首页 > 最新文献

Journal of Pharmacology and Experimental Therapeutics最新文献

英文 中文
Pharmacological effects of small molecule BCR-ABL tyrosine kinase inhibitors on platelet function. 小分子 BCR-ABL 酪氨酸激酶抑制剂对血小板功能的药理作用。
IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-10-08 DOI: 10.1124/jpet.124.002104
Yiheng Zhang, Chih-Jen Yang, Alexander R Melrose, Jiaqing Pang, Kirrali Schofield, Serena D Song, Iván Parra-Izquierdo, Tony J Zheng, Joseph P Lyssikatos, Stefan D Gross, Joseph J Shatzel, Owen J T McCarty, Joseph E Aslan

Tyrosine kinase inhibitors (TKIs) targeting the BCR-ABL fusion protein, such as imatinib (Gleevec), have revolutionized targeted cancer therapies. However, drug resistance and side effects, particularly those affecting hemostasis, continue to pose significant challenges for TKI therapies. As tyrosine kinases serve pivotal roles in platelet hemostatic function, we investigated the potential impact of both established and emerging ABL TKIs on human platelet activities ex vivo Our study included standard-of-care agents (e.g., imatinib and nilotinib), and second-generation ABL inhibitors including ponatinib and bosutinib designed to mitigate drug resistance. Additionally, we explored the effects of allosteric inhibitors targeting the myristoyl pocket of ABL (e.g., asciminib and GNF-2), and novel agents in preclinical development, including ELVN-919, which uniquely exhibits high specificity for the ABL kinase active site. Our findings reveal that while ABL inhibitors such as ponatinib and bosutinib impede platelet activity, highly specific new-generation ABL inhibitors, including first-in-class therapeutics, do not impact platelet function ex vivo Overall, these new insights around the effects of ABL TKIs on platelet function could inform the development of targeted therapies with reduced hematologic toxicities. Significance Statement This study examines the effects of clinically relevant small molecule BCR-ABL tyrosine kinase inhibitors (TKIs) on platelet activity. This analysis includes first-time assessments of agents such as asciminib and ELVN-919 on human platelet function ex vivo, alongside established therapies (e.g., imatinib, ponatinib) with well-characterized effects on platelet function, to discern potential anti-platelet and other effects of BCR-ABL TKIs and inform clinical safety.

以BCR-ABL融合蛋白为靶点的酪氨酸激酶抑制剂(TKIs),如伊马替尼(格列卫),彻底改变了癌症靶向疗法。然而,耐药性和副作用,尤其是影响止血的副作用,仍然是TKI疗法面临的重大挑战。由于酪氨酸激酶在血小板止血功能中起着举足轻重的作用,我们研究了已确立的和新出现的 ABL TKIs 对体内外人体血小板活动的潜在影响。此外,我们还探讨了靶向ABL肉豆蔻酰口袋的异位抑制剂(如阿西米尼和GNF-2)以及临床前开发中的新型药物(包括ELVN-919)的效果,ELVN-919对ABL激酶活性位点具有独特的高特异性。我们的研究结果表明,虽然波纳替尼和伯舒替尼等 ABL 抑制剂会阻碍血小板活性,但高度特异性的新一代 ABL 抑制剂(包括一流的治疗药物)不会影响体外血小板功能。 总体而言,这些有关 ABL TKIs 对血小板功能影响的新见解可为开发血液毒性更低的靶向疗法提供参考。意义声明 本研究探讨了与临床相关的小分子 BCR-ABL 酪氨酸激酶抑制剂 (TKIs) 对血小板活性的影响。这项分析包括首次评估 asciminib 和 ELVN-919 等药物对体内外人体血小板功能的影响,以及对血小板功能有良好表征作用的既有疗法(如伊马替尼、泊纳替尼),以鉴别 BCR-ABL TKIs 的潜在抗血小板作用和其他作用,并为临床安全性提供依据。
{"title":"<b>Pharmacological effects of small molecule BCR-ABL tyrosine kinase inhibitors on platelet <b>function</b></b>.","authors":"Yiheng Zhang, Chih-Jen Yang, Alexander R Melrose, Jiaqing Pang, Kirrali Schofield, Serena D Song, Iván Parra-Izquierdo, Tony J Zheng, Joseph P Lyssikatos, Stefan D Gross, Joseph J Shatzel, Owen J T McCarty, Joseph E Aslan","doi":"10.1124/jpet.124.002104","DOIUrl":"10.1124/jpet.124.002104","url":null,"abstract":"<p><p>Tyrosine kinase inhibitors (TKIs) targeting the BCR-ABL fusion protein, such as imatinib (Gleevec), have revolutionized targeted cancer therapies. However, drug resistance and side effects, particularly those affecting hemostasis, continue to pose significant challenges for TKI therapies. As tyrosine kinases serve pivotal roles in platelet hemostatic function, we investigated the potential impact of both established and emerging ABL TKIs on human platelet activities <i>ex vivo</i> Our study included standard-of-care agents (e.g., imatinib and nilotinib), and second-generation ABL inhibitors including ponatinib and bosutinib designed to mitigate drug resistance. Additionally, we explored the effects of allosteric inhibitors targeting the myristoyl pocket of ABL (e.g., asciminib and GNF-2), and novel agents in preclinical development, including ELVN-919, which uniquely exhibits high specificity for the ABL kinase active site. Our findings reveal that while ABL inhibitors such as ponatinib and bosutinib impede platelet activity, highly specific new-generation ABL inhibitors, including first-in-class therapeutics, do not impact platelet function <i>ex vivo</i> Overall, these new insights around the effects of ABL TKIs on platelet function could inform the development of targeted therapies with reduced hematologic toxicities. <b>Significance Statement</b> This study examines the effects of clinically relevant small molecule BCR-ABL tyrosine kinase inhibitors (TKIs) on platelet activity. This analysis includes first-time assessments of agents such as asciminib and ELVN-919 on human platelet function <i>ex vivo</i>, alongside established therapies (e.g., imatinib, ponatinib) with well-characterized effects on platelet function, to discern potential anti-platelet and other effects of BCR-ABL TKIs and inform clinical safety.</p>","PeriodicalId":16798,"journal":{"name":"Journal of Pharmacology and Experimental Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142391375","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}
引用次数: 0
Metformin in Overcoming Enzalutamide Resistance in Castration-Resistant Prostate Cancer. 二甲双胍克服阉割耐药前列腺癌患者对恩扎鲁胺的耐药性
IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-10-08 DOI: 10.1124/jpet.124.002424
Kendall Simpson, Derek B Allison, Daheng He, Jinpeng Liu, Chi Wang, Xiaoqi Liu

