Pub Date : 2025-02-01Epub Date: 2024-10-31DOI: 10.1007/s43440-024-00664-8
Qiming Yang, Rui Huan, Defeng Meng, Junwei Qi, Lei Xia
Vitexin (apigenin-8-C-beta-D-glucopyranoside) is a natural flavonoid derivative with anti-cancer, antioxidant, anti-inflammatory, antihypertensive, anti-asthma, anti-epilepsy, and other therapeutic effects. It is extracted from pearl millet, hawthorn, pigeon bean, mung bean, and other medicinal plants. Vitexin has received widespread attention because of its significant anti-tumor effect. It induces apoptosis and anti-tumor angiogenesis, inhibits tumor cell migration and invasion, regulates tumor cell autophagy and immunity, and increases patient sensitivity to radiotherapy and chemotherapy. It has a significant anti-tumor effect on breast, prostate, liver, cervical, and colon cancers, gliomas, and other malignant tumors. This review demonstrates the latest research progress on the anti-tumor effects and potential mechanisms of vitexin. It summarizes its anti-tumor mechanism to provide new theoretical support and reference for cancer treatment.
{"title":"Progress in the study of anti-tumor effects and mechanisms of vitexin.","authors":"Qiming Yang, Rui Huan, Defeng Meng, Junwei Qi, Lei Xia","doi":"10.1007/s43440-024-00664-8","DOIUrl":"10.1007/s43440-024-00664-8","url":null,"abstract":"<p><p>Vitexin (apigenin-8-C-beta-D-glucopyranoside) is a natural flavonoid derivative with anti-cancer, antioxidant, anti-inflammatory, antihypertensive, anti-asthma, anti-epilepsy, and other therapeutic effects. It is extracted from pearl millet, hawthorn, pigeon bean, mung bean, and other medicinal plants. Vitexin has received widespread attention because of its significant anti-tumor effect. It induces apoptosis and anti-tumor angiogenesis, inhibits tumor cell migration and invasion, regulates tumor cell autophagy and immunity, and increases patient sensitivity to radiotherapy and chemotherapy. It has a significant anti-tumor effect on breast, prostate, liver, cervical, and colon cancers, gliomas, and other malignant tumors. This review demonstrates the latest research progress on the anti-tumor effects and potential mechanisms of vitexin. It summarizes its anti-tumor mechanism to provide new theoretical support and reference for cancer treatment.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"124-134"},"PeriodicalIF":3.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142546682","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01Epub Date: 2024-09-04DOI: 10.1007/s43440-024-00643-z
Ana Mijušković, Susan Wray, Sarah Arrowsmith
Background: Spontaneous preterm birth is the leading cause of perinatal morbidity and mortality. Tocolytics are drugs used to inhibit uterine contractions in cases of imminent preterm birth, however, few are effective in stopping labour once initiated and all have side effects. Combination approaches involving drugs that target multiple signalling pathways that regulate contractions may increase efficacy, reduce dosage and improve tolerability. Both non-steroidal anti-inflammatory drugs (NSAIDs) and hydrogen sulphide (H2S)-releasing compounds can reduce myometrial contractions. In a novel approach we evaluated the tocolytic properties of ATB-346-a H2S-releasing derivative of the NSAID naproxen, shown clinically to reduce pain and inflammation in arthritis.
Methods: Using organ baths, paired strips of human myometrium were exposed to increasing concentrations of ATB-346, or equimolar concentrations (10µM and 30µM) of the parent drug, naproxen, or the H2S-releasing moiety, 4-hydroxy-thiobenzamide (TBZ), alone. The ability of ATB-346 versus the individual components of ATB-346 to decrease ex vivo spontaneous contractions was investigated, and the potency was compared to a known H2S donor, Na2S.
Results: Acute application of Na2S produced a concentration-dependent decrease in force amplitude and force integral (area under the curve) of contraction. ATB-346 produced a more profound decrease in contraction compared to equimolar concentrations of naproxen or TZB alone and was more potent than the equivalent concentration of Na2S.
Conclusions: ATB-346 exhibits potent tocolytic properties in human myometrium. These exciting results call for further exploration of ATB-346, with a view to repurposing this or similar drugs as novel therapies for delaying preterm labour.
