The pleiotropic and protective effects of melatonin have been demonstrated in a variety of animal models of renal injury. While coding RNAs regulated by melatonin in renal tissues are well identified, the functional involvement of long noncoding RNAs (lncRNAs) in melatonin signaling remains undefined. This study identified nuclear enriched abundant transcript 1 (NEAT1), a clock-controlled lncRNA that was upregulated by melatonin through the BMAL1/CLOCK heterodimer in renal tubular epithelial cells (TECs). Mechanistic studies showed that melatonin enhanced NEAT1 expression via increasing BMAL1 stability and thereby the enrichment of BMAL1 on NEAT1's promoter. Further studies have revealed that NEAT1 promotes the proliferation of TECs by increasing levels of H3K27ac and H3K4me1 at the promoter regions of the proliferation gene MKI67. Treatment of albumin-injured TECs with melatonin promoted proliferation by transactivating NEAT1 and restoring the expression levels of core clock genes and MKI67. Moreover, melatonin treatment ameliorated proteinuria, hypoalbuminemia, and fibrotic lesions, which was correlated with increased levels of core clock genes, H3K27ac, Mki67, and Neat1 in experimental MN kidneys. Melatonin mediates a novel regulatory axis, BMAL1-NEAT1-MKI67, in TEC proliferation, establishing potential therapeutic targets for MN and other renal diseases.
{"title":"Melatonin Alleviates Albumin-Induced Tubular Cell Injury by Activating Clock-Controlled Nuclear Enriched Abundant Transcript 1-Mediated Proliferation.","authors":"Yen-Sung Huang, Kuo-Cheng Lu, Yu-Tien Chang, Shuk-Man Ka, Cheng-Yi Guo, Hsin-Yi Hsieh, Hsiu-Ming Shih, Huey-Kang Sytwu, Chia-Chao Wu","doi":"10.1021/acsptsci.4c00495","DOIUrl":"10.1021/acsptsci.4c00495","url":null,"abstract":"<p><p>The pleiotropic and protective effects of melatonin have been demonstrated in a variety of animal models of renal injury. While coding RNAs regulated by melatonin in renal tissues are well identified, the functional involvement of long noncoding RNAs (lncRNAs) in melatonin signaling remains undefined. This study identified nuclear enriched abundant transcript 1 (<i>NEAT</i>1), a clock-controlled lncRNA that was upregulated by melatonin through the BMAL1/CLOCK heterodimer in renal tubular epithelial cells (TECs). Mechanistic studies showed that melatonin enhanced <i>NEAT</i>1 expression via increasing BMAL1 stability and thereby the enrichment of <i>BMAL</i>1 on <i>NEAT</i>1's promoter. Further studies have revealed that <i>NEAT</i>1 promotes the proliferation of TECs by increasing levels of H3K27ac and H3K4me1 at the promoter regions of the proliferation gene <i>MKI</i>67. Treatment of albumin-injured TECs with melatonin promoted proliferation by transactivating <i>NEAT</i>1 and restoring the expression levels of core clock genes and <i>MKI</i>67. Moreover, melatonin treatment ameliorated proteinuria, hypoalbuminemia, and fibrotic lesions, which was correlated with increased levels of core clock genes, H3K27ac, <i>Mki</i>67, and <i>Neat</i>1 in experimental MN kidneys. Melatonin mediates a novel regulatory axis, BMAL1-<i>NEAT</i>1-<i>MKI</i>67, in TEC proliferation, establishing potential therapeutic targets for MN and other renal diseases.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 11","pages":"3607-3617"},"PeriodicalIF":4.9,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555500/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142629127","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-07eCollection Date: 2024-11-08DOI: 10.1021/acsptsci.4c00490
Abdul Salam, Asif Ali, Umar Nishan, Noaman Khan, Mohamed A Ibrahim, Zafar Iqbal, Nawshad Muhammad, Anum Fayyaz, Fawad Muhammad, Abdul Mateen, Zhiyuan Wu, Saifullah Afridi
Pancreatic cancer is one of the most lethal and fast-growing cancers with a poor prognosis. Herein, we report the expression of programmed death ligand 1 (PD-L1) as a new prognostic biomarker in pancreatic cancer progression analysis at the clinical level. Immunohistochemistry was performed on 86 clinically proven cases of pancreatic cancer tissue microarrays (TMAs) using anti-PD-L1 antibodies. Histoscore was done, and a variety of cutoffs were identified for analyses of the results. The chi-square test and Kaplan-Meier method were used to find the association between pancreatic cancer and various clinicopathological variables and the overall survival of the patients. PD-L1 expression was associated with histological grade and recurrence of the disease for epithelial and stromal staining at 10 histoscores. In addition, PD-L1 expression was strongly associated with lymph node involvement at the stromal 20 histoscore. The tumor stage of pancreatic cancer had an association with PD-L1 expression with epithelial and stromal 20 histoscores for all comparisons. At a stromal 20 histoscore, overall survival in high-low expression of PD-L1 was 7-19 months, and at a nuclear/cytoplasmic 10 histoscore, it was 9-28 months (p = 0.0001), respectively. Overall, PD-L1 overexpression in subcellular compartments was associated with disease aggression phenotypes and poor patient survival. Overexpression of PD-L1 was directly linked to pancreatic cancer progression and a poor survival rate. Therefore, PD-L1 may be used as a prognostic biomarker in the diagnosis, treatment, and management of pancreatic cancer patients.
