Volatile oils (VOs), synonymously termed essential oils (EOs), are highly hydrophobic liquids obtained from aromatic plants, containing diverse organic compounds for example terpenes and terpenoids. These oils exhibit significant neuroprotective properties, containing antioxidant, anti-inflammatory, anti-apoptotic, glutamate activation, cholinesterase inhibitory action, and anti-protein aggregatory action, making them potential therapeutic agents in managing neurodegenerative diseases (NDs). VOs regulate glutamate activation, enhance synaptic plasticity, and inhibit oxidative stress through the stimulation of antioxidant enzymes. They also reduce inflammation by inhibiting key inflammatory mediators and enzymes. Furthermore, VOs prevent neuronal apoptosis by modulating apoptosis-related proteins and caspases. Their anti-protein aggregation potential helps mitigate the accumulation of misfolded proteins, a hallmark of neurodegenerative disorders. Additionally, VOs inhibit cholinesterase enzymes, increasing acetylcholine levels, and improving neuronal communication. In addition to their neuroprotective action, it also exerts some toxic effects, such as genotoxicity, hepatotoxicity, embryotoxicity, and hypersensitivity, which are most commonly caused by the presence of monoterpenes in the volatile oils. This review examines the diverse functions of vasoactive oxidants (VOs) in neuroprotection, underscoring their therapeutic promise for various neurological conditions, with a particular emphasis on Alzheimer's disease.
{"title":"The neuroprotective role of volatile oils: insights into chemical profiles, characteristics, neurochemical mechanisms, and preclinical studies in Alzheimer's disease.","authors":"Meghraj Suryawanshi, Pranjal Gujarathi, Bhupendra Prajapati, Kuldeep Vinchurkar, Piyush Gujarathi","doi":"10.1007/s10787-024-01626-5","DOIUrl":"https://doi.org/10.1007/s10787-024-01626-5","url":null,"abstract":"<p><p>Volatile oils (VOs), synonymously termed essential oils (EOs), are highly hydrophobic liquids obtained from aromatic plants, containing diverse organic compounds for example terpenes and terpenoids. These oils exhibit significant neuroprotective properties, containing antioxidant, anti-inflammatory, anti-apoptotic, glutamate activation, cholinesterase inhibitory action, and anti-protein aggregatory action, making them potential therapeutic agents in managing neurodegenerative diseases (NDs). VOs regulate glutamate activation, enhance synaptic plasticity, and inhibit oxidative stress through the stimulation of antioxidant enzymes. They also reduce inflammation by inhibiting key inflammatory mediators and enzymes. Furthermore, VOs prevent neuronal apoptosis by modulating apoptosis-related proteins and caspases. Their anti-protein aggregation potential helps mitigate the accumulation of misfolded proteins, a hallmark of neurodegenerative disorders. Additionally, VOs inhibit cholinesterase enzymes, increasing acetylcholine levels, and improving neuronal communication. In addition to their neuroprotective action, it also exerts some toxic effects, such as genotoxicity, hepatotoxicity, embryotoxicity, and hypersensitivity, which are most commonly caused by the presence of monoterpenes in the volatile oils. This review examines the diverse functions of vasoactive oxidants (VOs) in neuroprotection, underscoring their therapeutic promise for various neurological conditions, with a particular emphasis on Alzheimer's disease.</p>","PeriodicalId":13551,"journal":{"name":"Inflammopharmacology","volume":" ","pages":""},"PeriodicalIF":4.6,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143004819","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-17DOI: 10.1007/s10787-025-01639-8
Tao Jia, Yifan Xia, Mengyao Yi, Xinyue Zhang, Yi Zheng, Delu Che
Background: Rosacea is a chronic inflammatory disease characterized by persistent erythema, papules, and pustules, mainly on the skin of the face. Rosacea is difficult to treat; therefore, identifying new treatments is crucial. Mas-related G protein-coupled receptor X2 (MRGPRX2)-mediated mast cell (MC) activation is essential in the pathogenesis of rosacea. Casticin has been shown to exert anti-inflammatory effects; however, it remains unclear whether it can inhibit MRGPRX2 in treating rosacea. This study determined the therapeutic efficacy of casticin against rosacea by inhibiting MRGPRX2-mediated MC activation.
Methods: A mouse model of LL37-induced rosacea-like dermatitis was employed. The pathological changes were evaluated using hematoxylin and eosin (H&E) staining, and MCs and CD4+ T cells were observed. Inflammatory mediators were analyzed using ELISA. Mouse skin lesions were collected for transcriptomic sequencing. We used an MRGPRX2-mediated MC degranulation model to evaluate the inhibitory effects of casticin in vitro. Molecular docking analysis, molecular dynamics simulations, and surface plasmon resonance evaluated the binding between casticin and MRGPRX2.
Results: Casticin attenuated the LL37-induced inflammatory phenotype and reactions in rosacea-like dermatitis. RNA-seq data showed that casticin inhibited MC activation in a mouse model of rosacea. Furthermore, casticin significantly reduced CD4 + T-cell infiltration. Moreover, casticin inhibited MC activation as an MRGPRX2 antagonist in vitro and in vivo by influencing the NF-κB signaling pathway.