{"title":"A hydrogen sulphide-releasing non-steroidal anti-inflammatory, ATB-346, significantly attenuates human myometrial contractions.","authors":"Ana Mijušković, Susan Wray, Sarah Arrowsmith","doi":"10.1007/s43440-024-00643-z","DOIUrl":"10.1007/s43440-024-00643-z","url":null,"abstract":"<p><strong>Background: </strong>Spontaneous preterm birth is the leading cause of perinatal morbidity and mortality. Tocolytics are drugs used to inhibit uterine contractions in cases of imminent preterm birth, however, few are effective in stopping labour once initiated and all have side effects. Combination approaches involving drugs that target multiple signalling pathways that regulate contractions may increase efficacy, reduce dosage and improve tolerability. Both non-steroidal anti-inflammatory drugs (NSAIDs) and hydrogen sulphide (H<sub>2</sub>S)-releasing compounds can reduce myometrial contractions. In a novel approach we evaluated the tocolytic properties of ATB-346-a H<sub>2</sub>S-releasing derivative of the NSAID naproxen, shown clinically to reduce pain and inflammation in arthritis.</p><p><strong>Methods: </strong>Using organ baths, paired strips of human myometrium were exposed to increasing concentrations of ATB-346, or equimolar concentrations (10µM and 30µM) of the parent drug, naproxen, or the H<sub>2</sub>S-releasing moiety, 4-hydroxy-thiobenzamide (TBZ), alone. The ability of ATB-346 versus the individual components of ATB-346 to decrease ex vivo spontaneous contractions was investigated, and the potency was compared to a known H<sub>2</sub>S donor, Na<sub>2</sub>S.</p><p><strong>Results: </strong>Acute application of Na<sub>2</sub>S produced a concentration-dependent decrease in force amplitude and force integral (area under the curve) of contraction. ATB-346 produced a more profound decrease in contraction compared to equimolar concentrations of naproxen or TZB alone and was more potent than the equivalent concentration of Na<sub>2</sub>S.</p><p><strong>Conclusions: </strong>ATB-346 exhibits potent tocolytic properties in human myometrium. These exciting results call for further exploration of ATB-346, with a view to repurposing this or similar drugs as novel therapies for delaying preterm labour.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"287-294"},"PeriodicalIF":3.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11743401/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142133443","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01Epub Date: 2024-12-03DOI: 10.1007/s43440-024-00681-7
Łukasz Gąsior, Bartłomiej Pochwat, Monika Zaręba-Kozioł, Jakub Włodarczyk, Andreas Martin Grabrucker, Bernadeta Szewczyk
Background: Preclinical and clinical studies have shown that dietary zinc deficiency can lead to symptoms similar to those observed in major depressive disorder (MDD). However, the underlying molecular mechanisms remain unclear. To investigate these mechanisms, we examined proteomic changes in the prefrontal cortex (PFC) and hippocampus (HP) of rats, two critical brain regions implicated in the pathophysiology of depression.
Methods: Rats were fed diets either adequate in zinc (ZnA, 50 mg Zn/kg) or deficient in zinc (ZnD, <3 mg/kg) for four weeks. High-throughput proteomic analysis was used to detect changes in protein expression, supplemented by enzyme activity assay for mitochondrial complexes I and IV, examining their functional impacts.
Results: ZnD led to significant alterations in protein expression related to zinc transport and mitochondrial function. Proteomic analysis revealed changes in zinc transporter family members such as Slc30a1 (6.64 log2FC), Slc30a3 (-2.32 log2FC), Slc30a4 (2.87 log2FC), Slc30a5 (5.90 log2FC), Slc30a6 (1.50 log2FC), and Slc30a7 (2.17 log2FC) in the PFC, and Slc30a3 (-1.02 log2FC), Slc30a5 (-1.04 log2FC), and Slc30a7 (1.08 log2FC) in the HP of rats subjected to ZnD. Furthermore, ZnD significantly affected essential mitochondrial activity proteins, including Atp5pb (3.25 log2FC), Cox2 (2.28 log2FC), Atp5me (2.04 log2FC), Cyc1 (2.30 log2FC), Cox4i1 (1.23 log2FC), Cox7c (1.63 log2FC), and Cisd1 (1.55 log2FC), with a pronounced decrease in complex I activity in the PFC.
Conclusions: Our study demonstrates that ZnD leads to significant proteomic changes in the PFC and HP of rats. Specifically, ZnD alters the expression of zinc transporter proteins and proteins critical for mitochondrial function. The significant decrease in complex I activity in the PFC further underscores the impact of ZnD on mitochondrial function. These results highlight the molecular mechanisms by which ZnD can influence brain function and contribute to symptoms similar to those observed in depression.
{"title":"Proteomics analysis in rats reveals convergent mechanisms between major depressive disorder and dietary zinc deficiency.","authors":"Łukasz Gąsior, Bartłomiej Pochwat, Monika Zaręba-Kozioł, Jakub Włodarczyk, Andreas Martin Grabrucker, Bernadeta Szewczyk","doi":"10.1007/s43440-024-00681-7","DOIUrl":"10.1007/s43440-024-00681-7","url":null,"abstract":"<p><strong>Background: </strong>Preclinical and clinical studies have shown that dietary zinc deficiency can lead to symptoms similar to those observed in major depressive disorder (MDD). However, the underlying molecular mechanisms remain unclear. To investigate these mechanisms, we examined proteomic changes in the prefrontal cortex (PFC) and hippocampus (HP) of rats, two critical brain regions implicated in the pathophysiology of depression.</p><p><strong>Methods: </strong>Rats were fed diets either adequate in zinc (ZnA, 50 mg Zn/kg) or deficient in zinc (ZnD, <3 mg/kg) for four weeks. High-throughput proteomic analysis was used to detect changes in protein expression, supplemented by enzyme activity assay for mitochondrial complexes I and IV, examining their functional impacts.</p><p><strong>Results: </strong>ZnD led to significant alterations in protein expression related to zinc transport and mitochondrial function. Proteomic analysis revealed changes in zinc transporter family members such as Slc30a1 (6.64 log2FC), Slc30a3 (-2.32 log2FC), Slc30a4 (2.87 log2FC), Slc30a5 (5.90 log2FC), Slc30a6 (1.50 log2FC), and Slc30a7 (2.17 log2FC) in the PFC, and Slc30a3 (-1.02 log2FC), Slc30a5 (-1.04 log2FC), and Slc30a7 (1.08 log2FC) in the HP of rats subjected to ZnD. Furthermore, ZnD significantly affected essential mitochondrial activity proteins, including Atp5pb (3.25 log2FC), Cox2 (2.28 log2FC), Atp5me (2.04 log2FC), Cyc1 (2.30 log2FC), Cox4i1 (1.23 log2FC), Cox7c (1.63 log2FC), and Cisd1 (1.55 log2FC), with a pronounced decrease in complex I activity in the PFC.</p><p><strong>Conclusions: </strong>Our study demonstrates that ZnD leads to significant proteomic changes in the PFC and HP of rats. Specifically, ZnD alters the expression of zinc transporter proteins and proteins critical for mitochondrial function. The significant decrease in complex I activity in the PFC further underscores the impact of ZnD on mitochondrial function. These results highlight the molecular mechanisms by which ZnD can influence brain function and contribute to symptoms similar to those observed in depression.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"145-157"},"PeriodicalIF":3.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11743416/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142771312","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01Epub Date: 2024-11-29DOI: 10.1007/s43440-024-00680-8
