Pub Date : 2023-12-13DOI: 10.1007/s00210-023-02893-9
Ebtesam A. Mohamad, Abeer A. Ali, Marwa Sharaky, Reem H. El-Gebaly
Scientists are seeking to find an effective treatment for tumors that has no side effects. N-Acetyl-l-cysteine (NAC) is a thiol compound extracted from garlic. Current study explores the potential of NAC-loaded niosomes (NAC-NIO) for tumor treatment in mice. NAC-loaded niosomes’ efficiency, morphology, UV absorption, size distribution, zeta potential, release, and FTIR analysis were evaluated. For vivo study, 25 male BALB/c mice were divided to five groups: gp1 negative control (receive saline), gp2 positive control (tumor group), gp3 treated with NAC, gp4 treated with NAC-NIO at the same time of tumor injection, and gp5 treated with NAC-NIO after tumor growth (day 14). The impact of NAC-NIO on the tumor treatment was evaluated by measuring tumor size progress, comet assay, oxidative stress parameters (GSH, nitric oxide, MDA), western blot analysis, and histopathological investigation of tissues. NAC-NIO showed 72 ± 3% encapsulation efficiency and zeta potential − 5.95 mV with spherical shape. It was found that oral administration of NAC-NIO in a dose of 50 mg/kg provided significant protection against tumor cells. Our formulation decreases DNA injury significantly (P < 0.05). It was noticed that NAC-NIO can increase oxidative stress levels in tumor tissue. On the other hand, the caspase 3 and caspase 9 gene expression were upregulated significantly (P < 0.001) in mice administrated NAC-NIO compared with all other groups. Histological studies confirmed the protective effect of NAC-NIO against tumor especially for treatment during tumor growth protocol. The results suggested that oral delivery of NAC-NIO formulation improved antioxidant effect.
{"title":"Niosomes loading N-acetyl-L-cysteine for cancer treatment in vivo study","authors":"Ebtesam A. Mohamad, Abeer A. Ali, Marwa Sharaky, Reem H. El-Gebaly","doi":"10.1007/s00210-023-02893-9","DOIUrl":"https://doi.org/10.1007/s00210-023-02893-9","url":null,"abstract":"<p>Scientists are seeking to find an effective treatment for tumors that has no side effects. N-Acetyl-l-cysteine (NAC) is a thiol compound extracted from garlic. Current study explores the potential of NAC-loaded niosomes (NAC-NIO) for tumor treatment in mice. NAC-loaded niosomes’ efficiency, morphology, UV absorption, size distribution, zeta potential, release, and FTIR analysis were evaluated. For vivo study, 25 male BALB/c mice were divided to five groups: gp1 negative control (receive saline), gp2 positive control (tumor group), gp3 treated with NAC, gp4 treated with NAC-NIO at the same time of tumor injection, and gp5 treated with NAC-NIO after tumor growth (day 14). The impact of NAC-NIO on the tumor treatment was evaluated by measuring tumor size progress, comet assay, oxidative stress parameters (GSH, nitric oxide, MDA), western blot analysis, and histopathological investigation of tissues. NAC-NIO showed 72 ± 3% encapsulation efficiency and zeta potential − 5.95 mV with spherical shape. It was found that oral administration of NAC-NIO in a dose of 50 mg/kg provided significant protection against tumor cells. Our formulation decreases DNA injury significantly (<i>P</i> < 0.05). It was noticed that NAC-NIO can increase oxidative stress levels in tumor tissue. On the other hand, the caspase 3 and caspase 9 gene expression were upregulated significantly (<i>P</i> < 0.001) in mice administrated NAC-NIO compared with all other groups. Histological studies confirmed the protective effect of NAC-NIO against tumor especially for treatment during tumor growth protocol. The results suggested that oral delivery of NAC-NIO formulation improved antioxidant effect.</p>","PeriodicalId":18862,"journal":{"name":"Naunyn-schmiedebergs Archives of Pharmacology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138629040","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Non-alcoholic fatty liver disease (NAFLD) is a hepatic manifestation of metabolic syndrome. Vitamin E (VE) has antioxidant properties and can mediate lipid metabolism. Non-targeted metabolomics technology was employed to uncover comprehensively the metabolome of VE in NAFLD rats. NAFLD model was created with a high-fat and high-cholesterol diet (HFD) in rats. NAFLD rats in the VE group were given 75 mg/(kg day) VE. The metabolites in the serum of rats were identified via UPLC and Q-TOF/MS analysis. KEGG was applied for the pathway enrichment. VE improved the liver function, lipid metabolism, and oxidative stress in NAFLD rats induced by HFD. Based on the metabolite profile data, 132 differential metabolites were identified between VE group and the HFD group, mainly including pyridoxamine, betaine, and bretylium. According to the KEGG results, biosynthesis of cofactors was a key metabolic pathway of VE in NAFLD rats. VE can alleviate NAFLD induced by HFD, and the underlying mechanism is associated with the biosynthesis of cofactors, mainly including pyridoxine and betaine.
