Tacrolimus, a topical calcineurin inhibitor, is used to treat atopic dermatitis (AD). AD is classically T helper type (Th) 2-driven, but Th17 cells are implicated in chronic AD, yet its efficacy against Th17-dependent pathology remains unclear. We investigated the effects of tacrolimus using a murine model of Th17-mediated allergic skin inflammation. In CD4+ T cells from DO11.10/Rag2−/− mice, which express ovalbumin (OVA)-specific T cell receptor, tacrolimus differentially suppressed stimulation-induced cytokine expression by Th2 cells. At similar concentrations, tacrolimus suppressed Il21, but not Il17a or Il22, expression in Th17 cells. Subcutaneous OVA challenge elicited ear thickening in BALB/c mice after adoptive transfer of Th2 or Th17 cells, with a stronger response in Th17-transferred mice, but less than OVA-immunized controls. Topical tacrolimus reduced Th17-mediated ear swelling, corroborated by histopathology. In Th2-transferred mice, tacrolimus tended to reduce early skin thickening (day 3) but did not affect late responses (day 7). In Th17-transferred mice, tacrolimus significantly reduced allergen-specific T-cell accumulation in OVA-injected skin and tended to reduce Il21 expression, whereas Th2-cell accumulation and cytokine expression were unaffected. Tacrolimus exerts stronger inhibitory effects on Th17- than Th2-driven responses in this model, suggesting that suppression of Th17 pathways may contribute to its therapeutic benefit in AD.
{"title":"Tacrolimus attenuates Th17 cell-mediated allergic skin inflammation in mice","authors":"Fatemeh Beygom Mirkatouli , Yukiko Tao , Uyanga Enkhbaatar , Ryoken Yamanaka , Kento Miura , Norimasa Yamasaki , Sawako Ogata , Naohisa Hosomi , Akio Mori , Tomoharu Yasuda , Masaya Matsuda , Yuya Sannomiya , Takeshi Nabe , Tomofumi Numata , Kazumitsu Sugiura , Noriko Kitamura , Minoru Gotoh , Osamu Kaminuma","doi":"10.1016/j.jphs.2026.01.003","DOIUrl":"10.1016/j.jphs.2026.01.003","url":null,"abstract":"<div><div>Tacrolimus, a topical calcineurin inhibitor, is used to treat atopic dermatitis (AD). AD is classically T helper type (Th) 2-driven, but Th17 cells are implicated in chronic AD, yet its efficacy against Th17-dependent pathology remains unclear. We investigated the effects of tacrolimus using a murine model of Th17-mediated allergic skin inflammation. In CD4<sup>+</sup> T cells from DO11.10/Rag2<sup>−/−</sup> mice, which express ovalbumin (OVA)-specific T cell receptor, tacrolimus differentially suppressed stimulation-induced cytokine expression by Th2 cells. At similar concentrations, tacrolimus suppressed <em>Il21,</em> but not <em>Il17a</em> or <em>Il22</em>, expression in Th17 cells. Subcutaneous OVA challenge elicited ear thickening in BALB/c mice after adoptive transfer of Th2 or Th17 cells, with a stronger response in Th17-transferred mice, but less than OVA-immunized controls. Topical tacrolimus reduced Th17-mediated ear swelling, corroborated by histopathology. In Th2-transferred mice, tacrolimus tended to reduce early skin thickening (day 3) but did not affect late responses (day 7). In Th17-transferred mice, tacrolimus significantly reduced allergen-specific T-cell accumulation in OVA-injected skin and tended to reduce <em>Il21</em> expression, whereas Th2-cell accumulation and cytokine expression were unaffected. Tacrolimus exerts stronger inhibitory effects on Th17- than Th2-driven responses in this model, suggesting that suppression of Th17 pathways may contribute to its therapeutic benefit in AD.