Molecular Pharmacology最新文献

筛选
英文 中文
Therapeutic Potential for Metabotropic Glutamate Receptor 7 Modulators in Cognitive Disorders. 代谢型谷氨酸受体 7 (mGlu7) 调节剂在认知障碍中的治疗潜力。
IF 3.6 3区 医学
Molecular Pharmacology Pub Date : 2024-04-17 DOI: 10.1124/molpharm.124.000874
Harrison H Parent, Colleen M Niswender
{"title":"Therapeutic Potential for Metabotropic Glutamate Receptor 7 Modulators in Cognitive Disorders.","authors":"Harrison H Parent, Colleen M Niswender","doi":"10.1124/molpharm.124.000874","DOIUrl":"10.1124/molpharm.124.000874","url":null,"abstract":"<p><p>Metabotropic glutamate receptor 7 (mGlu<sub>7</sub>) is the most highly conserved and abundantly expressed mGlu receptor in the human brain. The presynaptic localization of mGlu<sub>7</sub>, coupled with its low affinity for its endogenous agonist, glutamate, are features that contribute to the receptor's role in modulating neuronal excitation and inhibition patterns, including long-term potentiation, in various brain regions. These characteristics suggest that mGlu<sub>7</sub> modulation may serve as a novel therapeutic strategy in disorders of cognitive dysfunction, including neurodevelopmental disorders that cause impairments in learning, memory, and attention. Primary mutations in the <i>GRM7</i> gene have recently been identified as novel causes of neurodevelopmental disorders, and these patients exhibit profound intellectual and cognitive disability. Pharmacological tools, such as agonists, antagonists, and allosteric modulators, have been the mainstay for targeting mGlu<sub>7</sub> in its endogenous homodimeric form to probe effects of its function and modulation in disease models. However, recent research has identified diversity in dimerization, as well as trans-synaptic interacting proteins, that also play a role in mGlu<sub>7</sub> signaling and pharmacological properties. These novel findings represent exciting opportunities in the field of mGlu receptor drug discovery and highlight the importance of further understanding the functions of mGlu<sub>7</sub> in complex neurologic conditions at both the molecular and physiologic levels. SIGNIFICANCE STATEMENT: Proper expression and function of mGlu<sub>7</sub> is essential for learning, attention, and memory formation at the molecular level within neural circuits. The pharmacological targeting of mGlu<sub>7</sub> is undergoing a paradigm shift by incorporating an understanding of receptor interaction with other <i>cis-</i> and <i>trans-</i> acting synaptic proteins, as well as various intracellular signaling pathways. Based upon these new findings, mGlu<sub>7</sub>'s potential as a drug target in the treatment of cognitive disorders and learning impairments is primed for exploration.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11026152/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139996761","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
Development of a Novel Assay for Direct Assessment of Selective Amylin Receptor Activation Reveals Novel Differences in Behavior of Selective and Nonselective Peptide Agonists. 为直接评估选择性淀粉样蛋白受体激活情况而开发的新型检测方法揭示了选择性和非选择性多肽激动剂在行为上的新差异。
IF 3.6 3区 医学
Molecular Pharmacology Pub Date : 2024-04-17 DOI: 10.1124/molpharm.123.000865
Peter Keov, George Christopoulos, Caroline A Hick, Tine Glendorf, Borja Ballarín-González, Denise Wootten, Patrick M Sexton
{"title":"Development of a Novel Assay for Direct Assessment of Selective Amylin Receptor Activation Reveals Novel Differences in Behavior of Selective and Nonselective Peptide Agonists.","authors":"Peter Keov, George Christopoulos, Caroline A Hick, Tine Glendorf, Borja Ballarín-González, Denise Wootten, Patrick M Sexton","doi":"10.1124/molpharm.123.000865","DOIUrl":"10.1124/molpharm.123.000865","url":null,"abstract":"<p><p>Dual amylin and calcitonin receptor agonists (DACRAs) show promise as efficacious therapeutics for treatment of metabolic disease, including obesity. However, differences in efficacy in vivo have been observed for individual DACRAs, indicating that detailed understanding of the pharmacology of these agents