{"title":"Study on the mechanism of vitamin E alleviating non-alcoholic fatty liver function based on non-targeted metabolomics analysis in rats","authors":"Baiyun Zhao, Jing Zhang, Kaiyue Zhao, Wenbin Zhao, Yajuan Shi, Jing Liu, Ling Zeng, Chaoxuan Wang, Xin Zeng, Junping Shi","doi":"10.1007/s00210-023-02864-0","DOIUrl":"https://doi.org/10.1007/s00210-023-02864-0","url":null,"abstract":"<p>Non-alcoholic fatty liver disease (NAFLD) is a hepatic manifestation of metabolic syndrome. Vitamin E (VE) has antioxidant properties and can mediate lipid metabolism. Non-targeted metabolomics technology was employed to uncover comprehensively the metabolome of VE in NAFLD rats. NAFLD model was created with a high-fat and high-cholesterol diet (HFD) in rats. NAFLD rats in the VE group were given 75 mg/(kg day) VE. The metabolites in the serum of rats were identified via UPLC and Q-TOF/MS analysis. KEGG was applied for the pathway enrichment. VE improved the liver function, lipid metabolism, and oxidative stress in NAFLD rats induced by HFD. Based on the metabolite profile data, 132 differential metabolites were identified between VE group and the HFD group, mainly including pyridoxamine, betaine, and bretylium. According to the KEGG results, biosynthesis of cofactors was a key metabolic pathway of VE in NAFLD rats. VE can alleviate NAFLD induced by HFD, and the underlying mechanism is associated with the biosynthesis of cofactors, mainly including pyridoxine and betaine.</p>","PeriodicalId":18862,"journal":{"name":"Naunyn-schmiedebergs Archives of Pharmacology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138629053","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Polymyxin E or colistin is an effective antibiotic against MDR Gram-negative bacteria. Due to unwanted side effects, the use of this antibiotic has been limited for a long time, but in recent years, the widespread of MDR Gram-negative bacteria infections has led to its reintroduction. Neurotoxicity and nephrotoxicity are the significant dose-limiting adverse effects of colistin. Several agents with anti-inflammatory and antioxidant properties have been used for the prevention of colistin-induced neurotoxicity. This study aims to review the preclinical studies in this field to prepare guidance for future human studies. The data was achieved by searching PubMed, Scopus, and Google Scholar databases. All eligible pre-clinical studies performed on neuroprotective agents against colistin-induced neurotoxicity, which were published up to September 2023, were included. Finally, 16 studies (ten in vitro and eight in vivo) are reviewed. Apoptosis (in 13 studies), inflammatory (in four studies), and oxidative stress (in 14 studies) pathways are the most commonly reported pathways involved in colistin-induced neurotoxicity. The assessed compounds include non-herbal (e.g., ascorbic acid, rapamycin, and minocycline) and herbal (e.g., curcumin, rutin, baicalein, salidroside, and ginsenoside) agents. Besides these compounds, some other measures like transplantation of mitochondria and the use of nerve growth factor and mesenchymal stem cells could be motivating subjects for future research. Based on the data from experimental (in vitro and animal) studies, a combination of colistin with neuroprotective agents could prevent or decrease colistin-induced neurotoxicity. However, well-designed randomized clinical trials and human studies are essential for demonstrating efficacy.