</div></div>","PeriodicalId":16786,"journal":{"name":"Journal of pharmacological sciences","volume":"160 3","pages":"Pages 152-157"},"PeriodicalIF":2.9,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145982078","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hypertensive disorders of pregnancy (HDPs) are serious complications that pose significant risks to both maternal and fetal health. In HDP, placental circulatory impairment is generally attributed to defective differentiation and invasion of the human extravillous trophoblasts (EVTs). Our previous RNA-seq analysis of placentas from early-onset HDP patients revealed decreased expression of dihydroorotate dehydrogenase (DHODH), a mitochondrial enzyme involved in pyrimidine biosynthesis. DHODH inhibition is hypothesized to induce mitochondrial dysfunction. In this study, we investigated whether DHODH inhibition affects mitochondrial homeostasis and invasive capacity in EVT cell line. Selective DHODH inhibitors resulted in decreased mitochondrial membrane potential, upregulation of mitochondrial degradation factors, and mitochondrial fragmentation. Inhibition of DHODH or mitochondrial complexes I, II, and IV led to impaired cell invasion, accompanied by an increase in SA-β-gal-positive senescent cells. Treatment with quercetin or riboflavin, enhancers of mitochondrial ATP functions, partially restored invasive capacity and reduced senescent cell accumulation under mitochondrial stress conditions. Thus, DHODH inhibition in EVTs induces cellular senescence and diminished capacity for invasion. Mitochondrial activators such as quercetin and riboflavin may hold therapeutic promise for ameliorating the pathology of HDPs.
{"title":"DHODH inhibition impairs mitochondrial function and extravillous trophoblast invasion: Protective roles of quercetin and riboflavin","authors":"Kanoko Yoshida, Kazuya Kusama, Kana Kanazawa, Yu Kawaguchi, Atsuya Tsuru, Mikihiro Yoshie, Kazuhiro Tamura","doi":"10.1016/j.jphs.2026.01.001","DOIUrl":"10.1016/j.jphs.2026.01.001","url":null,"abstract":"<div><div>Hypertensive disorders of pregnancy (HDPs) are serious complications that pose significant risks to both maternal and fetal health. In HDP, placental circulatory impairment is generally attributed to defective differentiation and invasion of the human extravillous trophoblasts (EVTs). Our previous RNA-seq analysis of placentas from early-onset HDP patients revealed decreased expression of dihydroorotate dehydrogenase (DHODH), a mitochondrial enzyme involved in pyrimidine biosynthesis. DHODH inhibition is hypothesized to induce mitochondrial dysfunction. In this study, we investigated whether DHODH inhibition affects mitochondrial homeostasis and invasive capacity in EVT cell line. Selective DHODH inhibitors resulted in decreased mitochondrial membrane potential, upregulation of mitochondrial degradation factors, and mitochondrial fragmentation. Inhibition of DHODH or mitochondrial complexes I, II, and IV led to impaired cell invasion, accompanied by an increase in SA-β-gal-positive senescent cells. Treatment with quercetin or riboflavin, enhancers of mitochondrial ATP functions, partially restored invasive capacity and reduced senescent cell accumulation under mitochondrial stress conditions. Thus, DHODH inhibition in EVTs induces cellular senescence and diminished capacity for invasion. Mitochondrial activators such as quercetin and riboflavin may hold therapeutic promise for ameliorating the pathology of HDPs.</div></div>","PeriodicalId":16786,"journal":{"name":"Journal of pharmacological sciences","volume":"160 3","pages":"Pages 143-151"},"PeriodicalIF":2.9,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145957726","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Recent studies have reported that sodium-glucose cotransporter (SGLT) 2 inhibitors ameliorate steatotic liver disease. We investigated the contribution of SGLT2 inhibitor-induced fluid loss due to glycosuria in hepatic ureagenesis for water conservation to the association between improvement of steatotic liver disease and the energy-consuming nature of hepatic ureagenesis.
General methods
ob/ob mice fed a high-fat diet without carbohydrate restriction were administered luseogliflozin, an SGLT2 inhibitor, for eight weeks.