{"title":"Investigation of Programmed Death Ligand-1 as a New Prognostic Biomarker in Pancreatic Cancer Patients.","authors":"Abdul Salam, Asif Ali, Umar Nishan, Noaman Khan, Mohamed A Ibrahim, Zafar Iqbal, Nawshad Muhammad, Anum Fayyaz, Fawad Muhammad, Abdul Mateen, Zhiyuan Wu, Saifullah Afridi","doi":"10.1021/acsptsci.4c00490","DOIUrl":"10.1021/acsptsci.4c00490","url":null,"abstract":"<p><p>Pancreatic cancer is one of the most lethal and fast-growing cancers with a poor prognosis. Herein, we report the expression of programmed death ligand 1 (PD-L1) as a new prognostic biomarker in pancreatic cancer progression analysis at the clinical level. Immunohistochemistry was performed on 86 clinically proven cases of pancreatic cancer tissue microarrays (TMAs) using anti-PD-L1 antibodies. Histoscore was done, and a variety of cutoffs were identified for analyses of the results. The chi-square test and Kaplan-Meier method were used to find the association between pancreatic cancer and various clinicopathological variables and the overall survival of the patients. PD-L1 expression was associated with histological grade and recurrence of the disease for epithelial and stromal staining at 10 histoscores. In addition, PD-L1 expression was strongly associated with lymph node involvement at the stromal 20 histoscore. The tumor stage of pancreatic cancer had an association with PD-L1 expression with epithelial and stromal 20 histoscores for all comparisons. At a stromal 20 histoscore, overall survival in high-low expression of PD-L1 was 7-19 months, and at a nuclear/cytoplasmic 10 histoscore, it was 9-28 months (<i>p</i> = 0.0001), respectively. Overall, PD-L1 overexpression in subcellular compartments was associated with disease aggression phenotypes and poor patient survival. Overexpression of PD-L1 was directly linked to pancreatic cancer progression and a poor survival rate. Therefore, PD-L1 may be used as a prognostic biomarker in the diagnosis, treatment, and management of pancreatic cancer patients.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 11","pages":"3585-3591"},"PeriodicalIF":4.9,"publicationDate":"2024-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555514/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142629120","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-03eCollection Date: 2024-10-11DOI: 10.1021/acsptsci.4c00432
Victor H Ruiz, David Encinas-Basurto, Neftali Ortega-Alarcon, Basanth Babu Eedara, Jeffrey R Fineman, Stephen M Black, Heidi M Mansour
Co-spray dried inhalable powder formulations of fasudil monohydrochloride salt (FMCS) and inhalable lung surfactant-based nanocarriers composed of synthetic phospholipids, zwitterionic DPPC (1,2-palmitoyl-sn-glycero-3-phosphocholine) and anionic DPPG (1,2-dipalmitoyl-sn-glycero-3-[phosphor-rac-1-glycerol]) sodium salt, were designed and optimized using organic solution advanced spray drying. FMCS can potentially be used for the treatment of various complex pulmonary diseases with this current work focusing on pulmonary arterial hypertension. Comprehensive physicochemical characterization, electron and optical microscopy imaging, thermal analysis, molecular fingerprinting spectroscopy, in vitro aerosol dispersion performance with human dry powder inhaler (DPI) devices, in vitro membrane permeation and drug release, and in vitro human cellular studies were conducted. Well-defined, small, and smooth nanoparticles/microparticles in the solid state were engineered at different molar ratios of FMCS/DPPC/DPPG (25:75, 50:50, and 75:25) and successfully produced as inhalable powders having the properties necessary for targeted pulmonary delivery as dry powder inhalers. In vitro aerosol performance demonstrated excellent aerosol dispersion with different DPI devices. The phospholipid bilayer biophysical properties were confirmed and retained following cospray drying. Sustained release of fasudil drug and in vitro biocompatibility were demonstrated on human lung cells from different airway regions.