{"title":"Prevention of colistin-induced neurotoxicity: a narrative review of preclinical data","authors":"Setareh Soroudi, Ghazal Mousavi, Fatemeh Jafari, Sepideh Elyasi","doi":"10.1007/s00210-023-02884-w","DOIUrl":"https://doi.org/10.1007/s00210-023-02884-w","url":null,"abstract":"<p>Polymyxin E or colistin is an effective antibiotic against MDR Gram-negative bacteria. Due to unwanted side effects, the use of this antibiotic has been limited for a long time, but in recent years, the widespread of MDR Gram-negative bacteria infections has led to its reintroduction. Neurotoxicity and nephrotoxicity are the significant dose-limiting adverse effects of colistin. Several agents with anti-inflammatory and antioxidant properties have been used for the prevention of colistin-induced neurotoxicity. This study aims to review the preclinical studies in this field to prepare guidance for future human studies. The data was achieved by searching PubMed, Scopus, and Google Scholar databases. All eligible pre-clinical studies performed on neuroprotective agents against colistin-induced neurotoxicity, which were published up to September 2023, were included. Finally, 16 studies (ten in vitro and eight in vivo) are reviewed. Apoptosis (in 13 studies), inflammatory (in four studies), and oxidative stress (in 14 studies) pathways are the most commonly reported pathways involved in colistin-induced neurotoxicity. The assessed compounds include non-herbal (e.g., ascorbic acid, rapamycin, and minocycline) and herbal (e.g., curcumin, rutin, baicalein, salidroside, and ginsenoside) agents. Besides these compounds, some other measures like transplantation of mitochondria and the use of nerve growth factor and mesenchymal stem cells could be motivating subjects for future research. Based on the data from experimental (in vitro and animal) studies, a combination of colistin with neuroprotective agents could prevent or decrease colistin-induced neurotoxicity. However, well-designed randomized clinical trials and human studies are essential for demonstrating efficacy.</p>","PeriodicalId":18862,"journal":{"name":"Naunyn-schmiedebergs Archives of Pharmacology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138629403","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-12-12DOI: 10.1007/s00210-023-02880-0
Penghui Bu, Weipeng Xie, Sicheng Wang, Zhi Yang, Kan Peng, Weisong Zhang, Shouye Hu
Recently, epidermal growth factor-like domain protein 6 (EGFL6) was proposed as a candidate gene for coupling angiogenesis to osteogenesis during bone repair; however, the exact role and underlying mechanism are largely unknown. Here, using immunohistochemical and Western blotting analyses, we found that EGFL6 was downregulated in the femoral head tissue of patients with steroid-induced osteonecrosis of the femoral head (SONFH) compared to patients with traumatic femoral neck fracture (FNF), accompanied by significantly downregulation of osteogenic and angiogenic marker genes. Then, bone marrow mesenchymal stem cells (BMSCs) were isolated from the FNF and the SONFH patients, respectively, and after identification by immunofluorescence staining surface markers, the effect of EGFL6 on their abilities of osteogenic differentiation and angiogenesis was evaluated. Our results of alizarin red staining and tubular formation experiment revealed that BMSCs from the SONFH patients (SONFH-BMSCs) displayed an obviously weaker ability of osteogenesis than FNF-BMSCs, and EGFL6 overexpression improved the abilities of osteogenic differentiation and angiogenesis of SONFH-BMSCs. Moreover, EGFL6 overexpression activated extracellular signal-regulated kinases 1/2 (ERK1/2). ERK1/2 inhibitor U0126 reversed the promoting effect of EGFL6 overexpression on the expression of osteogenesis and angiogenesis-related genes in the SONFH femoral head. In conclusion, EGFL6 plays a protective role in SONFH, it promotes osteogenesis and angiogenesis of BMSCs, and its effect is likely to be related to ERK1/2 activation.
最近,表皮生长因子样结构域蛋白6(EGFL6)被认为是骨修复过程中血管生成与骨生成耦合的候选基因,但其确切作用和内在机制尚不清楚。在此,我们通过免疫组化和 Western 印迹分析发现,与外伤性股骨颈骨折(FNF)患者相比,类固醇诱导的股骨头坏死(SONFH)患者股骨头组织中的 EGFL6 下调,同时伴随成骨和血管生成标记基因的显著下调。然后,分别从FNF和SONFH患者体内分离出骨髓间充质干细胞(BMSCs),经免疫荧光染色表面标记物鉴定后,评估EGFL6对其成骨分化和血管生成能力的影响。茜素红染色和小管形成实验结果显示,SONFH患者的BMSCs(SONFH-BMSCs)的成骨能力明显弱于FNF-BMSCs,而EGFL6的过表达提高了SONFH-BMSCs的成骨分化和血管生成能力。此外,EGFL6的过表达激活了细胞外信号调节激酶1/2(ERK1/2)。ERK1/2抑制剂U0126逆转了EGFL6过表达对SONFH股骨头成骨和血管生成相关基因表达的促进作用。总之,EGFL6在SONFH中起着保护作用,它能促进BMSCs的成骨和血管生成,其作用可能与ERK1/2的激活有关。