Androgen deprivation is the standard treatment for prostate cancer (PCa) patients. However, the disease eventually progresses as castration-resistant PCa (CRPC). Enzalutamide, an AR inhibitor, is a typical drug to treating CRPC and due to continuous reliance on the drug, can lead to Enzalutamide-resistance (ENZ-r). This highlights the necessity for developing novel therapeutic targets to combat the gain of resistance. Metformin has been recently investigated for its potential anti-tumorigenic effects in many cancer types. In this study, we used enzalutamide and metformin in combination to explore the possible rescued efficacy of enzalutamide in the treatment of ENZ-r CRPC. We first tested the effects of this combination treatment on cell viability, drug synergy, and cell proliferation in ENZ-r CRPC cell lines. After combination treatment, we observed a decrease in cell proliferation and viability as well as a synergistic effect of both enzalutamide and metformin in vitro Following these results, we sought to explore how combination treatment effected mitochondrial fitness utilizing mitochondrial stress test analysis and mitochondrial membrane potential (MMP) shifts due to metformin's action in inhibiting Complex I of oxidative phosphorylation. We employed 2 different strategies of in vivo testing using 22Rv1 and LuCaP35CR xenograft models. Finally, RNA sequencing revealed a potential link in the downregulation of Ras/MAPK signaling following combination treatment. Significance Statement Increasing evidence suggests that oxidative phosphorylation might play a critical role in the development of resistance to cancer therapy. We showed that targeting oxidative phosphorylation with metformin can enhance the efficacy of enzalutamide in castration-resistant prostate cancer in vitro.

雄激素剥夺是前列腺癌(PCa)患者的标准治疗方法。然而,这种疾病最终会发展成为耐阉割性前列腺癌(CRPC)。恩杂鲁胺是一种AR抑制剂,是治疗CRPC的典型药物,由于持续依赖该药物,可能导致恩杂鲁胺耐药性(ENZ-r)。这就凸显了开发新型治疗靶点以对抗耐药性的必要性。二甲双胍最近被研究用于多种癌症类型的潜在抗肿瘤作用。在本研究中,我们将恩杂鲁胺和二甲双胍联合使用,以探索恩杂鲁胺在治疗ENZ-r CRPC中可能的抢救疗效。我们首先检测了联合治疗对ENZ-r CRPC细胞系的细胞活力、药物协同作用和细胞增殖的影响。在联合治疗后,我们观察到细胞增殖和存活率下降,以及恩杂鲁胺和二甲双胍在体外的协同作用。根据这些结果,我们试图利用线粒体应激测试分析和二甲双胍抑制氧化磷酸化复合物 I 的作用所导致的线粒体膜电位(MMP)变化,探讨联合治疗如何影响线粒体的健康。我们使用 22Rv1 和 LuCaP35CR 异种移植模型进行了两种不同的体内测试。最后,RNA 测序揭示了联合治疗后 Ras/MAPK 信号下调的潜在联系。意义声明 越来越多的证据表明,氧化磷酸化可能在癌症治疗耐药性的形成过程中起到关键作用。我们的研究表明,用二甲双胍靶向氧化磷酸化可增强恩杂鲁胺对体外阉割耐药前列腺癌的疗效。
{"title":"Metformin in Overcoming Enzalutamide Resistance in Castration-Resistant Prostate Cancer.","authors":"Kendall Simpson, Derek B Allison, Daheng He, Jinpeng Liu, Chi Wang, Xiaoqi Liu","doi":"10.1124/jpet.124.002424","DOIUrl":"https://doi.org/10.1124/jpet.124.002424","url":null,"abstract":"<p><p>Androgen deprivation is the standard treatment for prostate cancer (PCa) patients. However, the disease eventually progresses as castration-resistant PCa (CRPC). Enzalutamide, an AR inhibitor, is a typical drug to treating CRPC and due to continuous reliance on the drug, can lead to Enzalutamide-resistance (ENZ-r). This highlights the necessity for developing novel therapeutic targets to combat the gain of resistance. Metformin has been recently investigated for its potential anti-tumorigenic effects in many cancer types. In this study, we used enzalutamide and metformin in combination to explore the possible rescued efficacy of enzalutamide in the treatment of ENZ-r CRPC. We first tested the effects of this combination treatment on cell viability, drug synergy, and cell proliferation in ENZ-r CRPC cell lines. After combination treatment, we observed a decrease in cell proliferation and viability as well as a synergistic effect of both enzalutamide and metformin <i>in vitro</i> Following these results, we sought to explore how combination treatment effected mitochondrial fitness utilizing mitochondrial stress test analysis and mitochondrial membrane potential (MMP) shifts due to metformin's action in inhibiting Complex I of oxidative phosphorylation. We employed 2 different strategies of <i>in vivo</i> testing using 22Rv1 and LuCaP35CR xenograft models. Finally, RNA sequencing revealed a potential link in the downregulation of Ras/MAPK signaling following combination treatment. <b>Significance Statement</b> Increasing evidence suggests that oxidative phosphorylation might play a critical role in the development of resistance to cancer therapy. We showed that targeting oxidative phosphorylation with metformin can enhance the efficacy of enzalutamide in castration-resistant prostate cancer in vitro.</p>","PeriodicalId":16798,"journal":{"name":"Journal of Pharmacology and Experimental Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142391379","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}
引用次数: 0
The protective effect of Amitriptyline on experimental colitis through inhibiting TLR4/MD2 signaling pathway. 阿米替林通过抑制 TLR4/MD2 信号通路对实验性结肠炎的保护作用
IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-10-08 DOI: 10.1124/jpet.124.002207
Chengcheng Zeng, Qingqing Zhu, Wu Peng, Chen Huang, Huiting Chen, Hongli Huang, Yongjian Zhou, Chong Zhao

Amitriptyline, a pleiotropic tricyclic antidepressant, possesses anti-oxidant and anti-inflammatory properties. Despite its diverse benefits, the specific effects of amitriptyline on IBD are not yet well defined. To explore this, we utilized a DSS-induced colitis model to examine the anti-inflammatory effects of amitriptyline and the underlying mechanisms by which it operates. Our research revealed that amitriptyline is effective in alleviating several pathological manifestations associated with colitis. This includes improvements in body weight retention, reductions in DAI, lessening of colon length shortening, and repair of colonic mucosal damage. Treatment with amitriptyline significantly protected mucosal injury by preserving the population of goblet cells and increasing the expression of tight junction proteins. Furthermore, we observed that amitriptyline effectively countered immune cell infiltration, specifically neutrophils and macrophages, while simultaneously lowering the levels of inflammatory cytokines such as TNF-α, IL-1β, and IL-6. Additionally, RNA sequencing analysis pointed to the potential involvement of the TLR pathway in the anti-colitic effects induced by amitriptyline. Subsequent Western blot analysis indicated that amitriptyline significantly inhibited the TLR4-mediated NF-κB signaling pathway. To bolster our findings, in vitro studies demonstrated that amitriptyline down-regulated the TLR4/NF-κB/MAPK signaling cascades in mouse macrophages stimulated with LPS. Further molecular investigations revealed that amitriptyline was able to suppress the elevated expression of MD-2 that LPS stimulation typically induces. In summary, our findings suggest that amitriptyline effectively mitigates DSS-induced colitis in mice through the inhibition of TLR4/MD-2 pathway signaling, indicating its potential repurposing for IBD treatment. Significance Statement The potential of utilizing amitriptyline in treating IBD appears promising, leveraging its established safety and dosing profile as an antidepressant. Our observations show that amitriptyline can alleviate pathological symptoms, inflammation, and intestinal mucosal damage in mice with colitis induced by DSS. The protective effect observed appear to be linked to the inhibition of the TLR4/MD2 signaling pathway. By exploring novel applications for existing medications, we can optimize amitriptyline's efficacy and broaden its impact in both medical and commercial contexts.