across target receptors is required for rational drug development. To date, such understanding has been hampered by lack of direct, subtype-selective, functional assays for the amylin receptors (AMYRs). Here, we describe the generation of receptor-specific assays for recruitment of Venus-tagged Gs protein through fusion of luciferase to either the human calcitonin receptor (CTR), human receptor activity-modifying protein (RAMP)-1, RAMP1 (AMY<sub>1</sub>R), human RAMP2 (AMY<sub>2</sub>R), or human RAMP3 (AMY<sub>3</sub>R). These assays revealed a complex pattern of receptor activation by calcitonin, amylin, or DACRA peptides that was distinct at each receptor subtype. Of particular note, although both of the CT-based DACRAs, sCT and AM1784, displayed relatively similar behaviors at CTR and AMY<sub>1</sub>R, they generated distinct responses at AMY<sub>2</sub>R and AMY<sub>3</sub>R. These data aid the rationalization of in vivo differences in response to DACRA peptides in rodent models of obesity. Direct assessment of the pharmacology of novel DACRAs at AMYR subtypes is likely to be important for development of optimized therapeutics for treatment of metabolic diseases. SIGNIFICANCE STATEMENT: Amylin receptors (AMYRs) are important obesity targets. Here we describe a novel assay that allows selective functional assessment of individual amylin receptor subtypes that provides unique insight into the pharmacology of potential therapeutic ligands. Direct assessment of the pharmacology of novel agonists at AMYR subtypes is likely to be important for development of optimized therapeutics for treatment of metabolic diseases.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140065544","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
Deorphanization of G Protein Coupled Receptors (GPCRs): a historical perspective. G 蛋白偶联受体(GPCR)的非形态化:历史视角。
IF 3.6 3区 医学
Molecular Pharmacology Pub Date : 2024-04-15 DOI: 10.1124/molpharm.124.000900
Luca Franchini, Cesare Orlandi
{"title":"Deorphanization of G Protein Coupled Receptors (GPCRs): a historical perspective.","authors":"Luca Franchini, Cesare Orlandi","doi":"10.1124/molpharm.124.000900","DOIUrl":"https://doi.org/10.1124/molpharm.124.000900","url":null,"abstract":"Counting over 800 members, G Protein Coupled Receptors (GPCRs) form the largest family of membrane receptors encoded in the human genome. Since the discovery of G proteins and GPCRs in the late 1970s and early 1980s, a significant portion of the GPCR research has been focused on identifying ligand/receptor pairs in parallel to studies related to their signaling properties. Despite significant advancements, about a fourth of the ~400 non-odorant GPCRs are still considered orphan because their natural or endogenous ligands have yet to be identified. We should consider that every GPCR was once an orphan and that endogenous ligands have often been associated with biological effects without a complete understanding of the molecular identity of their target receptors. Within this framework, this review offers a historical perspective on deorphanization processes for representative GPCRs, including Ghrelin receptor, GABAB receptor, Apelin receptor, Cannabinoid receptors, and GPR15. It explores three main scenarios encountered in deorphanization efforts and discusses key questions and methodologies employed in elucidating ligand-receptor interactions, providing insights for future research endeavors.","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-04-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140590013","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
Drugs Form Ternary Complexes with Human Liver Fatty Acid Binding Protein (FABP1) and FABP1 Binding Alters Drug Metabolism 药物与人类肝脏脂肪酸结合蛋白 (FABP1) 形成三元复合物,FABP1 结合改变药物代谢
IF 3.6 3区 医学
Molecular Pharmacology Pub Date : 2024-04-05 DOI: 10.1124/molpharm.124.000878
King Clyde B. Yabut, Alice Martynova, Abhinav Nath, Benjamin P Zercher, Matthew F. Bush, Nina Isoherranen
