Purpose: Diabetic cardiomyopathy (DCM), which is diabetes mellitus-induced cardiomyopathy, significantly elevates the risk of heart failure and sudden cardiac death. No specific treatments for DCM are currently available. Gallic acid (GA) is a polyhydroxyphenolic compound that has been shown to inhibit ferroptosis and maintain mitochondrial homeostasis, with potential therapeutic effects in various cardiac diseases. However, its specific role and underlying mechanisms in DCM remain unexplored.
Methods: An in vitro model was established using H9C2 cells pretreated with high glucose plus palmitate, and an in vivo type 2 diabetes mellitus model generated by treating rats with streptozotocin-induced and feeding a high-fat diet. The protective effects of GA and its mechanism of action were evaluated using various methods, including flow cytometry, Western blotting (WB), and transmission electron microscopy. Bioinformatics analysis identified potential target genes for GA's cardioprotection, which were subsequently validated using pAD/TSPO (for overexpression) and pAD/FTMT-shRNA (for silencing) constructs.
Results: GA treatment decreased PTGS2, lactate dehydrogenase, malondialdehyde, ferrous iron, ROS, and oxidized glutathione disulfide (GSSG) levels and increased cell viability, glutathione (GSH) levels, the GSH/GSSG ratio, and GPX4 protein levels in the injury models. GA markedly attenuated mitochondrial ultrastructural damage and promoted mitochondrial homeostasis. These protective effects were abrogated by TSPO overexpression and FTMT silencing.
Conclusion: GA was shown to attenuate diabetic cardiomyopathy by inhibiting ferroptosis and protecting mitochondria via the TSPO/FTMT signaling pathway.
{"title":"Gallic acid attenuates diabetic cardiomyopathy by inhibiting ferroptosis and protecting mitochondria via the TSPO/FTMT pathway.","authors":"Chenchao Zou, Huaihan Xu, Fajia Hu, Yuxian Yu, Lanxiang Liu, Xiuqi Wang, Zheyu Zhang, Huaxi Zou, Jichun Liu, Huang Huang, Songqing Lai","doi":"10.3389/fphar.2025.1661144","DOIUrl":"https://doi.org/10.3389/fphar.2025.1661144","url":null,"abstract":"<p><strong>Purpose: </strong>Diabetic cardiomyopathy (DCM), which is diabetes mellitus-induced cardiomyopathy, significantly elevates the risk of heart failure and sudden cardiac death. No specific treatments for DCM are currently available. Gallic acid (GA) is a polyhydroxyphenolic compound that has been shown to inhibit ferroptosis and maintain mitochondrial homeostasis, with potential therapeutic effects in various cardiac diseases. However, its specific role and underlying mechanisms in DCM remain unexplored.</p><p><strong>Methods: </strong>An <i>in vitro</i> model was established using H9C2 cells pretreated with high glucose plus palmitate, and an <i>in vivo</i> type 2 diabetes mellitus model generated by treating rats with streptozotocin-induced and feeding a high-fat diet. The protective effects of GA and its mechanism of action were evaluated using various methods, including flow cytometry, Western blotting (WB), and transmission electron microscopy. Bioinformatics analysis identified potential target genes for GA's cardioprotection, which were subsequently validated using pAD/TSPO (for overexpression) and pAD/FTMT-shRNA (for silencing) constructs.</p><p><strong>Results: </strong>GA treatment decreased PTGS2, lactate dehydrogenase, malondialdehyde, ferrous iron, ROS, and oxidized glutathione disulfide (GSSG) levels and increased cell viability, glutathione (GSH) levels, the GSH/GSSG ratio, and GPX4 protein levels in the injury models. GA markedly attenuated mitochondrial ultrastructural damage and promoted mitochondrial homeostasis. These protective effects were abrogated by TSPO overexpression and FTMT silencing.</p><p><strong>Conclusion: </strong>GA was shown to attenuate diabetic cardiomyopathy by inhibiting ferroptosis and protecting mitochondria via the TSPO/FTMT signaling pathway.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1661144"},"PeriodicalIF":4.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708275/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780902","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aim: This study aims to explore the impact of the distance between the tumor and the main trunk or first branch of the portal vein on the prognosis of hepatocellular carcinoma (HCC) patients undergoing radical resection.
Methods: This study retrospectively evaluated HCC patients who underwent radical resection between 1 January 2018 and 30 September 2024. Tumors were classified into two categories based on their location: central tumors and peripheral tumors. Central tumors were defined as those located within 2 cm of the main trunk or first branch of the portal vein, while the remaining tumors were classified as peripheral tumors. Recurrence-free survival (RFS) and overall survival (OS) were compared between the two groups. Univariate and multivariate COX analyses were conducted to identify factors associated with RFS and OS. Propensity score matching (PSM) was employed to eliminate intergroup differences for further validation.
Results: A total of 667 HCC patients undergoing radical resection were initially enrolled. Through PSM, 247 patients were successfully matched in each comparative group. In the PSM cohort, the median RFS (mRFS) for patients with central tumors was 23.00 months (95% CI, 18.01-27.99 months), while the mRFS for those with peripheral tumors was 30.50 months (95% CI, 26.17-34.83 months) (p = 0.004). The median OS was 56.00 months (95% CI, 52.10-59.90 months) for central tumors and 72.00 months (95% CI, 67.37-76.63 months) for peripheral tumors (p = 0.043). Multivariate COX analysis confirmed that the distance of less than 2 cm between the tumor and the main trunk or first branch of the portal vein was an independent risk factor for RFS and OS in patients undergoing radical resection for HCC (HR: 1.744, p < 0.001; 1.728, p < 0.001, respectively).
Conclusion: The distance between the tumor and the main portal vein trunk or first branch correlates with the prognosis of hepatocellular carcinoma patients undergoing radical resection.
