
2022 Impact Factor
Depression is one of the most common and serious psychiatric disorders affecting people of all ages (Ferrari
Several known mechanisms describe the pathophysiology of depression. One of which is the monoamine hypothesis, which predicts that the underlying cause of depression is the alteration of norepinephrine, dopamine, and serotonin levels in the brain (Jesulola
Neuroinflammation has been widely implicated in the study of depression with reports suggesting the involvement of immune stimulation and challenges (Raison
Type I interferons are innate immune molecules with strong pro-inflammatory responses and mediate inflammasome activation. Type I interferons are required for the upregulation of caspases, which resulted in the non-canonical nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome response or expression priming of NLRP3 components (Malireddi and Kanneganti, 2013). NLRP3 is an intracellular multiprotein complex responsible for the innate immune processes associated with infection, inflammation, and psychiatric conditions such as depression. We searched for a compound that can inhibit the type I interferon and inflammasome activity, and consequently, depressive-like behaviors. We found that hycanthone can inhibit both inflammation and inflammasome response in microglial cell lines.
In the present study, we aimed to investigate the antidepressant effects of hycanthone on the LPS-induced depression in mice model. Our results showed that the production of inflammatory cytokines including TNFα, IL-1β, and IL-18 is reduced by hycanthone in the microglial cell line. Moreover, we also demonstrated the antidepressant effects of hycanthone, at least in part, which is mediated via the modulation of type I interferon signaling pathway. These results imply that hycanthone can be used for the treatment of depression through the regulation of neuroinflammatory responses.
Male C57BL/6 mice (6 weeks) were obtained from the Orient Bio (Seongnam, Korea). The mice were housed in an optimal environment at a temperature range of 23 ± 2°C and humidity range of 50 ± 10% in a dark/light cycle (lights on at 12:00 AM; lights off at 12:00 PM). All procedures were approved by Konkuk IACUC, Seoul, Korea (KU20195).
Hycanthone (Cat No. HY-B1099) was purchased from MedChemExpress (NJ, USA) and LPS (Cat No. L2630) was obtained from Sigma-Aldrich (MO, USA). Hycanthone (25 and 50 mg/kg) was dissolved in 0.1% dimethyl sulfoxide (DMSO) for
The forced swim test (FST) is a widely used behavior test to identify depressive-like behaviors in rodents (Can
The mouse microglia cell line, BV-2 cells, were cultured in DMEM containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (PS) at 37°C in 5% CO2. The cells were pretreated with hycanthone (0.1, 1, 10 μM) 30 min before LPS (10 ng/mL) treatment. To observe the type I interferon-induced Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway activation, HEK-blue IFN α/β cells were used, which were obtained from InvivoGen. HEK-blue IFN α/β cells were cultured in DMEM supplemented with 10% FBS, 1% PS, blasticidin (30 μg/mL), and Zeocin™ (100 μg/mL).
BV-2 cells (1×105/well) were seeded in a 24-well plate and stabilized overnight. The cells were treated with various concentrations of hycanthone before LPS (10 ng/mL) stimulation. After 24 h, the cell supernatant was collected. Equal volumes of cell supernatant and Griess reagent (1% sulphanilamide and 0.1% N-1-napthylenediamine dihydrochloride) were mixed for 10 min at room temperature. The optical density was determined at 540 nm, using SpectraMax® ABS plus (Molecular Devices, CA, USA).
HEK-blue IFN α/β cells (3×105/well) were seeded in 96-well plate and stabilized overnight. The cells were pre-treated (1 h) by various concentration of hycanthone before IFN-α2b (0.1 ng/mL). After 17 h of stimulation, 190 µL of QUANTI-blue solution was added in 10 µL of cell supernatant. Subsequently, 200 µL of supernatant mixture is incubated at 37°C for 0.5 h. The optical density was determined at 655 nm using SpectraMax® ABS plus (Molecular Devices).
BV-2 cells (1×105/well) were seeded in a 24-well plate and stabilized overnight. The cells were treated with various concentrations of hycanthone before LPS (10 ng/mL) and adenosine 5’-triphosphate (ATP) (2 mM) treatment. Three h after adding LPS, the cells were co-stimulated with ATP for another hour. The cell supernatants were used to observe the change in the levels of inflammatory cytokines. ELISA kits (TNF-α, DY410-05; IL-1β, DY401; IL-18, DY7625-05) were purchased from R&D Systems (MN, USA). ELISA was performed according to the manufacturer’s instructions. The optical density was determined at 450 nm using SpectraMax® ABS Plus (Molecular Devices).
Quantitative real-time PCR was conducted to confirm the mRNA expression levels in the prefrontal cortex. The total RNA was extracted using Trizol reagent (Invitrogen, CA, USA) and measured by Nanodrop (Nanodrop Technologies, NC, USA). The cDNA was synthesized using 2 μg of total RNA and the RT reaction mixture containing RevertAid Reverse transcriptase, reaction buffer (Thermo Fisher Scientific, MA, USA), and dNTP (Promega, WI, USA). Template cDNA was amplified by QuantStudio3 (Thermo Fisher Scientific) using BlasTaq™ 2X qPCR MasterMix (Applied Biological Materials Inc., BC, Canada). The primer pairs used in qRT-PCR are mentioned in Table 1.