阿米替林是一种多效三环类抗抑郁药,具有抗氧化和抗炎特性。尽管阿米替林具有多种益处,但其对 IBD 的具体作用尚未明确。为了探讨这一问题,我们利用 DSS 诱导的结肠炎模型来研究阿米替林的抗炎作用及其潜在的作用机制。我们的研究发现,阿米替林能有效缓解结肠炎的多种病理表现。这包括改善体重保持、降低 DAI、减少结肠长度缩短以及修复结肠粘膜损伤。使用阿米替林治疗可通过保留鹅口疮细胞数量和增加紧密连接蛋白的表达来显著保护粘膜损伤。此外,我们还观察到阿米替林能有效抑制免疫细胞的浸润,特别是中性粒细胞和巨噬细胞,同时降低炎性细胞因子(如 TNF-α、IL-1β 和 IL-6)的水平。此外,RNA 测序分析表明 TLR 途径可能参与了阿米替林诱导的抗胆碱能效应。随后的 Western 印迹分析表明,阿米替林能显著抑制 TLR4 介导的 NF-κB 信号通路。为了证实我们的研究结果,体外研究表明,阿米替林可下调小鼠巨噬细胞在LPS刺激下的TLR4/NF-κB/MAPK信号级联。进一步的分子研究表明,阿米替林能够抑制LPS刺激通常会诱导的MD-2表达升高。综上所述,我们的研究结果表明,阿米替林可通过抑制 TLR4/MD-2 通路信号转导,有效缓解 DSS 诱导的小鼠结肠炎,这表明阿米替林有可能被重新用于 IBD 治疗。意义声明 利用阿米替林治疗 IBD 的潜力似乎很有希望,它作为一种抗抑郁药,具有公认的安全性和剂量特征。我们的观察结果表明,阿米替林可减轻DSS诱导的结肠炎小鼠的病理症状、炎症和肠粘膜损伤。观察到的保护作用似乎与抑制 TLR4/MD2 信号通路有关。通过探索现有药物的新应用,我们可以优化阿米替林的疗效,扩大其在医疗和商业领域的影响。
{"title":"The protective effect of Amitriptyline on experimental colitis through inhibiting TLR4/MD2 signaling pathway.","authors":"Chengcheng Zeng, Qingqing Zhu, Wu Peng, Chen Huang, Huiting Chen, Hongli Huang, Yongjian Zhou, Chong Zhao","doi":"10.1124/jpet.124.002207","DOIUrl":"https://doi.org/10.1124/jpet.124.002207","url":null,"abstract":"<p><p>Amitriptyline, a pleiotropic tricyclic antidepressant, possesses anti-oxidant and anti-inflammatory properties. Despite its diverse benefits, the specific effects of amitriptyline on IBD are not yet well defined. To explore this, we utilized a DSS-induced colitis model to examine the anti-inflammatory effects of amitriptyline and the underlying mechanisms by which it operates. Our research revealed that amitriptyline is effective in alleviating several pathological manifestations associated with colitis. This includes improvements in body weight retention, reductions in DAI, lessening of colon length shortening, and repair of colonic mucosal damage. Treatment with amitriptyline significantly protected mucosal injury by preserving the population of goblet cells and increasing the expression of tight junction proteins. Furthermore, we observed that amitriptyline effectively countered immune cell infiltration, specifically neutrophils and macrophages, while simultaneously lowering the levels of inflammatory cytokines such as TNF-α, IL-1β, and IL-6. Additionally, RNA sequencing analysis pointed to the potential involvement of the TLR pathway in the anti-colitic effects induced by amitriptyline. Subsequent Western blot analysis indicated that amitriptyline significantly inhibited the TLR4-mediated NF-κB signaling pathway. To bolster our findings, in vitro studies demonstrated that amitriptyline down-regulated the TLR4/NF-κB/MAPK signaling cascades in mouse macrophages stimulated with LPS. Further molecular investigations revealed that amitriptyline was able to suppress the elevated expression of MD-2 that LPS stimulation typically induces. In summary, our findings suggest that amitriptyline effectively mitigates DSS-induced colitis in mice through the inhibition of TLR4/MD-2 pathway signaling, indicating its potential repurposing for IBD treatment. <b>Significance Statement</b> The potential of utilizing amitriptyline in treating IBD appears promising, leveraging its established safety and dosing profile as an antidepressant. Our observations show that amitriptyline can alleviate pathological symptoms, inflammation, and intestinal mucosal damage in mice with colitis induced by DSS. The protective effect observed appear to be linked to the inhibition of the TLR4/MD2 signaling pathway. By exploring novel applications for existing medications, we can optimize amitriptyline's efficacy and broaden its impact in both medical and commercial contexts.</p>","PeriodicalId":16798,"journal":{"name":"Journal of Pharmacology and Experimental Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142391381","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}
引用次数: 0
The protective effect of irisin against hemorrhagic injury is mediated by PI3K and p38 pathways in hemorrhage/resuscitation. 鸢尾素对出血损伤的保护作用是由出血/复苏过程中的 PI3K 和 p38 通路介导的。
IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-10-08 DOI: 10.1124/jpet.124.002238
Huai Wen, Naohiro Yano, Thomas Zhao, Lei Wei, Ting C Zhao

The objective of this study is to investigate whether PI3kinase (PI3K) and p38 mitogen-activated kinase contributes to the protection of irisin during hemorrhage/resuscitation. Experimental groups were divided by receiving the different treatments during resuscitation: I) Hemorrhage: Adult male CD-1 mice were subjected to hemorrhage at a mean arterial blood pressure of 35~45 mmHg for 60 min followed by 120 min of resuscitation (n=13); II) Hemorrhage + Irisin: receiving irisin (5µg/kg) (n=13); III) Hemorrhage + Irisin + PI3K inhibitor: receiving both Ly294002 (1mg/kg, i.v.) and irisin (n=6); IV) Hemorrhage + Irisin + p38 inhibitor: receiving SB202190 (1mg/kg, i.v.) and irisin (n=6). As compared to hemorrhage/resuscitation control, irisin improved the cardiac function and recovery of hemodynamics in association with the decreased systemic IL-1, IL-6, and TNF-α, which was completely abrogated by PI3K or p38 inhibitions. Furthermore, inhibition of PI3K or p38 abolished irisin-induced reduction of the infiltration of inflammatory cells and TUNEL-positive apoptosis in the cardiac and skeletal muscles. Irisin reduced TNF-α and IL6 expression in cardiac and skeletal muscle, which was abrogated by inhibition of PI3K or p38. Irisin-treated hemorrhage increases the phosphorylation of PI3K and p38 in both cardiac and skeletal muscle, which was mitigated by inhibition of PI3K or p38. Conclusion: PI3K and p38 play a critical role in modulating the protective effect of irisin during the hemorrhage/resuscitation. Significance Statement 1). This study has identified a critical pathway in regulation of trauma/hemorrhage by using a preclinical and reproducible model, in which Irisin, as a hormone factor, stimulates PI3K and p38 pathways to induce the protection against traumatic conditions. 2). The study holds promise to develop a new therapeutic strategy to target irisin and its pathway related to PI3K and p38 to treat trauma and its comorbidities to reduce mortality for clinical implication.