Randall L Davis, Sascha Grotjahn, Burkhard Koenig, Daniel J Buck, Jimmie D Weaver
Background: Phytochemicals derived from the plant Cannabis sativa hold promise in terms of medicinal value. Cannabinoids such as Δ9-tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabinol (CBN) are arguably the best characterized and known to possess wide-ranging therapeutic benefits. The mechanism of action for these therapeutic effects remains to be fully elucidated, however, the anti-inflammatory actions are of particular interest. Maximizing therapeutic effects while limiting adverse effects is crucial in pharmaceutical development. Fluorination of natural products often yields molecules with enhanced biological properties and provides opportunities for intellectual property protection not available to the natural product.
Methods: Herein, we describe four novel cannabinoids (a deoxy trifluoroCBN analog (F3CBN), the racemic cis-deoxy-trifluoro-THC (F3THC), and truncated pyridine analogs of an intermediate in route to the THC and CBN, SG126 and SG154. Importantly, we provide the initial assessment of the biologic activity of these molecules, by investigating the in vitro effects on metabolic activity (via 3-[4,5-dimethylthiazol-2-yl]-2,5,-diphenyltetrazolium bromide, MTT assay) and cytokine expression (via enzyme linked immunosorbent assay, ELISA) in human C20 microglial cells.
Results: The cannabinoids examined had minimal to no effect on metabolic activity up to 10 µM. Notably, F3CBN and F3THC potentiated interleukin-1 β (IL-1β)-induced expression of interferon-γ inducible protein 10 (CXCL10) and IL-6 expression whereas, SG126 and SG154 were inhibitory.
Conclusions: These findings are foundational for new lines of investigation into the therapeutic potential of four novel fluorinated cannabinoids.
{"title":"Novel fluorinated cannabinoid analogs modulate cytokine expression in human C20 microglial cells.","authors":"Randall L Davis, Sascha Grotjahn, Burkhard Koenig, Daniel J Buck, Jimmie D Weaver","doi":"10.1007/s43440-024-00680-8","DOIUrl":"10.1007/s43440-024-00680-8","url":null,"abstract":"<p><strong>Background: </strong>Phytochemicals derived from the plant Cannabis sativa hold promise in terms of medicinal value. Cannabinoids such as Δ<sup>9</sup>-tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabinol (CBN) are arguably the best characterized and known to possess wide-ranging therapeutic benefits. The mechanism of action for these therapeutic effects remains to be fully elucidated, however, the anti-inflammatory actions are of particular interest. Maximizing therapeutic effects while limiting adverse effects is crucial in pharmaceutical development. Fluorination of natural products often yields molecules with enhanced biological properties and provides opportunities for intellectual property protection not available to the natural product.</p><p><strong>Methods: </strong>Herein, we describe four novel cannabinoids (a deoxy trifluoroCBN analog (F<sub>3</sub>CBN), the racemic cis-deoxy-trifluoro-THC (F<sub>3</sub>THC), and truncated pyridine analogs of an intermediate in route to the THC and CBN, SG126 and SG154. Importantly, we provide the initial assessment of the biologic activity of these molecules, by investigating the in vitro effects on metabolic activity (via 3-[4,5-dimethylthiazol-2-yl]-2,5,-diphenyltetrazolium bromide, MTT assay) and cytokine expression (via enzyme linked immunosorbent assay, ELISA) in human C20 microglial cells.</p><p><strong>Results: </strong>The cannabinoids examined had minimal to no effect on metabolic activity up to 10 µM. Notably, F<sub>3</sub>CBN and F<sub>3</sub>THC potentiated interleukin-1 β (IL-1β)-induced expression of interferon-γ inducible protein 10 (CXCL10) and IL-6 expression whereas, SG126 and SG154 were inhibitory.</p><p><strong>Conclusions: </strong>These findings are foundational for new lines of investigation into the therapeutic potential of four novel fluorinated cannabinoids.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"295-301"},"PeriodicalIF":3.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142751324","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Statins are widely utilized to reduce cholesterol levels, particularly in cardiovascular diseases. They interface with cholesterol synthesis by inhibiting the 3-hydroxy-3-methylglutaryl coenzyme-A (HMG-CoA) reductase enzyme. Besides their primary effect, statins demonstrate anti-inflammatory and immune-modulating properties in various diseases, highlighting the pleiotropic effect of these drugs. The CD40:CD40L signaling pathway is considered a prominent inflammatory pathway in multiple diseases, including autoimmune, inflammatory, and cardiovascular diseases. The findings from clinical trials and in vitro and in vivo studies suggest the potential anti-inflammatory effect of statins in modulating the CD40 signaling pathway and downstream inflammatory mediator. Accordingly, as its classic ligand, statins can suppress immune responses in autoimmune diseases by inhibiting CD40 expression and blocking its interaction with CD40L. Additionally, statins affect intracellular signaling and inhibit inflammatory mediator secretion in chronic inflammatory diseases like asthma and autoimmune disorders such as myasthenia gravis, multiple sclerosis, systemic lupus erymanthus, and cardiovascular diseases like atherosclerosis. However, it is essential to note that the anti-inflammatory effect of statins may vary depending on the specific type of statin used. In this study, we aim to explore the potential anti-inflammatory effects of statins in treating inflammatory diseases by examining their role in regulating immune responses, particularly their impact on the CD40:CD40L signaling pathway, through a comprehensive review of existing literature.
