Pub Date : 2026-01-13DOI: 10.1007/s11481-025-10271-w
Hossam A Raslan, Haidy E Michel, Esther T Menze, Amira A El-Gazar
Repetitive traumatic brain injury (RTBI) refers to brain injuries resulting from an external mechanical force causing cumulative and frequently severe neurological consequences. This study aimed to explore the neuroprotective effect of alogliptin (ALO) on RTBI-provoked endoplasmic reticulum (ER) stress and investigate the potential underlying mechanisms. For RTBI induction, rats were exposed to a sharp-edged weight at the right interior frontal area of the right cortex, one drop per day for five successive days. ALO (20 mg/kg/day, p.o.) was administered for one week. Results depicted that ALO recovered motor abnormalities and enhanced motor coordination in the open field test, decreased immobility and increased climbing time in the forced swimming test, and corrected histological aberrations. Moreover, ALO counteracted RTBI-triggered ER stress via suppression of activating transcription factor 6 (ATF6), glucose-regulated protein 78 (GRP78), aggregation of β-amyloid and Tau proteins, as well as elevation of the cortical content of brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin receptor kinase B (TrKB). ALO also exhibited an antioxidant and anti-inflammatory potential in addition to its effect on the gene expression of miRNAs (miRNA-322 and miRNA-125b). In conclusion, ALO exhibited a neuroprotective effect by mitigating ER stress induced in an RTBI rat model.
{"title":"Repositioning of Alogliptin to Mitigate Secondary Injury Induced by Repetitive TBI: Potential Role of its Antioxidant and Anti- Inflammatory Effects.","authors":"Hossam A Raslan, Haidy E Michel, Esther T Menze, Amira A El-Gazar","doi":"10.1007/s11481-025-10271-w","DOIUrl":"10.1007/s11481-025-10271-w","url":null,"abstract":"<p><p>Repetitive traumatic brain injury (RTBI) refers to brain injuries resulting from an external mechanical force causing cumulative and frequently severe neurological consequences. This study aimed to explore the neuroprotective effect of alogliptin (ALO) on RTBI-provoked endoplasmic reticulum (ER) stress and investigate the potential underlying mechanisms. For RTBI induction, rats were exposed to a sharp-edged weight at the right interior frontal area of the right cortex, one drop per day for five successive days. ALO (20 mg/kg/day, p.o.) was administered for one week. Results depicted that ALO recovered motor abnormalities and enhanced motor coordination in the open field test, decreased immobility and increased climbing time in the forced swimming test, and corrected histological aberrations. Moreover, ALO counteracted RTBI-triggered ER stress via suppression of activating transcription factor 6 (ATF6), glucose-regulated protein 78 (GRP78), aggregation of β-amyloid and Tau proteins, as well as elevation of the cortical content of brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin receptor kinase B (TrKB). ALO also exhibited an antioxidant and anti-inflammatory potential in addition to its effect on the gene expression of miRNAs (miRNA-322 and miRNA-125b). In conclusion, ALO exhibited a neuroprotective effect by mitigating ER stress induced in an RTBI rat model.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"6"},"PeriodicalIF":3.5,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12799724/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145960749","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sevoflurane (Sevo) anaesthesia in children is linked to an increased incidence of postoperative emergence agitation (EA) and potential neurotoxicity in developing brains. However, the specific risks of subanaesthetic foetal or neonatal exposure to Sevo remain unclear. This study evaluates the safety and efficacy of combining dexmedetomidine (Dex) with Sevo to manage EA in paediatric anaesthesia. A systematic review and meta-analysis of randomized controlled clinical trials involving children under 8 years old revealed that Dex significantly reduces EA incidence when administered via intravenous, perineural, and intranasal routes. Using in utero electroporation, we found that pregnant mice exposed to 2.5% Sevo at embryonic days 14.5 and 15.5 exhibited transient neuronal migration deficits, with 25% of neurons delayed in deeper cortical layers. However, these neurons migrated to the cortex by postnatal day 8. Neonatal mice exposed to 2.5% Sevo experienced a 10% reduction in dendritic spine density in adolescence, associated with impaired somatosensory function, as assessed by the Von Frey test. Remarkably, Dex pretreatment ameliorated these pathological and functional changes. Thus, foetal or neonatal Sevo exposure can delay neuronal migration and reduce dendritic spine density. Dex co-administration effectively mitigates these adverse outcomes, supporting its potential use in paediatric anaesthesia to protect developing brains.