{"title":"EGFL6 activates the ERK signaling to improve angiogenesis and osteogenesis of BMSCs in patients with steroid-induced osteonecrosis of the femoral head","authors":"Penghui Bu, Weipeng Xie, Sicheng Wang, Zhi Yang, Kan Peng, Weisong Zhang, Shouye Hu","doi":"10.1007/s00210-023-02880-0","DOIUrl":"https://doi.org/10.1007/s00210-023-02880-0","url":null,"abstract":"<p>Recently, epidermal growth factor-like domain protein 6 (<i>EGFL6</i>) was proposed as a candidate gene for coupling angiogenesis to osteogenesis during bone repair; however, the exact role and underlying mechanism are largely unknown. Here, using immunohistochemical and Western blotting analyses, we found that EGFL6 was downregulated in the femoral head tissue of patients with steroid-induced osteonecrosis of the femoral head (SONFH) compared to patients with traumatic femoral neck fracture (FNF), accompanied by significantly downregulation of osteogenic and angiogenic marker genes. Then, bone marrow mesenchymal stem cells (BMSCs) were isolated from the FNF and the SONFH patients, respectively, and after identification by immunofluorescence staining surface markers, the effect of EGFL6 on their abilities of osteogenic differentiation and angiogenesis was evaluated. Our results of alizarin red staining and tubular formation experiment revealed that BMSCs from the SONFH patients (SONFH-BMSCs) displayed an obviously weaker ability of osteogenesis than FNF-BMSCs, and EGFL6 overexpression improved the abilities of osteogenic differentiation and angiogenesis of SONFH-BMSCs. Moreover, EGFL6 overexpression activated extracellular signal-regulated kinases 1/2 (ERK1/2). ERK1/2 inhibitor U0126 reversed the promoting effect of EGFL6 overexpression on the expression of osteogenesis and angiogenesis-related genes in the SONFH femoral head. In conclusion, EGFL6 plays a protective role in SONFH, it promotes osteogenesis and angiogenesis of BMSCs, and its effect is likely to be related to ERK1/2 activation.</p>","PeriodicalId":18862,"journal":{"name":"Naunyn-schmiedebergs Archives of Pharmacology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138573929","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-12-12DOI: 10.1007/s00210-023-02881-z
Martha Ivonne Sánchez-Trigueros, Ivette Astrid Martínez-Vieyra, Elizabeth Arlen Pineda-Peña, Gilberto Castañeda-Hernández, Claudia Perez-Cruz, Doris Cerecedo, Aracely Evangelina Chávez-Piña
Therapeutic effect of non-steroidal anti-inflammatory drugs (NSAIDs) has been related with gastrointestinal injury. Docosahexaenoic acid (DHA), an omega-3 polyunsaturated fatty acid (PUFA), can prevent gastric and small intestinal damage. Nonetheless, contribution of antioxidative action in the protective effect of DHA has not been evaluated before in the small intestine injury after indomethacin treatment. Pathogenesis of NSAID-induced small intestinal injury is multifactorial, and reactive oxidative species have been related to indomethacin’s small intestinal damage. The present work aimed to evaluate antioxidative activity in the protective action of DHA in the indomethacin-induced small intestinal damage. Female Wistar rats were gavage with DHA (3 mg/kg) or omeprazole (3 mg/kg) for 10 days. Each rat received indomethacin (3 mg/kg, orally) daily to induce small intestinal damage. The total area of intestinal ulcers and histopathological analysis were performed. In DHA-treated rats, myeloperoxidase and superoxide dismutase activity, glutathione, malondialdehyde, leukotriene, and lipopolysaccharide (LPS) levels were measured. Furthermore, the relative abundance of selective bacteria was assessed. DHA administration (3 mg/kg, p.o.) caused a significant decrease in indomethacin-induced small intestinal injury in Wistar rats after 10 days of treatment. DHA’s enteroprotection resulted from the prevention of an increase in myeloperoxidase activity, and lipoperoxidation, as well as an improvement in the antioxidant defenses, such as glutathione levels and superoxide dismutase activity in the small intestine. Furthermore, we showed that DHA’s enteroprotective effect decreased significantly LPS levels in indomethacin-induced injury in small intestine. Our data suggest that DHA’s enteroprotective might be attributed to the prevention of oxidative stress.