{"title":"Drugs Form Ternary Complexes with Human Liver Fatty Acid Binding Protein (FABP1) and FABP1 Binding Alters Drug Metabolism","authors":"King Clyde B. Yabut, Alice Martynova, Abhinav Nath, Benjamin P Zercher, Matthew F. Bush, Nina Isoherranen","doi":"10.1124/molpharm.124.000878","DOIUrl":"https://doi.org/10.1124/molpharm.124.000878","url":null,"abstract":"Liver fatty acid binding protein (FABP1) binds diverse endogenous lipids and is highly expressed in the human liver. Binding to FABP1 alters the metabolism and homeostasis of endogenous lipids in the liver. Drugs have also been shown to bind to rat FABP1, but limited data is available for human FABP1 (hFABP1). FABP1 has a large binding pocket and up to two fatty acids can bind to FABP1 simultaneously. We hypothesized that drug binding to hFABP1 results in formation of ternary complexes and that FABP1 binding alters drug metabolism. To test these hypotheses, native protein mass spectrometry (MS) and fluorescent 11-(dansylamino)undecanoic acid (DAUDA) displacement assays were used to characterize drug binding to hFABP1, and diclofenac oxidation by cytochrome P450 2C9 (CYP2C9) was studied in the presence and absence of hFABP1. DAUDA binding to hFABP1 involved high (K<sub>d,1</sub>=0.2 µM) and low affinity (K<sub>d,2 </sub>&gt;10 µM) binding sites. Nine drugs bound to hFABP1 with K<sub>d</sub> values ranging from 1 to 20 µM. None of the tested drugs completely displaced DAUDA from hFABP1 and fluorescence spectra showed evidence of ternary complex formation. Formation of DAUDA-hFABP1-diclofenac ternary complex was verified with native MS. Docking predicted diclofenac binding in the portal region of FABP1 with DAUDA in the binding cavity. The k<sub>cat</sub> of diclofenac hydroxylation by CYP2C9 was decreased by ~50% (p&lt;0.01) in the presence of FABP1. Together, these results suggest that drugs form ternary complexes with hFABP1 and that hFABP1 binding in the liver will alter drug metabolism and clearance.","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140590205","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
Pharmacological Characterization and Radiolabeling of VUF15485, a High-Affinity Small-Molecule Agonist for the Atypical Chemokine Receptor ACKR3. 非典型趋化因子受体 ACKR3 的高亲和力小分子激动剂 VUF15485 的药理特征和放射性标记。
IF 3.6 3区 医学
Molecular Pharmacology Pub Date : 2024-03-14 DOI: 10.1124/molpharm.123.000835
Aurelien M Zarca, Ilze Adlere, Cristina P Viciano, Marta Arimont-Segura, Max Meyrath, Icaro A Simon, Jan Paul Bebelman, Dennis Laan, Hans G J Custers, Elwin Janssen, Kobus L Versteegh, Maurice C M L Buzink, Desislava N Nesheva, Reggie Bosma, Iwan J P de Esch, Henry F Vischer, Maikel Wijtmans, Martyna Szpakowska, Andy Chevigné, Carsten Hoffmann, Chris de Graaf, Barbara A Zarzycka, Albert D Windhorst, Martine J Smit, Rob Leurs
{"title":"Pharmacological Characterization and Radiolabeling of VUF15485, a High-Affinity Small-Molecule Agonist for the Atypical Chemokine Receptor ACKR3.","authors":"Aurelien M Zarca, Ilze Adlere, Cristina P Viciano, Marta Arimont-Segura, Max Meyrath, Icaro A Simon, Jan Paul Bebelman, Dennis Laan, Hans G J Custers, Elwin Janssen, Kobus L Versteegh, Maurice C M L Buzink, Desislava N Nesheva, Reggie Bosma, Iwan J P de Esch, Henry F Vischer, Maikel Wijtmans, Martyna Szpakowska, Andy Chevigné, Carsten Hoffmann, Chris de Graaf, Barbara A Zarzycka, Albert D Windhorst, Martine J Smit, Rob Leurs","doi":"10.1124/molpharm.123.000835","DOIUrl":"10.1124/molpharm.123.000835","url":null,"abstract":"<p><p>Atypical chemokine receptor 3 (ACKR3), formerly referred to as CXCR7, is considered to be an interesting drug target. In this study, we report on the synthesis, pharmacological characterization and radiolabeling of VUF15485, a new ACKR3 small-molecule agonist, that will serve as an important new tool to study this <i>β</i>-arrestin-biased chemokine receptor. VUF15485 binds with nanomolar affinity (pIC<sub>50</sub> = 8.3) to human ACKR3, as measured in [<sup>125</sup>I]CXCL12 competition binding experiments. Moreover, in a bioluminescence resonance energy transfer-based <i>β</i>-arrestin2 recruitment assay VUF15485 acts as a potent ACKR3 agonist (pEC<sub>50</sub> = 7.6) and shows a similar extent of receptor activation compared with CXCL12 when using a newly developed, fluorescence resonance energy transfer-based ACKR3 conformational sensor. Moreover, the ACKR3 agonist VUF15485, tested against a (atypical) chemokine receptor panel (agonist and antagonist mode), proves to be selective for ACKR3. VUF15485 labeled with tritium at one of its methoxy groups ([<sup>3</sup>H]VUF15485), binds ACKR3 saturably and with high affinity (<i>K</i> <sub>d</sub> = 8.2 nM). Additionally, [<sup>3</sup>H]VUF15485 shows rapid binding kinetics and consequently a short residence time (<2 minutes) for binding to ACKR3. The selectivity of [<sup>3</sup>H]VUF15485 for ACKR3, was confirmed by binding studies, whereupon CXCR3, CXCR4, and ACKR3 small-molecule ligands were competed for binding against the radiolabeled agonist. Interestingly, the chemokine ligands CXCL11 and CXCL12 are not able to displace the binding of [<sup>3</sup>H]VUF15485 to ACKR3. The radiolabeled VUF15485 was subsequently used to evaluate its binding pocket. Site-directed mutagenesis and docking studies using a recently solved cryo-EM structure propose that VUF15485 binds in the major and the minor binding pocket of ACKR3. SIGNIFICANCE STATEMENT: The atypical chemokine receptor atypical chemokine receptor 3 (ACKR3) is considered an interesting drug target in relation to cancer and multiple sclerosis. The study reports on new chemical biology tools for ACKR3, i.e., a new agonist that can also be radiolabeled and a new ACKR3 conformational sensor, that both can be used to directly study the interaction of ACKR3 ligands with the G protein-coupled receptor.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139723403","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
RGS4 Actions in Mouse Prefrontal Cortex Modulate Behavioral and Transcriptomic Responses to Chronic Stress and Ketamine. 小鼠前额叶皮层中的 RGS4 作用可调节对慢性应激和氯胺酮的行为和转录组反应。
IF 3.2 3区 医学
Molecular Pharmacology Pub Date : 2024-03-14 DOI: 10.1124/molpharm.123.000753
Vasiliki Mitsi, Anne Ruiz, Claire Polizu, Zahra Farzinpour, Aarthi Ramakrishnan, Randal A Serafini, Eric M Parise, Madeline Floodstrand, Omar K Sial, Sevasti Gaspari, Cheuk Y Tang, Eric J Nestler, Eric F Schmidt, Li Shen, Venetia Zachariou
{"title":"RGS4 Actions in Mouse Prefrontal Cortex Modulate Behavioral and Transcriptomic Responses to Chronic Stress and Ketamine.","authors":"Vasiliki Mitsi, Anne Ruiz, Claire Polizu, Zahra Farzinpour, Aarthi Ramakrishnan, Randal A Serafini, Eric M Parise, Madeline Floodstrand, Omar K Sial, Sevasti Gaspari, Cheuk Y Tang, Eric J Nestler, Eric F Schmidt, Li Shen, Venetia Zachariou","doi":"10.1124/molpharm.123.000753","DOIUrl":"10.1124/molpharm.123.000753","url":null,"abstract":"<p><p>The signal transduction protein, regulator of G protein signaling 4 (RGS4), plays a prominent role in physiologic and pharmacological responses by controlling multiple intracellular pathways. Our earlier work identified the dynamic but distinct roles of RGS4 in the efficacy of monoamine-targeting versus fast-acting antidepressants. Using a modified chronic variable stress (CVS) paradigm in mice, we demonstrate that stress-induced behavioral abnormalities are associated with the downregulation of RGS4 in the medial prefrontal cortex (mPFC). Knockout of RGS4 (RGS4KO) increases susceptibility to CVS, as mutant mice develop behavioral abnormalities as early as 2 weeks after CVS resting-state functional magnetic resonance imaging I (rs-fMRI) experiments indicate that stress susceptibility in RGS4KO mice is associated with changes in connectivity between the mediodorsal thalamus (MD-THL) and the mPFC. Notably, RGS4KO also paradoxically enhances the antidepressant efficacy of ketamine in the CVS paradigm. RNA-sequencing analysis of naive and CVS samples obtained from mPFC reveals that RGS4KO triggers unique gene expression signatures and affects several intracellular pathways associated with human major depressive disorder. Our analysis suggests that ketamine treatment in the RGS4KO group triggers changes in