Molecular Pharmacology最新文献

筛选
英文 中文
Isoprenaline shows unique kinase dependencies in stimulating β1AR-β-arrestin2 interaction compared to endogenous catecholamines. 与内源性儿茶酚胺相比,异丙肾上腺素在刺激β1AR-β-arrestin2相互作用方面表现出独特的激酶依赖性。
IF 3.2 3区 医学
Molecular Pharmacology Pub Date : 2025-06-01 Epub Date: 2025-04-21 DOI: 10.1016/j.molpha.2025.100041
Edda S F Matthees, Luca E Kletzin, Arnelle Löbbert, Jana S Hoffmann, Carolin Große, Alvar D Gossert, Carsten Hoffmann
{"title":"Isoprenaline shows unique kinase dependencies in stimulating β<sub>1</sub>AR-β-arrestin2 interaction compared to endogenous catecholamines.","authors":"Edda S F Matthees, Luca E Kletzin, Arnelle Löbbert, Jana S Hoffmann, Carolin Große, Alvar D Gossert, Carsten Hoffmann","doi":"10.1016/j.molpha.2025.100041","DOIUrl":"10.1016/j.molpha.2025.100041","url":null,"abstract":"<p><p>The β1-adrenergic receptor (β<sub>1</sub>AR) is an essential G protein-coupled receptor in the heart. Its dysregulation represents a hallmark of cardiac diseases. Studies have identified a unique mode of β-arrestin interaction, where β<sub>1</sub>AR briefly engages with β-arrestins before catalytically accumulating them at the plasma membrane (PM) independently of the receptor. Although receptor phosphorylation crucially impacts β-arrestins, the contributions of specific kinases vital in β<sub>1</sub>AR regulation remain unclear. Here, we employed G protein-coupled receptor kinase (GRK) GRK2/3/5/6 knockout cells and the protein kinase A inhibitor H89 in bioluminescence resonance energy transfer-based assays to systematically assess GRKs and protein kinase A in direct β-arrestin2 recruitment to β<sub>1</sub>AR and β-arrestin2 translocation to the PM. Furthermore, we compared the effects of the synthetic agonist isoprenaline with the endogenous catecholamines: epinephrine and norepinephrine. We observed pronounced differences in their kinase dependencies to mediate β-arrestin2 translocation to the PM. Upon isoprenaline stimulation, GRKs strongly influenced β-arrestin2 translocation to the PM but had no effect on direct β-arrestin2 recruitment to β<sub>1</sub>AR. Additionally, in a GRK2-specific context, protein kinase A inhibition primarily reduced the efficacy of isoprenaline for β-arrestin2 translocation, whereas for GRK5, it decreased potency. Strikingly, these kinase-dependent effects were absent for epinephrine and norepinephrine, suggesting distinct underlying molecular mechanisms for β-arrestin2 accumulation at the PM. This observation could be explained by agonist-specific differences in receptor conformational rearrangements, as suggested by distinct changes in the NMR spectra of β<sub>1</sub>AR. Our findings highlight that synthetic and endogenous ligands induce distinct molecular mechanisms in β<sub>1</sub>AR regulation, emphasizing the need to consider these differences when translating molecular insights into physiological contexts. SIGNIFICANCE STATEMENT: Our findings reveal mechanistic differences in β1-adrenergic receptor-mediated catalytic activation of β-arrestin2 by synthetic and endogenous agonists, driven by distinct G protein-coupled receptor kinases and protein kinase A dependencies. Although β-arrestin2 translocation to the PM occurred to similar extents with isoprenaline, epinephrine, and norepinephrine, kinase involvement was crucial only upon Iso stimulation of β1-adrenergic receptor. By elucidating these ligand-specific pathways, this study advances our understanding of β1-adrenergic receptor signaling and regulation while additionally highlighting the importance of considering these differences when translating molecular insights into pathophysiological contexts.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":"107 6","pages":"100041"},"PeriodicalIF":3.2,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12264555/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144028297","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
Corrigendum to "Targeting the Metastasis Suppressor, N-Myc Downstream Regulated Gene-1, with Novel Di-2- Pyridylketone Thiosemicarbazones: Suppression of Tumor Cell Migration and Cell-Collagen Adhesion by Inhibiting Focal Adhesion Kinase/Paxillin Signaling". 针对转移抑制因子N-Myc下游调控基因-1,使用新型的Di-2-吡啶基酮硫代氨基脲:通过抑制局灶黏附激酶/帕罗西林信号传导抑制肿瘤细胞迁移和细胞-胶原粘附