非甾体抗炎药(NSAIDs)的治疗效果与胃肠道损伤有关。二十二碳六烯酸(DHA)是一种欧米伽-3 多不饱和脂肪酸(PUFA),可预防胃和小肠损伤。然而,在吲哚美辛治疗后的小肠损伤中,DHA的保护作用中抗氧化作用的贡献尚未得到评估。非甾体抗炎药诱发小肠损伤的发病机制是多因素的,活性氧化物与吲哚美辛的小肠损伤有关。本研究旨在评估 DHA 对吲哚美辛诱导的小肠损伤的保护作用中的抗氧化活性。雌性 Wistar 大鼠连续 10 天灌胃 DHA(3 毫克/千克)或奥美拉唑(3 毫克/千克)。每只大鼠每天口服吲哚美辛(3 毫克/千克)以诱发小肠损伤。对肠溃疡的总面积和组织病理学进行分析。在 DHA 处理的大鼠中,测量了髓过氧化物酶和超氧化物歧化酶活性、谷胱甘肽、丙二醛、白三烯和脂多糖(LPS)水平。此外,还评估了选择性细菌的相对丰度。给 Wistar 大鼠服用 DHA(3 毫克/千克,口服)10 天后,吲哚美辛引起的小肠损伤明显减轻。DHA 的肠道保护作用来自于防止髓过氧化物酶活性和脂肪过氧化的增加,以及改善抗氧化防御功能,如谷胱甘肽水平和小肠中超氧化物歧化酶的活性。此外,我们还发现,在吲哚美辛诱导的小肠损伤中,DHA的肠道保护作用显著降低了LPS水平。我们的数据表明,DHA的肠道保护作用可能归因于对氧化应激的预防。
{"title":"Role of antioxidative activity in the docosahexaenoic acid’s enteroprotective effect in the indomethacin-induced small intestinal injury model","authors":"Martha Ivonne Sánchez-Trigueros, Ivette Astrid Martínez-Vieyra, Elizabeth Arlen Pineda-Peña, Gilberto Castañeda-Hernández, Claudia Perez-Cruz, Doris Cerecedo, Aracely Evangelina Chávez-Piña","doi":"10.1007/s00210-023-02881-z","DOIUrl":"https://doi.org/10.1007/s00210-023-02881-z","url":null,"abstract":"<p>Therapeutic effect of non-steroidal anti-inflammatory drugs (NSAIDs) has been related with gastrointestinal injury. Docosahexaenoic acid (DHA), an omega-3 polyunsaturated fatty acid (PUFA), can prevent gastric and small intestinal damage. Nonetheless, contribution of antioxidative action in the protective effect of DHA has not been evaluated before in the small intestine injury after indomethacin treatment. Pathogenesis of NSAID-induced small intestinal injury is multifactorial, and reactive oxidative species have been related to indomethacin’s small intestinal damage. The present work aimed to evaluate antioxidative activity in the protective action of DHA in the indomethacin-induced small intestinal damage. Female Wistar rats were gavage with DHA (3 mg/kg) or omeprazole (3 mg/kg) for 10 days. Each rat received indomethacin (3 mg/kg, orally) daily to induce small intestinal damage. The total area of intestinal ulcers and histopathological analysis were performed. In DHA-treated rats, myeloperoxidase and superoxide dismutase activity, glutathione, malondialdehyde, leukotriene, and lipopolysaccharide (LPS) levels were measured. Furthermore, the relative abundance of selective bacteria was assessed. DHA administration (3 mg/kg, p.o.) caused a significant decrease in indomethacin-induced small intestinal injury in Wistar rats after 10 days of treatment. DHA’s enteroprotection resulted from the prevention of an increase in myeloperoxidase activity, and lipoperoxidation, as well as an improvement in the antioxidant defenses, such as glutathione levels and superoxide dismutase activity in the small intestine. Furthermore, we showed that DHA’s enteroprotective effect decreased significantly LPS levels in indomethacin-induced injury in small intestine. Our data suggest that DHA’s enteroprotective might be attributed to the prevention of oxidative stress.</p>","PeriodicalId":18862,"journal":{"name":"Naunyn-schmiedebergs Archives of Pharmacology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138574154","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Alzheimer’s disease (AD), a neuro-degenerative disease that primarily affects the elderly, is a worldwide phenomenon. Loss of memory, cognitive decline, behavioural changes, and many other signs are used to classify it. Various hypotheses that may contribute to Alzheimer’s disease have been found during decades of survey, including tau theory, the amyloid theory, the cholinergic hypothesis, and the oxidative stress hypothesis. According to some theories, the two leading causes of AD are the accumulation of amyloid beta plaque and development of NFTs in the brain. The hippocampus and cerebral cortex are the primary sites where amyloid beta plaques gather in the body. NFT formation in the brain impairs the brain’s neurons’ potential of signalling. According to the age at which it manifests in a person, there are two subtypes of AD: ‘LOAD (Late Onset Alzheimer’s Disease)’ and ‘EOAD (Early Onset Alzheimer’s Disease)’. Long-term research into AD treatment has resulted in the introduction of some medications that provided symptomatic relief to patients but did not alter the disease’s pathophysiology, like cholinesterase inhibitors, inhibitors of tau aggregation, and monoclonal antibodies to Aβ aggregation. Even though the medications did not halt the progression of AD, researchers did not discontinue their work, which lead to the introduction of gene therapy — a recently created cutting-edge method of delivering genes to target sites where they can express the intended functionalities. Viral or non-viral vectors could be used to deliver the gene, each with advantages and limitations of their own. Gene therapy is proven to be a potential disease-modifying treatment for AD. This article discusses about gene therapy, its merits and demerits and the various ways of gene delivery. Additionally, it focuses on AD as the target for treatment through gene therapy, the pathophysiology of AD, and the multiple targets for gene therapy in the treatment of AD.