{"title":"The therapeutic efficacy of radical resection for hepatocellular carcinoma varies markedly by tumor location: a retrospective real-world study.","authors":"Xu Feng, Yi-Qiu Wei, Jia-Rui Liu, Zheng-Rong Shi, Kai Chen, Yong-Shuang Lv","doi":"10.3389/fphar.2025.1674998","DOIUrl":"https://doi.org/10.3389/fphar.2025.1674998","url":null,"abstract":"<p><strong>Aim: </strong>This study aims to explore the impact of the distance between the tumor and the main trunk or first branch of the portal vein on the prognosis of hepatocellular carcinoma (HCC) patients undergoing radical resection.</p><p><strong>Methods: </strong>This study retrospectively evaluated HCC patients who underwent radical resection between 1 January 2018 and 30 September 2024. Tumors were classified into two categories based on their location: central tumors and peripheral tumors. Central tumors were defined as those located within 2 cm of the main trunk or first branch of the portal vein, while the remaining tumors were classified as peripheral tumors. Recurrence-free survival (RFS) and overall survival (OS) were compared between the two groups. Univariate and multivariate COX analyses were conducted to identify factors associated with RFS and OS. Propensity score matching (PSM) was employed to eliminate intergroup differences for further validation.</p><p><strong>Results: </strong>A total of 667 HCC patients undergoing radical resection were initially enrolled. Through PSM, 247 patients were successfully matched in each comparative group. In the PSM cohort, the median RFS (mRFS) for patients with central tumors was 23.00 months (95% CI, 18.01-27.99 months), while the mRFS for those with peripheral tumors was 30.50 months (95% CI, 26.17-34.83 months) (p = 0.004). The median OS was 56.00 months (95% CI, 52.10-59.90 months) for central tumors and 72.00 months (95% CI, 67.37-76.63 months) for peripheral tumors (p = 0.043). Multivariate COX analysis confirmed that the distance of less than 2 cm between the tumor and the main trunk or first branch of the portal vein was an independent risk factor for RFS and OS in patients undergoing radical resection for HCC (HR: 1.744, p < 0.001; 1.728, p < 0.001, respectively).</p><p><strong>Conclusion: </strong>The distance between the tumor and the main portal vein trunk or first branch correlates with the prognosis of hepatocellular carcinoma patients undergoing radical resection.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1674998"},"PeriodicalIF":4.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708258/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145781037","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Darolutamide, a novel androgen receptor inhibitor, significantly improved outcomes in the ARASENS trial when combined with androgen deprivation therapy (ADT) and docetaxel chemotherapy for metastatic hormone-sensitive prostate cancer (mHSPC). In China, the regimen has been reimbursed since December 2023, expanding real-world accessibility. PSMA imaging-guided focal therapy enables precise localization of residual or oligometastatic lesions and may complement systemic control.
Objective: To evaluate the real-world efficacy and safety of darolutamide-based triplet therapy combined with PSMA imaging-guided focal treatment in patients with mHSPC.
Methods: This multicenter, retrospective study included 17 patients treated across three tertiary hospitals between June 2023 and June 2024. All patients received darolutamide (600 mg twice daily), ADT, and docetaxel (75 mg/m2 every 3 weeks for 4-6 cycles), followed 4-6 weeks later by focal therapy (surgery or radiotherapy) based on multidisciplinary team (MDT) assessment and PSMA IMAGING findings. Data collection followed STROBE recommendations. The primary endpoint was PSA90 response; secondary endpoints included PSA ≤0.2 ng/mL, PSA progression-free survival (PSA-PFS), radiographic PFS (rPFS), and safety.
Results: Seventeen patients with predominantly low- to intermediate-volume metastatic disease were analyzed. The PSA90 response rate was 94.1%, and 82.4% achieved PSA ≤0.2 ng/mL. Median PSA-PFS and rPFS were not reached at a median follow-up of 16 months. The 12-/18-month PSA-PFS rates were 88% and 65%, while the corresponding rPFS rates were 94% and 76%. Grade 1-2 myelosuppression (64.7%) and fatigue (47.1%) were most common; grade 3 neutropenia occurred in two patients (11.8%) without grade ≥4 events. Focal therapy was well tolerated, with no severe complications.
Conclusion: Darolutamide-based triplet therapy combined with PSMA-guided focal intervention achieved deep biochemical responses and durable disease control with manageable toxicity in real-world Chinese patients with mHSPC. The multicenter design and standardized methodology support the feasibility of this multimodal approach, which warrants validation in larger prospective studies.
{"title":"Darolutamide-based triple therapy combined with PSMA imaging guided focal treatment in patients with prostate cancer: a multicenter retrospective study.","authors":"Dewen Zhong, Ruochen Zhang, Jinling Wu, Zhixun Guo, Yu Zhang, Qinfei Zhou, Zijun Zou, Deng Lin, Zhonglei Lu, Jiawen Wang, Jinfeng Wu, Liefu Ye, Yongbao Wei","doi":"10.3389/fphar.2025.1692407","DOIUrl":"https://doi.org/10.3389/fphar.2025.1692407","url":null,"abstract":"<p><strong>Background: </strong>Darolutamide, a novel androgen receptor inhibitor, significantly improved outcomes in the ARASENS trial when combined with androgen deprivation therapy (ADT) and docetaxel chemotherapy for metastatic hormone-sensitive prostate cancer (mHSPC). In China, the regimen has been reimbursed since December 2023, expanding real-world accessibility. PSMA imaging-guided focal therapy enables precise localization of residual or oligometastatic lesions and may complement systemic control.</p><p><strong>Objective: </strong>To evaluate the real-world efficacy and safety of darolutamide-based triplet therapy combined with PSMA imaging-guided focal treatment in patients with mHSPC.</p><p><strong>Methods: </strong>This multicenter, retrospective study included 17 patients treated across three tertiary hospitals between June 2023 and June 2024. All patients received darolutamide (600 mg twice daily), ADT, and docetaxel (75 mg/m<sup>2</sup> every 3 weeks for 4-6 cycles), followed 4-6 weeks later by focal therapy (surgery or radiotherapy) based on multidisciplinary team (MDT) assessment and PSMA IMAGING findings. Data collection followed STROBE recommendations. The primary endpoint was PSA90 response; secondary endpoints included PSA ≤0.2 ng/mL, PSA progression-free survival (PSA-PFS), radiographic PFS (rPFS), and safety.</p><p><strong>Results: </strong>Seventeen patients with predominantly low- to intermediate-volume metastatic disease were analyzed. The PSA90 response rate was 94.1%, and 82.4% achieved PSA ≤0.2 ng/mL. Median PSA-PFS and rPFS were not reached at a median follow-up of 16 months. The 12-/18-month PSA-PFS rates were 88% and 65%, while the corresponding rPFS rates were 94% and 76%. Grade 1-2 myelosuppression (64.7%) and fatigue (47.1%) were most common; grade 3 neutropenia occurred in two patients (11.8%) without grade ≥4 events. Focal therapy was well tolerated, with no severe complications.</p><p><strong>Conclusion: </strong>Darolutamide-based triplet therapy combined with PSMA-guided focal intervention achieved deep biochemical responses and durable disease control with manageable toxicity in real-world Chinese patients with mHSPC. The multicenter design and standardized methodology support the feasibility of this multimodal approach, which warrants validation in larger prospective studies.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1692407"},"PeriodicalIF":4.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708570/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780843","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03eCollection Date: 2025-01-01DOI: 10.3389/fphar.2025.1667819
Jingjing Gao, Yao Zhang, Junhua Du, Qiang Liu, Yi Cheng, Wentao Yang, Daoyuan Wang, Wei Wang, Liang Zheng, Dong Wang, Lixin Wu, Xiaolei Jiang, Qunli Men, Chaozhao Liang, Xiansheng Zhang
Background: CP/CPPS is characterized by pain, the primary symptom, which significantly affects QoL and disrupts lower urinary tract function. Conventional monotherapies are often ineffective, necessitating multimodal treatments targeting key symptoms.