IF 3.2 3区 医学
Molecular Pharmacology Pub Date : 2025-05-01 Epub Date: 2025-04-15 DOI: 10.1016/j.molpha.2025.100038
{"title":"Corrigendum to \"Targeting the Metastasis Suppressor, N-Myc Downstream Regulated Gene-1, with Novel Di-2- Pyridylketone Thiosemicarbazones: Suppression of Tumor Cell Migration and Cell-Collagen Adhesion by Inhibiting Focal Adhesion Kinase/Paxillin Signaling\".","authors":"","doi":"10.1016/j.molpha.2025.100038","DOIUrl":"10.1016/j.molpha.2025.100038","url":null,"abstract":"","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":"107 5","pages":"100038"},"PeriodicalIF":3.2,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143971671","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
Discovery of RGS2-FBXO44 interaction inhibitors using a cell-based NanoBit assay. 使用基于细胞的NanoBit检测发现RGS2-FBXO44相互作用抑制剂。
IF 3.2 3区 医学
Molecular Pharmacology Pub Date : 2025-05-01 Epub Date: 2025-03-19 DOI: 10.1016/j.molpha.2025.100030
Sadikshya Aryal, Cindy Shi Yee Wong, Harrison J McNabb, Ahmad Junaid, Ryan A Altman, Benita Sjögren
{"title":"Discovery of RGS2-FBXO44 interaction inhibitors using a cell-based NanoBit assay.","authors":"Sadikshya Aryal, Cindy Shi Yee Wong, Harrison J McNabb, Ahmad Junaid, Ryan A Altman, Benita Sjögren","doi":"10.1016/j.molpha.2025.100030","DOIUrl":"10.1016/j.molpha.2025.100030","url":null,"abstract":"<p><p>Regulators of G protein signaling (RGS) proteins negatively regulate signaling through G protein-coupled receptors, and reduced RGS protein function is involved in numerous pathologies. However, therapeutic intervention is challenging, as RGS proteins lack druggable binding pockets and enzymatic activity. Instead, targeting mechanisms that control RGS protein expression show promise as an alternative. Pharmacological stabilization of RGS2 would be a feasible therapeutic strategy in pathologies associated with reduced RGS2 protein levels, such as hypertension, heart failure, and asthma. RGS2 is rapidly degraded through the ubiquitin-proteasomal system, and we recently identified the E3 ligase that recognizes RGS2. F-box Only Protein 44 (FBXO44) acts as the substrate recognition site for RGS2 in this E3 ligase complex, and we hypothesize that inhibiting the RGS2-FBXO44 interaction will lead to enhanced RGS2 levels. Here, we developed a NanoLuc Binary Technology (NanoBiT) assay that detects the interaction between RGS2 and FBXO44. This assay was used to screen 1600 compounds from the Life Chemicals protein-protein interaction fragment library. We identified a promising hit, denoted compound 10, that inhibits the RGS2-FBXO44 interaction with a potency of 19.6 μM, through direct binding to RGS2. The resulting increase in RGS2 protein levels is dependent on FBXO44, as siRNA-mediated FBXO44 knockdown attenuates the effect of compound 10. Altogether, compound 10 represents the first example of a small-molecule inhibitor of the RGS2-FBXO44 interaction and a first step toward the development of molecular probes with a defined mechanism to stabilize RGS2 protein levels. SIGNIFICANCE STATEMENT: This study provides a strategy to identify molecules that selectively inhibit RGS2 protein degradation as well as the first example of a compound with the ability to inhibit RGS2 interaction with the E3 ligase component FBXO44. This study provides proof of concept that a small-molecule RGS2-FBXO44 interaction inhibitor will increase RGS2 protein levels. Future development of compounds with this mechanism of action would be clinically useful in pathologies associated with low RGS2 protein levels, including hypertension, heart failure, and asthma.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":"107 5","pages":"100030"},"PeriodicalIF":3.2,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143811903","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
Multiwalled carbon nanotubes activate the NLRP3 inflammasome-dependent pyroptosis in macrophages. 多壁碳纳米管激活巨噬细胞NLRP3炎性体依赖性焦亡。
IF 3 3区 医学
Molecular Pharmacology Pub Date : 2025-05-01 Epub Date: 2025-04-02 DOI: 10.1016/j.molpha.2025.100031
Chol Seung Lim, Ja Kook Gu, Qiang Ma