{"title":"Dexmedetomidine Mitigates Sevoflurane-Induced Neurodevelopmental Effects in Paediatric Anaesthesia: A Meta-Analysis and Preclinical Study.","authors":"Hsuan-Chih Lao, Chia-Wei Huang, Ssu-Han Wang, Yen-Lin Su, Chien-Hui Chang, Cheng-Yen Liao, Jen-Chieh Wu, Ying-Chun Lin, Jin-Wu Tsai","doi":"10.1007/s11481-025-10273-8","DOIUrl":"10.1007/s11481-025-10273-8","url":null,"abstract":"<p><p>Sevoflurane (Sevo) anaesthesia in children is linked to an increased incidence of postoperative emergence agitation (EA) and potential neurotoxicity in developing brains. However, the specific risks of subanaesthetic foetal or neonatal exposure to Sevo remain unclear. This study evaluates the safety and efficacy of combining dexmedetomidine (Dex) with Sevo to manage EA in paediatric anaesthesia. A systematic review and meta-analysis of randomized controlled clinical trials involving children under 8 years old revealed that Dex significantly reduces EA incidence when administered via intravenous, perineural, and intranasal routes. Using in utero electroporation, we found that pregnant mice exposed to 2.5% Sevo at embryonic days 14.5 and 15.5 exhibited transient neuronal migration deficits, with 25% of neurons delayed in deeper cortical layers. However, these neurons migrated to the cortex by postnatal day 8. Neonatal mice exposed to 2.5% Sevo experienced a 10% reduction in dendritic spine density in adolescence, associated with impaired somatosensory function, as assessed by the Von Frey test. Remarkably, Dex pretreatment ameliorated these pathological and functional changes. Thus, foetal or neonatal Sevo exposure can delay neuronal migration and reduce dendritic spine density. Dex co-administration effectively mitigates these adverse outcomes, supporting its potential use in paediatric anaesthesia to protect developing brains.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"5"},"PeriodicalIF":3.5,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12779744/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145914028","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-03DOI: 10.1007/s11481-025-10270-x
Mennatallah Ibrahim, Asmaa M Khalil, Heba Attia, Saleh Alseekh, Ahmed F Mohamed, Mohammed F El-Yamany
Tauopathies are neurodegenerative diseases characterized by accumulation of hyperphosphorylated tau protein (P-tau). The gut microbiota (GM) is symbiotic with the host and altered in neurodegenerative diseases. Amitriptyline (AMI) is a functional inhibitor of acid sphingomyelinase (ASM) which is abnormally highly expressed in brains of Alzheimer patients. Little data is known about the role of colonic ASM in management of tauopathy. Therefore, the aim of this study was to investigate the role of AMI on reversing gut dysbiosis, ceramide levels, colonic inflammation and intestinal barrier disruption in tauopathy through the bidirectional gut-brain axis. P301S transgenic mice were administered AMI for 35 days. Colonic ASM, ceramides, inflammation and membrane integrity were assessed besides fecal microbiome analysis and serum lipopolysaccharides to assess intestinal membrane disruption. Levels of hippocampal P-tau, protein phosphatase 2 A and neurogenesis were assessed along with cognitive behavior. AMI treatment significantly reduced colonic ASM, ceramide levels, increased abundance of Harryflintia, Dubosiella, and Parasutterella and decreased abundance of Lactobacillus, Lachnoclostridium, Oscillibacter, Oscillospiracea UCG-003, Colidextribacter, Roseburia, Butyricicoccus, and Sphingomondales. In contrast, P301S mice displayed an altered GM profile with enriched Firmicutes and Clostridia, and low proportions of Bacteroidota- a phylum associated with intestinal barrier protection-, and Ruminococcaceae. Also, AMI treatment decreased inflammation and restored colonic membrane integrity with subsequent decrease in serum lipopolysaccharides, P-tau in hippocampus and improvement in cognitive behaviour and neurogenesis. The current results indicate that AMI has neuroprotective effects against tauopathy through modulation of ASM activity, associated ceramide levels, GM composition, colonic inflammation and membrane integrity through bidirectional gut-brain axis.
{"title":"Gut Microbiome-Sphingolipid Metabolism-Brain Axis Interactions: Neuroprotective Effects of Amitriptyline as Functional Inhibitor of Acid Sphingomyelinase in a Mouse Model of Tauopathy.","authors":"Mennatallah Ibrahim, Asmaa M Khalil, Heba Attia, Saleh Alseekh, Ahmed F Mohamed, Mohammed F El-Yamany","doi":"10.1007/s11481-025-10270-x","DOIUrl":"10.1007/s11481-025-10270-x","url":null,"abstract":"<p><p>Tauopathies are neurodegenerative diseases characterized by accumulation of hyperphosphorylated tau protein (P-tau). The gut microbiota (GM) is symbiotic with the host and altered in neurodegenerative diseases. Amitriptyline (AMI) is a functional inhibitor of acid sphingomyelinase (ASM) which is abnormally highly expressed in brains of Alzheimer patients. Little data is known about the role of colonic ASM in management of tauopathy. Therefore, the aim of this study was to investigate the role of AMI on reversing gut dysbiosis, ceramide levels, colonic inflammation and intestinal barrier disruption in tauopathy through the bidirectional gut-brain axis. P301S transgenic mice were administered AMI for 35 days. Colonic ASM, ceramides, inflammation and membrane integrity were assessed besides fecal microbiome analysis and serum lipopolysaccharides to assess intestinal membrane disruption. Levels of hippocampal P-tau, protein phosphatase 2 A and neurogenesis were assessed along with cognitive behavior. AMI treatment significantly reduced colonic ASM, ceramide levels, increased abundance of Harryflintia, Dubosiella, and Parasutterella and decreased abundance of Lactobacillus, Lachnoclostridium, Oscillibacter, Oscillospiracea UCG-003, Colidextribacter, Roseburia, Butyricicoccus, and Sphingomondales. In contrast, P301S mice displayed an altered GM profile with enriched Firmicutes and Clostridia, and low proportions of Bacteroidota- a phylum associated with intestinal barrier protection-, and Ruminococcaceae. Also, AMI treatment decreased inflammation and restored colonic membrane integrity with subsequent decrease in serum lipopolysaccharides, P-tau in hippocampus and improvement in cognitive behaviour and neurogenesis. The current results indicate that AMI has neuroprotective effects against tauopathy through modulation of ASM activity, associated ceramide levels, GM composition, colonic inflammation and membrane integrity through bidirectional gut-brain axis.