Findings
Luseogliflozin significantly decreased the liver weight, plasma aspartate aminotransferase and alanine aminotransferase levels, and fat content in mice with enhanced glycosuria (H-GlcV group). The ratio of urinary urea excretion decreased as a substitute for increased glucose excretion in the H-GlcV group. Luseogliflozin significantly increased liver urea content and tended to increase malate dehydrogenase (MDH)-1 activity. Liver MDH-1 activity was significantly positively correlated with liver urea content, suggesting that MDH-1-induced amino acid recruitment from the tricarboxylic acid cycle to the urea cycle may contribute to enhanced ureagenesis. In addition, liver weight was significantly negatively correlated with the liver urea content.
Conclusions
Our data suggest that enhanced hepatic ureagenesis as a compensatory water conservation mechanism for glycosuria-induced fluid loss may be associated with amelioration of steatotic liver disease in SGLT2 inhibitor-treated ob/ob mice.
{"title":"SGLT2 inhibitor-induced glycosuria improves steatotic liver disease in parallel with enhanced ureagenesis in ob/ob mice","authors":"Satoshi Kidoguchi , Kento Kitada , Asahiro Morishita , Yudai Tsuji , Hiroyuki Ohsaki , Daisuke Yamazaki , Takashi Morikawa , Yoshio Konishi , Hideki Kobara , Takashi Yokoo , Jens Titze , Kazuo Takahashi , Akira Nishiyama","doi":"10.1016/j.jphs.2025.12.006","DOIUrl":"10.1016/j.jphs.2025.12.006","url":null,"abstract":"<div><h3>Purpose</h3><div>Recent studies have reported that sodium-glucose cotransporter (SGLT) 2 inhibitors ameliorate steatotic liver disease. We investigated the contribution of SGLT2 inhibitor-induced fluid loss due to glycosuria in hepatic ureagenesis for water conservation to the association between improvement of steatotic liver disease and the energy-consuming nature of hepatic ureagenesis.</div></div><div><h3>General methods</h3><div><em>ob/ob</em> mice fed a high-fat diet without carbohydrate restriction were administered luseogliflozin, an SGLT2 inhibitor, for eight weeks.</div></div><div><h3>Findings</h3><div>Luseogliflozin significantly decreased the liver weight, plasma aspartate aminotransferase and alanine aminotransferase levels, and fat content in mice with enhanced glycosuria (H-GlcV group). The ratio of urinary urea excretion decreased as a substitute for increased glucose excretion in the H-GlcV group. Luseogliflozin significantly increased liver urea content and tended to increase malate dehydrogenase (MDH)-1 activity. Liver MDH-1 activity was significantly positively correlated with liver urea content, suggesting that MDH-1-induced amino acid recruitment from the tricarboxylic acid cycle to the urea cycle may contribute to enhanced ureagenesis. In addition, liver weight was significantly negatively correlated with the liver urea content.</div></div><div><h3>Conclusions</h3><div>Our data suggest that enhanced hepatic ureagenesis as a compensatory water conservation mechanism for glycosuria-induced fluid loss may be associated with amelioration of steatotic liver disease in SGLT2 inhibitor-treated <em>ob/ob</em> mice.</div></div>","PeriodicalId":16786,"journal":{"name":"Journal of pharmacological sciences","volume":"160 2","pages":"Pages 122-131"},"PeriodicalIF":2.9,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145837624","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-16DOI: 10.1016/j.jphs.2025.12.005
Ami Ono , Tatsunori Miyaoka , Daichi Koan , Zihao Jin , Lu Chen , Atsuko Hayata-Takano , Satoshi Asano , Rei Yokoyama , Kenji Ishimoto , Nobumasa Hino , Akihiro Harada , Takanobu Nakazawa , Hitoshi Hashimoto , James A. Waschek , Shinsaku Nakagawa , Kotaro Tanimoto , Yukio Ago
Accumulating evidence suggests that microduplications of the VIPR2 gene are strongly associated with schizophrenia. VIPR2 encodes vasoactive intestinal peptide receptor 2 (VPAC2). However, cell-type-specific actions of VPAC2 overexpression with respect to schizophrenia remain unclear. Therefore, we aimed to determine the effects of human VPAC2 overexpression in neurons on cognition-related behaviors and prefrontal cortex dendritic morphology in mice. We crossed a Tau-Cre mouse line, which targets neuronal recombinase activity, with a newly generated double transgenic mouse line containing tetracycline-responsive element-human VPAC2-IRES-mCherry and ROSA:LNL:tTA. Immunohistochemical and Western blot analyses revealed that VPAC2 was overexpressed in neurons throughout the brain. Mice that