{"title":"Impact of statin therapy on CD40:CD40L signaling: mechanistic insights and therapeutic opportunities.","authors":"Fatemeh Askarizadeh, Sercan Karav, Tannaz Jamialahmadi, Amirhossein Sahebkar","doi":"10.1007/s43440-024-00678-2","DOIUrl":"10.1007/s43440-024-00678-2","url":null,"abstract":"<p><p>Statins are widely utilized to reduce cholesterol levels, particularly in cardiovascular diseases. They interface with cholesterol synthesis by inhibiting the 3-hydroxy-3-methylglutaryl coenzyme-A (HMG-CoA) reductase enzyme. Besides their primary effect, statins demonstrate anti-inflammatory and immune-modulating properties in various diseases, highlighting the pleiotropic effect of these drugs. The CD40:CD40L signaling pathway is considered a prominent inflammatory pathway in multiple diseases, including autoimmune, inflammatory, and cardiovascular diseases. The findings from clinical trials and in vitro and in vivo studies suggest the potential anti-inflammatory effect of statins in modulating the CD40 signaling pathway and downstream inflammatory mediator. Accordingly, as its classic ligand, statins can suppress immune responses in autoimmune diseases by inhibiting CD40 expression and blocking its interaction with CD40L. Additionally, statins affect intracellular signaling and inhibit inflammatory mediator secretion in chronic inflammatory diseases like asthma and autoimmune disorders such as myasthenia gravis, multiple sclerosis, systemic lupus erymanthus, and cardiovascular diseases like atherosclerosis. However, it is essential to note that the anti-inflammatory effect of statins may vary depending on the specific type of statin used. In this study, we aim to explore the potential anti-inflammatory effects of statins in treating inflammatory diseases by examining their role in regulating immune responses, particularly their impact on the CD40:CD40L signaling pathway, through a comprehensive review of existing literature.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"43-71"},"PeriodicalIF":3.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142829610","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01Epub Date: 2024-12-16DOI: 10.1007/s43440-024-00686-2
Agnieszka Cios, Grażyna Chłoń-Rzepa, Magdalena Jastrzębska-Więsek, Krzysztof Pociecha, Katarzyna Wójcik-Pszczoła, Elżbieta Pękala, Anna Wesołowska
Background: Research on new candidates for antidepressant/anxiolytic drugs from the long-chain arylpiperazines (LCAPs) group containing a 1,3-dimethylpurine-2,6-dione as a terminal amide fragment fits into the modern exploration trend. This study aimed to examine, for the first time in male Wistar rats, pharmacodynamic (antidepressant- and anxiolytic-like) and pharmacokinetic properties of 7-(5-(4-(3-chlorophenyl)piperazin-1-yl)pentyl)-1,3-dimethyl-3,7-dihydro-1 H-purine-2,6-dione hydrochloride (GR-14).
Methods: Antidepressant- and anxiolytic-like activities have been assessed in the forced swim test (FST) and Vogel conflict drinking test, respectively. The pharmacokinetic characteristics of GR-14, its distribution into rat tissues, and several in vitro ADME-Tox parameters (hepatocytotoxic, neurocytotoxic, metabolic stability) have been defined.
Results: GR-14 produces strong and dose-dependent antidepressant- and anxiolytic-like effects in both tests used. Pharmacokinetic findings demonstrate that GR-14 reveals linear pharmacokinetics tested after intravenous (iv) and was rapidly absorbed after oral (po) administration. It rapidly crosses the blood-brain barrier (BBB) which is vital for therapeutic effects in vivo in psychiatric diseases, depression, and anxiety. Moreover, it is slowly eliminated from the brain, maintaining concentrations higher than those in plasma at the last time point measured. Further studies have also shown that GR-14 is an average high-clearance drug in rat liver microsomes and exerts neither hepatocytotoxic nor neurocytotoxic effects in vitro.
Conclusion: The tested derivative GR-14 presents prominent mood-modulating activity in rats and has promising pharmacokinetic parameters and a good safety profile. The beneficial pharmacology and pharmacokinetics of GR-14 in vivo are in high concordance with its profile in vitro, thus underlining very hopeful properties to support the early development process.