本研究旨在探讨PI3激酶(PI3K)和p38丝裂原活化激酶是否有助于在出血/复苏期间保护鸢尾素。实验组按复苏过程中接受的不同治疗进行划分:I) 大出血:成年雄性 CD-1 小鼠在平均动脉血压 35~45 mmHg 下大出血 60 分钟,然后复苏 120 分钟(n=13);II) 大出血 + 虹膜素:接受虹膜素(5µg/kg)(n=13);III) 大出血 + 虹膜素 + PI3K 抑制剂:同时接受 Ly294002(1mg/kg,i.静脉注射)和鸢尾素(n=6);IV)出血+鸢尾素+p38抑制剂:同时接受SB202190(1mg/kg,静脉注射)和鸢尾素(n=6)。与出血/复苏对照组相比,鸢尾素改善了心脏功能和血液动力学的恢复,同时降低了全身IL-1、IL-6和TNF-α,而PI3K或p38抑制剂则完全抑制了这一作用。此外,抑制 PI3K 或 p38 可抑制鸢尾素诱导的炎症细胞浸润和 TUNEL 阳性凋亡在心肌和骨骼肌中的减少。鸢尾素降低了心肌和骨骼肌中 TNF-α 和 IL6 的表达,而抑制 PI3K 或 p38 可抑制 TNF-α 和 IL6 的表达。鸢尾素处理的出血可增加心肌和骨骼肌中 PI3K 和 p38 的磷酸化,而抑制 PI3K 或 p38 可减轻这种磷酸化。结论PI3K 和 p38 在调节出血/复苏期间鸢尾素的保护作用方面起着关键作用。意义声明 1)。本研究通过使用临床前和可重复的模型,确定了调节创伤/出血的关键途径,其中鸢尾素作为一种激素因子,刺激 PI3K 和 p38 通路,诱导创伤条件下的保护作用。2).这项研究有望开发出一种新的治疗策略,以鸢尾素及其与 PI3K 和 p38 相关的通路为靶点,治疗创伤及其并发症,从而降低死亡率,达到临床目的。
{"title":"<b>The protective effect of irisin against hemorrhagic injury is mediated by PI3K and p38 pathways in hemorrhage/resuscitation.</b>","authors":"Huai Wen, Naohiro Yano, Thomas Zhao, Lei Wei, Ting C Zhao","doi":"10.1124/jpet.124.002238","DOIUrl":"https://doi.org/10.1124/jpet.124.002238","url":null,"abstract":"<p><p>The objective of this study is to investigate whether PI3kinase (PI3K) and p38 mitogen-activated kinase contributes to the protection of irisin during hemorrhage/resuscitation. Experimental groups were divided by receiving the different treatments during resuscitation: <b>I</b>) Hemorrhage: Adult male CD-1 mice were subjected to hemorrhage at a mean arterial blood pressure of 35~45 mmHg for 60 min followed by 120 min of resuscitation (n=13); <b>II</b>) Hemorrhage + Irisin: receiving irisin (5µg/kg) (n=13); <b>III</b>) Hemorrhage + Irisin + PI3K inhibitor: receiving both Ly294002 (1mg/kg, i.v.) and irisin (n=6); <b>IV</b>) Hemorrhage + Irisin + p38 inhibitor: receiving SB202190 (1mg/kg, i.v.) and irisin (n=6). As compared to hemorrhage/resuscitation control, irisin improved the cardiac function and recovery of hemodynamics in association with the decreased systemic IL-1, IL-6, and TNF-α, which was completely abrogated by PI3K or p38 inhibitions. Furthermore, inhibition of PI3K or p38 abolished irisin-induced reduction of the infiltration of inflammatory cells and TUNEL-positive apoptosis in the cardiac and skeletal muscles. Irisin reduced TNF-α and IL6 expression in cardiac and skeletal muscle, which was abrogated by inhibition of PI3K or p38. Irisin-treated hemorrhage increases the phosphorylation of PI3K and p38 in both cardiac and skeletal muscle, which was mitigated by inhibition of PI3K or p38<b>. Conclusion:</b> PI3K and p38 play a critical role in modulating the protective effect of irisin during the hemorrhage/resuscitation. <b>Significance Statement</b> 1). This study has identified a critical pathway in regulation of trauma/hemorrhage by using a preclinical and reproducible model, in which Irisin, as a hormone factor, stimulates PI3K and p38 pathways to induce the protection against traumatic conditions. 2). The study holds promise to develop a new therapeutic strategy to target irisin and its pathway related to PI3K and p38 to treat trauma and its comorbidities to reduce mortality for clinical implication.</p>","PeriodicalId":16798,"journal":{"name":"Journal of Pharmacology and Experimental Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142391376","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}
引用次数: 0
New potential ligand-receptor axis involved in tissue repair as therapeutic targets in progressive multiple sclerosis. 新的潜在配体-受体轴参与组织修复,可作为进行性多发性硬化症的治疗靶点。
IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-10-08 DOI: 10.1124/jpet.124.002254
Eugenio Antonio Carrera Silva, Jorge Correale, Carla Rothlin, Juan Manuel Ortiz Wilczyñski

Progressive multiple sclerosis (PMS) represents the worsening phase of the disease by accumulative neurodegeneration and disability, mainly refractory to current treatments. The therapeutic options remain challenging based partially on the lack of understanding of the pathogenic mechanisms but also because the early dogma was centered on neuroinflammation, overshadowing the critical role of the tissue repair process. The tissue repair target should necessarily start early in disease development and PMS should combine anti-inflammatory and neuroprotective therapeutic strategies. Increasing preclinical evidence, together with the new era of omics applied on frozen human brain tissue, shed light on some ligand receptors axis, such as GAS6/TYRO3 and PROS1/AXL required to dampen inflammation, promote tissue repair and engage remyelination, at the early stages of multiple sclerosis (MS) as a critical step in preventing or stopping neurodegeneration. Here, we will discuss those receptor/ligand pairs that could be targetable for therapeutic intervention in progressive MS disease. Significance Statement The aim for PMS should be to combine anti-inflammatory and neuroprotective therapeutic strategies based on early intervention. The TYRO3, AXL, and MERTK (TAM) signaling axis, particularly GAS6/TYRO3 and PROS1/AXL, which are involved in tempering inflammation, promoting tissue repair, and engaging remyelination, could significantly benefit patients at the early PMS.