pathways implicated in synaptic activity and responses to stress, including pathways associated with axonal guidance and myelination. Overall, we show that reducing RGS4 activity triggers unique gene expression adaptations that contribute to chronic stress disorders and that RGS4 is a negative modulator of ketamine actions. SIGNIFICANCE STATEMENT: Chronic stress promotes robust maladaptation in the brain, but the exact intracellular pathways contributing to stress vulnerability and mood disorders have not been thoroughly investigated. In this study, the authors used murine models of chronic stress and multiple methodologies to demonstrate the critical role of the signal transduction modulator regulator of G protein signaling 4 in the medial prefrontal cortex in vulnerability to chronic stress and the efficacy of the fast-acting antidepressant ketamine.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.2,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10949159/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139730032","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
Prodigiosin Inhibits Transforming Growth Factor β Signaling by Interfering Receptor Recycling and Subcellular Translocation in Epithelial Cells. 原薯蓣皂苷能通过干扰上皮细胞中的受体循环和亚细胞转运来抑制转化生长因子β的信号传导。
IF 3.6 3区 医学
Molecular Pharmacology Pub Date : 2024-03-14 DOI: 10.1124/molpharm.123.000776
Shun-Ban Tai, Chih-Yin Huang, Chih-Ling Chung, Ping-Jyun Sung, Zhi-Hong Wen, Chun-Lin Chen
{"title":"Prodigiosin Inhibits Transforming Growth Factor <i>β</i> Signaling by Interfering Receptor Recycling and Subcellular Translocation in Epithelial Cells.","authors":"Shun-Ban Tai, Chih-Yin Huang, Chih-Ling Chung, Ping-Jyun Sung, Zhi-Hong Wen, Chun-Lin Chen","doi":"10.1124/molpharm.123.000776","DOIUrl":"10.1124/molpharm.123.000776","url":null,"abstract":"<p><p>Prodigiosin (PG) is a naturally occurring polypyrrole red pigment produced by numerous microorganisms including some <i>Serratia</i> and <i>Streptomyces</i> strains. PG has exhibited promising anticancer activity; however, the molecular mechanisms of action of PG on malignant cells remain ambiguous. Transforming growth factor-<i>β</i> (TGF-<i>β</i>) is a multifunctional cytokine that governs a wide array of cellular processes in development and tissue homeostasis. Malfunctions of TGF-<i>β</i> signaling are associated with numerous human cancers. Emerging evidence underscores the significance of internalized TGF-<i>β</i> receptors and their intracellular trafficking in initiating signaling cascades. In this study, we identified PG as a potent inhibitor of the TGF-<i>β</i> pathway. PG blocked TGF-<i>β</i> signaling by targeting multiple sites of this pathway, including facilitating the sequestering of TGF-<i>β</i> receptors in the cytoplasm by impeding the recycling of type II TGF-<i>β</i> receptors to the cell surface. Additionally, PG prompts a reduction in the abundance of receptors on the cell surface through the disruption of the receptor glycosylation. In human Caucasian lung carcinoma cells and human hepatocellular cancer cell line cells, nanomolar concentrations of PG substantially diminish TGF-<i>β</i>-triggered phosphorylation of Smad2 protein. This attenuation is further reflected in the suppression of downstream target gene expression, including those encoding fibronectin, plasminogen activator inhibitor-1, and N-cadherin. SIGNIFICANCE STATEMENT: Prodigiosin (PG) emerges from this study as a potent TGF-β pathway inhibitor, disrupting receptor trafficking and glycosylation and reducing TGF-β signaling and downstream gene expression. These findings not only shed light on PG's potential therapeutic role but also present a captivating avenue towards future anti-TGF-β strategies.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139567280","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
Circadian Regulation of Endocrine Fibroblast Growth Factors on Systemic Energy Metabolism. 内分泌成纤维细胞生长因子对全身能量代谢的昼夜节律调控
IF 3.6 3区 医学
Molecular Pharmacology Pub Date : 2024-02-15 DOI: 10.1124/molpharm.123.000831