背景:从以1,3-二甲基嘌呤-2,6-二酮为末端酰胺片段的长链芳基哌嗪(LCAPs)基团中寻找抗抑郁/抗焦虑药物的新候选物符合现代探索的趋势。本研究首次在雄性Wistar大鼠中研究了7-(5-(4-(3-氯苯基)哌嗪-1-基)戊基)-1,3-二甲基-3,7-二氢-1 h -嘌呤-2,6-二酮盐酸(GR-14)的药效学(抗抑郁和抗焦虑样)和药代动力学特性。方法:分别用强迫游泳试验(FST)和Vogel冲突饮酒试验评估抗抑郁药和焦虑样活动。确定了GR-14的药代动力学特征、在大鼠组织中的分布以及ADME-Tox的几个体外参数(肝细胞毒性、神经细胞毒性、代谢稳定性)。结果:GR-14在使用的两种测试中产生强烈的剂量依赖性抗抑郁药和抗焦虑药样作用。药代动力学结果表明,GR-14在静脉注射(iv)后呈线性药代动力学,口服(po)后迅速吸收。它迅速穿过血脑屏障(BBB),这对精神疾病、抑郁症和焦虑症的体内治疗效果至关重要。此外,它会慢慢地从大脑中消失,维持比上次测量时血浆中的浓度更高的浓度。进一步的研究也表明,GR-14在大鼠肝微粒体中是一种平均高清除率的药物,在体外不产生肝细胞毒性和神经细胞毒性作用。结论:所测衍生物GR-14在大鼠体内具有明显的情绪调节活性,具有良好的药动学参数和安全性。GR-14在体内的有益药理学和药代动力学与其在体外的情况高度一致,因此强调了非常有希望支持早期发育过程的特性。
{"title":"Advanced pharmacological and pharmacokinetic evaluation of 1,3 dimethylpurine-2,6-dione derivative (GR-14) with prominent mood-modulating activity in rats.","authors":"Agnieszka Cios, Grażyna Chłoń-Rzepa, Magdalena Jastrzębska-Więsek, Krzysztof Pociecha, Katarzyna Wójcik-Pszczoła, Elżbieta Pękala, Anna Wesołowska","doi":"10.1007/s43440-024-00686-2","DOIUrl":"10.1007/s43440-024-00686-2","url":null,"abstract":"<p><strong>Background: </strong>Research on new candidates for antidepressant/anxiolytic drugs from the long-chain arylpiperazines (LCAPs) group containing a 1,3-dimethylpurine-2,6-dione as a terminal amide fragment fits into the modern exploration trend. This study aimed to examine, for the first time in male Wistar rats, pharmacodynamic (antidepressant- and anxiolytic-like) and pharmacokinetic properties of 7-(5-(4-(3-chlorophenyl)piperazin-1-yl)pentyl)-1,3-dimethyl-3,7-dihydro-1 H-purine-2,6-dione hydrochloride (GR-14).</p><p><strong>Methods: </strong>Antidepressant- and anxiolytic-like activities have been assessed in the forced swim test (FST) and Vogel conflict drinking test, respectively. The pharmacokinetic characteristics of GR-14, its distribution into rat tissues, and several in vitro ADME-Tox parameters (hepatocytotoxic, neurocytotoxic, metabolic stability) have been defined.</p><p><strong>Results: </strong>GR-14 produces strong and dose-dependent antidepressant- and anxiolytic-like effects in both tests used. Pharmacokinetic findings demonstrate that GR-14 reveals linear pharmacokinetics tested after intravenous (iv) and was rapidly absorbed after oral (po) administration. It rapidly crosses the blood-brain barrier (BBB) which is vital for therapeutic effects in vivo in psychiatric diseases, depression, and anxiety. Moreover, it is slowly eliminated from the brain, maintaining concentrations higher than those in plasma at the last time point measured. Further studies have also shown that GR-14 is an average high-clearance drug in rat liver microsomes and exerts neither hepatocytotoxic nor neurocytotoxic effects in vitro.</p><p><strong>Conclusion: </strong>The tested derivative GR-14 presents prominent mood-modulating activity in rats and has promising pharmacokinetic parameters and a good safety profile. The beneficial pharmacology and pharmacokinetics of GR-14 in vivo are in high concordance with its profile in vitro, thus underlining very hopeful properties to support the early development process.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"158-171"},"PeriodicalIF":3.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142829604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Pancreatic cancer (PC) is one of the most aggressive cancers and is the seventh leading cause of cancer-related death worldwide. PC is characterized by rapid progression and resistance to conventional treatments. Mutations in KRAS, CDKN2A, TP53, SMAD4/DPC4, and MYC are major genetic alterations associated with poor treatment outcomes in patients with PC. Therefore, optimizing PC therapy is a tremendous challenge. Unsymmetrical bisacridines (UAs), synthesized by our group, are new promising compounds that have exhibited high cytotoxicity and antitumor activity against several solid tumors, including pancreatic cancer.
Methods: The cellular effects induced by UAs in PC cells were evaluated by MTT assay (cell growth inhibition), flow cytometry, and fluorescence and light microscopy (cell cycle distribution, apoptosis, and senescence detection). Analysis of the effects of UAs on the levels of proteins (c-Myc, p53, SMAD4, p21, and p16) was performed by Western blotting.
Results: Apoptosis was the main triggered mechanism of death after UAs treatment, and induction of the SMAD4 protein can facilitate this process. c-Myc, which is one of the molecular targets of UAs, can participate in the induction of cell death in a p53-independent manner. Moreover, UAs can also induce accelerated senescence through the upregulation of p21. Notably, senescent cells can die via apoptosis after prolonged exposure to UAs.