进行性多发性硬化症(PMS)代表着疾病的恶化阶段,表现为神经退行性变和残疾的累积,主要是对当前治疗方法的难治性。治疗方案仍然具有挑战性,部分原因是对致病机制缺乏了解,另一部分原因是早期的教条以神经炎症为中心,掩盖了组织修复过程的关键作用。组织修复目标必须在疾病发展的早期开始,PMS 应结合抗炎和神经保护治疗策略。越来越多的临床前证据以及在冷冻人类脑组织上应用的新时代全息技术,揭示了一些配体受体轴,如 GAS6/TYRO3 和 PROS1/AXL,它们在多发性硬化症(MS)的早期阶段需要抑制炎症、促进组织修复和参与髓鞘再形成,这是预防或阻止神经退行性变的关键步骤。在此,我们将讨论那些可能成为进行性多发性硬化疾病治疗干预靶点的受体/配体对。意义声明 对进展期多发性硬化症的治疗目标应该是在早期干预的基础上,将抗炎和神经保护治疗策略结合起来。TYRO3、AXL和MERTK(TAM)信号轴,尤其是GAS6/TYRO3和PROS1/AXL,参与炎症缓解、促进组织修复和髓鞘再形成,可使早期多发性硬化症患者显著受益。
{"title":"New potential ligand-receptor axis involved in tissue repair as therapeutic targets in progressive multiple sclerosis.","authors":"Eugenio Antonio Carrera Silva, Jorge Correale, Carla Rothlin, Juan Manuel Ortiz Wilczyñski","doi":"10.1124/jpet.124.002254","DOIUrl":"https://doi.org/10.1124/jpet.124.002254","url":null,"abstract":"<p><p>Progressive multiple sclerosis (PMS) represents the worsening phase of the disease by accumulative neurodegeneration and disability, mainly refractory to current treatments. The therapeutic options remain challenging based partially on the lack of understanding of the pathogenic mechanisms but also because the early dogma was centered on neuroinflammation, overshadowing the critical role of the tissue repair process. The tissue repair target should necessarily start early in disease development and PMS should combine anti-inflammatory and neuroprotective therapeutic strategies. Increasing preclinical evidence, together with the new era of omics applied on frozen human brain tissue, shed light on some ligand receptors axis, such as GAS6/TYRO3 and PROS1/AXL required to dampen inflammation, promote tissue repair and engage remyelination, at the early stages of multiple sclerosis (MS) as a critical step in preventing or stopping neurodegeneration. Here, we will discuss those receptor/ligand pairs that could be targetable for therapeutic intervention in progressive MS disease. <b>Significance Statement</b> The aim for PMS should be to combine anti-inflammatory and neuroprotective therapeutic strategies based on early intervention. The TYRO3, AXL, and MERTK (TAM) signaling axis, particularly GAS6/TYRO3 and PROS1/AXL, which are involved in tempering inflammation, promoting tissue repair, and engaging remyelination, could significantly benefit patients at the early PMS.</p>","PeriodicalId":16798,"journal":{"name":"Journal of Pharmacology and Experimental Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142391380","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}
引用次数: 0
Correction to “Comments on: Increasing Enzyme Mannose-6-Phosphate Levels but Not Miglustat Coadministration Enhances the Efficacy of Enzyme Replacement Therapy in Pompe Mice” 更正 "关于:增加酶甘露糖-6-磷酸水平而非米格鲁司他联合用药可增强庞贝氏症小鼠酶替代疗法的疗效"
IF 3.5 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-10-01 DOI: 10.1124/jpet.123.002014err
American Society for Pharmacology and Experimental Therapeutics
{"title":"Correction to “Comments on: Increasing Enzyme Mannose-6-Phosphate Levels but Not Miglustat Coadministration Enhances the Efficacy of Enzyme Replacement Therapy in Pompe Mice”","authors":"American Society for Pharmacology and Experimental Therapeutics","doi":"10.1124/jpet.123.002014err","DOIUrl":"https://doi.org/10.1124/jpet.123.002014err","url":null,"abstract":"","PeriodicalId":16798,"journal":{"name":"Journal of Pharmacology and Experimental Therapeutics","volume":"31 1","pages":""},"PeriodicalIF":3.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142255507","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}
引用次数: 0
Contrasting the Reinforcing Effects of the Novel Dopamine Transport Inhibitors JJC8-088 and JJC8-091 in Monkeys: Potential Translation to Medication Assisted Treatment. 对比新型多巴胺转运抑制剂 JJC8-088 和 JJC8-091 对猴子的强化作用:药物辅助治疗的潜在转化。
IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-09-20 DOI: 10.1124/jpet.124.002356
Mia I Allen, Omeed Rahimi, Bernard N Johnson, Jianjing Cao, Amy Hauck Newman, Michael A Nader

Despite considerable efforts, there remains no FDA-approved medications for cocaine use disorder (CUD). One strategy to mitigate cocaine craving and relapse is to elevate dopamine (DA). The DA transport inhibitor and releaser d-amphetamine has been shown to decrease cocaine self-administration (SA), although it has abuse liability. Recently, several modafinil analogues reduced cocaine SA in rats and monkeys, including JJC8-088, characterized as "cocaine like" in rats, and JJC8-091, characterized as "atypical" and not SA by rats. The present studies evaluated the reinforcing effects of both compounds in monkeys under several conditions. For Experiment 1, four male cocaine-experienced rhesus monkeys self-administered cocaine (0.001-0.3 mg/kg/injection), JJC8-088 (0.001-0.3 mg/kg/injection), and JJC8-091 (0.1-3.0 mg/kg/injection) under a progressive-ratio (PR) schedule of reinforcement. Both JJC compounds functioned as reinforcers with equal reinforcing strength to cocaine. Although JJC8-091 was less potent than cocaine, JJC8-088 and cocaine had similar potencies. For Experiment 2, one male and two females drug-naïve cynomolgus monkeys responded on a fixed-ratio schedule of food reinforcement. JJC8-091 was self-administered at rates higher than saline in all three monkeys. In Experiment 3, monkeys from Experiment 2 responded under a concurrent drug vs. food choice paradigm and given access to cocaine or JJC8-091 under these conditions. At doses equal to or one-half log-units higher than doses used in Experiment 2, cocaine, but not JJC8-091, was chosen over food. Together, these results demonstrate that while JJC8-091 may be reinforcing under some conditions, its reinforcing strength, in the presence of an alternative reinforcer, is substantially less than cocaine. Significance Statement JJC8-088 and JJC8-091 have shown efficacy is reducing cocaine self-administration in rats and in nonhuman primates. This study found that both compounds maintained self-administration in monkeys responding under several conditions. However, when given access to an alternative reinforcer during the self-administration session, JJC8-091 was not reinforcing, suggesting that JJC8-091 may be a viable candidate for CUD since, in the human population, alternatives to drug use are often available.