{"title":"Inhalable Advanced Co-Spray Dried Microparticles/Nanoparticles of a Novel RhoA/Rho Kinase Inhibitor with Lung Surfactant Biomimetic Phospholipids for Targeted Lung Delivery.","authors":"Victor H Ruiz, David Encinas-Basurto, Neftali Ortega-Alarcon, Basanth Babu Eedara, Jeffrey R Fineman, Stephen M Black, Heidi M Mansour","doi":"10.1021/acsptsci.4c00432","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00432","url":null,"abstract":"<p><p>Co-spray dried inhalable powder formulations of fasudil monohydrochloride salt (FMCS) and inhalable lung surfactant-based nanocarriers composed of synthetic phospholipids, zwitterionic DPPC (1,2-palmitoyl-<i>sn</i>-glycero-3-phosphocholine) and anionic DPPG (1,2-dipalmitoyl-<i>sn</i>-glycero-3-[phosphor-rac-1-glycerol]) sodium salt, were designed and optimized using organic solution advanced spray drying. FMCS can potentially be used for the treatment of various complex pulmonary diseases with this current work focusing on pulmonary arterial hypertension. Comprehensive physicochemical characterization, electron and optical microscopy imaging, thermal analysis, molecular fingerprinting spectroscopy, in vitro aerosol dispersion performance with human dry powder inhaler (DPI) devices, in vitro membrane permeation and drug release, and in vitro human cellular studies were conducted. Well-defined, small, and smooth nanoparticles/microparticles in the solid state were engineered at different molar ratios of FMCS/DPPC/DPPG (25:75, 50:50, and 75:25) and successfully produced as inhalable powders having the properties necessary for targeted pulmonary delivery as dry powder inhalers. In vitro aerosol performance demonstrated excellent aerosol dispersion with different DPI devices. The phospholipid bilayer biophysical properties were confirmed and retained following cospray drying. Sustained release of fasudil drug and in vitro biocompatibility were demonstrated on human lung cells from different airway regions.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 10","pages":"3241-3254"},"PeriodicalIF":4.9,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11475283/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142476709","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-02eCollection Date: 2024-10-11DOI: 10.1021/acsptsci.4c00310
Deepali Kumari, Simranjit Kaur, Manoj P Dandekar
The secondary insult in the aftermath of traumatic brain injury (TBI) causes detrimental and self-perpetuating alteration in cells, resulting in aberrant function and the death of neuronal cells. The secondary insult is mainly driven by activation of the neuroinflammatory pathway. Among several classical pathways, the cGAS-STING pathway, a primary neuroinflammatory route, encompasses the cyclic GMP-AMP synthase (cGAS), stimulator of interferon genes (STING), and downstream signaling adaptor. Recently, the cGAS-STING research domain has gained exponential attention. The aberrant stimulation of cGAS-STING machinery and corresponding neuroinflammation have also been reported after TBI. In addition to the critical contribution to neuroinflammation, the cGAS-STING signaling also provokes neuronal cell death through various cell death mechanisms. This review highlights the structural and molecular mechanisms of the cGAS-STING machinery associated with TBI. We also focus on the intricate relationship and framework between cGAS-STING signaling and cell death mechanisms (autophagy, apoptosis, pyroptosis, ferroptosis, and necroptosis) in the aftermath of TBI. We suggest that the targeting of cGAS-STING signaling may open new therapeutic strategies to combat neuroinflammation and neurodegeneration in TBI.
{"title":"Intricate Role of the Cyclic Guanosine Monophosphate Adenosine Monophosphate Synthase-Stimulator of Interferon Genes (cGAS-STING) Pathway in Traumatic Brain Injury-Generated Neuroinflammation and Neuronal Death.","authors":"Deepali Kumari, Simranjit Kaur, Manoj P Dandekar","doi":"10.1021/acsptsci.4c00310","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00310","url":null,"abstract":"<p><p>The secondary insult in the aftermath of traumatic brain injury (TBI) causes detrimental and self-perpetuating alteration in cells, resulting in aberrant function and the death of neuronal cells. The secondary insult is mainly driven by activation of the neuroinflammatory pathway. Among several classical pathways, the cGAS-STING pathway, a primary neuroinflammatory route, encompasses the cyclic GMP-AMP synthase (cGAS), stimulator of interferon genes (STING), and downstream signaling adaptor. Recently, the cGAS-STING research domain has gained exponential attention. The aberrant stimulation of cGAS-STING machinery and corresponding neuroinflammation have also been reported after TBI. In addition to the critical contribution to neuroinflammation, the cGAS-STING signaling also provokes neuronal cell death through various cell death mechanisms. This review highlights the structural and molecular mechanisms of the cGAS-STING machinery associated with TBI. We also focus on the intricate relationship and framework between cGAS-STING signaling and cell death mechanisms (autophagy, apoptosis, pyroptosis, ferroptosis, and necroptosis) in the aftermath of TBI. We suggest that the targeting of cGAS-STING signaling may open new therapeutic strategies to combat neuroinflammation and neurodegeneration in TBI.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 10","pages":"2936-2950"},"PeriodicalIF":4.9,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11475349/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142476710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Although the coronavirus disease 2019 (COVID-19) crisis has passed, there remains a necessity for continuous efforts toward developing more targeted drugs and preparing for potential future virus attacks. Currently, most of the drugs received authorization for the treatment of COVID-19 have exhibited several limitations, such as poor metabolic stability, formidable preparation, and uncertain effectiveness. It is still significant to develop novel, structurally diverse small-molecule antiviral drugs targeting SARS-CoV-2 3-chymotrypsin-like protease (3CLpro). Herein, we report a class of alkynylamide-based nonpeptidic 3CLpro inhibitors that can be prepared conveniently by our previously developed one-pot synthetic method. The structure-activity relationships of alkynylamides as SARS-CoV-2 3CLpro inhibitors have been carefully investigated and discussed in this study. The two stereoisomers of the resulting molecules exhibit stereoselective interaction with 3CLpro, and the optimized compound (S,R)-4y inhibits 3CLpro with high potency (IC50 = 0.43 μM), low cytotoxicity, and acceptable cell permeability. Compound (S,R)-4y presents as a noncovalent inhibitor of 3CLpro against SARS-CoV-2 by the time-dependent inhibition assay (TDI) and mass spectrometry analysis. The Lineweaver-Burk plots, binding energy, surface plasmon resonance, and molecular docking studies suggest that (S,R)-4y specifically binds to an allosteric pocket of the SARS-CoV-2 3CLpro. These findings provide a novel class of nonpeptidic alkynylamide-based allosteric inhibitors with high selectivity against SARS-CoV-2 3CLpro featured by a simplified one-pot synthesis at room temperature in air.