Conclusions: UAs have emerged as potent anticancer agents that induce apoptosis by inhibiting c-Myc protein and triggering cellular senescence in a dose-dependent manner by increasing p21 levels. Thus, UAs exhibit desirable features as promising candidates for future pancreatic anticancer therapies.
背景:胰腺癌(PC)是侵袭性最强的癌症之一,也是全球第七大癌症致死原因。胰腺癌的特点是进展迅速和对常规治疗产生抗药性。KRAS、CDKN2A、TP53、SMAD4/DPC4 和 MYC 基因突变是与 PC 患者治疗效果不佳相关的主要基因改变。因此,优化 PC 治疗是一项巨大的挑战。我们小组合成的不对称双吖啶(UAs)是一种新的有前途的化合物,对包括胰腺癌在内的多种实体瘤具有很高的细胞毒性和抗肿瘤活性:方法:通过 MTT 试验(细胞生长抑制)、流式细胞术、荧光和光学显微镜(细胞周期分布、细胞凋亡和衰老检测)评估 UAs 对 PC 细胞的细胞效应。用 Western 印迹法分析了 UAs 对蛋白质(c-Myc、p53、SMAD4、p21 和 p16)水平的影响:c-Myc是UAs的分子靶标之一,能以与p53无关的方式参与诱导细胞死亡。此外,UAs 还能通过 p21 的上调诱导加速衰老。值得注意的是,衰老细胞在长期接触 UAs 后可通过凋亡死亡:UAs已成为一种有效的抗癌剂,它通过抑制c-Myc蛋白诱导细胞凋亡,并通过提高p21水平以剂量依赖的方式引发细胞衰老。因此,UAs 具有理想的特性,有望成为未来胰腺癌抗癌疗法的候选药物。
{"title":"c-Myc inhibition and p21 modulation contribute to unsymmetrical bisacridines-induced apoptosis and senescence in pancreatic cancer cells.","authors":"Agnieszka Kurdyn, Monika Pawłowska, Ewa Paluszkiewicz, Mirosława Cichorek, Ewa Augustin","doi":"10.1007/s43440-024-00658-6","DOIUrl":"10.1007/s43440-024-00658-6","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic cancer (PC) is one of the most aggressive cancers and is the seventh leading cause of cancer-related death worldwide. PC is characterized by rapid progression and resistance to conventional treatments. Mutations in KRAS, CDKN2A, TP53, SMAD4/DPC4, and MYC are major genetic alterations associated with poor treatment outcomes in patients with PC. Therefore, optimizing PC therapy is a tremendous challenge. Unsymmetrical bisacridines (UAs), synthesized by our group, are new promising compounds that have exhibited high cytotoxicity and antitumor activity against several solid tumors, including pancreatic cancer.</p><p><strong>Methods: </strong>The cellular effects induced by UAs in PC cells were evaluated by MTT assay (cell growth inhibition), flow cytometry, and fluorescence and light microscopy (cell cycle distribution, apoptosis, and senescence detection). Analysis of the effects of UAs on the levels of proteins (c-Myc, p53, SMAD4, p21, and p16) was performed by Western blotting.</p><p><strong>Results: </strong>Apoptosis was the main triggered mechanism of death after UAs treatment, and induction of the SMAD4 protein can facilitate this process. c-Myc, which is one of the molecular targets of UAs, can participate in the induction of cell death in a p53-independent manner. Moreover, UAs can also induce accelerated senescence through the upregulation of p21. Notably, senescent cells can die via apoptosis after prolonged exposure to UAs.</p><p><strong>Conclusions: </strong>UAs have emerged as potent anticancer agents that induce apoptosis by inhibiting c-Myc protein and triggering cellular senescence in a dose-dependent manner by increasing p21 levels. Thus, UAs exhibit desirable features as promising candidates for future pancreatic anticancer therapies.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"182-209"},"PeriodicalIF":3.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11743403/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142366182","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01Epub Date: 2024-10-23DOI: 10.1007/s43440-024-00668-4
Annamaria Mascolo, Alessia Zinzi, Mario Gaio, Donatella Ruggiero, Cristina Scavone, Francesco Rossi, Annalisa Capuano
Introduction: Dapagliflozin and empagliflozin are sodium-glucose cotransporter-2 (SGLT2) inhibitors initially approved for the treatment of type 2 diabetes mellitus (DM), and later for heart failure (HF). Considering this differential therapeutic use, we decided to evaluate cases related to these agents by comparing those with DM, HF, or both (DM and HF).
Methods: A retrospective, pharmacovigilance study was conducted by using data contained in the EudraVigilance from January 1st, 2021, to December 31st, 2023. Cases were classified into those with DM, HF, or both diseases. The Reporting Odds Ratio (ROR), and its 95% confidence interval (95%CI) were computed to compare the reporting probability of the four most reported adverse events. The following comparisons were performed: DM vs. HF; both DM and HF vs. HF; both DM and HF vs. DM. Analyses were adjusted for age, sex, and time between the approval date of the SGLT2 inhibitor and the reporting date.