{"title":"Multiwalled carbon nanotubes activate the NLRP3 inflammasome-dependent pyroptosis in macrophages.","authors":"Chol Seung Lim, Ja Kook Gu, Qiang Ma","doi":"10.1016/j.molpha.2025.100031","DOIUrl":"10.1016/j.molpha.2025.100031","url":null,"abstract":"<p><p>Macrophages are major innate immune cells for the clearance of inhaled nanoparticles but may undergo cell death upon phagocytosis of certain nanoparticles due to their resistance to lysosomal degradation and high toxicity to the cell. Here we investigated the pyroptotic effect of exposure to fibrogenic multiwalled carbon nanotubes (MWCNTs) on macrophages, an inflammatory form of cell death. We first evaluated MWCNT-induced cell death in M1 and M2 macrophages that mediate the temporal inflammatory response to MWCNTs in mammalian lungs. Macrophages were differentiated from human monocytic THP-1 cells, followed by polarization to M1 or M2 cells. MWCNTs caused concentration- and time-dependent cytotoxicity in M1 and, to a lesser extent, M2 cells. Carbon black, an amorphous carbonous material control for CNTs, did not cause apparent toxicity in the cells. MWCNTs increased the production and secretion of IL-1β, accompanied by activation of caspase-1, in M1, but not M2, cells. Moreover, MWCNTs induced the formation of apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain specks and the release of cathepsin B in M1 cells, revealing activation of the nucleotide-binding, oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome via lysosomal damage. MWCNTs induced the cleavage of gasdermin D (GSDMD) to form the 31 kDa N-terminal fragment (GSDMD-N), the pore-forming peptide causing pyroptotic cell death. Increased IL-1β release was completely suppressed by AC-YVAD-CMK (a caspase-1 inhibitor), MCC-950 (an NLRP3 inflammasome inhibitor), or CA-074 Me (a cathepsin B inhibitor), alongside the blockage of MWCNT-induced cleavage of GSDMD. The study demonstrates that MWCNTs trigger pyroptosis in M1 macrophages and boost sterile inflammation by activating the NLRP3 inflammasome pathway. SIGNIFICANCE STATEMENT: The nucleotide-binding, oligomerization domain-like receptor family pyrin domain containing 3 inflammasome mediates the inflammatory response to fibrogenic nanoparticles in the lung via multiple means. The current study uncovers the induction of pyroptotic death of macrophages as a major means of nanotoxicity and sterile inflammation via the nucleotide-binding, oligomerization domain-like receptor family pyrin domain containing 3 pathway by nanoparticles.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":"107 5","pages":"100031"},"PeriodicalIF":3.0,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12305632/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144010182","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
Non-nutritive sweeteners in food-drug interactions: An overview of current evidence. 食品药物相互作用中的非营养性甜味剂:当前证据综述。
IF 3.2 3区 医学
Molecular Pharmacology Pub Date : 2025-05-01 Epub Date: 2025-04-09 DOI: 10.1016/j.molpha.2025.100035
Laura Danner, Kale Kroenke, Stephanie Olivier-Van Stichelen
{"title":"Non-nutritive sweeteners in food-drug interactions: An overview of current evidence.","authors":"Laura Danner, Kale Kroenke, Stephanie Olivier-Van Stichelen","doi":"10.1016/j.molpha.2025.100035","DOIUrl":"10.1016/j.molpha.2025.100035","url":null,"abstract":"<p><p>Food-drug interactions occur when the presence of foods interferes with the absorption, distribution, metabolism, or excretion of pharmaceuticals. Specific compounds within foods, like certain phytochemicals from grapefruit, have been known to precipitate food-drug interactions for decades, leading to guidance from physicians and pharmacists about patients' dietary restrictions while taking certain drugs. Although approved by the Food and Drug Administration, high-intensity non-nutritive sweeteners (NNS) share qualities with drugs that suggest the potential for similar interactions. In this minireview, we have reviewed 5 of the most popular NNS, including saccharin, aspartame, acesulfame potassium, sucralose, and stevia, and detail their drug-like qualities, regulatory status, pharmacokinetics, and primary research articles containing evidence of NNS interacting