尽管付出了巨大努力,但美国食品及药物管理局(FDA)仍未批准治疗可卡因使用障碍(CUD)的药物。缓解可卡因渴求和复吸的一种策略是提高多巴胺(DA)。DA转运抑制剂和释放剂d-苯丙胺已被证明可以减少可卡因自我给药,但它也有滥用的风险。最近,几种莫达非尼类似物减少了大鼠和猴子的可卡因自吸,其中包括在大鼠中被描述为 "类似可卡因 "的 JJC8-088,以及在大鼠中被描述为 "非典型 "且不自吸的 JJC8-091。本研究评估了这两种化合物在几种条件下对猴子的强化作用。在实验 1 中,四只有可卡因经验的雄性恒河猴在累进比率(PR)强化计划下自我注射了可卡因(0.001-0.3 毫克/千克/注射)、JJC8-088(0.001-0.3 毫克/千克/注射)和 JJC8-091(0.1-3.0 毫克/千克/注射)。这两种JJC化合物作为强化剂的强化强度与可卡因相当。虽然JJC8-091的效力低于可卡因,但JJC8-088和可卡因的效力相似。在实验 2 中,一只雄性和两只雌性对药物不敏感的犬科猴按照固定比例的食物强化时间表做出反应。三只猴子自我给药 JJC8-091 的比率均高于生理盐水。在实验 3 中,实验 2 中的猴子在同时进行的药物与食物选择范式下做出反应,并在这些条件下获得可卡因或 JJC8-091 。在剂量等于或高于实验 2 所用剂量对数单位的情况下,猴子选择可卡因而不是 JJC8-091 而不是食物。总之,这些结果表明,虽然 JJC8-091 在某些条件下可能具有强化作用,但在有替代强化物的情况下,其强化强度大大低于可卡因。意义声明 JJC8-088 和 JJC8-091 在减少大鼠和非人灵长类动物的可卡因自我给药方面具有疗效。这项研究发现,这两种化合物都能维持猴子在多种条件下的自我给药反应。这表明,JJC8-091 可能是一种可行的 CUD 候选药物,因为在人类群体中,吸毒的替代品通常是存在的。
{"title":"<b>Contrasting the Reinforcing Effects of the Novel Dopamine Transport Inhibitors JJC8-088 and JJC8-091 in Monkeys: Potential Translation to Medication Assisted Treatment</b>.","authors":"Mia I Allen, Omeed Rahimi, Bernard N Johnson, Jianjing Cao, Amy Hauck Newman, Michael A Nader","doi":"10.1124/jpet.124.002356","DOIUrl":"https://doi.org/10.1124/jpet.124.002356","url":null,"abstract":"<p><p>Despite considerable efforts, there remains no FDA-approved medications for cocaine use disorder (CUD). One strategy to mitigate cocaine craving and relapse is to elevate dopamine (DA). The DA transport inhibitor and releaser <i>d</i>-amphetamine has been shown to decrease cocaine self-administration (SA), although it has abuse liability. Recently, several modafinil analogues reduced cocaine SA in rats and monkeys, including JJC8-088, characterized as \"cocaine like\" in rats, and JJC8-091, characterized as \"atypical\" and not SA by rats. The present studies evaluated the reinforcing effects of both compounds in monkeys under several conditions. For Experiment 1, four male cocaine-experienced rhesus monkeys self-administered cocaine (0.001-0.3 mg/kg/injection), JJC8-088 (0.001-0.3 mg/kg/injection), and JJC8-091 (0.1-3.0 mg/kg/injection) under a progressive-ratio (PR) schedule of reinforcement. Both JJC compounds functioned as reinforcers with equal reinforcing strength to cocaine. Although JJC8-091 was less potent than cocaine, JJC8-088 and cocaine had similar potencies. For Experiment 2, one male and two females drug-naïve cynomolgus monkeys responded on a fixed-ratio schedule of food reinforcement. JJC8-091 was self-administered at rates higher than saline in all three monkeys. In Experiment 3, monkeys from Experiment 2 responded under a concurrent drug vs. food choice paradigm and given access to cocaine or JJC8-091 under these conditions. At doses equal to or one-half log-units higher than doses used in Experiment 2, cocaine, but not JJC8-091, was chosen over food. Together, these results demonstrate that while JJC8-091 may be reinforcing under some conditions, its reinforcing strength, in the presence of an alternative reinforcer, is substantially less than cocaine. <b>Significance Statement</b> JJC8-088 and JJC8-091 have shown efficacy is reducing cocaine self-administration in rats and in nonhuman primates. This study found that both compounds maintained self-administration in monkeys responding under several conditions. However, when given access to an alternative reinforcer during the self-administration session, JJC8-091 was not reinforcing, suggesting that JJC8-091 may be a viable candidate for CUD since, in the human population, alternatives to drug use are often available.</p>","PeriodicalId":16798,"journal":{"name":"Journal of Pharmacology and Experimental Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142289740","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}
引用次数: 0
Efficacy of Fluoxetine and (R,S) Ketamine in Attenuating Conditioned Fear Behaviors in Male Mice. 氟西汀和(R,S)氯胺酮对减轻雄性小鼠条件反射恐惧行为的功效
IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-09-20 DOI: 10.1124/jpet.124.002252
Megan Wells, Jan Hoffmann, Autumn Stage, Isabella Enger, Jayme Pomper, Lily Briggs, Amber LaCrosse

Post-traumatic stress disorder (PTSD) is caused by exposure to a traumatic or stressful event. Symptoms related to this disorder include persistent re-experiencing of memories and fear generalization. Current pharmacological treatments for PTSD are insufficient, with fewer than 30% of patients reporting symptom remission. This study aims to determine the efficacy of acute (R,S) ketamine and chronic fluoxetine (FLX) in reducing fear memory and fear generalization. In rodents, fear conditioning (FC) is commonly used in the literature to induce behaviors related to symptoms of PTSD, and the open field test (OFT) can assess anxiety and fear generalization behaviors during the exploration of a novel environment. In this study, FC consisted of a white noise cue stimulus and four inescapable foot shocks. Treatments began 4 hours after FC. Fear and anxiety behaviors were recorded during re-exposure to the FC stimuli at 24 hours and 2 weeks. The OFT was conducted one day before the last FC re-exposure. Results support the combined use of acute ketamine and chronic FLX as a treatment for reducing behaviors indicative of fear memory during re-exposure at 2 weeks, but not behaviors indicative of anxiety and fear generalization in the OFT. FLX alone was most effective in reducing behaviors related to fear generalization. This study contributes to the existing literature on pharmacological treatment for fear and anxiety behaviors relating to fear memory and fear generalization. Continued research is necessary to replicate results, optimize treatment protocols, and investigate the molecular adaptations to trauma and treatment. Significance Statement Up to 6% of people in the United States will develop PTSD within their lifetime, and less than half of those individuals will find relief from their symptoms given the current therapeutic options. This study offers preliminary support for the efficacy of ketamine and FLX in reducing PTSD-like behaviors induced by fear-conditioning in mice. Compared to current standard treatments, results indicate the potential for a more effective therapeutic option for those with stress-related disorders, such as PTSD.