Methods: This multicenter, prospective, randomized, parallel, positive-controlled trial enrolled 323 men aged 18-60 years with CP/CPPS across 14 centers in China. Participants were randomly assigned to three groups: tamsulosin 0.2 mg daily, Ningmitai capsule (NMT) 1.52 g thrice daily, or a combination of both, for 8 weeks. The primary endpoint was the change in the total NIH-CPSI score from baseline to week 8. Secondary endpoints included changes in pain, urinary, QoL subdomains, the percentage of patients achieving a ≥25% reduction in NIH-CPSI total score, and pain scores. Safety was monitored through adverse events and liver function tests.
Results: Of the 323 patients, 108 received tamsulosin, 109 NMT, and 106 combination therapy. After 8 weeks, both NMT and combination therapy demonstrated significantly greater reductions in total NIH-CPSI scores compared to tamsulosin (-11.44 vs. -8.58, P < 0.001; -11.94 vs. -8.58, P < 0.001). Combination therapy also significantly reduced pain (P = 0.001) and improved QoL (P < 0.001) compared to tamsulosin. The NMT group showed greater improvements in pain scores at both 4 and 8 weeks compared to tamsulosin (P < 0.05). At week 8, the percentage of patients achieving a ≥25% reduction in NIH-CPSI total score was significantly higher in the NMT (78.64% vs. 55.91%, P < 0.001) and combination groups (82.83% vs. 55.91%, P < 0.001). Subgroup analyses indicated that NMT was most effective in patients with mild to moderate CP/CPPS, younger age (18-34 years), and disease duration of <12 months. No serious adverse events occurred, and NMT was well-tolerated across all groups.
Conclusion: NMT demonstrated superior efficacy over tamsulosin in reducing pain and improving QoL in CP/CPPS patients. Combination therapy provided enhanced symptom relief, particularly in micturition and QoL domains, supporting a multimodal approach for more effective CP/CPPS management. These findings validate NMT as a promising treatment, either alone or in combination, for CP/CPPS. Further long-term studies are needed to optimize its use.
背景:CP/CPPS以疼痛为主要症状,显著影响生活质量并干扰下尿路功能。传统的单一疗法往往无效,需要针对关键症状进行多模式治疗。方法:这项多中心、前瞻性、随机、平行、阳性对照试验在中国14个中心招募了323名年龄在18-60岁的CP/CPPS患者。参与者被随机分为三组:坦索罗辛每天0.2 mg,宁米泰胶囊(NMT) 1.52 g,每天三次,或两者联合,持续8周。主要终点是NIH-CPSI总评分从基线到第8周的变化。次要终点包括疼痛、泌尿、生活质量亚域的变化、NIH-CPSI总分降低≥25%的患者百分比和疼痛评分。通过不良事件和肝功能检测来监测安全性。结果:323例患者中,108例接受坦索罗辛治疗,109例接受NMT治疗,106例接受联合治疗。8周后,与坦索罗辛相比,NMT和联合治疗均显示NIH-CPSI总分显著降低(-11.44比-8.58,P < 0.001; -11.94比-8.58,P < 0.001)。与坦索罗辛相比,联合治疗也显著减轻了疼痛(P = 0.001),改善了生活质量(P < 0.001)。与坦索罗辛相比,NMT组在第4周和第8周的疼痛评分均有较大改善(P < 0.05)。在第8周,NMT组(78.64% vs. 55.91%, P < 0.001)和联合组(82.83% vs. 55.91%, P < 0.001)达到NIH-CPSI总分降低≥25%的患者比例显著高于NMT组(78.64% vs. 55.91%, P < 0.001)。亚组分析显示,NMT在轻至中度CP/CPPS、年龄较小(18-34岁)和病程较长的患者中最有效。结论:NMT在减轻CP/CPPS患者疼痛和改善生活质量方面优于坦索罗辛。联合治疗提供了增强的症状缓解,特别是在排尿和生活质量领域,支持更有效的CP/CPPS管理的多模式方法。这些发现证实,无论是单独治疗还是联合治疗,NMT都是一种很有希望的治疗CP/CPPS的方法。需要进一步的长期研究来优化其使用。临床试验注册:ClinicalTrials.gov,标识符NCT05890235。
{"title":"Ningmitai capsule in patients with chronic prostatitis/chronic pelvic pain syndrome: a multicenter, prospective, randomized, parallel, positive-controlled study.","authors":"Jingjing Gao, Yao Zhang, Junhua Du, Qiang Liu, Yi Cheng, Wentao Yang, Daoyuan Wang, Wei Wang, Liang Zheng, Dong Wang, Lixin Wu, Xiaolei Jiang, Qunli Men, Chaozhao Liang, Xiansheng Zhang","doi":"10.3389/fphar.2025.1667819","DOIUrl":"https://doi.org/10.3389/fphar.2025.1667819","url":null,"abstract":"<p><strong>Background: </strong>CP/CPPS is characterized by pain, the primary symptom, which significantly affects QoL and disrupts lower urinary tract function. Conventional monotherapies are often ineffective, necessitating multimodal treatments targeting key symptoms.</p><p><strong>Methods: </strong>This multicenter, prospective, randomized, parallel, positive-controlled trial enrolled 323 men aged 18-60 years with CP/CPPS across 14 centers in China. Participants were randomly assigned to three groups: tamsulosin 0.2 mg daily, Ningmitai capsule (NMT) 1.52 g thrice daily, or a combination of both, for 8 weeks. The primary endpoint was the change in the total NIH-CPSI score from baseline to week 8. Secondary endpoints included changes in pain, urinary, QoL subdomains, the percentage of patients achieving a ≥25% reduction in NIH-CPSI total score, and pain scores. Safety was monitored through adverse events and liver function tests.</p><p><strong>Results: </strong>Of the 323 patients, 108 received tamsulosin, 109 NMT, and 106 combination therapy. After 8 weeks, both NMT and combination therapy demonstrated significantly greater reductions in total NIH-CPSI scores compared to tamsulosin (-11.44 vs. -8.58, <i>P</i> < 0.001; -11.94 vs. -8.58, <i>P</i> < 0.001). Combination therapy also significantly reduced pain (<i>P</i> = 0.001) and improved QoL (<i>P</i> < 0.001) compared to tamsulosin. The NMT group showed greater improvements in pain scores at both 4 and 8 weeks compared to tamsulosin (<i>P</i> < 0.05). At week 8, the percentage of patients achieving a ≥25% reduction in NIH-CPSI total score was significantly higher in the NMT (78.64% vs. 55.91%, <i>P</i> < 0.001) and combination groups (82.83% vs. 55.91%, <i>P</i> < 0.001). Subgroup analyses indicated that NMT was most effective in patients with mild to moderate CP/CPPS, younger age (18-34 years), and disease duration of <12 months. No serious adverse events occurred, and NMT was well-tolerated across all groups.</p><p><strong>Conclusion: </strong>NMT demonstrated superior efficacy over tamsulosin in reducing pain and improving QoL in CP/CPPS patients. Combination therapy provided enhanced symptom relief, particularly in micturition and QoL domains, supporting a multimodal approach for more effective CP/CPPS management. These findings validate NMT as a promising treatment, either alone or in combination, for CP/CPPS. Further long-term studies are needed to optimize its use.</p><p><strong>Clinical trial registration: </strong>ClinicalTrials.gov, identifier NCT05890235.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1667819"},"PeriodicalIF":4.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708510/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780872","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03eCollection Date: 2025-01-01DOI: 10.3389/fphar.2025.1673462