with drug absorption, distribution, metabolism, and excretion. Although studies varied widely in concentration ranges for NNS, model systems, and methods, all NNS included in this review were found to have known interactions with mediators of absorption, distribution, metabolism, and excretion from studies conducted after their Food and Drug Administration approval or generally recognized as safe designation. We have highlighted essential gaps in the literature and recommend the scientific community actively research NNS as food additives that may interact with drugs. SIGNIFICANCE STATEMENT: Food-drug interactions are a growing concern in Western societies where polypharmacy and ultraprocessed foods and beverages are increasingly common. High-intensity non-nutritive sweeteners bear structural similarities to pharmaceuticals, and evidence suggests they interact with mediators of drug pharmacokinetics. This minireview highlights the interactions uncovered thus far and serves as a call to action for the scientific community to establish rigorous, consistent testing that will enable updated safety guidelines for consumers.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":"107 5","pages":"100035"},"PeriodicalIF":3.2,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143972015","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
Corrigendum to "Novel Thiosemicarbazones Inhibit Lysine-Rich Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 (CEACAM1) Coisolated (LYRIC) and the LYRIC-Induced Epithelial- Mesenchymal Transition via Upregulation of N-Myc Downstream-Regulated Gene 1 (NDRG1)". “新型硫代氨基脲类抑制富含赖氨酸的癌胚抗原相关细胞粘附分子1 (CEACAM1)共分离物(LYRIC)和LYRIC通过上调N-Myc下游调节基因1 (NDRG1)诱导的上皮-间质转化”的更正。
IF 3.2 3区 医学
Molecular Pharmacology Pub Date : 2025-05-01 Epub Date: 2025-04-15 DOI: 10.1016/j.molpha.2025.100037
{"title":"Corrigendum to \"Novel Thiosemicarbazones Inhibit Lysine-Rich Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 (CEACAM1) Coisolated (LYRIC) and the LYRIC-Induced Epithelial- Mesenchymal Transition via Upregulation of N-Myc Downstream-Regulated Gene 1 (NDRG1)\".","authors":"","doi":"10.1016/j.molpha.2025.100037","DOIUrl":"10.1016/j.molpha.2025.100037","url":null,"abstract":"","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":"107 5","pages":"100037"},"PeriodicalIF":3.2,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144013405","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
Single-molecule localization microscopy as a tool to quantify di/oligomerization of receptor tyrosine kinases and G protein-coupled receptors. 单分子定位显微镜作为定量受体酪氨酸激酶和G蛋白偶联受体二/寡聚化的工具。
IF 3.2 3区 医学
Molecular Pharmacology Pub Date : 2025-05-01 Epub Date: 2025-03-26 DOI: 10.1016/j.molpha.2025.100033
Katie L Sharrocks, Aisha M Swaih, Aylin C Hanyaloglu
{"title":"Single-molecule localization microscopy as a tool to quantify di/oligomerization of receptor tyrosine kinases and G protein-coupled receptors.","authors":"Katie L Sharrocks, Aisha M Swaih, Aylin C Hanyaloglu","doi":"10.1016/j.molpha.2025.100033","DOIUrl":"10.1016/j.molpha.2025.100033","url":null,"abstract":"<p><p>Dimerization and oligomerization of membrane receptors, including G protein-coupled receptors and receptor tyrosine kinases, are fundamental for regulating cell signaling and diversifying downstream responses to mediate a range of physiological processes. Receptor di/oligomers play roles in diverse facets of receptor function. Changes in receptor di/oligomers have been implicated in a range of diseases; therefore, better understanding of the specific composition and interactions between receptors in complexes is essential, especially for the development of di/oligomer-specific drugs. Previously, different optical microscopy approaches and proximity-based biophysical assays have been used to demonstrate di/oligomerization of membrane receptors. However, in recent years, single-molecule super-resolution microscopy techniques have allowed researchers to quantify and uncover the precise dynamics and stoichiometry of specific receptor complexes. This allows the organization of membrane protein receptors to be mapped across the plasma membrane to explore the effects of factors such as