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"3"},"PeriodicalIF":3.5,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12764700/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145897151","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Maternal immune activation (MIA) during pregnancy has been implicated as a key environmental risk factor in autism spectrum disorder (ASD). Interferon-alpha (IFN-α), a type I interferon, may disrupt fetal neurodevelopment, yet its mechanistic impact remains insufficiently understood. This study explores the effects of maternal IFN-α exposure on neurobehavioral and neurobiological outcomes in a Wistar rat model. Pregnant rats received IFN-α on gestational day 10, and offspring were evaluated through behavioral assays, neurochemical analyses, and histopathological assessments. IFN-α exposure resulted in significant reductions in GABA, 5-HIAA, and GAD-67 levels, particularly in male offspring, indicating neurotransmitter dysregulation. Histologically, neuronal loss was observed in the hippocampal CA1 and CA3 regions and cerebellar Purkinje cells. Astrocyte activation, reflected by increased GFAP immunoreactivity, was prominent, suggesting a neuroinflammatory response. Additionally, reduced brain-derived neurotrophic factor (BDNF) and elevated tumor necrosis factor-alpha (TNF-α) levels support the presence of inflammation-induced synaptic dysfunction and impaired neuroplasticity. Behaviorally, male offspring exhibited reduced sociability and impaired social novelty recognition. Both sexes demonstrated deficits in motor coordination and exploratory activity. These findings align with core ASD phenotypes and underscore a heightened male vulnerability. Overall, the study provides compelling evidence that prenatal IFN-α exposure leads to persistent neuroimmune, neurochemical, and structural alterations resembling ASD. The results highlight the need for further research into immune-mediated neurodevelopmental disruptions and sex-specific vulnerabilities, offering potential pathways for preventive and therapeutic interventions targeting MIA-related risk mechanisms.
{"title":"Prenatal Interferon-Alpha Exposure Induces Autism-Like Neurobehavioral and Neurochemical Alterations in Male Offspring.","authors":"Güliz Otkıran, Mümin Alper Erdoğan, Yiğit Uyanıkgil, Oytun Erbaş","doi":"10.1007/s11481-025-10275-6","DOIUrl":"10.1007/s11481-025-10275-6","url":null,"abstract":"<p><p>Maternal immune activation (MIA) during pregnancy has been implicated as a key environmental risk factor in autism spectrum disorder (ASD). Interferon-alpha (IFN-α), a type I interferon, may disrupt fetal neurodevelopment, yet its mechanistic impact remains insufficiently understood. This study explores the effects of maternal IFN-α exposure on neurobehavioral and neurobiological outcomes in a Wistar rat model. Pregnant rats received IFN-α on gestational day 10, and offspring were evaluated through behavioral assays, neurochemical analyses, and histopathological assessments. IFN-α exposure resulted in significant reductions in GABA, 5-HIAA, and GAD-67 levels, particularly in male offspring, indicating neurotransmitter dysregulation. Histologically, neuronal loss was observed in the hippocampal CA1 and CA3 regions and cerebellar Purkinje cells. Astrocyte activation, reflected by increased GFAP immunoreactivity, was prominent, suggesting a neuroinflammatory response. Additionally, reduced brain-derived neurotrophic factor (BDNF) and elevated tumor necrosis factor-alpha (TNF-α) levels support the presence of inflammation-induced synaptic dysfunction and impaired neuroplasticity. Behaviorally, male offspring exhibited reduced sociability and impaired social novelty recognition. Both sexes demonstrated deficits in motor coordination and exploratory activity. These findings align with core ASD phenotypes and underscore a heightened male vulnerability. Overall, the study provides compelling evidence that prenatal IFN-α exposure leads to persistent neuroimmune, neurochemical, and structural alterations resembling ASD. The results highlight the need for further research into immune-mediated neurodevelopmental disruptions and sex-specific vulnerabilities, offering potential pathways for preventive and therapeutic interventions targeting MIA-related risk mechanisms.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"4"},"PeriodicalIF":3.5,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145897156","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-02DOI: 10.1007/s11481-025-10269-4
Kamini R Shirasath, Kartik T Nakhate, Sameer N Goyal, Sanjay N Awathale