{"title":"Novel Alkynylamide-Based Nonpeptidic Allosteric Inhibitors for SARS-CoV-2 3-Chymotrypsin-like Protease.","authors":"Jian Xue, Hongtao Li, Ruyu Wang, Meiting Wang, Xixiang Chen, Yaqi Deng, Jiani Lu, Yexi Li, Yuheng Song, Jianrong Xu, Tong Zhu, Lili Chen, Shunying Liu","doi":"10.1021/acsptsci.4c00369","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00369","url":null,"abstract":"<p><p>Although the coronavirus disease 2019 (COVID-19) crisis has passed, there remains a necessity for continuous efforts toward developing more targeted drugs and preparing for potential future virus attacks. Currently, most of the drugs received authorization for the treatment of COVID-19 have exhibited several limitations, such as poor metabolic stability, formidable preparation, and uncertain effectiveness. It is still significant to develop novel, structurally diverse small-molecule antiviral drugs targeting SARS-CoV-2 3-chymotrypsin-like protease (3CL<sup>pro</sup>). Herein, we report a class of alkynylamide-based nonpeptidic 3CL<sup>pro</sup> inhibitors that can be prepared conveniently by our previously developed one-pot synthetic method. The structure-activity relationships of alkynylamides as SARS-CoV-2 3CL<sup>pro</sup> inhibitors have been carefully investigated and discussed in this study. The two stereoisomers of the resulting molecules exhibit stereoselective interaction with 3CL<sup>pro</sup>, and the optimized compound (<i>S</i>,<i>R</i>)-<b>4y</b> inhibits 3CL<sup>pro</sup> with high potency (IC<sub>50</sub> = 0.43 μM), low cytotoxicity, and acceptable cell permeability. Compound (<i>S</i>,<i>R</i>)-<b>4y</b> presents as a noncovalent inhibitor of 3CL<sup>pro</sup> against SARS-CoV-2 by the time-dependent inhibition assay (TDI) and mass spectrometry analysis. The Lineweaver-Burk plots, binding energy, surface plasmon resonance, and molecular docking studies suggest that (<i>S</i>,<i>R</i>)-<b>4y</b> specifically binds to an allosteric pocket of the SARS-CoV-2 3CL<sup>pro</sup>. These findings provide a novel class of nonpeptidic alkynylamide-based allosteric inhibitors with high selectivity against SARS-CoV-2 3CL<sup>pro</sup> featured by a simplified one-pot synthesis at room temperature in air.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 10","pages":"3170-3191"},"PeriodicalIF":4.9,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11481521/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142476713","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-02eCollection Date: 2024-11-08DOI: 10.1021/acsptsci.4c00415
Tengfei Bian, Allison Lynch, Kayleigh Ballas, Jessica Mamallapalli, Breanne Freeman, Alexander Scala, Yifan Wang, Hussein Traboulsi, Ranjith Kumar Chellian, Amy Fagan, Zhixin Tang, Haocheng Ding, Umasankar De, Kristianna M Fredenburg, Zhiguang Huo, Carolyn J Baglole, Weizhou Zhang, Leah R Reznikov, Adriaan W Bruijnzeel, Chengguo Xing
Tobacco smoke remains a serious global issue, resulting in serious health complications, contributing to the onset of numerous preventive diseases and imposing significant health burdens. Despite regulatory policies and cessation measures aimed at curbing its usage, novel interventions are urgently needed for effective damage reduction. Our preclinical and pilot clinical studies showed that AB-free kava has the potential to reduce tobacco-smoking-induced lung cancer risk, mitigate tobacco dependence, and reduce tobacco use. To understand the scope of its benefits in damage reduction and potential limitations, this study evaluated the effects of AB-free kava on a panel of health indicators in mice exposed to 2-4 weeks of daily tobacco smoke exposure. Our assessments included global transcriptional profiling of the lung and liver tissues, analysis of lung inflammation, evaluation of lung function, exploration of tobacco nicotine withdrawal, and characterization of the causal protein kinase A (PKA) signaling pathway. As expected, tobacco smoke exposure perturbed a wide range of biological processes and compromised multiple functions in mice. Remarkably, AB-free kava demonstrated the ability to globally mitigate tobacco smoke-induced deficits at the molecular and functional levels with promising safety profiles, offering AB-free kava unique promise to mitigate tobacco smoke-related health damages. Further preclinical evaluations are warranted to fully harness the potential of AB-free kava in combating tobacco smoke-related harms in the preparation of its clinical translation.