创伤后应激障碍(PTSD)是由创伤或应激事件引起的。这种疾病的相关症状包括持续的记忆重现和恐惧泛化。目前针对创伤后应激障碍的药物治疗效果不佳,只有不到 30% 的患者症状得到缓解。本研究旨在确定急性(R,S)氯胺酮和慢性氟西汀(FLX)在减少恐惧记忆和恐惧泛化方面的疗效。在啮齿类动物中,恐惧条件反射(FC)是文献中常用的诱导创伤后应激障碍症状相关行为的方法,而开放场试验(OFT)可以评估探索新环境时的焦虑和恐惧泛化行为。在本研究中,FC 包括一个白噪声提示刺激和四个无法逃脱的脚震。治疗在 FC 4 小时后开始。分别在 24 小时和 2 周后再次接触 FC 刺激时记录恐惧和焦虑行为。OFT 在最后一次 FC 再次暴露前一天进行。结果表明,联合使用急性氯胺酮和慢性 FLX 可以减少 2 周后再次暴露时的恐惧记忆行为,但不能减少 OFT 中的焦虑和恐惧泛化行为。单独使用 FLX 对减少与恐惧泛化有关的行为最为有效。这项研究为现有的有关恐惧记忆和恐惧泛化的恐惧和焦虑行为的药物治疗文献做出了贡献。有必要继续开展研究,以复制研究结果、优化治疗方案并调查分子对创伤和治疗的适应性。意义声明 在美国,多达 6% 的人会在一生中患上创伤后应激障碍,而在目前的治疗方案下,只有不到一半的人能缓解症状。这项研究初步证实了氯胺酮和 FLX 在减少小鼠由恐惧条件反射诱发的创伤后应激障碍类似行为方面的疗效。与目前的标准疗法相比,研究结果表明,氯胺酮和FLX有可能为创伤后应激障碍等压力相关疾病患者提供更有效的治疗方案。
{"title":"<b>Efficacy of Fluoxetine and (<i>R,S)</i> Ketamine in Attenuating Conditioned Fear Behaviors in Male Mice</b>.","authors":"Megan Wells, Jan Hoffmann, Autumn Stage, Isabella Enger, Jayme Pomper, Lily Briggs, Amber LaCrosse","doi":"10.1124/jpet.124.002252","DOIUrl":"https://doi.org/10.1124/jpet.124.002252","url":null,"abstract":"<p><p>Post-traumatic stress disorder (PTSD) is caused by exposure to a traumatic or stressful event. Symptoms related to this disorder include persistent re-experiencing of memories and fear generalization. Current pharmacological treatments for PTSD are insufficient, with fewer than 30% of patients reporting symptom remission. This study aims to determine the efficacy of acute (<i>R,S)</i> ketamine and chronic fluoxetine (FLX) in reducing fear memory and fear generalization. In rodents, fear conditioning (FC) is commonly used in the literature to induce behaviors related to symptoms of PTSD, and the open field test (OFT) can assess anxiety and fear generalization behaviors during the exploration of a novel environment. In this study, FC consisted of a white noise cue stimulus and four inescapable foot shocks. Treatments began 4 hours after FC. Fear and anxiety behaviors were recorded during re-exposure to the FC stimuli at 24 hours and 2 weeks. The OFT was conducted one day before the last FC re-exposure. Results support the combined use of acute ketamine and chronic FLX as a treatment for reducing behaviors indicative of fear memory during re-exposure at 2 weeks, but not behaviors indicative of anxiety and fear generalization in the OFT. FLX alone was most effective in reducing behaviors related to fear generalization. This study contributes to the existing literature on pharmacological treatment for fear and anxiety behaviors relating to fear memory and fear generalization. Continued research is necessary to replicate results, optimize treatment protocols, and investigate the molecular adaptations to trauma and treatment. <b>Significance Statement</b> Up to 6% of people in the United States will develop PTSD within their lifetime, and less than half of those individuals will find relief from their symptoms given the current therapeutic options. This study offers preliminary support for the efficacy of ketamine and FLX in reducing PTSD-like behaviors induced by fear-conditioning in mice. Compared to current standard treatments, results indicate the potential for a more effective therapeutic option for those with stress-related disorders, such as PTSD.</p>","PeriodicalId":16798,"journal":{"name":"Journal of Pharmacology and Experimental Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142289741","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}
引用次数: 0
Methocinnamox is a Potent and Long-Acting Antagonist that can Prevent and Reverse Opioid-Induced Respiratory Depression. 甲氧肉桂醛是一种强效长效拮抗剂,可预防和逆转阿片类药物引起的呼吸抑制。
IF 3.5 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-09-18 DOI: 10.1124/jpet.124.002205
James A Carr,Daniel J Morgan
{"title":"Methocinnamox is a Potent and Long-Acting Antagonist that can Prevent and Reverse Opioid-Induced Respiratory Depression.","authors":"James A Carr,Daniel J Morgan","doi":"10.1124/jpet.124.002205","DOIUrl":"https://doi.org/10.1124/jpet.124.002205","url":null,"abstract":"","PeriodicalId":16798,"journal":{"name":"Journal of Pharmacology and Experimental Therapeutics","volume":"39 1","pages":"1-3"},"PeriodicalIF":3.5,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142255551","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}
引用次数: 0
Levetiracetam Prevents Neurophysiological Changes and Preserves Cognitive Function in the Human Immunodeficiency Virus (HIV)-1 Transactivator of Transcription Transgenic Mouse Model of HIV-Associated Neurocognitive Disorder. 在艾滋病毒相关神经认知障碍的 HIV-1 TAT 转基因小鼠模型中,左乙拉西坦能预防神经生理学变化并保护认知功能。
IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-09-18 DOI: 10.1124/jpet.124.002272
Ashley N Ewens, Alexander Pilski, Shayne D Hastings, Chris Krook-Magnuson, Steven M Graves, Esther Krook-Magnuson, Stanley A Thayer