Rafael Martins de Oliveira, Natália Carine Lima Dos Santos, Marcos Paulo Oliveira Almeida, Joed Pires De Lima Júnior, Guilherme De Souza, Matheus Carvalho Barbosa, Guilherme Vieira Faria, Marina Paschoalino, Rosiane Nascimento Alves, Angelica Oliveira Gomes, Juliana Quero Reimão, Bellisa Freitas Barbosa, Samuel Cota Teixeira, Eloisa Amália Vieira Ferro
Introduction: Toxoplasmosis is a disease caused by the protozoan Toxoplasma gondii. Infection during pregnancy can lead to congenital toxoplasmosis, which is associated with severe outcomes such as fetal abnormalities, stillbirths, and miscarriage. Current treatment options for congenital toxoplasmosis have several limitations, including low efficacy in the chronic phase of the disease and concerns about potential fetal toxicity, such as bone marrow suppression and teratogenic effects. Therefore, there is an urgent need for more effective and safer therapeutic alternatives. The Medicines for Malaria Venture (MMV) offers a collection of bioactive compounds with antiparasitic potential for drug repurposing.
Methods: In this study, we evaluated three MMV Pathogen Box compounds-MMV675968, MMV022478, and MMV021013-for their ability to control T. gondii infection in human trophoblastic cells (BeWo) and third-trimester placental villous explants. We assessed compound toxicity, effects on the parasite's lytic cycle (including adhesion and infection), alterations in parasite morphology, and the host immune response through cytokine quantification.
Results: Nontoxic concentrations of all the three compounds irreversibly inhibited parasite proliferation and interfered with early stages of the lytic cycle, including adhesion and infection. Moreover, treated T. gondii tachyzoites exhibited membrane disruption, cytoplasmic degradation, and organelle disorganization. The cytokine profile indicated that compound treatment promoted an anti-inflammatory immune response, primarily by reducing IL-8 levels. Importantly, these compounds also effectively controlled T. gondii infection in human placental explants without inducing cytotoxicity.
Conclusion: Taken together, our findings support the potential of MMV675968, MMV022478, and MMV021013 as promising drug candidates for the treatment of congenital toxoplasmosis. MMV021013 stands out as the most promising compound, combining high predicted gastrointestinal (GI) absorption and blood-brain barrier (BBB) permeability, with no predicted mutagenic, tumorigenic, irritant, or reproductive effects.
{"title":"Novel drug candidates targeting <i>Toxoplasma gondii</i> in maternal-fetal interface models.","authors":"Rafael Martins de Oliveira, Natália Carine Lima Dos Santos, Marcos Paulo Oliveira Almeida, Joed Pires De Lima Júnior, Guilherme De Souza, Matheus Carvalho Barbosa, Guilherme Vieira Faria, Marina Paschoalino, Rosiane Nascimento Alves, Angelica Oliveira Gomes, Juliana Quero Reimão, Bellisa Freitas Barbosa, Samuel Cota Teixeira, Eloisa Amália Vieira Ferro","doi":"10.3389/fphar.2025.1673462","DOIUrl":"https://doi.org/10.3389/fphar.2025.1673462","url":null,"abstract":"<p><strong>Introduction: </strong>Toxoplasmosis is a disease caused by the protozoan <i>Toxoplasma gondii</i>. Infection during pregnancy can lead to congenital toxoplasmosis, which is associated with severe outcomes such as fetal abnormalities, stillbirths, and miscarriage. Current treatment options for congenital toxoplasmosis have several limitations, including low efficacy in the chronic phase of the disease and concerns about potential fetal toxicity, such as bone marrow suppression and teratogenic effects. Therefore, there is an urgent need for more effective and safer therapeutic alternatives. The Medicines for Malaria Venture (MMV) offers a collection of bioactive compounds with antiparasitic potential for drug repurposing.</p><p><strong>Methods: </strong>In this study, we evaluated three MMV Pathogen Box compounds-MMV675968, MMV022478, and MMV021013-for their ability to control <i>T. gondii</i> infection in human trophoblastic cells (BeWo) and third-trimester placental villous explants. We assessed compound toxicity, effects on the parasite's lytic cycle (including adhesion and infection), alterations in parasite morphology, and the host immune response through cytokine quantification.</p><p><strong>Results: </strong>Nontoxic concentrations of all the three compounds irreversibly inhibited parasite proliferation and interfered with early stages of the lytic cycle, including adhesion and infection. Moreover, treated <i>T. gondii</i> tachyzoites exhibited membrane disruption, cytoplasmic degradation, and organelle disorganization. The cytokine profile indicated that compound treatment promoted an anti-inflammatory immune response, primarily by reducing IL-8 levels. Importantly, these compounds also effectively controlled <i>T. gondii</i> infection in human placental explants without inducing cytotoxicity.</p><p><strong>Conclusion: </strong>Taken together, our findings support the potential of MMV675968, MMV022478, and MMV021013 as promising drug candidates for the treatment of congenital toxoplasmosis. MMV021013 stands out as the most promising compound, combining high predicted gastrointestinal (GI) absorption and blood-brain barrier (BBB) permeability, with no predicted mutagenic, tumorigenic, irritant, or reproductive effects.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1673462"},"PeriodicalIF":4.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708611/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780901","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03eCollection Date: 2025-01-01DOI: 10.3389/fphar.2025.1665897
Gonzalo Fuentes-Barros, Sebastián Castro-Saavedra, Nicolás Montalva, Marco Mellado, Antonia Diaz-Valdés, Claudia Guerrero-Rodríguez, Javier Echeverría
Background: Cryptocarya alba (Mol.) Looser [Lauraceae], known as peumo, is an endemic species of the central Chilean landscape. C. alba has an essential ecological role in the threatened sclerophyllous forest, with traditional uses of leaves, bark, and fruits, and the biotechnological and pharmacological potential of its phytochemicals.