烟草烟雾仍然是一个严重的全球性问题,会导致严重的健康并发症,引发多种预防性疾病,造成巨大的健康负担。尽管有旨在遏制烟草使用的监管政策和戒烟措施,但仍迫切需要新型干预措施来有效减少烟草危害。我们的临床前和试验性临床研究表明,不含 AB 的卡瓦具有降低吸烟诱发肺癌风险、减轻烟草依赖和减少烟草使用的潜力。为了了解不含 AB 的卡瓦在减少损害方面的益处范围和潜在局限性,本研究评估了不含 AB 的卡瓦对每天暴露于烟草烟雾 2-4 周的小鼠的一系列健康指标的影响。我们的评估包括肺部和肝脏组织的全局转录谱分析、肺部炎症分析、肺功能评估、烟草尼古丁戒断探索以及致病蛋白激酶 A(PKA)信号通路的特征描述。不出所料,烟草烟雾暴露扰乱了小鼠的多种生物过程,损害了多种功能。值得注意的是,不含 AB 的卡瓦在分子和功能水平上全面减轻了烟草烟雾引起的缺陷,而且安全性良好,这为不含 AB 的卡瓦减轻烟草烟雾相关的健康损害提供了独特的前景。为了充分利用无 AB 卡瓦在消除烟草烟雾相关危害方面的潜力,我们有必要进一步开展临床前评估,为临床转化做准备。
{"title":"Flavokavains A- and B-Free Kava Enhances Resilience against the Adverse Health Effects of Tobacco Smoke in Mice.","authors":"Tengfei Bian, Allison Lynch, Kayleigh Ballas, Jessica Mamallapalli, Breanne Freeman, Alexander Scala, Yifan Wang, Hussein Traboulsi, Ranjith Kumar Chellian, Amy Fagan, Zhixin Tang, Haocheng Ding, Umasankar De, Kristianna M Fredenburg, Zhiguang Huo, Carolyn J Baglole, Weizhou Zhang, Leah R Reznikov, Adriaan W Bruijnzeel, Chengguo Xing","doi":"10.1021/acsptsci.4c00415","DOIUrl":"10.1021/acsptsci.4c00415","url":null,"abstract":"<p><p>Tobacco smoke remains a serious global issue, resulting in serious health complications, contributing to the onset of numerous preventive diseases and imposing significant health burdens. Despite regulatory policies and cessation measures aimed at curbing its usage, novel interventions are urgently needed for effective damage reduction. Our preclinical and pilot clinical studies showed that AB-free kava has the potential to reduce tobacco-smoking-induced lung cancer risk, mitigate tobacco dependence, and reduce tobacco use. To understand the scope of its benefits in damage reduction and potential limitations, this study evaluated the effects of AB-free kava on a panel of health indicators in mice exposed to 2-4 weeks of daily tobacco smoke exposure. Our assessments included global transcriptional profiling of the lung and liver tissues, analysis of lung inflammation, evaluation of lung function, exploration of tobacco nicotine withdrawal, and characterization of the causal protein kinase A (PKA) signaling pathway. As expected, tobacco smoke exposure perturbed a wide range of biological processes and compromised multiple functions in mice. Remarkably, AB-free kava demonstrated the ability to globally mitigate tobacco smoke-induced deficits at the molecular and functional levels with promising safety profiles, offering AB-free kava unique promise to mitigate tobacco smoke-related health damages. Further preclinical evaluations are warranted to fully harness the potential of AB-free kava in combating tobacco smoke-related harms in the preparation of its clinical translation.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 11","pages":"3502-3517"},"PeriodicalIF":4.9,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555507/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142629111","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-30eCollection Date: 2024-10-11DOI: 10.1021/acsptsci.4c00525
Zongkai Peng, Yunpeng Lan, Susan L Nimmo, Richard Hoang Bui, Doris M Benbrook, Anthony W G Burgett, Zhibo Yang
Although most advanced-stage ovarian cancers initially respond to platinum- and taxane-based chemotherapy, the majority of them will recur and eventually develop chemoresistance. Among all drug resistance mechanisms, reduced drug uptake in tumors is regarded as an important pathway acquired by drug-resistant cancer cells. For patients with ovarian cancer, chemoresistant cells can develop into multicellular spheroids and spread through ascite fluid that accumulates in their abdomen. These spheroids consist of 3D structures that are highly heterogeneous with different shapes, sizes, and compositions of cell types. Thus, studying drug uptake at the single spheroid level is important for understanding chemosensitivity and chemoresistance; however, drug-uptake studies in single spheroids have not been previously reported due to the lack of a suitable analytical technique. In this study, we cultured spheroids using the ovarian cancer cell line (OVCAR-8) and treated them using paclitaxel or OSW-1, a natural compound with anticancer properties. We then developed a method of quantifying drug uptake in single spheroids using LC/MS measurements and then normalized the drug amount in each spheroid to its size and total protein content. Our method can be used in translational studies of drug development, treatment, and prediction of drug efficacy prior to chemotherapy.