Human immunodeficiency virus (HIV)-associated neurocognitive disorder (HAND) affects nearly half of the 39 million people living with HIV. HAND symptoms range from subclinical cognitive impairment to dementia; the mechanisms that underlie HAND remain unclear and there is no treatment. The HIV protein transactivator of transcription (TAT) is thought to contribute to HAND because it persists in the central nervous system and elicits neurotoxicity in animal models. Network hyperexcitability is associated with accelerated cognitive decline in neurodegenerative disorders. Here we show that the antiepileptic drug levetiracetam (LEV) attenuated aberrant excitatory synaptic transmission, protected synaptic plasticity, reduced seizure susceptibility, and preserved cognition in inducible TAT (iTAT) transgenic male mice. iTAT mice had an increased frequency of spontaneous excitatory postsynaptic currents in hippocampal slice recordings and impaired long-term potentiation, a form of synaptic plasticity that underlies learning and memory. Two-week administration of LEV by osmotic minipump prevented both impairments. Kainic acid administered to iTAT mice induced a higher maximum behavioral seizure score, longer seizure duration, and shorter latency to first seizure, consistent with a lower seizure threshold. LEV treatment prevented these in vivo signs of hyperexcitability. Lastly, in the Barnes maze, iTAT mice required more time to reach the goal, committed more errors, and received lower cognitive scores relative to iTAT mice treated with LEV. Thus, TAT expression drives functional deficits, suggesting a causative role in HAND. As LEV not only prevented aberrant synaptic activity in iTAT mice but also prevented cognitive dysfunction, it may provide a promising pharmacological approach to the treatment of HAND. SIGNIFICANCE STATEMENT: Approximately half of people living with human immunodeficiency virus (HIV) also suffer from HIV-associated neurocognitive disorder (HAND), for which there is no treatment. The HIV protein transactivator of transcription (TAT) causes toxicity that is thought to contribute to HAND. Here, the antiepileptic drug levetiracetam (LEV) prevented synaptic and cognitive impairments in a TAT-expressing mouse. LEV is widely used to treat seizures and is well-tolerated in humans, including those with HIV. This study supports further investigation of LEV-mediated neuroprotection in HAND.

在 3900 万艾滋病毒感染者中,近一半人患有艾滋病毒相关神经认知障碍(HAND)。HAND 的症状从亚临床认知障碍到痴呆不等;HAND 的发病机制尚不清楚,也没有治疗方法。艾滋病毒蛋白转录激活因子(TAT)被认为是导致 HAND 的原因,因为它在中枢神经系统中持续存在,并在动物模型中引起神经毒性。神经网络过度兴奋与神经退行性疾病的认知能力加速下降有关。在这里,我们发现抗癫痫药物左乙拉西坦(LEV)可以减轻异常兴奋性突触传递,保护突触可塑性,降低癫痫发作的易感性,并保护诱导性TAT(iTAT)转基因雄性小鼠的认知能力。iTAT小鼠在海马切片记录中的自发兴奋性突触后电流频率增加,长期电位(一种突触可塑性形式,是学习和记忆的基础)受损。通过渗透性微型泵给药两周的LEV可防止这两种损害。给 iTAT 小鼠注射凯尼酸会诱发较高的最大行为发作评分、较长的发作持续时间和较短的首次发作潜伏期,这与较低的发作阈值相符。LEV治疗可防止这些体内过度兴奋的迹象。最后,在巴恩斯迷宫中,与接受 LEV 治疗的 iTAT 小鼠相比,接受 LEV 治疗的 iTAT 小鼠需要更多时间才能到达目标,犯的错误更多,认知得分更低。因此,TAT的表达驱动了功能缺陷,表明它在手足徐动症中起着致病作用。由于LEV不仅能防止iTAT小鼠的突触活动异常,还能防止认知功能障碍,因此它可能为治疗手足徐动症提供一种很有前景的药理学方法。意义声明 约有一半的艾滋病病毒感染者患有艾滋病相关神经认知障碍(HAND),目前尚无治疗方法。艾滋病病毒蛋白 TAT 会引起毒性,被认为是导致 HAND 的原因之一。在这里,我们展示了一种抗癫痫药物--左乙拉西坦(LEV)--能防止TAT表达小鼠出现突触和认知障碍。左乙拉西坦被广泛用于治疗癫痫发作,在人类(包括艾滋病毒感染者)中的耐受性良好。这项研究支持进一步研究 LEV 治疗对 HAND 神经保护的作用。
{"title":"Levetiracetam Prevents Neurophysiological Changes and Preserves Cognitive Function in the Human Immunodeficiency Virus (HIV)-1 Transactivator of Transcription Transgenic Mouse Model of HIV-Associated Neurocognitive Disorder.","authors":"Ashley N Ewens, Alexander Pilski, Shayne D Hastings, Chris Krook-Magnuson, Steven M Graves, Esther Krook-Magnuson, Stanley A Thayer","doi":"10.1124/jpet.124.002272","DOIUrl":"10.1124/jpet.124.002272","url":null,"abstract":"<p><p>Human immunodeficiency virus (HIV)-associated neurocognitive disorder (HAND) affects nearly half of the 39 million people living with HIV. HAND symptoms range from subclinical cognitive impairment to dementia; the mechanisms that underlie HAND remain unclear and there is no treatment. The HIV protein transactivator of transcription (TAT) is thought to contribute to HAND because it persists in the central nervous system and elicits neurotoxicity in animal models. Network hyperexcitability is associated with accelerated cognitive decline in neurodegenerative disorders. Here we show that the antiepileptic drug levetiracetam (LEV) attenuated aberrant excitatory synaptic transmission, protected synaptic plasticity, reduced seizure susceptibility, and preserved cognition in inducible TAT (iTAT) transgenic male mice. iTAT mice had an increased frequency of spontaneous excitatory postsynaptic currents in hippocampal slice recordings and impaired long-term potentiation, a form of synaptic plasticity that underlies learning and memory. Two-week administration of LEV by osmotic minipump prevented both impairments. Kainic acid administered to iTAT mice induced a higher maximum behavioral seizure score, longer seizure duration, and shorter latency to first seizure, consistent with a lower seizure threshold. LEV treatment prevented these in vivo signs of hyperexcitability. Lastly, in the Barnes maze, iTAT mice required more time to reach the goal, committed more errors, and received lower cognitive scores relative to iTAT mice treated with LEV. Thus, TAT expression drives functional deficits, suggesting a causative role in HAND. As LEV not only prevented aberrant synaptic activity in iTAT mice but also prevented cognitive dysfunction, it may provide a promising pharmacological approach to the treatment of HAND. SIGNIFICANCE STATEMENT: Approximately half of people living with human immunodeficiency virus (HIV) also suffer from HIV-associated neurocognitive disorder (HAND), for which there is no treatment. The HIV protein transactivator of transcription (TAT) causes toxicity that is thought to contribute to HAND. Here, the antiepileptic drug levetiracetam (LEV) prevented synaptic and cognitive impairments in a TAT-expressing mouse. LEV is widely used to treat seizures and is well-tolerated in humans, including those with HIV. This study supports further investigation of LEV-mediated neuroprotection in HAND.</p>","PeriodicalId":16798,"journal":{"name":"Journal of Pharmacology and Experimental Therapeutics","volume":" ","pages":"104-118"},"PeriodicalIF":3.1,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11413936/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141766411","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}
引用次数: 0
期刊
Journal of Pharmacology and Experimental Therapeutics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1