Purpose: The aim is to present the first comprehensive review of the current state of knowledge regarding traditional uses, ethnopharmacology, chemical composition, pharmacokinetic profile, and biological activities of C. alba.
Methodology: Literature data on the traditional uses, ethnopharmacology, chemistry, and bioactivity of C. alba were primarily obtained from digital databases, including Scopus®, ScienceDirect®, SciFinder®, PubMed®, SciELO, and Google Scholar®, as well as from the scientific journal publishers' platforms associated with these databases.
Results and discussion: Traditional uses include its role as a food source for prehistoric populations and ethnomedicinal applications for liver diseases, rheumatism, and infections. The aerial parts are rich in polyphenols, including chlorogenic acid, epicatechin, procyanidins, quercitrin, rutin, and hyperoside, as well as essential oils derived from the leaves. While it contains various alkaloids, only reticuline is present in significant amounts, contributing to the species' highly variable chemical composition. Studies evaluating the biological and pharmacological properties of its extracts and constituents are limited to a few in vitro and in vivo studies; to date, no preliminary or clinical studies are available.
Conclusion: The review highlights the entire existing ethnographic and cultural context of C. alba, revealing a significant gap in information about the species. Although there is a strong historical component, it supports the bioactivity of its main secondary metabolites, given its chemical and pharmacological profile. Given the limited nature of current biological and pharmacological evaluation studies, future research should focus on advancing preclinical and clinical trials, as well as toxicology studies, to ensure the safe and effective use of this approach.
背景:Cryptocarya alba (Mol.) Looser[樟科],被称为peumo,是智利中部景观的特有物种。白桦在濒危的硬叶林中具有重要的生态作用,其叶、树皮和果实的传统用途以及其植物化学物质的生物技术和药理潜力。目的:目的是首次全面回顾关于白刺的传统用途、民族药理学、化学成分、药代动力学特征和生物活性的知识现状。研究方法:关于白骨草的传统用途、民族药理学、化学和生物活性的文献数据主要来自数字数据库,包括Scopus®、ScienceDirect®、SciFinder®、PubMed®、SciELO和谷歌Scholar®,以及与这些数据库相关的科学期刊出版商平台。结果和讨论:其传统用途包括作为史前人群的食物来源,以及用于治疗肝病、风湿病和感染的民族医学应用。空中部分含有丰富的多酚,包括绿原酸、表儿茶素、原花青素、槲皮苷、芦丁和金丝桃苷,以及从叶子中提取的精油。虽然它含有各种生物碱,但只有网状碱大量存在,这导致了该物种的化学成分变化很大。评估其提取物和成分的生物学和药理学特性的研究仅限于少数体外和体内研究;迄今为止,尚无初步或临床研究。结论:这篇综述突出了阿尔巴的整个现有人种学和文化背景,揭示了该物种的重要信息空白。虽然有一个强大的历史成分,它支持其主要次生代谢物的生物活性,鉴于其化学和药理学概况。鉴于目前生物学和药理学评价研究的有限性,未来的研究应侧重于推进临床前和临床试验,以及毒理学研究,以确保该方法的安全有效使用。
{"title":"<i>Cryptocarya alba</i> (Peumo): an endemic Chilean tree with phytochemicals with bioactive potential.","authors":"Gonzalo Fuentes-Barros, Sebastián Castro-Saavedra, Nicolás Montalva, Marco Mellado, Antonia Diaz-Valdés, Claudia Guerrero-Rodríguez, Javier Echeverría","doi":"10.3389/fphar.2025.1665897","DOIUrl":"https://doi.org/10.3389/fphar.2025.1665897","url":null,"abstract":"<p><strong>Background: </strong><i>Cryptocarya alba</i> (Mol.) Looser [Lauraceae], known as <i>peumo</i>, is an endemic species of the central Chilean landscape. <i>C. alba</i> has an essential ecological role in the threatened sclerophyllous forest, with traditional uses of leaves, bark, and fruits, and the biotechnological and pharmacological potential of its phytochemicals.</p><p><strong>Purpose: </strong>The aim is to present the first comprehensive review of the current state of knowledge regarding traditional uses, ethnopharmacology, chemical composition, pharmacokinetic profile, and biological activities of <i>C. alba</i>.</p><p><strong>Methodology: </strong>Literature data on the traditional uses, ethnopharmacology, chemistry, and bioactivity of <i>C. alba</i> were primarily obtained from digital databases, including Scopus®, ScienceDirect®, SciFinder®, PubMed®, SciELO, and Google Scholar®, as well as from the scientific journal publishers' platforms associated with these databases.</p><p><strong>Results and discussion: </strong>Traditional uses include its role as a food source for prehistoric populations and ethnomedicinal applications for liver diseases, rheumatism, and infections. The aerial parts are rich in polyphenols, including chlorogenic acid, epicatechin, procyanidins, quercitrin, rutin, and hyperoside, as well as essential oils derived from the leaves. While it contains various alkaloids, only reticuline is present in significant amounts, contributing to the species' highly variable chemical composition. Studies evaluating the biological and pharmacological properties of its extracts and constituents are limited to a few <i>in vitro</i> and <i>in vivo</i> studies; to date, no preliminary or clinical studies are available.</p><p><strong>Conclusion: </strong>The review highlights the entire existing ethnographic and cultural context of <i>C. alba</i>, revealing a significant gap in information about the species. Although there is a strong historical component, it supports the bioactivity of its main secondary metabolites, given its chemical and pharmacological profile. Given the limited nature of current biological and pharmacological evaluation studies, future research should focus on advancing preclinical and clinical trials, as well as toxicology studies, to ensure the safe and effective use of this approach.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1665897"},"PeriodicalIF":4.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12709134/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780690","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03eCollection Date: 2025-01-01DOI: 10.3389/fphar.2025.1702234
Tonghui Jin, Yu Du, Chaoyue Liu, Jingming Zhao, Tiejun Liu
Common kidney diseases include acute kidney injury, diabetic kidney disease, kidney cancer, and other related conditions. Ginsenosides, the principal bioactive constituents of ginseng, have been widely reported as therapeutic agents against these disorders. However, recent advances regarding their efficacy in kidney diseases have not been comprehensively synthesized. This review addresses this gap by summarizing current findings on the mechanisms and therapeutic targets of ginsenosides. Literature from PubMed, Web of Science, and other databases was systematically retrieved using keywords such as ginsenosides, acute kidney injury, diabetic nephropathy, renal cell carcinoma, lupus nephritis, and aging-related kidney injury. Evidence from cell-based and animal studies demonstrates that ginsenoside compound K, Rg1, Rg3, Rh2, Rb1, Rb3, Rg2, and Rg5 are the most frequently reported for kidney protection. Mechanistically, ginsenosides modulate multiple signalling networks, including NF-κB, PI3K/AKT, MAPK, TGF-β/Smads, PPAR, SIRT1, NLRP3, and Nrf2, to mitigate inflammation, oxidative stress, apoptosis, epithelial-mesenchymal transition, pyroptosis, autophagy, and endoplasmic reticulum stress. Taken together, these findings provide valuable insights into the therapeutic potential of ginsenosides and underscore their promise as candidates for the prevention and treatment of kidney diseases.