Table 1 Sequence of qRT-PCR primers
Gene | Accession number | Forward | Reverse |
---|---|---|---|
TNFa | NM_013693 | TTC GAG TGA CAA GCC TGT AG | AGA TTG ACC TCA GCG CTG AGT |
IL1b | NM_008361 | GTC TTT CCC GTG GAC CTT CC | TCG GAG CCT GTA GTG CAG TTG |
IL18 | NM_008360 | TGG CTG CCA TGT CAG AAG ACT | CAG TGA AGT CGG CCA AAG TTG |
NLRP3 | NM_145827 | AAG ATT ACC CGC CCG AGA AAG | TCG CAG CAA AGA TCC ACA CA |
iNOS | NM_010927 | CAA GAG TTT GAC CAG AGG ACC | TGG AAC CAC TCG TAC TTG GGA |
COX2 | NM_011198 | TGC TGT ACA AGC AGT GGC AA | AGG TGC TCG GCT TCC AGT AT |
IDO1 | NM_008324 | AGT CGG AAG AGC CCT CAA AT | GGT GTT TTC TGT GCC CTG AT |
Ifi44 | NM_133871 | ATG CAC TCT TCT GAG CTG GTG G | TCA GAT CCA GGC TAT CCA CGT G |
Gbp7 | NM_145545 | GGT GTC ATC ACA GCA GAC GAG T | CCG TCT TGG AAA GAA GTG CCT G |
All values were described as the mean ± standard error of the mean (SEM). Data were analyzed using one-way ANOVA followed by Tukey’s multiple comparisons in the GraphPad Prism 7 software (GraphPad, CA, USA). The level of statistical significance was considered at a
To evaluate the anti-interferon activity of hycanthone, we performed the QUANTI-blue assay in HEK-blue IFN α/β cells. Secreted alkaline phosphatase (SEAP) reporter system level was significantly increased by the stimulation of interferon-α2b and reduced by treatment of hycanthone at 10 μM (Fig. 1). Ruxolitinib has an inhibitory effect on JAK1/2 and type 1 interferon signaling pathways and was used here as a positive control. The result suggests that hycanthone have modulatory effects on IFN-induced inflammation response.
To investigate whether hycanthone affects the neuroinflammatory response, we first stimulated the microglial BV2 cells with LPS. Nitric oxide (NO) production was increased by LPS stimulation and was counteracted by hycanthone in a dose-dependent manner (Fig. 2A). The elevation of TNF-α expression after LPS stimulation was also reduced by 10 μM hycanthone (Fig. 2B). To activate the NLRP3 inflammasome (Coll
Various studies have demonstrated that neuroinflammation is involved in the pathophysiology of depression by enhancing the expression of inflammatory cytokines (Jeon and Kim, 2018). To determine the antidepressant effect of hycanthone, we treated C57BL/6N mice with hycanthone for 10 days and subsequently induced depressive-like behaviors via LPS stimulation (O’Connor
To investigate whether hycanthone affects the expression of interferon-regulated genes in the prefrontal cortex (PFC) from the mice of depression models, qRT-PCR was performed on interferon induced protein 44 (Ifi44) and guanylate binding protein 7 (Gbp7) genes. LPS increased the expression of Ifi44 (Fig. 4A) and Gbp7 (Fig. 4B), and both were normalized by hycanthone treatment, especially at 50 mg/kg. Both genes are classified into the interferon regulated genes, which are critical in controlling inflammation response. The expression of inflammatory factors and cytokines such as TNF-α and IL-1β was increased in LPS model and hycanthone reduced the expression of both cytokines, although statistical significance was absent (Supplementary Fig. 1). In this study, LPS-induced increased expression of COX-2 was significantly inhibited by hycanthone treatment (Fig. 5), reflecting that hycanthone would inhibit the interferon activity in depressive animal models.
Since indoleamine 2,3-dioxygenase 1 (IDO1) is one of the critical mediators in inflammation-mediated depressive-like behaviors (Jeon
The etiology of depression has been focused mostly on the monoamine deficiency hypothesis (Delgado, 2000) which has progressed to include genetic factors (Shyn and Hamilton, 2010), neuroplasticity and neurogenesis abnormalities (Boku
Various approaches have been used in rodents such as restraint stress, unpredicted chronic mild stress, corticosterone injection, and LPS injection to mimic a human phenotype of depression (O’Connor
Among various gene family members involved in interferons, Ifi44 is over-expressed after stimulation of LPS (Ovstebø
The cytokine-activated JAK-STAT pathway plays a crucial role in the regulation of the immune system (Shuai and Liu, 2003) and is activated by inflammatory cytokines such as IFN-α, IFN-γ, and TNF-α (Schindler
In conclusion, hycanthone may be a potential candidate drug for neuroinflammatory disorders, especially depression, potentially via modulating type-1 interferone pathways as well as cytokine production. Potential downstream targets could be Ifi44, Gbp7 and IDO-1, which deserve further attention. Understanding the mechanisms of hycanthone activity related to the interferon signaling pathway would provide new insights into targeting CNS diseases.
This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (NRF-2016R1A5A2012284 and NRF-2020M3E5D9080165 to Dr. Shin) and by the Korea Drug Development Fund, funded by Ministry of Science and ICT, Ministry of Trade, Industry, and Energy, and Ministry of Health and Welfare (HN21C1076 to Dr. Jeon).
![]() |
![]() |