Although transient receptor potential melastatin 3 (TRPM3) channels are primarily known for their role in spinal nociception, emerging evidence suggests their involvement in psychiatric conditions and central reward processing. Menopause, characterized by estrogen decline, induces neuroimmune activation and increases pro-inflammatory factors such as ciliary neurotrophic factor (CNTF). While a direct regulatory effect of CNTF on TRPM3 is not well established, both are involved in inflammation-related signaling, suggesting potential crosstalk. TRPM3 responds to neuroinflammatory and neurotrophic signals and may contribute to postmenopausal cognitive decline. However, this link remains unexplored. Naringenin, a natural flavonoid with estrogen-like properties, has been reported to inhibit TRPM3 channels and may help to alleviate postmenopausal memory impairment. This study aimed to investigate the role of elevated CNTF levels in increasing TRPM3 expression in the dentate gyrus (DG), contributing to cognitive deficits, and to assess the potential of naringenin in reversing these effects. Bilateral ovariectomy (OVX) was performed on female Sprague-Dawley rats, followed by treatment with naringenin (2.5, 5 and 10 mg/kg, intraperitoneal) for 14 days. Cognitive functions were assessed using the novel object recognition and passive avoidance tests. CNTF levels in the plasma and the DG, along with TRPM3 expression in the DG, were measured using ELISA and immunohistochemistry, respectively. Dendritic arborization in DG neurons was analyzed using Golgi-Cox staining. OVX rats showed impaired cognition, elevated CNTF and TRPM3 expression, and reduced dendritic complexity. Naringenin treatment reversed these changes, suggesting its potential to improve postmenopausal cognitive decline by modulating CNTF levels and TRPM3 activity in the DG.
{"title":"Modulation of Cognitive Function by TRPM3 Channels in the Dentate Gyrus of a Menopausal Rat Model: Effects of Naringenin Treatment and Ciliary Neurotrophic Factor.","authors":"Kamini R Shirasath, Kartik T Nakhate, Sameer N Goyal, Sanjay N Awathale","doi":"10.1007/s11481-025-10269-4","DOIUrl":"10.1007/s11481-025-10269-4","url":null,"abstract":"<p><p>Although transient receptor potential melastatin 3 (TRPM3) channels are primarily known for their role in spinal nociception, emerging evidence suggests their involvement in psychiatric conditions and central reward processing. Menopause, characterized by estrogen decline, induces neuroimmune activation and increases pro-inflammatory factors such as ciliary neurotrophic factor (CNTF). While a direct regulatory effect of CNTF on TRPM3 is not well established, both are involved in inflammation-related signaling, suggesting potential crosstalk. TRPM3 responds to neuroinflammatory and neurotrophic signals and may contribute to postmenopausal cognitive decline. However, this link remains unexplored. Naringenin, a natural flavonoid with estrogen-like properties, has been reported to inhibit TRPM3 channels and may help to alleviate postmenopausal memory impairment. This study aimed to investigate the role of elevated CNTF levels in increasing TRPM3 expression in the dentate gyrus (DG), contributing to cognitive deficits, and to assess the potential of naringenin in reversing these effects. Bilateral ovariectomy (OVX) was performed on female Sprague-Dawley rats, followed by treatment with naringenin (2.5, 5 and 10 mg/kg, intraperitoneal) for 14 days. Cognitive functions were assessed using the novel object recognition and passive avoidance tests. CNTF levels in the plasma and the DG, along with TRPM3 expression in the DG, were measured using ELISA and immunohistochemistry, respectively. Dendritic arborization in DG neurons was analyzed using Golgi-Cox staining. OVX rats showed impaired cognition, elevated CNTF and TRPM3 expression, and reduced dendritic complexity. Naringenin treatment reversed these changes, suggesting its potential to improve postmenopausal cognitive decline by modulating CNTF levels and TRPM3 activity in the DG.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"1"},"PeriodicalIF":3.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145893550","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-02DOI: 10.1007/s11481-025-10272-9
Ying Zhu, Benqiao Wang, Chunxiang Hu, Ruixia Zhu
Generalized myasthenia gravis (gMG) is an antibody mediated autoimmune neuromuscular junction disorder characterized by muscle weakness and fatigue as well as acetylcholine receptor antibody (AChR-Ab) as the main presence. A proportion of patients fail to achieve minimal symptom expression (MSE), furthermore 10-20% of them develop into refractory under conventional immunotherapy. We conducted a retrospective study to explore the effectiveness and safety of telitacicept in gMG and thymoma-associated MG (TAMG) patients. The treatment response was assessed by the variation of QMG, MG-ADL and MG-QOL-15 scores. Time to MSE as well as usage of corticosteroid were also evaluated. In this retrospective study, we included 22 AChR-gMG patients (15 women, 7 men), including 7 refractory and 12 TAMG, who were treated with telitacicept by following-up at least 6 months. Compared to the baseline, a significant decrease in QMG, ADL and MG-QOL-15 scores was observed at every visit, especially for the QMG score with at least 3 points decline in all the patients in week 4. Twenty patients attained MSE and the time to MSE was 4 months during the observed period. At the last follow-up, the dose of prednisone of all the patients treated with telitacicept was ≤ 5 mg/d. The AChR-Ab titers and CD19+ B cells significantly decreased from baseline to week 24. Telitacicept is generally well tolerated, the most common (18%) adverse effect was mild and transient injected site swelling. Our study provides evidence to support that telitacicept is beneficial and well tolerated in the management of gMG especially in refractory MG and TAMG. Clinical outcomes showed increased efficacy of telitacicept when used earlier in the disease course, which leads to a sparing of prednisone.