常见的肾脏疾病包括急性肾损伤、糖尿病肾病、肾癌和其他相关疾病。人参皂苷是人参的主要生物活性成分,已被广泛报道为治疗这些疾病的药物。然而,关于它们在肾脏疾病中的疗效的最新进展尚未全面综合。这篇综述通过总结目前关于人参皂苷的机制和治疗靶点的发现来解决这一空白。使用人参皂苷、急性肾损伤、糖尿病肾病、肾细胞癌、狼疮性肾炎、衰老相关肾损伤等关键词,系统检索PubMed、Web of Science等数据库的文献。基于细胞和动物研究的证据表明,人参皂苷化合物K, Rg1, Rg3, Rh2, Rb1, Rb3, Rg2和Rg5是最常报道的肾脏保护作用。在机制上,人参皂苷调节多种信号网络,包括NF-κB、PI3K/AKT、MAPK、TGF-β/Smads、PPAR、SIRT1、NLRP3和Nrf2,以减轻炎症、氧化应激、细胞凋亡、上皮-间质转化、焦亡、自噬和内质网应激。综上所述,这些发现为人参皂苷的治疗潜力提供了有价值的见解,并强调了它们作为预防和治疗肾脏疾病的候选药物的前景。
{"title":"Advances in ginsenoside treatment for common kidney diseases: pharmacological evaluation and potential mechanisms.","authors":"Tonghui Jin, Yu Du, Chaoyue Liu, Jingming Zhao, Tiejun Liu","doi":"10.3389/fphar.2025.1702234","DOIUrl":"https://doi.org/10.3389/fphar.2025.1702234","url":null,"abstract":"<p><p>Common kidney diseases include acute kidney injury, diabetic kidney disease, kidney cancer, and other related conditions. Ginsenosides, the principal bioactive constituents of ginseng, have been widely reported as therapeutic agents against these disorders. However, recent advances regarding their efficacy in kidney diseases have not been comprehensively synthesized. This review addresses this gap by summarizing current findings on the mechanisms and therapeutic targets of ginsenosides. Literature from PubMed, Web of Science, and other databases was systematically retrieved using keywords such as ginsenosides, acute kidney injury, diabetic nephropathy, renal cell carcinoma, lupus nephritis, and aging-related kidney injury. Evidence from cell-based and animal studies demonstrates that ginsenoside compound K, Rg1, Rg3, Rh2, Rb1, Rb3, Rg2, and Rg5 are the most frequently reported for kidney protection. Mechanistically, ginsenosides modulate multiple signalling networks, including NF-κB, PI3K/AKT, MAPK, TGF-β/Smads, PPAR, SIRT1, NLRP3, and Nrf2, to mitigate inflammation, oxidative stress, apoptosis, epithelial-mesenchymal transition, pyroptosis, autophagy, and endoplasmic reticulum stress. Taken together, these findings provide valuable insights into the therapeutic potential of ginsenosides and underscore their promise as candidates for the prevention and treatment of kidney diseases.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1702234"},"PeriodicalIF":4.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12709171/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780659","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Objective: Although farrerol exhibits promising antitumor properties against various cancers, its potential therapeutic effects on colorectal cancer (CRC) remain unexplored, and the underlying mechanisms are still unclear. Based on network pharmacology, molecular docking, molecular dynamics simulations, and in vitro experiments, this study aims to investigate the molecular mechanisms of farrerol in the treatment of CRC, thereby providing new research directions for CRC therapy.
Methods: This study employed network pharmacology to screen for potential therapeutic targets and pathways of farrerol in CRC, followed by preliminary validation of target validity through molecular docking and molecular dynamics simulations. Finally, in vitro experiments were conducted to verify the antitumor effects of farrerol against CRC.
Results: Network pharmacology identified 12 key targets: CCNA1, CCNA2, CCNE1, CDC25B, CDK2, CYP19A1, ESR1, ESR2, HSP90AA1, PTPN1, RAF1, and SRC. The molecular docking results revealed that the binding energies of farrerol with all target proteins were as follows: farrerol-CCNA1 (-8.6 kcal·mol-1), farrerol-CCNA2 (-7.0 kcal·mol-1), farrerol-CCNE1 (-7.4 kcal·mol1), farrerol-CDC25B (-7.3 kcal·mol-1), farrerol-CDK2 (-10.1 kcal·mol-1), farrerol-CYP19A1 (-8.4 kcal·mol-1), farrerol-ESR1 (-3.3 kcal·mol-1), farrerol-ESR2 (-8.9 kcal·mol-1), farrerol-HSP90AA1 (-10.4 kcal·mol-1), farrerol-PTPN1 (-7.6 kcal·mol-1), farrerol-RAF1 (-4.5 kcal·mol-1), farrerol-SRC (-9.9 kcal·mol-1), farrerol-VEGFA (-8.5 kcal·mol-1), and farrerol-KDR (-9.0 kcal·mol-1). These data indicate that farrerol can spontaneously bind to the target proteins. Molecular dynamics simulations demonstrated favorable interactions within the KDR-Farrerol and VEGFA-Farrerol complexes. In vitro experimental results demonstrated that farrerol could inhibit the proliferation and migration of CRC cells, induce cell cycle arrest at the G0/G1 phase, and suppress the protein expression of VEGFA, VEGFR2, and p-VEGFR2.