Conclusion: Our study demonstrated that casticin exhibits therapeutic efficacy against rosacea by inhibiting MC activation via MRGPRX2.
{"title":"Casticin reduces rosacea-related inflammation by inhibiting mast cell activation via Mas-related G protein-coupled receptor X2.","authors":"Tao Jia, Yifan Xia, Mengyao Yi, Xinyue Zhang, Yi Zheng, Delu Che","doi":"10.1007/s10787-025-01639-8","DOIUrl":"https://doi.org/10.1007/s10787-025-01639-8","url":null,"abstract":"<p><strong>Background: </strong>Rosacea is a chronic inflammatory disease characterized by persistent erythema, papules, and pustules, mainly on the skin of the face. Rosacea is difficult to treat; therefore, identifying new treatments is crucial. Mas-related G protein-coupled receptor X2 (MRGPRX2)-mediated mast cell (MC) activation is essential in the pathogenesis of rosacea. Casticin has been shown to exert anti-inflammatory effects; however, it remains unclear whether it can inhibit MRGPRX2 in treating rosacea. This study determined the therapeutic efficacy of casticin against rosacea by inhibiting MRGPRX2-mediated MC activation.</p><p><strong>Methods: </strong>A mouse model of LL37-induced rosacea-like dermatitis was employed. The pathological changes were evaluated using hematoxylin and eosin (H&E) staining, and MCs and CD4<sup>+</sup> T cells were observed. Inflammatory mediators were analyzed using ELISA. Mouse skin lesions were collected for transcriptomic sequencing. We used an MRGPRX2-mediated MC degranulation model to evaluate the inhibitory effects of casticin in vitro. Molecular docking analysis, molecular dynamics simulations, and surface plasmon resonance evaluated the binding between casticin and MRGPRX2.</p><p><strong>Results: </strong>Casticin attenuated the LL37-induced inflammatory phenotype and reactions in rosacea-like dermatitis. RNA-seq data showed that casticin inhibited MC activation in a mouse model of rosacea. Furthermore, casticin significantly reduced CD4 + T-cell infiltration. Moreover, casticin inhibited MC activation as an MRGPRX2 antagonist in vitro and in vivo by influencing the NF-κB signaling pathway.</p><p><strong>Conclusion: </strong>Our study demonstrated that casticin exhibits therapeutic efficacy against rosacea by inhibiting MC activation via MRGPRX2.</p>","PeriodicalId":13551,"journal":{"name":"Inflammopharmacology","volume":" ","pages":""},"PeriodicalIF":4.6,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143004816","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-14DOI: 10.1007/s10787-024-01638-1
Shilpa Kumari, Kajal Bagri, Rahul Deshmukh
Alzheimer's Disease (AD), a progressive and age-associated neurodegenerative disorder, is primarily characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles. Despite advances in targeting Aβ-mediated neuronal damage with anti-Aβ antibodies, these treatments provide only symptomatic relief and fail to address the multifactorial pathology of the disease. This necessitates the exploration of novel therapeutic approaches and a deeper understanding of molecular signaling mechanisms underlying AD. Phosphodiesterases (PDEs), particularly Phosphodiesterase 4 (PDE4), play a pivotal role in regulating cyclic adenosine monophosphate (cAMP), a key molecule involved in memory consolidation and cognitive function. PDE4 inhibitors have demonstrated potential in enhancing memory and cognition in preclinical models of AD by modulating cAMP signaling. However, their clinical translation has been limited due to challenges such as adverse effects, narrow therapeutic windows, and low specificity in mechanism of action. This review bridges the gap between preclinical discoveries and clinical applications of PDE4 inhibitors in AD. It highlights preclinical evidence supporting the neuroprotective and anti-inflammatory effects of PDE4 inhibitors while addressing challenges in their clinical development, including issues of safety, efficacy, and disease-specific targeting. By integrating findings from both preclinical and clinical studies, we provide a comprehensive understanding of the therapeutic potential of PDE4 inhibitors in AD. Furthermore, this review outlines future research directions aimed at optimizing PDE4 inhibition strategies for AD treatment, offering a roadmap to translate foundational insights into clinical realities.