ligands, effectors, membrane environment, and therapeutic agents. Quantification of receptor complexes is required to better understand the intricate balance of distinct receptor complexes in cells. In this brief review, we provide an overview of single-molecule approaches for the quantification of receptor di/oligomerization. We will discuss the techniques commonly employed to study membrane receptor di/oligomerization and their relative advantages and limitations. SIGNIFICANCE STATEMENT: Receptor di/oligomerization plays an important role in their function. For some receptors, di/oligomerization is essential for functional signaling, whereas for others, it acts as a mechanism to achieve signaling pleiotropy. Aberrant receptor di/oligomerization has been implicated in a wide range of diseases. Single-molecule super-resolution microscopy techniques provide convincing methods to precisely quantify receptor complexes at the plasma membrane. Understanding receptor complex organization in disease models can also influence the targeting of specific monomeric or oligomeric complexes in therapeutic strategies.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":"107 5","pages":"100033"},"PeriodicalIF":3.2,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12163491/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144028295","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
Doxorubicin-induced apoptosis is exacerbated by MG53 and associated with altered Akt signaling in H9c2 cells. MG53可加重阿霉素诱导的H9c2细胞凋亡,并与Akt信号通路改变有关。
IF 3.2 3区 医学
Molecular Pharmacology Pub Date : 2025-05-01 Epub Date: 2025-03-27 DOI: 10.1016/j.molpha.2025.100032
Bowen Xu, Hongye Li, Hongping Chen, Yanxin Ren, Jun Li, Lei Gong, Lin Zhong, Jun Yang
{"title":"Doxorubicin-induced apoptosis is exacerbated by MG53 and associated with altered Akt signaling in H9c2 cells.","authors":"Bowen Xu, Hongye Li, Hongping Chen, Yanxin Ren, Jun Li, Lei Gong, Lin Zhong, Jun Yang","doi":"10.1016/j.molpha.2025.100032","DOIUrl":"10.1016/j.molpha.2025.100032","url":null,"abstract":"<p><p>Chemotherapy drugs such as doxorubicin (DOX) are frequently used to treat cancer, but its negative impact on the heart reduces its effectiveness. Among the members of the TRIM protein family, mitsugumin (MG)53, also known as TRIM72, is unique. It is primarily present in the plasma membrane of cardiac and skeletal muscle cells and has been demonstrated to participate in mending cellular membrane damage while protecting against heart ischemia/reperfusion injury. This research investigated the role of MG53 in DOX-induced apoptosis using H9c2 cells, a cardiomyoblast cell line, as an experimental model. Our findings indicate that DOX treatment statistically significantly upregulates MG53 expression in H9c2 cells. Furthermore, MG53 overexpression exacerbated DOX-induced apoptosis, as confirmed by elevated levels of cleaved-caspase3 and BAX and reduced expression of Bcl-2. Flow cytometry analysis supported the elevated cell death rate in cells overexpressing MG53. Additionally, MG53 overexpression was associated with reduced phosphorylation levels of protein kinase B (AKT), as indicated by the decreased phosphorylation levels of AKT. Conversely, silencing MG53 through siRNA increased the phosphorylation levels of AKT. These results imply that MG53 exacerbates DOX-induced apoptosis, related to reduced AKT phosphorylation. Our investigation sheds light on the detrimental effects of MG53 in DOX-induced myocardial damage and underscores its potential as a therapeutic target for alleviating DOX treatment-related heart toxicity. SIGNIFICANCE STATEMENT: This study reveals that mitsugumin 53 exacerbates doxorubicin-induced apoptosis in H9c2 cells, associated with altered protein kinase B signaling. Targeting mitsugumin 53 may offer a novel therapeutic approach to mitigate doxorubicin-associated cardiotoxicity.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":"107 5","pages":"100032"},"PeriodicalIF":3.2,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144017951","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