Conclusion: This study, for the first time, validated the antitumor effect of farrerol against CRC through network pharmacology, molecular docking, molecular dynamics simulations, and in vitro experiments. The findings indicate that the ability of farrerol to inhibit the proliferation and migration of colorectal cancer cells may be associated with the induction of G0/G1 phase cell cycle arrest and the regulation of VEGF signaling pathway activation via binding to VEGFA and KDR.
{"title":"Farrerol inhibits proliferation and migration of colorectal cancer via the VEGF signaling pathway: evidence from network pharmacology, molecular docking, molecular dynamics simulation, and <i>in vitro</i> experiments.","authors":"Longhui Zhang, Qijing Xu, Guanduo Sun, Xiaokang Zhang, Jialong Xue, Chun Yao, Dechun Liu, Jingming Zhai","doi":"10.3389/fphar.2025.1717293","DOIUrl":"https://doi.org/10.3389/fphar.2025.1717293","url":null,"abstract":"<p><strong>Objective: </strong>Although farrerol exhibits promising antitumor properties against various cancers, its potential therapeutic effects on colorectal cancer (CRC) remain unexplored, and the underlying mechanisms are still unclear. Based on network pharmacology, molecular docking, molecular dynamics simulations, and <i>in vitro</i> experiments, this study aims to investigate the molecular mechanisms of farrerol in the treatment of CRC, thereby providing new research directions for CRC therapy.</p><p><strong>Methods: </strong>This study employed network pharmacology to screen for potential therapeutic targets and pathways of farrerol in CRC, followed by preliminary validation of target validity through molecular docking and molecular dynamics simulations. Finally, <i>in vitro</i> experiments were conducted to verify the antitumor effects of farrerol against CRC.</p><p><strong>Results: </strong>Network pharmacology identified 12 key targets: CCNA1, CCNA2, CCNE1, CDC25B, CDK2, CYP19A1, ESR1, ESR2, HSP90AA1, PTPN1, RAF1, and SRC. The molecular docking results revealed that the binding energies of farrerol with all target proteins were as follows: farrerol-CCNA1 (-8.6 kcal·mol<sup>-1</sup>), farrerol-CCNA2 (-7.0 kcal·mol<sup>-1</sup>), farrerol-CCNE1 (-7.4 kcal·mol<sup>1</sup>), farrerol-CDC25B (-7.3 kcal·mol<sup>-1</sup>), farrerol-CDK2 (-10.1 kcal·mol<sup>-1</sup>), farrerol-CYP19A1 (-8.4 kcal·mol<sup>-1</sup>), farrerol-ESR1 (-3.3 kcal·mol<sup>-1</sup>), farrerol-ESR2 (-8.9 kcal·mol<sup>-1</sup>), farrerol-HSP90AA1 (-10.4 kcal·mol<sup>-1</sup>), farrerol-PTPN1 (-7.6 kcal·mol<sup>-1</sup>), farrerol-RAF1 (-4.5 kcal·mol<sup>-1</sup>), farrerol-SRC (-9.9 kcal·mol<sup>-1</sup>), farrerol-VEGFA (-8.5 kcal·mol<sup>-1</sup>), and farrerol-KDR (-9.0 kcal·mol<sup>-1</sup>). These data indicate that farrerol can spontaneously bind to the target proteins. Molecular dynamics simulations demonstrated favorable interactions within the KDR-Farrerol and VEGFA-Farrerol complexes. <i>In vitro</i> experimental results demonstrated that farrerol could inhibit the proliferation and migration of CRC cells, induce cell cycle arrest at the G0/G1 phase, and suppress the protein expression of VEGFA, VEGFR2, and p-VEGFR2.</p><p><strong>Conclusion: </strong>This study, for the first time, validated the antitumor effect of farrerol against CRC through network pharmacology, molecular docking, molecular dynamics simulations, and <i>in vitro</i> experiments. The findings indicate that the ability of farrerol to inhibit the proliferation and migration of colorectal cancer cells may be associated with the induction of G0/G1 phase cell cycle arrest and the regulation of VEGF signaling pathway activation via binding to VEGFA and KDR.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1717293"},"PeriodicalIF":4.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708557/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780908","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03eCollection Date: 2025-01-01DOI: 10.3389/fphar.2025.1694723
Letizia Spataro, Maurizio Ragni, Agnese Segala, Alice Vetturi, Giulia Sofia Marcotto, Luca Canciani, Michele O Carruba, Roberto Aquilani, Ginetta Collo, Alessandra Valerio, Enzo Nisoli, Chiara Ruocco
Objective: Obesity disrupts intestinal homeostasis, leading to increased permeability ("leaky gut"), mucosal inflammation, and systemic metabolic dysfunction. Mitochondrial impairment in intestinal epithelial cells (IECs) is a central driver of this process. Essential amino acids (EAAs) improve mitochondrial function in metabolic tissues, but their impact on intestinal health remains underexplored. Here, we investigated whether dietary EAAs preserve gut barrier integrity through mitochondrial protection in obesity and inflammation.
Methods: Male C57BL/6N mice were fed a high-fat diet (HFD) or an isocaloric, isonitrogenous EAA-substituted HFD (HFD-EAA) for 33 weeks to assess metabolic outcomes, intestinal barrier function, inflammation, and mitochondrial biogenesis. Parallel, in vitro studies in differentiated Caco-2 cells tested an EAA formula enriched with Krebs cycle intermediates (E7), under basal and pro-inflammatory conditions (IL-1β, TNF-α, LPS).
Results: HFD-EAA supplementation prevented and reversed obesity, improved glucose tolerance, reduced mesenteric fat expansion, and preserved intestinal barrier integrity while attenuating inflammation. EAAs restored intestinal length and weight, lowered plasma calprotectin, and normalized citrulline, a biomarker of enterocyte mass. Tight and adherens junction proteins (zonulin-1, occludin, E-cadherin, claudins) were maintained, while pore-forming claudin-2 was reduced. EAAs also upregulated PGC-1α and mitochondrial electron transport chain genes in intestinal epithelial cells (IECs). Their direct effects were confirmed in vitro in Caco-2 cells, where E7 increased transepithelial electrical resistance (TEER), enhanced mitochondrial respiration, suppressed inflammation-induced glycolytic reprogramming, activated antioxidant defenses, and reduced IL8 secretion. Mechanistically, E7 promoted eNOS phosphorylation and inhibited mTORC1 signaling.