{"title":"Connecting dots: Preclinical foundations to clinical realities of PDE4 inhibitors in Alzheimer's disease.","authors":"Shilpa Kumari, Kajal Bagri, Rahul Deshmukh","doi":"10.1007/s10787-024-01638-1","DOIUrl":"https://doi.org/10.1007/s10787-024-01638-1","url":null,"abstract":"<p><p>Alzheimer's Disease (AD), a progressive and age-associated neurodegenerative disorder, is primarily characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles. Despite advances in targeting Aβ-mediated neuronal damage with anti-Aβ antibodies, these treatments provide only symptomatic relief and fail to address the multifactorial pathology of the disease. This necessitates the exploration of novel therapeutic approaches and a deeper understanding of molecular signaling mechanisms underlying AD. Phosphodiesterases (PDEs), particularly Phosphodiesterase 4 (PDE4), play a pivotal role in regulating cyclic adenosine monophosphate (cAMP), a key molecule involved in memory consolidation and cognitive function. PDE4 inhibitors have demonstrated potential in enhancing memory and cognition in preclinical models of AD by modulating cAMP signaling. However, their clinical translation has been limited due to challenges such as adverse effects, narrow therapeutic windows, and low specificity in mechanism of action. This review bridges the gap between preclinical discoveries and clinical applications of PDE4 inhibitors in AD. It highlights preclinical evidence supporting the neuroprotective and anti-inflammatory effects of PDE4 inhibitors while addressing challenges in their clinical development, including issues of safety, efficacy, and disease-specific targeting. By integrating findings from both preclinical and clinical studies, we provide a comprehensive understanding of the therapeutic potential of PDE4 inhibitors in AD. Furthermore, this review outlines future research directions aimed at optimizing PDE4 inhibition strategies for AD treatment, offering a roadmap to translate foundational insights into clinical realities.</p>","PeriodicalId":13551,"journal":{"name":"Inflammopharmacology","volume":" ","pages":""},"PeriodicalIF":4.6,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142978343","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Osteoarthritis, the most common arthritic condition, is an age-related progressive disease characterized by the loss of cartilage and synovial inflammation in the knees and hips. Development of pain, stiffness, and considerably restricted mobility of the joints are responsible for the production of matrix metalloproteinases and cytokines. Although several treatments are available for the management of this disease condition, they possess limitations at different levels. Recently, efforts have focused on regulating the production of the NLRP3 inflammasome, which plays a critical role in the disease's progression due to its dysregulation. Inhibition of NLRP3 inflammasome has shown the potential to modulate the production of MMP-13, caspase-1, IL-1β, etc., which has been reflected by positive responses in different preclinical and clinical studies. Aiming inhibition of this NLRP3 inflammasome, several compounds are in different stages of research owing to bring a novel agent for the treatment of osteoarthritis. This review summarizes the mechanistic pathways linking NLRP3 activation to osteoarthritis development and discusses the progress in new therapeutics aimed at effective treatment.
{"title":"Targeting the NLRP3 inflammasome as a novel therapeutic target for osteoarthritis.","authors":"Varnita Karmakar, Mayukh Chain, Ankit Majie, Arya Ghosh, Pallav Sengupta, Sulagna Dutta, Papiya Mitra Mazumder, Bapi Gorain","doi":"10.1007/s10787-024-01629-2","DOIUrl":"https://doi.org/10.1007/s10787-024-01629-2","url":null,"abstract":"<p><p>Osteoarthritis, the most common arthritic condition, is an age-related progressive disease characterized by the loss of cartilage and synovial inflammation in the knees and hips. Development of pain, stiffness, and considerably restricted mobility of the joints are responsible for the production of matrix metalloproteinases and cytokines. Although several treatments are available for the management of this disease condition, they possess limitations at different levels. Recently, efforts have focused on regulating the production of the NLRP3 inflammasome, which plays a critical role in the disease's progression due to its dysregulation. Inhibition of NLRP3 inflammasome has shown the potential to modulate the production of MMP-13, caspase-1, IL-1β, etc., which has been reflected by positive responses in different preclinical and clinical studies. Aiming inhibition of this NLRP3 inflammasome, several compounds are in different stages of research owing to bring a novel agent for the treatment of osteoarthritis. This review summarizes the mechanistic pathways linking NLRP3 activation to osteoarthritis development and discusses the progress in new therapeutics aimed at effective treatment.</p>","PeriodicalId":13551,"journal":{"name":"Inflammopharmacology","volume":" ","pages":""},"PeriodicalIF":4.6,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142978345","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-13DOI: 10.1007/s10787-024-01637-2
Yongjun Liu, Hongwei Gao, Bin Du, Qianwen Huo
Objective: The aim of this study was to comprehensively investigate the clinical efficacy of intraoperative local joint injection and intramuscular injection of betamethasone in patients with severe traumatic knee osteoarthritis (KOA).
Methods: 80 patients with severe traumatic KOA undergoing total knee arthroplasty were retrospectively recruited and rolled into S1 group (intra-articular injection of ropivacaine + betamethasone and isotonic saline mixture at joint incision), S2 group (muscle local injection of betamethasone before incision closure, simultaneously intra-articular injection of ropivacaine + isotonic saline mixture at joint incision), and D group (intra-articular injection of ropivacaine + isotonic saline mixture at the joint incision). Visual analog scale (VAS) score, serum inflammatory factors (IFs), hospital for special surgery (HSS)score, Pittsburgh sleep quality index (PSQI), and adverse reaction events (AREs) were analyzed.
Results: Pain scores of patients in all three groups decreased drastically over time on postoperative days (PDs) 1, 2, and 3, with the scores in S1 and S2 groups markedly inferior to D group (P < 0.05). HSS scores of patients in S1 and S2 groups at postoperative months 1, 3, and 6 were considerably superior to those in D group (P < 0.05). PSQI scores of patients in S1 and S2 groups at postoperative months 1, 3, and 6 were notably inferior to those in D group (P < 0.05).
Conclusion: Both intraoperative local joint injection and muscle injection of betamethasone are effective in patients with severe traumatic KOA.