Results: A total of 14,594 (50.5%) cases were classifiable for DM (N = 11,962; 82.0%), HF (N = 2,100; 14.4%), or both DM and HF (N = 532; 3.64%). The empagliflozin was the most reported SGLT2 inhibitor (60.1%), and only 15 cases (0.1%) reported both empagliflozin and dapagliflozin. Cases with DM and both DM and HF were associated with a higher reporting probability of ketoacidosis (ROR: 5.95, 95%CI: 4.87-7.26; ROR: 3.05, 95%CI: 2.27-4.09) and Fournier's gangrene (ROR: 2.30, 95%CI: 1.65-3.20; ROR: 2.30, 95%CI: 1.38-3.82) than HF. These results were also confirmed by adjusted analyses.
Conclusion: We found that ketoacidosis and Fournier's gangrene had a higher reporting in cases with DM. Further studies are warranted.
{"title":"Safety of dapagliflozin and empagliflozin in cases with diabetes mellitus or/and heart failure: a retrospective pharmacovigilance study conducted on the eudravigilance database.","authors":"Annamaria Mascolo, Alessia Zinzi, Mario Gaio, Donatella Ruggiero, Cristina Scavone, Francesco Rossi, Annalisa Capuano","doi":"10.1007/s43440-024-00668-4","DOIUrl":"10.1007/s43440-024-00668-4","url":null,"abstract":"<p><strong>Introduction: </strong>Dapagliflozin and empagliflozin are sodium-glucose cotransporter-2 (SGLT2) inhibitors initially approved for the treatment of type 2 diabetes mellitus (DM), and later for heart failure (HF). Considering this differential therapeutic use, we decided to evaluate cases related to these agents by comparing those with DM, HF, or both (DM and HF).</p><p><strong>Methods: </strong>A retrospective, pharmacovigilance study was conducted by using data contained in the EudraVigilance from January 1st, 2021, to December 31st, 2023. Cases were classified into those with DM, HF, or both diseases. The Reporting Odds Ratio (ROR), and its 95% confidence interval (95%CI) were computed to compare the reporting probability of the four most reported adverse events. The following comparisons were performed: DM vs. HF; both DM and HF vs. HF; both DM and HF vs. DM. Analyses were adjusted for age, sex, and time between the approval date of the SGLT2 inhibitor and the reporting date.</p><p><strong>Results: </strong>A total of 14,594 (50.5%) cases were classifiable for DM (N = 11,962; 82.0%), HF (N = 2,100; 14.4%), or both DM and HF (N = 532; 3.64%). The empagliflozin was the most reported SGLT2 inhibitor (60.1%), and only 15 cases (0.1%) reported both empagliflozin and dapagliflozin. Cases with DM and both DM and HF were associated with a higher reporting probability of ketoacidosis (ROR: 5.95, 95%CI: 4.87-7.26; ROR: 3.05, 95%CI: 2.27-4.09) and Fournier's gangrene (ROR: 2.30, 95%CI: 1.65-3.20; ROR: 2.30, 95%CI: 1.38-3.82) than HF. These results were also confirmed by adjusted analyses.</p><p><strong>Conclusion: </strong>We found that ketoacidosis and Fournier's gangrene had a higher reporting in cases with DM. Further studies are warranted.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"274-286"},"PeriodicalIF":3.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142505741","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01Epub Date: 2024-11-14DOI: 10.1007/s43440-024-00677-3
Agnieszka Rusak, Marlena Gąsior-Głogowska, Azzurra Sargenti, Edward Krzyżak, Krzysztof Kotowski, Monika Mrozowska, Tomasz Górnicki, Krzysztof Kujawa, Piotr Dzięgiel
Background: The protein CHI3L1 contributes to cancer development by several mechanisms, including stimulation of angiogenesis and invasion as well as immunomodulatory effects. These properties make it a potential target for the development of targeted therapies in precision medicine. In this context, the particular potential of CHI3L1 inhibition could be considered in glioblastoma multiforme (GBM), whose tumors exhibit high levels of angiogenesis and increased CHI3L1 expression. This study aims to investigate whether inhibition of CHI3L1 in spheroids used as a GBM model affects the mechanisms of invasiveness; METHODS: We analyzed the interactions between CHI3L1 and the inhibitor G721-0282 in molecular docking and molecular dynamics (in silico) and infrared spectroscopy. Uptake of G721-0282 in GBM spheroids was measured using a label-free physical cytometer. Changes in E-, N- and VE-cadherins, VCAM-1, and EGFR were analyzed by immunohistochemical reactions, Western blot, and ddPCR methods in U-87 MG cells and GBM spheroids consisting of U-87 MG glioblastoma cells, HMEC-1 endothelial cells and macrophages; RESULTS: A direct interaction between CHI3L1 and G721-0282 was confirmed. G721-0282 decreased N-cadherins and VCAM-1 in GBM spheroids, but the changes in the 2D model of U-87 MG glioblastoma cells were different; CONCLUSION: Inhibition of CHI3L1 has the potential to reduce the invasiveness of GBM tumors. The 3D model of GBM spheroids is of great significance for investigating changes in membrane proteins and the tumor microenvironment.