阿尔茨海默病(AD)是一种主要影响老年人的神经退行性疾病,是一种世界性现象。记忆力丧失、认知能力下降、行为改变和许多其他迹象都被用来对其进行分类。在数十年的调查中,人们发现了可能导致阿尔茨海默病的各种假说,包括 tau 理论、淀粉样蛋白理论、胆碱能假说和氧化应激假说。根据一些理论,导致阿兹海默症的两个主要原因是淀粉样蛋白β斑块在大脑中的积累和NFT的发展。海马和大脑皮层是体内淀粉样β斑块聚集的主要部位。脑内 NFT 的形成会损害大脑神经元的信号潜能。根据患者的发病年龄,阿兹海默症可分为两种亚型:"晚发阿兹海默症(LOAD)"和 "早发阿兹海默症(EOAD)"。经过对阿尔茨海默病治疗的长期研究,一些药物(如胆碱酯酶抑制剂、tau 聚集抑制剂和 Aβ 聚集单克隆抗体)已经问世,它们能缓解患者的症状,但不能改变疾病的病理生理学。尽管这些药物并不能阻止注意力缺失症的发展,但研究人员并没有停止他们的工作,这导致了基因疗法的问世--一种新近创造的将基因传递到目标位点并使其表达预期功能的尖端方法。可以使用病毒或非病毒载体来传递基因,每种载体都有自己的优势和局限性。基因疗法已被证明是一种潜在的改变AD疾病的治疗方法。本文将讨论基因疗法的优缺点以及各种基因传递方式。此外,文章还重点介绍了基因疗法的治疗靶点--AD、AD 的病理生理学以及基因疗法治疗 AD 的多种靶点。
{"title":"Gene therapy: an alternative to treat Alzheimer’s disease","authors":"Vanshika Doshi, Garima Joshi, Sanjay Sharma, Deepak Choudhary","doi":"10.1007/s00210-023-02873-z","DOIUrl":"https://doi.org/10.1007/s00210-023-02873-z","url":null,"abstract":"<p>Alzheimer’s disease (AD), a neuro-degenerative disease that primarily affects the elderly, is a worldwide phenomenon. Loss of memory, cognitive decline, behavioural changes, and many other signs are used to classify it. Various hypotheses that may contribute to Alzheimer’s disease have been found during decades of survey, including tau theory, the amyloid theory, the cholinergic hypothesis, and the oxidative stress hypothesis. According to some theories, the two leading causes of AD are the accumulation of amyloid beta plaque and development of NFTs in the brain. The hippocampus and cerebral cortex are the primary sites where amyloid beta plaques gather in the body. NFT formation in the brain impairs the brain’s neurons’ potential of signalling. According to the age at which it manifests in a person, there are two subtypes of AD: ‘LOAD (Late Onset Alzheimer’s Disease)’ and ‘EOAD (Early Onset Alzheimer’s Disease)’. Long-term research into AD treatment has resulted in the introduction of some medications that provided symptomatic relief to patients but did not alter the disease’s pathophysiology, like cholinesterase inhibitors, inhibitors of tau aggregation, and monoclonal antibodies to Aβ aggregation. Even though the medications did not halt the progression of AD, researchers did not discontinue their work, which lead to the introduction of gene therapy — a recently created cutting-edge method of delivering genes to target sites where they can express the intended functionalities. Viral or non-viral vectors could be used to deliver the gene, each with advantages and limitations of their own. Gene therapy is proven to be a potential disease-modifying treatment for AD. This article discusses about gene therapy, its merits and demerits and the various ways of gene delivery. Additionally, it focuses on AD as the target for treatment through gene therapy, the pathophysiology of AD, and the multiple targets for gene therapy in the treatment of AD.</p>","PeriodicalId":18862,"journal":{"name":"Naunyn-schmiedebergs Archives of Pharmacology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138574321","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Licochalcone A (Lico A), a flavonoid found in licorice, possesses multiple pharmacological activities in modulating oxidative stress, glycemia, inflammation, and lipid metabolism. This study aimed to explore the potential mechanism of Lico A in mitigating ferroptosis associated with doxorubicin-induced cardiotoxicity (DIC). Initially, network pharmacology analysis was applied to identify the active components present in licorice and their targeted genes associated with DIC. Subsequently, to assess the role of Lico A in a DIC mouse model, electrocardiograms, myocardial injury markers, and myocardial histopathological changes were measured. Additionally, cell viability, reactive oxygen species (ROS), ferrous iron, glutathione/glutathione disulfide (GSH/GSSG), and malondialdehyde (MDA) were measured in the cell model as hallmarks of ferroptosis. Finally, the PI3K/AKT/MDM2/p53 signaling pathway and ferroptosis-related proteins were measured in vitro and in vivo. Bioinformatics results revealed that 8 major compounds of licorice, including Lico A, primarily regulated targets such as p53 and the PI3K/AKT signaling pathways in DIC. In the mouse model of DIC, Lico A significantly ameliorated serum biomarkers, histopathology, and electrocardiogram abnormalities. Pretreatment with Lico A enhanced the viability of H9C2 cells treated with doxorubicin. Furthermore, Lico A administration resulted in decreased levels of ROS, ferrous iron, and MDA and increased levels of GSH/GSSG. At the protein level, Lico A increased the phosphorylation of PI3K/AKT/MDM2, reduced p53 accumulation, and induced the upregulation of SLC7A11 and GPX4 expression. However, selective inhibition of PI3K/AKT and plasmid-based overexpression of p53 significantly abolished the anti-ferroptosis functions of Lico A. In conclusion, Lico A attenuates DIC by suppressing p53-mediated ferroptosis through activating PI3K/AKT/MDM2 signaling.