{"title":"Effect of intramuscular vs intra-articular betamethasone injection on pain and inflammatory factors among patients with severe traumatic knee osteoarthritis.","authors":"Yongjun Liu, Hongwei Gao, Bin Du, Qianwen Huo","doi":"10.1007/s10787-024-01637-2","DOIUrl":"https://doi.org/10.1007/s10787-024-01637-2","url":null,"abstract":"<p><strong>Objective: </strong>The aim of this study was to comprehensively investigate the clinical efficacy of intraoperative local joint injection and intramuscular injection of betamethasone in patients with severe traumatic knee osteoarthritis (KOA).</p><p><strong>Methods: </strong>80 patients with severe traumatic KOA undergoing total knee arthroplasty were retrospectively recruited and rolled into S1 group (intra-articular injection of ropivacaine + betamethasone and isotonic saline mixture at joint incision), S2 group (muscle local injection of betamethasone before incision closure, simultaneously intra-articular injection of ropivacaine + isotonic saline mixture at joint incision), and D group (intra-articular injection of ropivacaine + isotonic saline mixture at the joint incision). Visual analog scale (VAS) score, serum inflammatory factors (IFs), hospital for special surgery (HSS)score, Pittsburgh sleep quality index (PSQI), and adverse reaction events (AREs) were analyzed.</p><p><strong>Results: </strong>Pain scores of patients in all three groups decreased drastically over time on postoperative days (PDs) 1, 2, and 3, with the scores in S1 and S2 groups markedly inferior to D group (P < 0.05). HSS scores of patients in S1 and S2 groups at postoperative months 1, 3, and 6 were considerably superior to those in D group (P < 0.05). PSQI scores of patients in S1 and S2 groups at postoperative months 1, 3, and 6 were notably inferior to those in D group (P < 0.05).</p><p><strong>Conclusion: </strong>Both intraoperative local joint injection and muscle injection of betamethasone are effective in patients with severe traumatic KOA.</p>","PeriodicalId":13551,"journal":{"name":"Inflammopharmacology","volume":" ","pages":""},"PeriodicalIF":4.6,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142978344","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Maejo 341 Sweet potato (MSP) is a new purple sweet potato variety cultivated in Northern Thailand, but its health benefits are unknown. This study aimed to investigate its antioxidant, anti-inflammatory, and anti-osteoporotic activities, as well as its anthocyanin content. The peel and flesh of MSP were extracted with ethanol and water. Compared with the flesh extracts, the peel extracts presented greater antioxidant capacity and were rich in phenolics, flavonoids, and anthocyanins, namely, cyanidin-3-O-glucoside, peonidin-3-O-glucoside, pelargonidin-3-O-glucoside, cyanidin, and peonidin. The peel extracts suppressed lipopolysaccharide-induced inflammation by inhibiting the secretion of proinflammatory cytokines and enzymes, including TNF-α, IL-1β, IL-6, COX-2, and iNOS, as well as reducing nitric oxide and matrix metalloproteinase-9 secretion. The extracts inhibited the RANKL-induced NF-κB and MAPK pathways and downregulated osteoclastogenic marker expression. Under LPS and RANKL treatment, the peel extracts notably reduced reactive oxygen species production while increasing antioxidant gene expression. Furthermore, they increased osteoblast viability and slightly raise alkaline phosphatase activity. These findings suggest that MSP peel could be used as a functional food to alleviate oxidative stress and inflammation-related osteoporosis.
Maejo 341甘薯(MSP)是泰国北部栽培的紫色甘薯新品种,但其健康益处尚不清楚。本研究旨在探讨其抗氧化、抗炎、抗骨质疏松活性及花青素含量。用乙醇和水提取MSP的果皮和果肉。与果肉提取物相比,果皮提取物具有更强的抗氧化能力,并含有丰富的酚类、黄酮类和花青素,即花青素-3- o -葡萄糖苷、芍药苷-3- o -葡萄糖苷、芍药苷-3- o -葡萄糖苷、花青素和芍药苷。果皮提取物通过抑制TNF-α、IL-1β、IL-6、COX-2、iNOS等促炎细胞因子和酶的分泌,以及减少一氧化氮和基质金属蛋白酶-9的分泌来抑制脂多糖诱导的炎症。提取物抑制rankl诱导的NF-κB和MAPK通路,下调破骨细胞标志物的表达。在LPS和RANKL处理下,果皮提取物显著降低活性氧的产生,同时增加抗氧化基因的表达。此外,它们增加了成骨细胞的活力,并轻微提高了碱性磷酸酶的活性。这些结果表明,MSP果皮可作为一种功能性食品来缓解氧化应激和炎症相关性骨质疏松症。
{"title":"Anthocyanins from a new hybrid sweet potato peel cultivated in Northern Thailand mitigate LPS-induced inflammation and RANKL-induced osteoporosis by regulating ROS-mediated pathways.","authors":"Chalermpong Saenjum, Arthid Thim-Uam, Chakkraphong Khonthun, Panida Oonlao, Piyawan Nuntaboon, Young-Joon Surh, Kanokkarn Phromnoi","doi":"10.1007/s10787-024-01634-5","DOIUrl":"https://doi.org/10.1007/s10787-024-01634-5","url":null,"abstract":"<p><p>Maejo 341 Sweet potato (MSP) is a new purple sweet potato variety cultivated in Northern Thailand, but its health benefits are unknown. This study aimed to investigate its antioxidant, anti-inflammatory, and anti-osteoporotic activities, as well as its anthocyanin content. The peel and flesh of MSP were extracted with ethanol and water. Compared with the flesh extracts, the peel extracts presented greater antioxidant capacity and were rich in phenolics, flavonoids, and anthocyanins, namely, cyanidin-3-O-glucoside, peonidin-3-O-glucoside, pelargonidin-3-O-glucoside, cyanidin, and peonidin. The peel extracts suppressed lipopolysaccharide-induced inflammation by inhibiting the secretion of proinflammatory cytokines and enzymes, including TNF-α, IL-1β, IL-6, COX-2, and iNOS, as well as reducing nitric oxide and matrix metalloproteinase-9 secretion. The extracts inhibited the RANKL-induced NF-κB and MAPK pathways and downregulated osteoclastogenic marker expression. Under LPS and RANKL treatment, the peel extracts notably reduced reactive oxygen species production while increasing antioxidant gene expression. Furthermore, they increased osteoblast viability and slightly raise alkaline phosphatase activity. These findings suggest that MSP peel could be used as a functional food to alleviate oxidative stress and inflammation-related osteoporosis.</p>","PeriodicalId":13551,"journal":{"name":"Inflammopharmacology","volume":" ","pages":""},"PeriodicalIF":4.6,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142978332","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-09DOI: 10.1007/s10787-024-01628-3