overexpressed VPAC2 showed impaired performance in the novel object recognition test. Furthermore, VPAC2-overexpressing mice exhibited significant reductions in brain weight and the length, branch number, and complexity of arborization of prefrontal cortex pyramidal neuron dendrites. RNA sequencing analysis revealed that VPAC2 overexpression may affect signaling pathways involved in regulating stem cell pluripotency, cell cycle, and actin cytoskeleton. Quantitative PCR analysis also confirmed increased expression of the X-linked lymphocyte-regulated 3B gene, which regulates dendritic morphogenesis and spine assembly. These results suggest that VPAC2 overexpression in neurons has a detrimental effect on brain development, which leads to impaired neural circuitry and cognitive function.
{"title":"Neuron-specific overexpression of human vasoactive intestinal peptide receptor 2 in mice causes cognitive dysfunction and abnormal dendritic morphology in the prefrontal cortex","authors":"Ami Ono , Tatsunori Miyaoka , Daichi Koan , Zihao Jin , Lu Chen , Atsuko Hayata-Takano , Satoshi Asano , Rei Yokoyama , Kenji Ishimoto , Nobumasa Hino , Akihiro Harada , Takanobu Nakazawa , Hitoshi Hashimoto , James A. Waschek , Shinsaku Nakagawa , Kotaro Tanimoto , Yukio Ago","doi":"10.1016/j.jphs.2025.12.005","DOIUrl":"10.1016/j.jphs.2025.12.005","url":null,"abstract":"<div><div>Accumulating evidence suggests that microduplications of the <em>VIPR2</em> gene are strongly associated with schizophrenia. <em>VIPR2</em> encodes vasoactive intestinal peptide receptor 2 (VPAC2). However, cell-type-specific actions of VPAC2 overexpression with respect to schizophrenia remain unclear. Therefore, we aimed to determine the effects of human VPAC2 overexpression in neurons on cognition-related behaviors and prefrontal cortex dendritic morphology in mice. We crossed a Tau-Cre mouse line, which targets neuronal recombinase activity, with a newly generated double transgenic mouse line containing tetracycline-responsive element-human VPAC2-IRES-mCherry and ROSA:LNL:tTA. Immunohistochemical and Western blot analyses revealed that VPAC2 was overexpressed in neurons throughout the brain. Mice that overexpressed VPAC2 showed impaired performance in the novel object recognition test. Furthermore, VPAC2-overexpressing mice exhibited significant reductions in brain weight and the length, branch number, and complexity of arborization of prefrontal cortex pyramidal neuron dendrites. RNA sequencing analysis revealed that VPAC2 overexpression may affect signaling pathways involved in regulating stem cell pluripotency, cell cycle, and actin cytoskeleton. Quantitative PCR analysis also confirmed increased expression of the X-linked lymphocyte-regulated 3B gene, which regulates dendritic morphogenesis and spine assembly. These results suggest that VPAC2 overexpression in neurons has a detrimental effect on brain development, which leads to impaired neural circuitry and cognitive function.</div></div>","PeriodicalId":16786,"journal":{"name":"Journal of pharmacological sciences","volume":"160 2","pages":"Pages 111-121"},"PeriodicalIF":2.9,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145837570","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Triple-negative breast cancer (TNBC), which lacks expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2), is associated with poor prognosis. Immune checkpoint inhibitors (ICIs) have emerged as a promising therapeutic option for TNBC, but their efficacy remains limited due to resistance. In this study, we investigated whether the non-receptor tyrosine kinase Src (Src tyrosine kinase) regulates interferon-gamma (IFNγ)-induced expression of programmed death-ligand 1 (PD-L1). IFNγ stimulation of TNBC and luminal A breast cancer cell lines induced time-dependent phosphorylation of Src at its activation site (Y419). Pharmacological inhibition of Src significantly suppressed IFNγ–induced PD-L1 mRNA and protein expression, as well as activation of PD-L1–related transcription factors, suggesting transcriptional regulation. In co-culture assays with CD8+ T-cells, TNBC cells were more susceptible to T-cell–mediated cytotoxicity compared with luminal A cells, and Src inhibition further enhanced this cytotoxicity. These findings indicate that Src plays a crucial role in IFNγ–mediated PD-L1 expression and immune evasion in TNBC cell lines. Src inhibition may represent a promising combinatorial strategy to enhance antitumor immunity in TNBC cell lines.