{"title":"Mass Spectrometry Quantification of Anticancer Drug Uptake in Single Multicellular Tumor Spheroids.","authors":"Zongkai Peng, Yunpeng Lan, Susan L Nimmo, Richard Hoang Bui, Doris M Benbrook, Anthony W G Burgett, Zhibo Yang","doi":"10.1021/acsptsci.4c00525","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00525","url":null,"abstract":"<p><p>Although most advanced-stage ovarian cancers initially respond to platinum- and taxane-based chemotherapy, the majority of them will recur and eventually develop chemoresistance. Among all drug resistance mechanisms, reduced drug uptake in tumors is regarded as an important pathway acquired by drug-resistant cancer cells. For patients with ovarian cancer, chemoresistant cells can develop into multicellular spheroids and spread through ascite fluid that accumulates in their abdomen. These spheroids consist of 3D structures that are highly heterogeneous with different shapes, sizes, and compositions of cell types. Thus, studying drug uptake at the single spheroid level is important for understanding chemosensitivity and chemoresistance; however, drug-uptake studies in single spheroids have not been previously reported due to the lack of a suitable analytical technique. In this study, we cultured spheroids using the ovarian cancer cell line (OVCAR-8) and treated them using paclitaxel or OSW-1, a natural compound with anticancer properties. We then developed a method of quantifying drug uptake in single spheroids using LC/MS measurements and then normalized the drug amount in each spheroid to its size and total protein content. Our method can be used in translational studies of drug development, treatment, and prediction of drug efficacy prior to chemotherapy.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 10","pages":"3011-3016"},"PeriodicalIF":4.9,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11475282/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142476712","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-26eCollection Date: 2024-10-11DOI: 10.1021/acsptsci.4c00457
Rohit R Doke, Ganesh J Lamkhade, Kuldeep Vinchurkar, Sudarshan Singh
Neuroinflammatory mediators play a pivotal role in the pathogenesis of Alzheimer's Disease (AD), influencing its onset, progression, and severity. The precise mechanisms behind AD are still not fully understood, leading current treatments to focus mainly on managing symptoms rather than preventing or curing the condition. The amyloid and tau hypotheses are the most widely accepted explanations for AD pathology; however, they do not completely account for the neuronal degeneration observed in AD. Growing evidence underscores the crucial role of neuroinflammation in the pathology of AD. The neuroinflammatory hypothesis presents a promising new approach to understanding the mechanisms driving AD. This review examines the importance of neuroinflammatory biomarkers in the diagnosis, prognosis, and treatment of AD. It delves into the mechanisms underlying neuroinflammation in AD, highlighting the involvement of various mediators such as cytokines, chemokines, and ROS. Additionally, this review discusses the potential of neuroinflammatory biomarkers as diagnostic tools, prognostic indicators, and therapeutic targets for AD management. By understanding the intricate interplay between neuroinflammation and AD pathology, this review aims to help in the development of efficient diagnostic and treatment plans to fight this debilitating neurological condition. Furthermore, it elaborates recent advancements in neuroimaging techniques and biofluid analysis for the identification and monitoring of neuroinflammatory biomarkers in AD patients.