Sahar M El-Haggar, Dina S Attalla, Mostafa Elhelbawy, Dalia R El-Afify
Objective: This study aimed to assess the potential antifibrotic impact of zinc sulfate in chronic Hepatitis C Virus (HCV) patients receiving direct-acting antiviral therapy.
Methods: This randomized controlled study included 50 chronic HCV-infected patients with fibrosis stage (F1 & F2). Participants were randomly assigned to two groups: Group 1 (Control group, n = 25) received standard direct-acting antiviral therapy for 3 months, while Group 2 (Zinc group, n = 25) received 50 mg/day of zinc sulfate in addition to the standard direct-acting antiviral therapy for the same duration. Baseline and 3-month post-intervention assessments included evaluating serum levels of hyaluronic acid, transforming growth factor beta-1, and fibronectin. Furthermore, indices of liver fibrosis, such as the Fibrosis Index based on the 4 factors (FIB-4) and the Aspartate Transaminase-to-Platelet-Ratio Index (APRI), were calculated during these assessments.
Results: At baseline, the two studied groups had no statistical difference in demographic and laboratory data. After treatment, serum zinc levels significantly increased in the zinc-treated group compared to the control group. Additionally, serum fibronectin and hyaluronic acid levels were significantly reduced in group 2 (zinc group) compared to group 1 (control group). Moreover, zinc group showed lower APRI scores than the control group after a 3-month follow-up period, but there was non-significant difference in FIB-4 scores between the two groups after treatment. Furthermore, total bilirubin levels were reduced after zinc therapy for 3 months.
Conclusions: Administering zinc sulfate could potentially serve as a safe and efficient therapeutic strategy for the management of hepatic fibrosis in individuals with chronic hepatitis C virus.
Trial registration: ClinicalTrials.gov identifier: NCT05465434, On 19/7/2022.
{"title":"A randomized clinical study to evaluate the possible antifibrotic effect of zinc sulfate in chronic HCV patient receiving direct-acting anti-viral therapy.","authors":"Sahar M El-Haggar, Dina S Attalla, Mostafa Elhelbawy, Dalia R El-Afify","doi":"10.1007/s10787-024-01628-3","DOIUrl":"https://doi.org/10.1007/s10787-024-01628-3","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to assess the potential antifibrotic impact of zinc sulfate in chronic Hepatitis C Virus (HCV) patients receiving direct-acting antiviral therapy.</p><p><strong>Methods: </strong>This randomized controlled study included 50 chronic HCV-infected patients with fibrosis stage (F1 & F2). Participants were randomly assigned to two groups: Group 1 (Control group, n = 25) received standard direct-acting antiviral therapy for 3 months, while Group 2 (Zinc group, n = 25) received 50 mg/day of zinc sulfate in addition to the standard direct-acting antiviral therapy for the same duration. Baseline and 3-month post-intervention assessments included evaluating serum levels of hyaluronic acid, transforming growth factor beta-1, and fibronectin. Furthermore, indices of liver fibrosis, such as the Fibrosis Index based on the 4 factors (FIB-4) and the Aspartate Transaminase-to-Platelet-Ratio Index (APRI), were calculated during these assessments.</p><p><strong>Results: </strong>At baseline, the two studied groups had no statistical difference in demographic and laboratory data. After treatment, serum zinc levels significantly increased in the zinc-treated group compared to the control group. Additionally, serum fibronectin and hyaluronic acid levels were significantly reduced in group 2 (zinc group) compared to group 1 (control group). Moreover, zinc group showed lower APRI scores than the control group after a 3-month follow-up period, but there was non-significant difference in FIB-4 scores between the two groups after treatment. Furthermore, total bilirubin levels were reduced after zinc therapy for 3 months.</p><p><strong>Conclusions: </strong>Administering zinc sulfate could potentially serve as a safe and efficient therapeutic strategy for the management of hepatic fibrosis in individuals with chronic hepatitis C virus.</p><p><strong>Trial registration: </strong>ClinicalTrials.gov identifier: NCT05465434, On 19/7/2022.</p>","PeriodicalId":13551,"journal":{"name":"Inflammopharmacology","volume":" ","pages":""},"PeriodicalIF":4.6,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142948033","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-08DOI: 10.1007/s10787-024-01616-7
Ahmed Ramzi, Subhia Maya, Nadeen Balousha, Mufreh Amin, Robert Charles Powell, Mostafa Ramzi Shiha
Background: Chronic inflammation has been linked to many psychiatric disorders, and therefore, pertinent anti-inflammatory therapies have been empirically evaluated for management. An enduring example of long-term safety, attainability, and versatility has been pentoxifylline (PTX). PTX is a phosphodiesterase inhibitor that modulates inflammatory mediators and affects most blood components and the blood vessels.