{"title":"Src-dependent modulation of IFNγ-induced PD-L1 expression in human breast cancer cell lines","authors":"Chihiro Hayashi , Yuto Mizuno , Yu Iida , Akane Nagasako , Michiko Endo , Wakana Fukae , Eriko Yamashita , Yoshihiro Ishikawa , Masanari Umemura","doi":"10.1016/j.jphs.2025.12.004","DOIUrl":"10.1016/j.jphs.2025.12.004","url":null,"abstract":"<div><div>Triple-negative breast cancer (TNBC), which lacks expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2), is associated with poor prognosis. Immune checkpoint inhibitors (ICIs) have emerged as a promising therapeutic option for TNBC, but their efficacy remains limited due to resistance. In this study, we investigated whether the non-receptor tyrosine kinase Src (Src tyrosine kinase) regulates interferon-gamma (IFNγ)-induced expression of programmed death-ligand 1 (PD-L1). IFNγ stimulation of TNBC and luminal A breast cancer cell lines induced time-dependent phosphorylation of Src at its activation site (Y419). Pharmacological inhibition of Src significantly suppressed IFNγ–induced PD-L1 mRNA and protein expression, as well as activation of PD-L1–related transcription factors, suggesting transcriptional regulation. In co-culture assays with CD8<sup>+</sup> T-cells, TNBC cells were more susceptible to T-cell–mediated cytotoxicity compared with luminal A cells, and Src inhibition further enhanced this cytotoxicity. These findings indicate that Src plays a crucial role in IFNγ–mediated PD-L1 expression and immune evasion in TNBC cell lines. Src inhibition may represent a promising combinatorial strategy to enhance antitumor immunity in TNBC cell lines.</div></div>","PeriodicalId":16786,"journal":{"name":"Journal of pharmacological sciences","volume":"160 2","pages":"Pages 132-141"},"PeriodicalIF":2.9,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145837623","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Magnesium (Mg) is an essential cation critical for neuronal and cardiac homeostasis, principally by modulating N-methyl-d-aspartate (NMDA) receptor function. While magnesium deficiency (MgD) is known to impair memory and autonomic function, the systemic interplay remains poorly understood.
As a preliminary step to investigating neural activity during memory tasks, this study investigated the effects of 3 weeks of MgD on neural activity patterns and cardiac function in rats. Simultaneous electrophysiological recordings from the dorsal hippocampus and medial prefrontal cortex, along with electrocardiogram and electromyogram (EMG) monitoring, revealed that MgD selectively altered neural oscillations without affecting cardiac function or sleep architecture. Notably, frequency-specific changes in local field potentials (LFPs) were most pronounced during quiet wakefulness under dark conditions. This excessive enhancement of gamma wave activity may contribute to memory impairment.
This 3-week deficiency was characterized by altered neural excitability, which could lead to cognitive impairment and partial cardiac arrhythmia.
Our findings demonstrate that spontaneous neural activity is altered even in the 3-week of MgD and support a model in which the deficiency progressively disrupts physiological regulation.
