5 young FRL; n = 9 old FRL; * p < 0.05, ** p < 0.01.

Figure 1

Fig. 2. Relative mRNA levels of the Sirt1, Sirt2 and Npy genes in the hippocampi of young FSL and FRL rats. (A) Sirt1 levels were significantly decreased in young FSL compared to young FRL. (B) No differences in Sirt2 levels between young FRL and young FSL were detected. n = 8 FSL; n = 7 FRL (one outlier in the FSL group in the Sirt1 gene has been excluded) *p < 0.05, **p < 0.01. (C) Npy levels were significantly decreased in young FSL compared to young FRL. The relative quantification (R.Q) bars represent mean values and the error bars represent the standard error of the mean (SEM) n = 8 FSL; n = 7 FRL (one outlier in the FSL group in the Sirt1 gene has been excluded) *p < 0.05, ** p < 0.01.

Figure 2

Table 1. Significant outcomes and limitations