神经炎症介质在阿尔茨海默病(AD)的发病机制中起着关键作用,影响着其发病、进展和严重程度。人们对阿尔茨海默病的确切发病机制仍不完全清楚,因此目前的治疗方法主要集中在控制症状上,而不是预防或治愈该病。淀粉样蛋白和tau假说是最广为接受的AD病理学解释;然而,它们并不能完全解释AD中观察到的神经元变性。越来越多的证据强调了神经炎症在AD病理学中的关键作用。神经炎症假说为理解AD的驱动机制提供了一种很有希望的新方法。这篇综述探讨了神经炎症生物标志物在 AD 诊断、预后和治疗中的重要性。它深入探讨了 AD 神经炎症的内在机制,强调了细胞因子、趋化因子和 ROS 等各种介质的参与。此外,这篇综述还讨论了神经炎症生物标志物作为诊断工具、预后指标和 AD 治疗靶点的潜力。通过了解神经炎症与注意力缺失症病理之间错综复杂的相互作用,本综述旨在帮助制定有效的诊断和治疗计划,以对抗这种使人衰弱的神经系统疾病。此外,它还阐述了神经成像技术和生物流体分析在鉴定和监测 AD 患者神经炎症生物标志物方面的最新进展。
{"title":"Demystifying the Role of Neuroinflammatory Mediators as Biomarkers for Diagnosis, Prognosis, and Treatment of Alzheimer's Disease: A Review.","authors":"Rohit R Doke, Ganesh J Lamkhade, Kuldeep Vinchurkar, Sudarshan Singh","doi":"10.1021/acsptsci.4c00457","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00457","url":null,"abstract":"<p><p>Neuroinflammatory mediators play a pivotal role in the pathogenesis of Alzheimer's Disease (AD), influencing its onset, progression, and severity. The precise mechanisms behind AD are still not fully understood, leading current treatments to focus mainly on managing symptoms rather than preventing or curing the condition. The amyloid and tau hypotheses are the most widely accepted explanations for AD pathology; however, they do not completely account for the neuronal degeneration observed in AD. Growing evidence underscores the crucial role of neuroinflammation in the pathology of AD. The neuroinflammatory hypothesis presents a promising new approach to understanding the mechanisms driving AD. This review examines the importance of neuroinflammatory biomarkers in the diagnosis, prognosis, and treatment of AD. It delves into the mechanisms underlying neuroinflammation in AD, highlighting the involvement of various mediators such as cytokines, chemokines, and ROS. Additionally, this review discusses the potential of neuroinflammatory biomarkers as diagnostic tools, prognostic indicators, and therapeutic targets for AD management. By understanding the intricate interplay between neuroinflammation and AD pathology, this review aims to help in the development of efficient diagnostic and treatment plans to fight this debilitating neurological condition. Furthermore, it elaborates recent advancements in neuroimaging techniques and biofluid analysis for the identification and monitoring of neuroinflammatory biomarkers in AD patients.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 10","pages":"2987-3003"},"PeriodicalIF":4.9,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11475310/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142476708","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-26eCollection Date: 2024-10-11DOI: 10.1021/acsptsci.4c00489
Zhiwei Wen, Asmita Pramanik, Sarah A Lewicki, Young-Hwan Jung, Zhan-Guo Gao, John C R Randle, Sylvie Breton, Zhoumou Chen, Gregory S Whitehead, Daniela Salvemini, Donald N Cook, Kenneth A Jacobson
Tethered glycoconjugates of a naphthalene- and piperidine-containing antagonist of the P2Y14 receptor (PPTN) were synthesized, and their nM receptor binding affinity was determined using a fluorescent tracer in hP2Y14R-expressing whole CHO cells. The rationale for preparing mono- and disaccharide conjugates of the antagonists was to explore the receptor binding site, which we know recognizes a glucose moiety on the native agonist (UDP-glucose), as well as enhance aqueous solubility and pharmacokinetics, including kidney excretion to potentially counteract sterile inflammation. Glycoconjugates with varied linker length, including PEG chains, were compared in hP2Y14R binding, suggesting that an optimal affinity (IC50, nM) in the piperidine series was achieved for triazolyl N-linked glucose conjugates having one (8a, MRS4872, 3.21) or two (7a, MRS4865, 2.40) methylene spacers. In comparison of different carbohydrate conjugates lacking a piperidine moiety but containing triazole spacers, optimal hP2Y14R affinity (IC50, nM) was achieved with N-linked glycosides of fucose 10f (6.19) and lactose 10h (1.88), and C-linked glucose 11a (5.30). Selected compounds were examined in mouse models of conditions known to be ameliorated by P2Y14R antagonists. Two glycoconjugates that lacked a piperidine moiety, N-linked glucose derivative 10a and the isomeric C-linked glucose derivative 11a, were protective in a mouse model of allergic asthma. Piperidine-containing glucose conjugate 7a of intermediate linker length and corresponding glucuronide 7b (MRS4866) protected against neuropathic pain. Thus, glycoconjugation of a known antagonist scaffold has produced less hydrophobic P2Y14R antagonists having substantial in vitro and in vivo activity.