{"title":"Magnesium deficiency differentially modulates hippocampal and prefrontal oscillations and cardiac rhythms","authors":"Sora Inoue , Yuji Ikegaya , Nobuyoshi Matsumoto , Tetsuhiko Kashima","doi":"10.1016/j.jphs.2025.12.002","DOIUrl":"10.1016/j.jphs.2025.12.002","url":null,"abstract":"<div><div>Magnesium (Mg) is an essential cation critical for neuronal and cardiac homeostasis, principally by modulating <em>N</em>-methyl-d-aspartate (NMDA) receptor function. While magnesium deficiency (MgD) is known to impair memory and autonomic function, the systemic interplay remains poorly understood.</div><div>As a preliminary step to investigating neural activity during memory tasks, this study investigated the effects of 3 weeks of MgD on neural activity patterns and cardiac function in rats. Simultaneous electrophysiological recordings from the dorsal hippocampus and medial prefrontal cortex, along with electrocardiogram and electromyogram (EMG) monitoring, revealed that MgD selectively altered neural oscillations without affecting cardiac function or sleep architecture. Notably, frequency-specific changes in local field potentials (LFPs) were most pronounced during quiet wakefulness under dark conditions. This excessive enhancement of gamma wave activity may contribute to memory impairment.</div><div>This 3-week deficiency was characterized by altered neural excitability, which could lead to cognitive impairment and partial cardiac arrhythmia.</div><div>Our findings demonstrate that spontaneous neural activity is altered even in the 3-week of MgD and support a model in which the deficiency progressively disrupts physiological regulation.</div></div>","PeriodicalId":16786,"journal":{"name":"Journal of pharmacological sciences","volume":"160 2","pages":"Pages 97-110"},"PeriodicalIF":2.9,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145711962","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Appropriate processing of aversive information is essential for survival. We previously demonstrated that serotonin neurons in the median raphe nucleus (MRN) play a key role in such processing; however, MRN responses to predictive cues of aversive events remain unclear. Here, we found that the MRN serotonin neurons were activated by aversive air-puff stimuli but not by auditory cues predicting the air puff. Moreover, delayed activation of the ventral hippocampus-projecting MRN serotonin neurons, together with subsequent 5-HT2A receptor signaling, was required for aversion. These findings shed light on the roles of the hippocampus-projecting MRN serotonin neurons and elucidate their molecular mechanisms.
{"title":"Delayed response of the median raphe serotonin neurons projecting to the ventral hippocampus to aversive stimuli","authors":"Hinako Morishita , Harune Hori , Hiroyuki Kawai , Hisashi Shirakawa , Hitoshi Hashimoto , Kazuki Nagayasu","doi":"10.1016/j.jphs.2025.12.003","DOIUrl":"10.1016/j.jphs.2025.12.003","url":null,"abstract":"<div><div>Appropriate processing of aversive information is essential for survival. We previously demonstrated that serotonin neurons in the median raphe nucleus (MRN) play a key role in such processing; however, MRN responses to predictive cues of aversive events remain unclear. Here, we found that the MRN serotonin neurons were activated by aversive air-puff stimuli but not by auditory cues predicting the air puff. Moreover, delayed activation of the ventral hippocampus-projecting MRN serotonin neurons, together with subsequent 5-HT<sub>2A</sub> receptor signaling, was required for aversion. These findings shed light on the roles of the hippocampus-projecting MRN serotonin neurons and elucidate their molecular mechanisms.</div></div>","PeriodicalId":16786,"journal":{"name":"Journal of pharmacological sciences","volume":"160 2","pages":"Pages 91-96"},"PeriodicalIF":2.9,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145711963","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pericytes are perivascular cells that contribute to maintaining vascular integrity and central nervous system homeostasis. β2-microglobulin (B2M) is a component of the major histocompatibility complex class I molecule; it has recently been implicated in age-related and injury-associated inflammation. Here, we investigated the phenotypic and transcriptomic effects of B2M on mouse brain pericytes in vitro. B2M treatment increased Bromodeoxyuridine (BrdU) incorporation into the cultured pericytes as well as the number of Ki67-positive pericytes. Morphologically, B2M promoted pericyte extension. Toll-like receptor 4 (TLR4), one of the key molecules that regulates B2M function, was involved in the B2M-dependent pericyte proliferation. These findings were consistent with RNA-seq results showing differential expression of genes related to cell proliferation. These results suggest that B2M directly acts on pericytes and regulates part of their functional responses through TLR4 signaling.