甘草中的黄酮类化合物甘草查尔酮 A(Lico A)具有多种药理活性,可调节氧化应激、血糖、炎症和脂质代谢。本研究旨在探索 Lico A 在减轻与多柔比星诱导的心脏毒性(DIC)相关的铁变态反应方面的潜在机制。首先,应用网络药理学分析确定了甘草中的活性成分及其与 DIC 相关的靶基因。随后,为了评估甘草 A 在 DIC 小鼠模型中的作用,测量了心电图、心肌损伤标志物和心肌组织病理学变化。此外,还测量了细胞模型中的细胞活力、活性氧(ROS)、亚铁、谷胱甘肽/二硫化谷胱甘肽(GSH/GSSG)和丙二醛(MDA)等铁中毒标志物。最后,在体外和体内测量了 PI3K/AKT/MDM2/p53 信号通路和铁变态相关蛋白。生物信息学结果显示,包括Lico A在内的8种主要甘草化合物主要调控DIC中的p53和PI3K/AKT信号通路等靶标。在 DIC 小鼠模型中,Lico A 能明显改善血清生物标志物、组织病理学和心电图异常。用 Lico A 预处理可提高用多柔比星处理的 H9C2 细胞的存活率。此外,服用 Lico A 还降低了 ROS、亚铁和 MDA 的水平,提高了 GSH/GSSG 的水平。在蛋白质水平上,Lico A 增加了 PI3K/AKT/MDM2 的磷酸化,减少了 p53 的积累,并诱导了 SLC7A11 和 GPX4 表达的上调。总之,Lico A 可通过激活 PI3K/AKT/MDM2 信号抑制 p53 介导的铁变态反应,从而减轻 DIC。
{"title":"Licochalcone A alleviates ferroptosis in doxorubicin-induced cardiotoxicity via the PI3K/AKT/MDM2/p53 pathway","authors":"Ganxiao Chen, Shunxiang Luo, Hongdou Guo, Jiayi Lin, Shanghua Xu","doi":"10.1007/s00210-023-02863-1","DOIUrl":"https://doi.org/10.1007/s00210-023-02863-1","url":null,"abstract":"<p>Licochalcone A (Lico A), a flavonoid found in licorice, possesses multiple pharmacological activities in modulating oxidative stress, glycemia, inflammation, and lipid metabolism. This study aimed to explore the potential mechanism of Lico A in mitigating ferroptosis associated with doxorubicin-induced cardiotoxicity (DIC). Initially, network pharmacology analysis was applied to identify the active components present in licorice and their targeted genes associated with DIC. Subsequently, to assess the role of Lico A in a DIC mouse model, electrocardiograms, myocardial injury markers, and myocardial histopathological changes were measured. Additionally, cell viability, reactive oxygen species (ROS), ferrous iron, glutathione/glutathione disulfide (GSH/GSSG), and malondialdehyde (MDA) were measured in the cell model as hallmarks of ferroptosis. Finally, the PI3K/AKT/MDM2/p53 signaling pathway and ferroptosis-related proteins were measured in vitro and in vivo. Bioinformatics results revealed that 8 major compounds of licorice, including Lico A, primarily regulated targets such as p53 and the PI3K/AKT signaling pathways in DIC. In the mouse model of DIC, Lico A significantly ameliorated serum biomarkers, histopathology, and electrocardiogram abnormalities. Pretreatment with Lico A enhanced the viability of H9C2 cells treated with doxorubicin. Furthermore, Lico A administration resulted in decreased levels of ROS, ferrous iron, and MDA and increased levels of GSH/GSSG. At the protein level, Lico A increased the phosphorylation of PI3K/AKT/MDM2, reduced p53 accumulation, and induced the upregulation of SLC7A11 and GPX4 expression. However, selective inhibition of PI3K/AKT and plasmid-based overexpression of p53 significantly abolished the anti-ferroptosis functions of Lico A. In conclusion, Lico A attenuates DIC by suppressing p53-mediated ferroptosis through activating PI3K/AKT/MDM2 signaling.</p>","PeriodicalId":18862,"journal":{"name":"Naunyn-schmiedebergs Archives of Pharmacology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138574141","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-12-11DOI: 10.1007/s00210-023-02882-y
Nisha Gulati, Dinesh Kumar Chellappan, Ronan MacLoughlin, Gaurav Gupta, Sachin Kumar Singh, Brian G. Oliver, Kamal Dua, Harish Dureja
Asthma, lung cancer, cystic fibrosis, tuberculosis, acute respiratory distress syndrome, chronic obstructive pulmonary disease, and COVID-19 are few examples of inflammatory lung conditions that cause cytokine release syndrome. It can initiate a widespread inflammatory response and may activate several inflammatory pathways that cause multiple organ failures leading to increased number of deaths and increased prevalence rates around the world. Nanotechnology-based therapeutic modalities such as nanoparticles, liposomes, nanosuspension, monoclonal antibodies, and vaccines can be used in the effective treatment of inflammatory lung diseases at both cellular