{"title":"Telitacicept as a New Therapeutic Avenue for Generalized Myasthenia Gravis and Thymoma-Associated Myasthenia Gravis.","authors":"Ying Zhu, Benqiao Wang, Chunxiang Hu, Ruixia Zhu","doi":"10.1007/s11481-025-10272-9","DOIUrl":"https://doi.org/10.1007/s11481-025-10272-9","url":null,"abstract":"<p><p>Generalized myasthenia gravis (gMG) is an antibody mediated autoimmune neuromuscular junction disorder characterized by muscle weakness and fatigue as well as acetylcholine receptor antibody (AChR-Ab) as the main presence. A proportion of patients fail to achieve minimal symptom expression (MSE), furthermore 10-20% of them develop into refractory under conventional immunotherapy. We conducted a retrospective study to explore the effectiveness and safety of telitacicept in gMG and thymoma-associated MG (TAMG) patients. The treatment response was assessed by the variation of QMG, MG-ADL and MG-QOL-15 scores. Time to MSE as well as usage of corticosteroid were also evaluated. In this retrospective study, we included 22 AChR-gMG patients (15 women, 7 men), including 7 refractory and 12 TAMG, who were treated with telitacicept by following-up at least 6 months. Compared to the baseline, a significant decrease in QMG, ADL and MG-QOL-15 scores was observed at every visit, especially for the QMG score with at least 3 points decline in all the patients in week 4. Twenty patients attained MSE and the time to MSE was 4 months during the observed period. At the last follow-up, the dose of prednisone of all the patients treated with telitacicept was ≤ 5 mg/d. The AChR-Ab titers and CD19<sup>+</sup> B cells significantly decreased from baseline to week 24. Telitacicept is generally well tolerated, the most common (18%) adverse effect was mild and transient injected site swelling. Our study provides evidence to support that telitacicept is beneficial and well tolerated in the management of gMG especially in refractory MG and TAMG. Clinical outcomes showed increased efficacy of telitacicept when used earlier in the disease course, which leads to a sparing of prednisone.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"2"},"PeriodicalIF":3.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145893540","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-19DOI: 10.1007/s11481-025-10256-9
Danielle Germundson-Hermanson, Marilyn G Klug, Kumi Nagamoto-Combs
Mood and behavior-related comorbidities are often reported with food allergies, an atopic condition that elevates histamine (HA) levels in tissues and circulation. However, whether allergy-induced HA directly affects the central nervous system is unclear. Previously, we demonstrated that the levels of HA and its receptor subtype, H3 receptor (H3R), were elevated in the brains of mice with subclinical cow's milk allergy (CMA) generated by sensitizing C57BL/6J mice to a bovine whey allergen, β-lactoglobulin (BLG, Bos d 5). Furthermore, these BLG-sensitized CMA mice showed depression-like behavior associated with mast cell activation, neuroinflammation, and cortical demyelination, leading us to postulate that peripheral immune responses raised brain HA and dysregulated the neuronal histaminergic system. Hypothesizing that the autoregulatory function of H3R signaling is pivotal in eliciting altered behavior and neuropathologies, we investigated whether thioperamide, a brain-permeable H3R-selective antagonist, would attenuate the changes observed in CMA mice. Male and female CMA mice were fed a whey-containing diet for 2 weeks without or with thioperamide. While sensorimotor functions were not impaired in CMA mice of either sex, some aspects of affective and cognitive behaviors were significantly altered in males. Male CMA mice also showed more IgE-immunopositive, degranulated mast cells in the dura mater than females, regardless of thioperamide treatment. Importantly, thioperamide reduced CMA-associated behavioral and neuropathological changes in male mice, although it also uniquely affected female mice. Our results suggest that thioperamide ameliorates CMA-associated behavioral changes and neuropathologies via H3R inhibition in a sex-dependent manner.