{"title":"β2 microglobulin promotes pericyte proliferation through toll-like receptor 4","authors":"Yoshino Yonezu , Akiko Uyeda , Hidemi Misawa , Rieko Muramatsu","doi":"10.1016/j.jphs.2025.12.001","DOIUrl":"10.1016/j.jphs.2025.12.001","url":null,"abstract":"<div><div>Pericytes are perivascular cells that contribute to maintaining vascular integrity and central nervous system homeostasis. β<sub>2</sub>-microglobulin (B2M) is a component of the major histocompatibility complex class I molecule; it has recently been implicated in age-related and injury-associated inflammation. Here, we investigated the phenotypic and transcriptomic effects of B2M on mouse brain pericytes <em>in vitro</em>. B2M treatment increased Bromodeoxyuridine (BrdU) incorporation into the cultured pericytes as well as the number of Ki67-positive pericytes. Morphologically, B2M promoted pericyte extension. Toll-like receptor 4 (TLR4), one of the key molecules that regulates B2M function, was involved in the B2M-dependent pericyte proliferation. These findings were consistent with RNA-seq results showing differential expression of genes related to cell proliferation. These results suggest that B2M directly acts on pericytes and regulates part of their functional responses through TLR4 signaling.</div></div>","PeriodicalId":16786,"journal":{"name":"Journal of pharmacological sciences","volume":"160 1","pages":"Pages 81-89"},"PeriodicalIF":2.9,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145733260","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-02DOI: 10.1016/j.jphs.2025.11.007
Olesia F. Moroz , Viktoriia I. Kravchenko , Alexander V. Zholos
Essential oils have long been recognized for their therapeutic potential, with growing interest in their molecular mechanisms of action in neurological health. Among emerging targets, polymodal Ca2+-permeable Transient Receptor Potential (TRP) cation channels have gained particular attention for their roles in neuronal signaling, synaptic plasticity, and modulation of cognitive, neurodegenerative, and psychological disorders. This review explores the ability of essential oil constituents to modulate TRP channels. The major channels to be discussed here include TRPV1, TRPM8, and TRPA1 and some other TRPVs and TRPMs. Some TRPC members have also been reviewed, albeit more briefly. Key bioactive compounds – including menthol, linalool, and eugenol – are highlighted for their ability to interact with TRP channels, while influencing neurophysiological pathways related to learning, memory, neuroinflammation, and emotional regulation. Preclinical evidence suggests these interactions may offer neuroprotective, anxiolytic, and antidepressant effects. However, challenges such as bioavailability, standardization, and safety remain barriers to clinical translation. This review underscores the therapeutic promise of essential oil constituents as modulators of TRP channels and outlines future directions for their integration into neurotherapeutic strategies.