and molecular levels. This would also help significantly in the reduction of patient mortality. Therefore, nanotechnology could be a potent platform for repurposing current medications in the treatment of inflammatory lung diseases. The aim and approach of this article are to highlight the clinical manifestations of cytokine storm in inflammatory lung diseases along with the advances and potential applications of nanotechnology-based therapeutics in the management of cytokine storm. Further in-depth studies are required to understand the molecular pathophysiology, and how nanotechnology-based therapeutics can help to effectively combat this problem.
{"title":"Advances in nano-based drug delivery systems for the management of cytokine influx-mediated inflammation in lung diseases","authors":"Nisha Gulati, Dinesh Kumar Chellappan, Ronan MacLoughlin, Gaurav Gupta, Sachin Kumar Singh, Brian G. Oliver, Kamal Dua, Harish Dureja","doi":"10.1007/s00210-023-02882-y","DOIUrl":"https://doi.org/10.1007/s00210-023-02882-y","url":null,"abstract":"<p>Asthma, lung cancer, cystic fibrosis, tuberculosis, acute respiratory distress syndrome, chronic obstructive pulmonary disease, and COVID-19 are few examples of inflammatory lung conditions that cause cytokine release syndrome. It can initiate a widespread inflammatory response and may activate several inflammatory pathways that cause multiple organ failures leading to increased number of deaths and increased prevalence rates around the world. Nanotechnology-based therapeutic modalities such as nanoparticles, liposomes, nanosuspension, monoclonal antibodies, and vaccines can be used in the effective treatment of inflammatory lung diseases at both cellular and molecular levels. This would also help significantly in the reduction of patient mortality. Therefore, nanotechnology could be a potent platform for repurposing current medications in the treatment of inflammatory lung diseases. The aim and approach of this article are to highlight the clinical manifestations of cytokine storm in inflammatory lung diseases along with the advances and potential applications of nanotechnology-based therapeutics in the management of cytokine storm. Further in-depth studies are required to understand the molecular pathophysiology, and how nanotechnology-based therapeutics can help to effectively combat this problem.</p>","PeriodicalId":18862,"journal":{"name":"Naunyn-schmiedebergs Archives of Pharmacology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138573920","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2007-01-01DOI: 10.1007/S00210-006-0116-8
P. Giussani, T. Colleoni, R. Bassi, L. Brioschi, G. Tettamanti, K. Hanada, L. Riboni, P. Viani
{"title":"Metabolic and functional role of the ceramide binding protein CERT in glioma cells","authors":"P. Giussani, T. Colleoni, R. Bassi, L. Brioschi, G. Tettamanti, K. Hanada, L. Riboni, P. Viani","doi":"10.1007/S00210-006-0116-8","DOIUrl":"https://doi.org/10.1007/S00210-006-0116-8","url":null,"abstract":"","PeriodicalId":18862,"journal":{"name":"Naunyn-schmiedebergs Archives of Pharmacology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2007-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"76323382","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
R. Schroeder, C. Meyer-Burgdorff, D. Rott, O. Brahms
{"title":"[Comparative studies on the effect of ADH, hypertensin and renin on the renal excretion of water and electrolytes in the rat].","authors":"R. Schroeder, C. Meyer-Burgdorff, D. Rott, O. Brahms","doi":"10.1007/BF00247698","DOIUrl":"https://doi.org/10.1007/BF00247698","url":null,"abstract":"","PeriodicalId":18862,"journal":{"name":"Naunyn-schmiedebergs Archives of Pharmacology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2004-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83972287","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}