情绪和行为相关的合并症通常与食物过敏一起报道,食物过敏是一种升高组织和循环中组胺(HA)水平的特应性疾病。然而,过敏诱导的血凝素是否直接影响中枢神经系统尚不清楚。先前,我们证明了HA及其受体亚型H3受体(H3R)的水平在C57BL/6J小鼠对牛乳清过敏原β-乳球蛋白(BLG, bod 5)致敏后产生的亚临床牛奶过敏(CMA)小鼠的大脑中升高。此外,这些blg致敏的CMA小鼠表现出与肥大细胞激活、神经炎症和皮质脱髓鞘相关的抑郁样行为,这使我们假设外周免疫反应提高了脑HA并调节了神经元组胺能系统。假设H3R信号的自调节功能在引发行为改变和神经病理中起关键作用,我们研究了硫哌丁胺,一种脑渗透性H3R选择性拮抗剂,是否会减弱CMA小鼠中观察到的变化。雄性和雌性CMA小鼠分别饲喂不含硫哌丁胺或含硫哌丁胺的含乳清饲料2周。虽然CMA小鼠的感觉运动功能没有受到损害,但雄性的情感和认知行为的某些方面却发生了显著改变。无论硫哌丁胺治疗,雄性CMA小鼠的硬脑膜中也显示出更多的ige免疫阳性、脱颗粒肥大细胞。重要的是,硫哌丁胺减少了雄性小鼠与cma相关的行为和神经病理变化,尽管它也只影响雌性小鼠。我们的研究结果表明,硫哌丁胺通过抑制H3R以性别依赖的方式改善cma相关的行为改变和神经病理。
{"title":"Histamine H3 Receptor Antagonist, Thioperamide, Improves Behavioral and Neuropathological Changes Associated with Subclinical Hypersensitivity to a Cow's Milk Allergen.","authors":"Danielle Germundson-Hermanson, Marilyn G Klug, Kumi Nagamoto-Combs","doi":"10.1007/s11481-025-10256-9","DOIUrl":"10.1007/s11481-025-10256-9","url":null,"abstract":"<p><p>Mood and behavior-related comorbidities are often reported with food allergies, an atopic condition that elevates histamine (HA) levels in tissues and circulation. However, whether allergy-induced HA directly affects the central nervous system is unclear. Previously, we demonstrated that the levels of HA and its receptor subtype, H3 receptor (H3R), were elevated in the brains of mice with subclinical cow's milk allergy (CMA) generated by sensitizing C57BL/6J mice to a bovine whey allergen, β-lactoglobulin (BLG, Bos d 5). Furthermore, these BLG-sensitized CMA mice showed depression-like behavior associated with mast cell activation, neuroinflammation, and cortical demyelination, leading us to postulate that peripheral immune responses raised brain HA and dysregulated the neuronal histaminergic system. Hypothesizing that the autoregulatory function of H3R signaling is pivotal in eliciting altered behavior and neuropathologies, we investigated whether thioperamide, a brain-permeable H3R-selective antagonist, would attenuate the changes observed in CMA mice. Male and female CMA mice were fed a whey-containing diet for 2 weeks without or with thioperamide. While sensorimotor functions were not impaired in CMA mice of either sex, some aspects of affective and cognitive behaviors were significantly altered in males. Male CMA mice also showed more IgE-immunopositive, degranulated mast cells in the dura mater than females, regardless of thioperamide treatment. Importantly, thioperamide reduced CMA-associated behavioral and neuropathological changes in male mice, although it also uniquely affected female mice. Our results suggest that thioperamide ameliorates CMA-associated behavioral changes and neuropathologies via H3R inhibition in a sex-dependent manner.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"110"},"PeriodicalIF":3.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12717242/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145795630","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-12DOI: 10.1007/s11481-025-10268-5
Lily Mohammadipoor-Ghasemabad, Khadijeh Esmaeilpour, Manzumeh Shamsi Meymandi, Farhad Iranmanesh, Sheida Amiri Khorasani, Vahid Sheibani, Farahnaz Taheri
Tramadol (TM) abuse negatively affects the central nervous system, especially brain regions like the hippocampus involved in cognition. Recent studies have demonstrated neuroprotective effects of Vitamin C (Vit C) in various neurological diseases. No study has yet examined the effects of Vit C on tramadol-induced synaptic plasticity impairment. Therefore, we aimed to investigate the neuroprotective effects of Vit C on cognitive performance and synaptic plasticity in tramadol-exposed rats. Fifty-two juvenile male rats (30 days old) were divided into four groups: TM (30 mg/kg/day, intraperitoneally in the first week, 40 mg/kg/day in the second week and 50 mg/kg/day in third and fourth weeks), Vit.C (200 mg/kg/day, orally for 4 weeks), TM + Vit.C (as in the TM and Vit C groups, Vit C administered half an hour prior to TM), and Ctrl (0.25 mL saline/day for 4 weeks). Behavioral tests (open field, Morris water maze, novel object recognition) assessed locomotor activity and memory. In vivo recordings evaluated synaptic plasticity, and hippocampal oxidative stress markers [malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), total antioxidant capacity (TAC)] were measured according to the manufacturers' protocols with ELISA. TM caused learning and memory deficits, reduced long-term potentiation (LTP) induction, and disrupted the oxidative stress balance in the hippocampus. In contrast, Vit C inhibited these changes. These findings suggest that Vit C can attenuate cognitive impairments associated with chronic TM consumption, likely through modulation of hippocampal oxidative stress and enhancement of LTP induction. Therefore, Vit C could be a promising candidate for further investigation as a potential therapeutic agent to mitigate cognitive dysfunction associated with TM use.