Methods: Major databases were systematically searched to identify randomized controlled trials (RCTs) on PTX in psychiatric and neuropsychiatric disorders until September 25, 2024.
Results: 21 RCTs were included. Five studies evaluated clinical depression: four on major depressive disorder (MDD) and one on bipolar patients experiencing treatment-resistant depression. PTX significantly reduced depressive symptoms in MDD in the four double-blind, randomized, placebo-controlled trials, with the three studies combining PTX and SSRIs showing statistically significant improvements in response rates. Ten RCTs on cognitive impairment reported beneficial effects, particularly in vascular dementia. Meta-analyses support its efficacy in reducing depressive symptoms, cognitive decline, asthenia, and inflammatory markers.
Conclusion: Exploring the effects of PTX on psychiatric and neuropsychiatric conditions has provided considerable support for its utility across various disorders, most notably in moderate to severe major depressive disorder (as adjunctive therapy with SSRIs) and cognitive impairment in vascular dementia (as monotherapy). Relevantly, the potential of PTX across a wide range of conditions might prove beneficial in cases of co-occurrence.
{"title":"Effects of the anti-inflammatory pentoxifylline on psychiatric and neuropsychiatric conditions: exploring various off-label utilities with meta-analyses.","authors":"Ahmed Ramzi, Subhia Maya, Nadeen Balousha, Mufreh Amin, Robert Charles Powell, Mostafa Ramzi Shiha","doi":"10.1007/s10787-024-01616-7","DOIUrl":"https://doi.org/10.1007/s10787-024-01616-7","url":null,"abstract":"<p><strong>Background: </strong>Chronic inflammation has been linked to many psychiatric disorders, and therefore, pertinent anti-inflammatory therapies have been empirically evaluated for management. An enduring example of long-term safety, attainability, and versatility has been pentoxifylline (PTX). PTX is a phosphodiesterase inhibitor that modulates inflammatory mediators and affects most blood components and the blood vessels.</p><p><strong>Methods: </strong>Major databases were systematically searched to identify randomized controlled trials (RCTs) on PTX in psychiatric and neuropsychiatric disorders until September 25, 2024.</p><p><strong>Results: </strong>21 RCTs were included. Five studies evaluated clinical depression: four on major depressive disorder (MDD) and one on bipolar patients experiencing treatment-resistant depression. PTX significantly reduced depressive symptoms in MDD in the four double-blind, randomized, placebo-controlled trials, with the three studies combining PTX and SSRIs showing statistically significant improvements in response rates. Ten RCTs on cognitive impairment reported beneficial effects, particularly in vascular dementia. Meta-analyses support its efficacy in reducing depressive symptoms, cognitive decline, asthenia, and inflammatory markers.</p><p><strong>Conclusion: </strong>Exploring the effects of PTX on psychiatric and neuropsychiatric conditions has provided considerable support for its utility across various disorders, most notably in moderate to severe major depressive disorder (as adjunctive therapy with SSRIs) and cognitive impairment in vascular dementia (as monotherapy). Relevantly, the potential of PTX across a wide range of conditions might prove beneficial in cases of co-occurrence.</p>","PeriodicalId":13551,"journal":{"name":"Inflammopharmacology","volume":" ","pages":""},"PeriodicalIF":4.6,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142947967","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-07DOI: 10.1007/s10787-024-01632-7
Mazaghul Basar, Muhammad Imran Khan, Muhammad Furqan Akhtar, Fareeha Anwar, Ammara Saleem, Asadullah Madni, Zulcaif Ahmad, Ali Sharif, Bushra Akhtar, Uzma Shakoor, Aslam Khan
<p><p>Flurbiprofen (FBP) is poorly water-soluble BCS class II drug with anti-inflammatory and analgesic effects, used to treat arthritis and degenerative joint diseases. This study was aimed to develop SNEDDS loaded with FBP. Six SNEDDS using two oils olive oil (F<sub>1OLV</sub>, F<sub>2OLV</sub>, F<sub>3OLV</sub>) and castor oil (F<sub>4CAS</sub>, F<sub>5CAS</sub>, F<sub>6CAS</sub>) with three different Smix ratios consisting of Tween 20 and PEG 400 (1:1, 1:2, 2:1) were prepared and characterized. Compatibility between FBP and polymers was investigated using FTIR. SNEDDS were characterized for physicochemical attributes. Two optimized formulations were investigated at 10 mg/kg dose given orally in Wistar rats for analgesic activity by