{"title":"Chimeras Derived from a P2Y<sub>14</sub> Receptor Antagonist and UDP-Sugar Agonists for Potential Treatment of Inflammation.","authors":"Zhiwei Wen, Asmita Pramanik, Sarah A Lewicki, Young-Hwan Jung, Zhan-Guo Gao, John C R Randle, Sylvie Breton, Zhoumou Chen, Gregory S Whitehead, Daniela Salvemini, Donald N Cook, Kenneth A Jacobson","doi":"10.1021/acsptsci.4c00489","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00489","url":null,"abstract":"<p><p>Tethered glycoconjugates of a naphthalene- and piperidine-containing antagonist of the P2Y<sub>14</sub> receptor (PPTN) were synthesized, and their nM receptor binding affinity was determined using a fluorescent tracer in hP2Y<sub>14</sub>R-expressing whole CHO cells. The rationale for preparing mono- and disaccharide conjugates of the antagonists was to explore the receptor binding site, which we know recognizes a glucose moiety on the native agonist (UDP-glucose), as well as enhance aqueous solubility and pharmacokinetics, including kidney excretion to potentially counteract sterile inflammation. Glycoconjugates with varied linker length, including PEG chains, were compared in hP2Y<sub>14</sub>R binding, suggesting that an optimal affinity (IC<sub>50</sub>, nM) in the piperidine series was achieved for triazolyl <i>N</i>-linked glucose conjugates having one (<b>8a</b>, MRS4872, 3.21) or two (<b>7a</b>, MRS4865, 2.40) methylene spacers. In comparison of different carbohydrate conjugates lacking a piperidine moiety but containing triazole spacers, optimal hP2Y<sub>14</sub>R affinity (IC<sub>50</sub>, nM) was achieved with <i>N</i>-linked glycosides of fucose <b>10f</b> (6.19) and lactose <b>10h</b> (1.88), and <i>C</i>-linked glucose <b>11a</b> (5.30). Selected compounds were examined in mouse models of conditions known to be ameliorated by P2Y<sub>14</sub>R antagonists. Two glycoconjugates that lacked a piperidine moiety, <i>N</i>-linked glucose derivative <b>10a</b> and the isomeric <i>C</i>-linked glucose derivative <b>11a</b>, were protective in a mouse model of allergic asthma. Piperidine-containing glucose conjugate <b>7a</b> of intermediate linker length and corresponding glucuronide <b>7b</b> (MRS4866) protected against neuropathic pain. Thus, glycoconjugation of a known antagonist scaffold has produced less hydrophobic P2Y<sub>14</sub>R antagonists having substantial in vitro and in vivo activity.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 10","pages":"3255-3278"},"PeriodicalIF":4.9,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11480895/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142476610","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-25eCollection Date: 2024-10-11DOI: 10.1021/acsptsci.4c00384
Dick Yan Tam, Wendy K M Lau, Yosephine Tania Limanto, Dennis K P Ng
Combination therapy has been proven as an effective strategy for cancer treatment. To this end, we report herein a self-assembled nucleic acid-based complex for dual photodynamic and antisense therapy. It contains a nucleolin-targeting As1411-based G-quadruplex platform, a partially hybridized antisense oligonucleotide 4625, which can inhibit the antiapoptotic protein B cell lymphoma-xL inducing apoptotic cell death, and a zinc(II) phthalocyanine (ZnPc)-based photosensitizer held by noncovalent interactions. Through a series of in vitro experiments, we have demonstrated that this DNA complex can be internalized selectively to nucleolin-overexpressed MCF-7 and A549 cells through receptor-mediated endocytosis and is localized in the lysosomes. Upon light irradiation, the photosensitization of ZnPc triggers the formation of reactive oxygen species for cell killing and promotes the lysosomal escape of 4625 for antisense therapy. The combined therapeutic effect can eliminate the cancer cells effectively with a half maximal inhibitory concentration of ca. 0.5 μM.
联合疗法已被证明是一种有效的癌症治疗策略。为此,我们在此报告一种基于核酸的自组装复合物,用于光动力和反义双重疗法。它包含一个以 As1411 为基础的核素靶向 G-四联体平台、一个部分杂交的反义寡核苷酸 4625(可抑制诱导细胞凋亡的抗凋亡蛋白 B 细胞淋巴瘤-xL)和一个以锌(II)酞菁(ZnPc)为基础、通过非共价相互作用保持的光敏剂。通过一系列体外实验,我们证明了这种 DNA 复合物可通过受体介导的内吞作用选择性地内化到过表达的 MCF-7 和 A549 细胞中,并定位于溶酶体中。在光照射下,ZnPc 的光敏化会引发活性氧的形成,从而杀死细胞,并促进 4625 在溶酶体中的逃逸,从而达到反义治疗的目的。联合治疗效果可有效消灭癌细胞,半数最大抑制浓度约为 0.5 μM。
{"title":"Light-Promoted Lysosomal Escape of a Phthalocyanine and Antisense Oligonucleotide-Complexed G-Quadruplex for Dual Photodynamic and Antisense Therapy.","authors":"Dick Yan Tam, Wendy K M Lau, Yosephine Tania Limanto, Dennis K P Ng","doi":"10.1021/acsptsci.4c00384","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00384","url":null,"abstract":"<p><p>Combination therapy has been proven as an effective strategy for cancer treatment. To this end, we report herein a self-assembled nucleic acid-based complex for dual photodynamic and antisense therapy. It contains a nucleolin-targeting As1411-based G-quadruplex platform, a partially hybridized antisense oligonucleotide 4625, which can inhibit the antiapoptotic protein B cell lymphoma-xL inducing apoptotic cell death, and a zinc(II) phthalocyanine (ZnPc)-based photosensitizer held by noncovalent interactions. Through a series of in vitro experiments, we have demonstrated that this DNA complex can be internalized selectively to nucleolin-overexpressed MCF-7 and A549 cells through receptor-mediated endocytosis and is localized in the lysosomes. Upon light irradiation, the photosensitization of ZnPc triggers the formation of reactive oxygen species for cell killing and promotes the lysosomal escape of 4625 for antisense therapy. The combined therapeutic effect can eliminate the cancer cells effectively with a half maximal inhibitory concentration of ca. 0.5 μM.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 10","pages":"3216-3227"},"PeriodicalIF":4.9,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11475320/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142476711","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}