{"title":"Ameliorative Effect of Vitamin C Against Tramadol-Induced Learning and Memory Impairment in Juvenile Rat Via Attenuation of Oxidative Stress and Dysfunctional Synaptic Plasticity.","authors":"Lily Mohammadipoor-Ghasemabad, Khadijeh Esmaeilpour, Manzumeh Shamsi Meymandi, Farhad Iranmanesh, Sheida Amiri Khorasani, Vahid Sheibani, Farahnaz Taheri","doi":"10.1007/s11481-025-10268-5","DOIUrl":"https://doi.org/10.1007/s11481-025-10268-5","url":null,"abstract":"<p><p>Tramadol (TM) abuse negatively affects the central nervous system, especially brain regions like the hippocampus involved in cognition. Recent studies have demonstrated neuroprotective effects of Vitamin C (Vit C) in various neurological diseases. No study has yet examined the effects of Vit C on tramadol-induced synaptic plasticity impairment. Therefore, we aimed to investigate the neuroprotective effects of Vit C on cognitive performance and synaptic plasticity in tramadol-exposed rats. Fifty-two juvenile male rats (30 days old) were divided into four groups: TM (30 mg/kg/day, intraperitoneally in the first week, 40 mg/kg/day in the second week and 50 mg/kg/day in third and fourth weeks), Vit.C (200 mg/kg/day, orally for 4 weeks), TM + Vit.C (as in the TM and Vit C groups, Vit C administered half an hour prior to TM), and Ctrl (0.25 mL saline/day for 4 weeks). Behavioral tests (open field, Morris water maze, novel object recognition) assessed locomotor activity and memory. In vivo recordings evaluated synaptic plasticity, and hippocampal oxidative stress markers [malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), total antioxidant capacity (TAC)] were measured according to the manufacturers' protocols with ELISA. TM caused learning and memory deficits, reduced long-term potentiation (LTP) induction, and disrupted the oxidative stress balance in the hippocampus. In contrast, Vit C inhibited these changes. These findings suggest that Vit C can attenuate cognitive impairments associated with chronic TM consumption, likely through modulation of hippocampal oxidative stress and enhancement of LTP induction. Therefore, Vit C could be a promising candidate for further investigation as a potential therapeutic agent to mitigate cognitive dysfunction associated with TM use.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"109"},"PeriodicalIF":3.5,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145745990","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-09DOI: 10.1007/s11481-025-10260-z
Nahla E El-Ashmawy, Naglaa F Khedr, Nada N Helmy, Amera O Ibrahim
Multiple sclerosis (MS) is a long-lasting autoimmune condition characterized by myelin destruction and neurodegeneration. Research indicates that ferroptosis significantly influences MS pathogenesis, exacerbating neuronal tissue damage. Our study intended to explore the possible neuroprotective role of fisetin (FIS) in cuprizone (CPZ) model of MS and the associated molecular mechanisms. The 9-week experiment comprised a 5-week demyelination period in which C57BL/6 mice were provided with 0.2% w/w CPZ added to rodent chow, followed by a 4-week remyelination period in which mice were fed CPZ-free chow. FIS (80 mg/kg/day) was given by oral gavage to mice daily for 4 weeks starting in the 2nd week of demyelination. For remyelination, FIS was administered daily during the 4 weeks recovery. During demyelination, FIS significantly improved CPZ-induced behavioral and locomotor deficits, as demonstrated by tail suspension test and inverted screen grip strength test. LFB and H & E staining, MBP, GFAP and vimentin immunostaining revealed that FIS treatment significantly improved myelination, alleviated astrogliosis and neuronal injury in CPZ-fed mice throughout both phases. FIS attenuated ferroptosis and neuroinflammation during de- and remyelination as supported by reduced brain iron deposits, IL-1 β, MDA concentrations and restored GPX4. Moreover, FIS significantly downregulated NCOA4 and TfR1 gene expression and TfR1 protein level but upregulated FTH1 gene expression and ferritin protein level. Additionally, FIS upregulated Olig-1 during demyelination, but not remyelination. Fisetin has a potential neuroprotective effect in CPZ model of MS and can be studied as a promising adjuvant therapy to enhance remyelination and mitigate disability in MS patients possibly by modulating ferroptosis pathway.