{"title":"Inhibition of CHI3L1 decreases N-cadherin and VCAM-1 levels in glioblastoma.","authors":"Agnieszka Rusak, Marlena Gąsior-Głogowska, Azzurra Sargenti, Edward Krzyżak, Krzysztof Kotowski, Monika Mrozowska, Tomasz Górnicki, Krzysztof Kujawa, Piotr Dzięgiel","doi":"10.1007/s43440-024-00677-3","DOIUrl":"10.1007/s43440-024-00677-3","url":null,"abstract":"<p><strong>Background: </strong>The protein CHI3L1 contributes to cancer development by several mechanisms, including stimulation of angiogenesis and invasion as well as immunomodulatory effects. These properties make it a potential target for the development of targeted therapies in precision medicine. In this context, the particular potential of CHI3L1 inhibition could be considered in glioblastoma multiforme (GBM), whose tumors exhibit high levels of angiogenesis and increased CHI3L1 expression. This study aims to investigate whether inhibition of CHI3L1 in spheroids used as a GBM model affects the mechanisms of invasiveness; METHODS: We analyzed the interactions between CHI3L1 and the inhibitor G721-0282 in molecular docking and molecular dynamics (in silico) and infrared spectroscopy. Uptake of G721-0282 in GBM spheroids was measured using a label-free physical cytometer. Changes in E-, N- and VE-cadherins, VCAM-1, and EGFR were analyzed by immunohistochemical reactions, Western blot, and ddPCR methods in U-87 MG cells and GBM spheroids consisting of U-87 MG glioblastoma cells, HMEC-1 endothelial cells and macrophages; RESULTS: A direct interaction between CHI3L1 and G721-0282 was confirmed. G721-0282 decreased N-cadherins and VCAM-1 in GBM spheroids, but the changes in the 2D model of U-87 MG glioblastoma cells were different; CONCLUSION: Inhibition of CHI3L1 has the potential to reduce the invasiveness of GBM tumors. The 3D model of GBM spheroids is of great significance for investigating changes in membrane proteins and the tumor microenvironment.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"210-228"},"PeriodicalIF":3.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11743419/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142740119","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01Epub Date: 2024-10-21DOI: 10.1007/s43440-024-00662-w
Yousef A Abdelrady, Hayam S Thabet, Ahmed M Sayed
Metronomic chemotherapy (MC), long-term continuous administration of anticancer drugs, is gaining attention as an alternative to the traditional maximum tolerated dose (MTD) chemotherapy. By combining MC with other treatments, the therapeutic efficacy is enhanced while minimizing toxicity. MC employs multiple mechanisms, making it a versatile approach against various cancers. However, drug resistance limits the long-term effectiveness of MC, necessitating ongoing development of anticancer drugs. Traditional drug discovery is lengthy and costly due to processes like target protein identification, virtual screening, lead optimization, and safety and efficacy evaluations. Drug repurposing (DR), which screens FDA-approved drugs for new uses, is emerging as a cost-effective alternative. Both experimental and computational methods, such as protein binding assays, in vitro cytotoxicity tests, structure-based screening, and several types of association analyses (Similarity-Based, Network-Based, and Target Gene), along with retrospective clinical analyses, are employed for virtual screening. This review covers the mechanisms of MC, its application in various cancers, DR strategies, examples of repurposed drugs, and the associated challenges and future directions.
作为传统的最大耐受剂量化疗(MTD)的替代疗法,长期连续服用抗癌药物的 "剂量化疗"(MC)正受到越来越多的关注。通过将 MC 与其他治疗方法相结合,可在提高疗效的同时将毒性降至最低。MC 采用了多种机制,因此是治疗各种癌症的通用方法。然而,耐药性限制了 MC 的长期有效性,因此需要不断开发抗癌药物。传统的药物研发过程漫长而昂贵,需要经过靶蛋白鉴定、虚拟筛选、先导物优化以及安全性和有效性评估等过程。药物再利用(Drug repurposing,DR),即筛选美国食品及药物管理局批准的药物用于新用途,正在成为一种具有成本效益的替代方法。虚拟筛选采用了实验和计算方法,如蛋白质结合测定、体外细胞毒性测试、结构筛选、几种关联分析(基于相似性、基于网络和目标基因)以及回顾性临床分析。本综述涉及 MC 的机制、在各种癌症中的应用、DR 策略、再利用药物的实例以及相关挑战和未来方向。
{"title":"The future of metronomic chemotherapy: experimental and computational approaches of drug repurposing.","authors":"Yousef A Abdelrady, Hayam S Thabet, Ahmed M Sayed","doi":"10.1007/s43440-024-00662-w","DOIUrl":"10.1007/s43440-024-00662-w","url":null,"abstract":"<p><p>Metronomic chemotherapy (MC), long-term continuous administration of anticancer drugs, is gaining attention as an alternative to the traditional maximum tolerated dose (MTD) chemotherapy. By combining MC with other treatments, the therapeutic efficacy is enhanced while minimizing toxicity. MC employs multiple mechanisms, making it a versatile approach against various cancers. However, drug resistance limits the long-term effectiveness of MC, necessitating ongoing development of anticancer drugs. Traditional drug discovery is lengthy and costly due to processes like target protein identification, virtual screening, lead optimization, and safety and efficacy evaluations. Drug repurposing (DR), which screens FDA-approved drugs for new uses, is emerging as a cost-effective alternative. Both experimental and computational methods, such as protein binding assays, in vitro cytotoxicity tests, structure-based screening, and several types of association analyses (Similarity-Based, Network-Based, and Target Gene), along with retrospective clinical analyses, are employed for virtual screening. This review covers the mechanisms of MC, its application in various cancers, DR strategies, examples of repurposed drugs, and the associated challenges and future directions.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"1-20"},"PeriodicalIF":3.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142472215","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}