Conclusion: EAAs protect gut barrier integrity by sustaining mitochondrial biogenesis and function in IECs, thereby reducing obesity- and stress-induced inflammation. These findings highlight EAAs as a promising nutritional strategy to counteract mitochondrial dysfunction and prevent or reverse gut barrier disruption in obesity-related and inflammatory disorders.
{"title":"Essential amino acids preserve intestinal barrier integrity <i>via</i> mitochondrial protection in obesity and gut inflammation.","authors":"Letizia Spataro, Maurizio Ragni, Agnese Segala, Alice Vetturi, Giulia Sofia Marcotto, Luca Canciani, Michele O Carruba, Roberto Aquilani, Ginetta Collo, Alessandra Valerio, Enzo Nisoli, Chiara Ruocco","doi":"10.3389/fphar.2025.1694723","DOIUrl":"https://doi.org/10.3389/fphar.2025.1694723","url":null,"abstract":"<p><strong>Objective: </strong>Obesity disrupts intestinal homeostasis, leading to increased permeability (\"leaky gut\"), mucosal inflammation, and systemic metabolic dysfunction. Mitochondrial impairment in intestinal epithelial cells (IECs) is a central driver of this process. Essential amino acids (EAAs) improve mitochondrial function in metabolic tissues, but their impact on intestinal health remains underexplored. Here, we investigated whether dietary EAAs preserve gut barrier integrity through mitochondrial protection in obesity and inflammation.</p><p><strong>Methods: </strong>Male C57BL/6N mice were fed a high-fat diet (HFD) or an isocaloric, isonitrogenous EAA-substituted HFD (HFD-EAA) for 33 weeks to assess metabolic outcomes, intestinal barrier function, inflammation, and mitochondrial biogenesis. Parallel, <i>in vitro</i> studies in differentiated Caco-2 cells tested an EAA formula enriched with Krebs cycle intermediates (E7), under basal and pro-inflammatory conditions (IL-1β, TNF-α, LPS).</p><p><strong>Results: </strong>HFD-EAA supplementation prevented and reversed obesity, improved glucose tolerance, reduced mesenteric fat expansion, and preserved intestinal barrier integrity while attenuating inflammation. EAAs restored intestinal length and weight, lowered plasma calprotectin, and normalized citrulline, a biomarker of enterocyte mass. Tight and adherens junction proteins (zonulin-1, occludin, E-cadherin, claudins) were maintained, while pore-forming claudin-2 was reduced. EAAs also upregulated PGC-1α and mitochondrial electron transport chain genes in intestinal epithelial cells (IECs). Their direct effects were confirmed <i>in vitro</i> in Caco-2 cells, where E7 increased transepithelial electrical resistance (TEER), enhanced mitochondrial respiration, suppressed inflammation-induced glycolytic reprogramming, activated antioxidant defenses, and reduced IL8 secretion. Mechanistically, E7 promoted eNOS phosphorylation and inhibited mTORC1 signaling.</p><p><strong>Conclusion: </strong>EAAs protect gut barrier integrity by sustaining mitochondrial biogenesis and function in IECs, thereby reducing obesity- and stress-induced inflammation. These findings highlight EAAs as a promising nutritional strategy to counteract mitochondrial dysfunction and prevent or reverse gut barrier disruption in obesity-related and inflammatory disorders.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1694723"},"PeriodicalIF":4.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708562/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780927","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03eCollection Date: 2025-01-01DOI: 10.3389/fphar.2025.1731165
Fuwei Bai, Dongyang Wang, Yingying Wu
Dapansutrile, an orally active and selective NLRP3 inflammasome inhibitor, exerts its effects by directly binding to the NLRP3 NACHT domain. This action disrupts inflammasome assembly and caspase-1 activation, thereby inhibiting the maturation and release of the pro-inflammatory cytokines IL-1β and IL-18. Beyond this core inhibition, dapansutrile modulates immune cell chemotaxis and inhibits pyroptosis. Preclinical and clinical studies demonstrate its efficacy in mitigating pathology in diverse conditions, including gouty arthritis, cardiovascular diseases, neurodegenerative disorders, inflammatory bowel disease, and periodontitis. A favorable safety profile distinguishes it from other NLRP3 inhibitors like MCC950, with no significant hepatotoxicity reported in trials. Furthermore, dapansutrile exhibits synergistic effects when combined with agents such as lonafarnib or immune checkpoint inhibitors, enhancing anti-inflammatory and anti-tumor responses. This review consolidates evidence on dapansutrile's molecular mechanisms, therapeutic applications, and biosafety, highlighting its potential as a novel, well-tolerated, and versatile anti-inflammatory agent. Future research should focus on optimizing its delivery, particularly to the central nervous system, and leveraging artificial intelligence to predict effective drug combinations.
{"title":"Dapansutrile in multidisciplinary therapeutic applications: mechanisms and clinical perspectives.","authors":"Fuwei Bai, Dongyang Wang, Yingying Wu","doi":"10.3389/fphar.2025.1731165","DOIUrl":"https://doi.org/10.3389/fphar.2025.1731165","url":null,"abstract":"<p><p>Dapansutrile, an orally active and selective NLRP3 inflammasome inhibitor, exerts its effects by directly binding to the NLRP3 NACHT domain. This action disrupts inflammasome assembly and caspase-1 activation, thereby inhibiting the maturation and release of the pro-inflammatory cytokines IL-1β and IL-18. Beyond this core inhibition, dapansutrile modulates immune cell chemotaxis and inhibits pyroptosis. Preclinical and clinical studies demonstrate its efficacy in mitigating pathology in diverse conditions, including gouty arthritis, cardiovascular diseases, neurodegenerative disorders, inflammatory bowel disease, and periodontitis. A favorable safety profile distinguishes it from other NLRP3 inhibitors like MCC950, with no significant hepatotoxicity reported in trials. Furthermore, dapansutrile exhibits synergistic effects when combined with agents such as lonafarnib or immune checkpoint inhibitors, enhancing anti-inflammatory and anti-tumor responses. This review consolidates evidence on dapansutrile's molecular mechanisms, therapeutic applications, and biosafety, highlighting its potential as a novel, well-tolerated, and versatile anti-inflammatory agent. Future research should focus on optimizing its delivery, particularly to the central nervous system, and leveraging artificial intelligence to predict effective drug combinations.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1731165"},"PeriodicalIF":4.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708538/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780754","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}