{"title":"Fisetin Mitigates Ferroptosis and Promotes Remyelination in a Cuprizone Model of Multiple Sclerosis.","authors":"Nahla E El-Ashmawy, Naglaa F Khedr, Nada N Helmy, Amera O Ibrahim","doi":"10.1007/s11481-025-10260-z","DOIUrl":"10.1007/s11481-025-10260-z","url":null,"abstract":"<p><p>Multiple sclerosis (MS) is a long-lasting autoimmune condition characterized by myelin destruction and neurodegeneration. Research indicates that ferroptosis significantly influences MS pathogenesis, exacerbating neuronal tissue damage. Our study intended to explore the possible neuroprotective role of fisetin (FIS) in cuprizone (CPZ) model of MS and the associated molecular mechanisms. The 9-week experiment comprised a 5-week demyelination period in which C57BL/6 mice were provided with 0.2% w/w CPZ added to rodent chow, followed by a 4-week remyelination period in which mice were fed CPZ-free chow. FIS (80 mg/kg/day) was given by oral gavage to mice daily for 4 weeks starting in the 2nd week of demyelination. For remyelination, FIS was administered daily during the 4 weeks recovery. During demyelination, FIS significantly improved CPZ-induced behavioral and locomotor deficits, as demonstrated by tail suspension test and inverted screen grip strength test. LFB and H & E staining, MBP, GFAP and vimentin immunostaining revealed that FIS treatment significantly improved myelination, alleviated astrogliosis and neuronal injury in CPZ-fed mice throughout both phases. FIS attenuated ferroptosis and neuroinflammation during de- and remyelination as supported by reduced brain iron deposits, IL-1 β, MDA concentrations and restored GPX4. Moreover, FIS significantly downregulated NCOA4 and TfR1 gene expression and TfR1 protein level but upregulated FTH1 gene expression and ferritin protein level. Additionally, FIS upregulated Olig-1 during demyelination, but not remyelination. Fisetin has a potential neuroprotective effect in CPZ model of MS and can be studied as a promising adjuvant therapy to enhance remyelination and mitigate disability in MS patients possibly by modulating ferroptosis pathway.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"108"},"PeriodicalIF":3.5,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12685980/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145710436","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Diabetes mellitus exacerbates cerebral ischemic damage by potentiating neuroinflammation. We hypothesized that activation of the bradykinin type 2 receptor, a mediator of inflammation and vascular dynamics, might be detrimental to ischemic injury development in diabetic animals. We monitored the acute phase of cerebral ischemia in type 1 diabetic mice, diabetic bradykinin type 2 receptor knock-out mice, and their non-diabetic controls using neurological assessment, magnetic resonance imaging, and a comprehensive immuno-histochemical and morphological analysis to quantify changes in microglial, neutrophil, and neuronal populations. Our findings reveal that bradykinin type 2 receptor deficiency ameliorates neurological deficit in non-diabetic mice, despite similar ischemic lesion volumes across all investigated groups. Furthermore, in non-diabetic animals, the bradykinin type 2 receptor plays a discernible role in edema resolution, neuroprotection, and regulation of microglial response to ischemia. However, diabetes, as a stroke comorbidity, alters the involvement of the bradykinin type 2 receptor in ischemic injury development. Bradykinin type 2 receptor-deficient diabetic animals demonstrate delayed microglial cell loss and reduced microglial reactivity following ischemia compared to diabetic animals with functional bradykinin type 2 receptors. The attenuated immune response is accompanied by a marked absence of infiltrating neutrophils within the ischemic territory and improved neuronal survival. This study demonstrates that diabetes profoundly modifies the role of bradykinin type 2 receptor in cerebral ischemic injury, influencing both acute neuroinflammation and cell survival. These findings support the potential of the bradykinin type 2 receptor as a therapeutic target for stroke in diabetic population, warranting further investigation.
{"title":"Bradykinin Type 2 Receptor Deficiency Reshapes Acute Neuroinflammation and Improves Cell Survival after Ischemic Stroke in Diabetic Mice.","authors":"Anja Barić, Dinko Smilović, Helena Justić, Iva Šimunić, Siniša Škokić, Marina Dobrivojević Radmilović","doi":"10.1007/s11481-025-10267-6","DOIUrl":"https://doi.org/10.1007/s11481-025-10267-6","url":null,"abstract":"<p><p>Diabetes mellitus exacerbates cerebral ischemic damage by potentiating neuroinflammation. We hypothesized that activation of the bradykinin type 2 receptor, a mediator of inflammation and vascular dynamics, might be detrimental to ischemic injury development in diabetic animals. We monitored the acute phase of cerebral ischemia in type 1 diabetic mice, diabetic bradykinin type 2 receptor knock-out mice, and their non-diabetic controls using neurological assessment, magnetic resonance imaging, and a comprehensive immuno-histochemical and morphological analysis to quantify changes in microglial, neutrophil, and neuronal populations. Our findings reveal that bradykinin type 2 receptor deficiency ameliorates neurological deficit in non-diabetic mice, despite similar ischemic lesion volumes across all investigated groups. Furthermore, in non-diabetic animals, the bradykinin type 2 receptor plays a discernible role in edema resolution, neuroprotection, and regulation of microglial response to ischemia. However, diabetes, as a stroke comorbidity, alters the involvement of the bradykinin type 2 receptor in ischemic injury development. Bradykinin type 2 receptor-deficient diabetic animals demonstrate delayed microglial cell loss and reduced microglial reactivity following ischemia compared to diabetic animals with functional bradykinin type 2 receptors. The attenuated immune response is accompanied by a marked absence of infiltrating neutrophils within the ischemic territory and improved neuronal survival. This study demonstrates that diabetes profoundly modifies the role of bradykinin type 2 receptor in cerebral ischemic injury, influencing both acute neuroinflammation and cell survival. These findings support the potential of the bradykinin type 2 receptor as a therapeutic target for stroke in diabetic population, warranting further investigation.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"107"},"PeriodicalIF":3.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145650276","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}