hot plate and tail flick methods, and anti-inflammatory activity by carrageenan induced paw edema method. Anti-inflammatory activity was further explored by motor coordination and motility by Rota rod and cage activity tests. Following anesthesia blood samples were collected before dissection to measure inflammatory mediators and oxidative stress markers. Sciatica nerves and hind paws of rats were also removed for histopathological evaluation. FTIR studies revealed compatibility of FBP with other components. Droplet size of F<sub>1OLV</sub>, F<sub>2OLV</sub>, F<sub>3OLV</sub> was 128.5 ± 0.7 nm, 202.5 ± 1.3 nm, and 541.5 ± 1.7 nm, whereas it was 142.5 ± 1.1 nm, 215.4 ± 1.2 nm and 349.9 ± 1.8 nm for F<sub>4CAS</sub>, F<sub>5CAS</sub>, F<sub>6CAS</sub>. %EE of F<sub>1OLV</sub>, F<sub>2OLV</sub>, F<sub>3OLV</sub> was found 85 ± 4.89%-91 ± 4.67%, whereas the %EE F<sub>4CAS</sub>, F<sub>5CAS</sub>, F<sub>6CAS</sub> was 84 ± 4.15%-90 ± 4.21%. DSC curves of F<sub>1OLV</sub> and F<sub>4CAS</sub> revealed amorphous nature of the FBP. SEM showed spherical shape of globules. % of drug released in the pH medium 1.2 for plain FBP, F<sub>1OLV</sub> and F<sub>4CAS</sub> was 25%, 59% and 57%. % drug released in the pH 6.8 for plain FBP, F<sub>1OLV</sub> and F<sub>4CAS</sub> was 59%, 85% and 83%. Oral administration of FBP-loaded SNEDDS (F<sub>1OLV</sub> and F<sub>4CAS</sub>) significantly decreased paw diameter and enhanced motor coordination in rats when compared to the disease control group. This was linked to the ability of FBP to reduce inflammation and oxidative stress, with histological studies indicating decreased tissue damage in SNEDDS treated groups, implying the possibility of tissue recovery. Administration of both formulations started to demonstrate analgesic and anti-inflammatory effects after one hour of administration. In addition to anti-inflammatory effect, both formulations improved motor coordination, motility, and reduced infiltration of inflammatory cells in the inflamed paws. The anti-inflammatory and analgesic activities were attributed to decreased serum levels of IL-6 and TNF-α, increased activity of SOD and reduced nitrite content in sciatic nerves. Histopathological evaluation revealed reduced vasculari
{"title":"Olive oil and castor oil-based self-nanoemulsifying drug delivery system of flurbiprofen can relieve peripheral pain and inflammation through reduction of oxidative stress and inflammatory biomarkers: a comprehensive formulation and pharmacological insights.","authors":"Mazaghul Basar, Muhammad Imran Khan, Muhammad Furqan Akhtar, Fareeha Anwar, Ammara Saleem, Asadullah Madni, Zulcaif Ahmad, Ali Sharif, Bushra Akhtar, Uzma Shakoor, Aslam Khan","doi":"10.1007/s10787-024-01632-7","DOIUrl":"https://doi.org/10.1007/s10787-024-01632-7","url":null,"abstract":"<p><p>Flurbiprofen (FBP) is poorly water-soluble BCS class II drug with anti-inflammatory and analgesic effects, used to treat arthritis and degenerative joint diseases. This study was aimed to develop SNEDDS loaded with FBP. Six SNEDDS using two oils olive oil (F<sub>1OLV</sub>, F<sub>2OLV</sub>, F<sub>3OLV</sub>) and castor oil (F<sub>4CAS</sub>, F<sub>5CAS</sub>, F<sub>6CAS</sub>) with three different Smix ratios consisting of Tween 20 and PEG 400 (1:1, 1:2, 2:1) were prepared and characterized. Compatibility between FBP and polymers was investigated using FTIR. SNEDDS were characterized for physicochemical attributes. Two optimized formulations were investigated at 10 mg/kg dose given orally in Wistar rats for analgesic activity by hot plate and tail flick methods, and anti-inflammatory activity by carrageenan induced paw edema method. Anti-inflammatory activity was further explored by motor coordination and motility by Rota rod and cage activity tests. Following anesthesia blood samples were collected before dissection to measure inflammatory mediators and oxidative stress markers. Sciatica nerves and hind paws of rats were also removed for histopathological evaluation. FTIR studies revealed compatibility of FBP with other components. Droplet size of F<sub>1OLV</sub>, F<sub>2OLV</sub>, F<sub>3OLV</sub> was 128.5 ± 0.7 nm, 202.5 ± 1.3 nm, and 541.5 ± 1.7 nm, whereas it was 142.5 ± 1.1 nm, 215.4 ± 1.2 nm and 349.9 ± 1.8 nm for F<sub>4CAS</sub>, F<sub>5CAS</sub>, F<sub>6CAS</sub>. %EE of F<sub>1OLV</sub>, F<sub>2OLV</sub>, F<sub>3OLV</sub> was found 85 ± 4.89%-91 ± 4.67%, whereas the %EE F<sub>4CAS</sub>, F<sub>5CAS</sub>, F<sub>6CAS</sub> was 84 ± 4.15%-90 ± 4.21%. DSC curves of F<sub>1OLV</sub> and F<sub>4CAS</sub> revealed amorphous nature of