Corrigendum to "Targeting the Metastasis Suppressor, NDRG1, Using Novel Iron Chelators: Regulation of Stress Fiber-Mediated Tumor Cell Migration via Modulation of the ROCK1/pMLC2 Signaling Pathway". “使用新型铁螯合剂靶向转移抑制因子NDRG1:通过调节ROCK1/pMLC2信号通路调节应激纤维介导的肿瘤细胞迁移”的更正。
IF 3.2 3区 医学
Molecular Pharmacology Pub Date : 2025-05-01 Epub Date: 2025-04-15 DOI: 10.1016/j.molpha.2025.100036
{"title":"Corrigendum to \"Targeting the Metastasis Suppressor, NDRG1, Using Novel Iron Chelators: Regulation of Stress Fiber-Mediated Tumor Cell Migration via Modulation of the ROCK1/pMLC2 Signaling Pathway\".","authors":"","doi":"10.1016/j.molpha.2025.100036","DOIUrl":"10.1016/j.molpha.2025.100036","url":null,"abstract":"","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":"107 5","pages":"100036"},"PeriodicalIF":3.2,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144031891","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
HDAC6 inhibition ameliorates sensory hypersensitivity and reduces immune cell signatures in the dorsal root ganglia in murine chronic pain models. 抑制HDAC6可改善小鼠慢性疼痛模型中感觉超敏反应并降低背根神经节中的免疫细胞特征。
IF 3.2 3区 医学
Molecular Pharmacology Pub Date : 2025-05-01 Epub Date: 2025-03-27 DOI: 10.1016/j.molpha.2025.100034
Ilinca M Giosan, Randal A Serafini, Aarthi Ramakrishnan, Madden J Tuffy, Jeffrey Zimering, Alexandru Babes, Li Shen, Venetia Zachariou
{"title":"HDAC6 inhibition ameliorates sensory hypersensitivity and reduces immune cell signatures in the dorsal root ganglia in murine chronic pain models.","authors":"Ilinca M Giosan, Randal A Serafini, Aarthi Ramakrishnan, Madden J Tuffy, Jeffrey Zimering, Alexandru Babes, Li Shen, Venetia Zachariou","doi":"10.1016/j.molpha.2025.100034","DOIUrl":"10.1016/j.molpha.2025.100034","url":null,"abstract":"<p><p>Histone deacetylase (HDAC)6 is a broadly expressed class IIb HDAC that regulates cytoskeletal dynamics and some nuclear processes. Previously research has shown that HDAC6 enzymatic inhibition has analgesic properties in models of chemotherapy-induced peripheral neuropathy. Here, we evaluated the effects of genetic and pharmacologic inhibition of HDAC6 on the development of sensory hypersensitivity in mouse models of peripheral nerve injury and peripheral inflammation. Daily administration of the peripherally restricted HDAC6 inhibitor, ACY1215 (Regenacy Pharmaceuticals, Inc), attenuated mechanical allodynia in the von Frey assay within 2 days of treatment initiation, with no signs of analgesic tolerance after 21 days of administration. We observed a similar antiallodynic effect across the implemented injury models after conditionally knocking down Hdac6 in the adult dorsal root ganglia (DRGs). Bioinformatic analysis of whole-transcriptome RNA-sequencing data predicted that ACY1215 treatment predominantly attenuated proinflammatory mechanisms, such as the suppression of immune cell infiltration into the DRG after injury. Accordingly, we demonstrated a reduction in the expression of various immune cell markers in the DRG after pharmacologic and genetic HDAC6 inhibition in both neuropathic and inflammatory pain models. We identified a direct relationship between Ccl5/Ccr5 and Hdac6 downregulation, as well as reduced hypersensitivity after hind paw CCL5 administration upon Hdac6 knockdown in the DRG. Our findings highlight that peripheral inhibition of HDAC6 ameliorates sensory hypersensitivity in models of postoperative inflammatory and neuropathic pain through mechanisms beyond reduction of tubulin deacetylation. SIGNIFICANCE STATEMENT: Recent studies highlight the role of histone deacetylase (HDAC)6 in chemotherapy-induced peripheral neuropathy, through mechanisms of action including tubulin acetylation and mitochondrial trafficking. In this study, various murine models of acute and chronic pain are applied to show that inhibition of HDAC6 activity in the periphery, using the clinically tested ACY1215 compound, and genetic inactivation of the Hdac6 gene in the dorsal root ganglia, alleviated mechanical hypersensitivity in male and in female mice through mechanisms that include targeting injury-induced inflammation.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":"107 5","pages":"100034"},"PeriodicalIF":3.2,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144003209","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信