{"title":"Constituents of essential oils as modulators of TRP channels: Focus on cognitive functions, neurodegenerative, and psychological diseases","authors":"Olesia F. Moroz , Viktoriia I. Kravchenko , Alexander V. Zholos","doi":"10.1016/j.jphs.2025.11.007","DOIUrl":"10.1016/j.jphs.2025.11.007","url":null,"abstract":"<div><div>Essential oils have long been recognized for their therapeutic potential, with growing interest in their molecular mechanisms of action in neurological health. Among emerging targets, polymodal Ca<sup>2+</sup>-permeable Transient Receptor Potential (TRP) cation channels have gained particular attention for their roles in neuronal signaling, synaptic plasticity, and modulation of cognitive, neurodegenerative, and psychological disorders. This review explores the ability of essential oil constituents to modulate TRP channels. The major channels to be discussed here include TRPV1, TRPM8, and TRPA1 and some other TRPVs and TRPMs. Some TRPC members have also been reviewed, albeit more briefly. Key bioactive compounds – including menthol, linalool, and eugenol – are highlighted for their ability to interact with TRP channels, while influencing neurophysiological pathways related to learning, memory, neuroinflammation, and emotional regulation. Preclinical evidence suggests these interactions may offer neuroprotective, anxiolytic, and antidepressant effects. However, challenges such as bioavailability, standardization, and safety remain barriers to clinical translation. This review underscores the therapeutic promise of essential oil constituents as modulators of TRP channels and outlines future directions for their integration into neurotherapeutic strategies.</div></div>","PeriodicalId":16786,"journal":{"name":"Journal of pharmacological sciences","volume":"160 1","pages":"Pages 69-80"},"PeriodicalIF":2.9,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145681539","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-29DOI: 10.1016/j.jphs.2025.11.006
Yasushi Yabuki , Norifumi Shioda
Neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease are on the rise in super-aging societies. However, the mechanisms underlying the aggregation and propagation of prion-like proteins such as α-synuclein and Tau that contribute to the pathogenesis of neurodegeneration remain poorly understood. Although prion-like proteins are known to undergo liquid–liquid phase separation (LLPS) followed by a sol–gel transition in vitro, the key factors governing their phase transition remain to be elucidated in vivo. Most prion-like proteins are classified as RNA-binding proteins, and recent studies suggest that RNA plays a critical role in mediating both LLPS and the subsequent sol–gel transition of these proteins. In the review, we summarized our findings on RNA G-quadruplexes (rG4s) as a pathological key molecule in neurodegenerative disorders and introduce recent advances in RNA-induced phase transition of prion-like proteins.
神经退行性疾病,如阿尔茨海默病和帕金森病,在超老龄化社会中呈上升趋势。然而,朊病毒样蛋白(如α-突触核蛋白和Tau)聚集和繁殖的机制仍不清楚,这些蛋白与神经变性的发病机制有关。虽然已知朊病毒样蛋白在体外经历液-液相分离(LLPS),然后是溶胶-凝胶转变,但控制其相变的关键因素仍有待于在体内阐明。大多数朊病毒样蛋白被归类为RNA结合蛋白,最近的研究表明RNA在介导LLPS和随后的这些蛋白的溶胶-凝胶转化中起着关键作用。本文综述了RNA g -四重复合物(rG4s)作为神经退行性疾病病理关键分子的研究进展,并介绍了RNA诱导朊蛋白样蛋白相变的最新进展。
{"title":"RNA-mediated aggregation mechanism of prion-like proteins and its application to drug discovery","authors":"Yasushi Yabuki , Norifumi Shioda","doi":"10.1016/j.jphs.2025.11.006","DOIUrl":"10.1016/j.jphs.2025.11.006","url":null,"abstract":"<div><div>Neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease are on the rise in super-aging societies. However, the mechanisms underlying the aggregation and propagation of prion-like proteins such as α-synuclein and Tau that contribute to the pathogenesis of neurodegeneration remain poorly understood. Although prion-like proteins are known to undergo liquid–liquid phase separation (LLPS) followed by a sol–gel transition <em>in vitro</em>, the key factors governing their phase transition remain to be elucidated <em>in vivo</em>. Most prion-like proteins are classified as RNA-binding proteins, and recent studies suggest that RNA plays a critical role in mediating both LLPS and the subsequent sol–gel transition of these proteins. In the review, we summarized our findings on RNA G-quadruplexes (rG4s) as a pathological key molecule in neurodegenerative disorders and introduce recent advances in RNA-induced phase transition of prion-like proteins.</div></div>","PeriodicalId":16786,"journal":{"name":"Journal of pharmacological sciences","volume":"160 1","pages":"Pages 64-68"},"PeriodicalIF":2.9,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145681538","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}