the FBP. SEM showed spherical shape of globules. % of drug released in the pH medium 1.2 for plain FBP, F<sub>1OLV</sub> and F<sub>4CAS</sub> was 25%, 59% and 57%. % drug released in the pH 6.8 for plain FBP, F<sub>1OLV</sub> and F<sub>4CAS</sub> was 59%, 85% and 83%. Oral administration of FBP-loaded SNEDDS (F<sub>1OLV</sub> and F<sub>4CAS</sub>) significantly decreased paw diameter and enhanced motor coordination in rats when compared to the disease control group. This was linked to the ability of FBP to reduce inflammation and oxidative stress, with histological studies indicating decreased tissue damage in SNEDDS treated groups, implying the possibility of tissue recovery. Administration of both formulations started to demonstrate analgesic and anti-inflammatory effects after one hour of administration. In addition to anti-inflammatory effect, both formulations improved motor coordination, motility, and reduced infiltration of inflammatory cells in the inflamed paws. The anti-inflammatory and analgesic activities were attributed to decreased serum levels of IL-6 and TNF-α, increased activity of SOD and reduced nitrite content in sciatic nerves. Histopathological evaluation revealed reduced vasculari","PeriodicalId":13551,"journal":{"name":"Inflammopharmacology","volume":" ","pages":""},"PeriodicalIF":4.6,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142948102","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Alzheimer's disease (AD) is a type of neurodegenerative disease that describes cognitive decline and memory loss resulting in disability in movement, memory, speech etc. Which first affects the hippocampal and entorhinal cortex regions of brain. Pathogenesis of AD depends on Amyloid-β, hyper-phosphorylation of tau protein, mitochondrial dysfunction, cholinergic hypothesis and oxidative stress. In comparison with males, females are more prone to AD due to reduced estrogen level. Some of the FDA-approved drugs and their conventional formulations available in the market are discussed in this review. Nose-to-brain delivery system provides the target specific drug delivery via olfactory and trigeminal nerve (active and passive drug targeting strategies) and bypassing the Blood Brain Barrier. Mucoadhesive agents and permeation enhancers are mostly utilized to enhance the retention time and bioavailability of the drugs. Liposomes, niosomes, cubosomes, solid lipid nanoparticles, nanoemulsions, micelles, and many more nanocarriers for nose-to-brain delivery of drugs are also described thoroughly in this review. It also covers the clinical trials and patents for nose-to-brain delivery. In this article, we investigate the nose-to-brain pathways for AD treatment strategies.
{"title":"Advancements in nose-to-brain drug targeting for Alzheimer's disease: a review of nanocarriers and clinical insights.","authors":"Kumari Komal, Rashmi Ghosh, Debayan Sil, Rohit Sharma, Sourabh Kumar, Prachi Pandey, Manish Kumar","doi":"10.1007/s10787-024-01636-3","DOIUrl":"https://doi.org/10.1007/s10787-024-01636-3","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is a type of neurodegenerative disease that describes cognitive decline and memory loss resulting in disability in movement, memory, speech etc. Which first affects the hippocampal and entorhinal cortex regions of brain. Pathogenesis of AD depends on Amyloid-β, hyper-phosphorylation of tau protein, mitochondrial dysfunction, cholinergic hypothesis and oxidative stress. In comparison with males, females are more prone to AD due to reduced estrogen level. Some of the FDA-approved drugs and their conventional formulations available in the market are discussed in this review. Nose-to-brain delivery system provides the target specific drug delivery via olfactory and trigeminal nerve (active and passive drug targeting strategies) and bypassing the Blood Brain Barrier. Mucoadhesive agents and permeation enhancers are mostly utilized to enhance the retention time and bioavailability of the drugs. Liposomes, niosomes, cubosomes, solid lipid nanoparticles, nanoemulsions, micelles, and many more nanocarriers for nose-to-brain delivery of drugs are also described thoroughly in this review. It also covers the clinical trials and patents for nose-to-brain delivery. In this article, we investigate the nose-to-brain pathways for AD treatment strategies.</p>","PeriodicalId":13551,"journal":{"name":"Inflammopharmacology","volume":" ","pages":""},"PeriodicalIF":4.6,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142947966","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}