Zhenning Yang, Helmut Zarbl, Grace L Guo
{"title":"Circadian Regulation of Endocrine Fibroblast Growth Factors on Systemic Energy Metabolism.","authors":"Zhenning Yang, Helmut Zarbl, Grace L Guo","doi":"10.1124/molpharm.123.000831","DOIUrl":"10.1124/molpharm.123.000831","url":null,"abstract":"<p><p>The circadian clock is an endogenous biochemical timing system that coordinates the physiology and behavior of organisms to earth's ∼24-hour circadian day/night cycle. The central circadian clock synchronized by environmental cues hierarchically entrains peripheral clocks throughout the body. The circadian system modulates a wide variety of metabolic signaling pathways to maintain whole-body metabolic homeostasis in mammals under changing environmental conditions. Endocrine fibroblast growth factors (FGFs), namely FGF15/19, FGF21, and FGF23, play an important role in regulating systemic metabolism of bile acids, lipids, glucose, proteins, and minerals. Recent evidence indicates that endocrine FGFs function as nutrient sensors that mediate multifactorial interactions between peripheral clocks and energy homeostasis by regulating the expression of metabolic enzymes and hormones. Circadian disruption induced by environmental stressors or genetic ablation is associated with metabolic dysfunction and diurnal disturbances in FGF signaling pathways that contribute to the pathogenesis of metabolic diseases. Time-restricted feeding strengthens the circadian pattern of metabolic signals to improve metabolic health and prevent against metabolic diseases. Chronotherapy, the strategic timing of medication administration to maximize beneficial effects and minimize toxic effects, can provide novel insights into linking biologic rhythms to drug metabolism and toxicity within the therapeutical regimens of diseases. Here we review the circadian regulation of endocrine FGF signaling in whole-body metabolism and the potential effect of circadian dysfunction on the pathogenesis and development of metabolic diseases. We also discuss the potential of chrononutrition and chronotherapy for informing the development of timing interventions with endocrine FGFs to optimize whole-body metabolism in humans. SIGNIFICANCE STATEMENT: The circadian timing system governs physiological, metabolic, and behavioral functions in living organisms. The endocrine fibroblast growth factor (FGF) family (FGF15/19, FGF21, and FGF23) plays an important role in regulating energy and mineral metabolism. Endocrine FGFs function as nutrient sensors that mediate multifactorial interactions between circadian clocks and metabolic homeostasis. Chronic disruption of circadian rhythms increases the risk of metabolic diseases. Chronological interventions such as chrononutrition and chronotherapy provide insights into linking biological rhythms to disease prevention and treatment.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10877735/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139491573","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
Coexpressed δ-, μ-, and κ-Opioid Receptors Modulate Voltage-Gated Ca2+ Channels in Gastric-Projecting Vagal Afferent Neurons. 共表达的δ-、μ-和卡帕-阿片受体可调节胃射入迷走神经传入神经元的电压门控 Ca2+ 通道。
IF 3.6 3区 医学
Molecular Pharmacology Pub Date : 2024-02-15 DOI: 10.1124/molpharm.123.000774
Hannah J Goudsward, Victor Ruiz-Velasco, Salvatore L Stella, Lisa B Willing, Gregory M Holmes
{"title":"Coexpressed <i>δ</i>-, <i>μ</i>-, and <i>κ</i>-Opioid Receptors Modulate Voltage-Gated Ca<sup>2+</sup> Channels in Gastric-Projecting Vagal Afferent Neurons.","authors":"Hannah J Goudsward, Victor Ruiz-Velasco, Salvatore L Stella, Lisa B Willing, Gregory M Holmes","doi":"10.1124/molpharm.123.000774","DOIUrl":"10.1124/molpharm.123.000774","url":null,"abstract":"<p><p>Opioid analgesics are frequently associated with gastrointestinal side effects, including constipation, nausea, dysphagia, and reduced gastric motility. Though it has been shown that stimulation of opioid receptors expressed in enteric motor neurons contributes to opioid-induced constipation, it remains unclear whether activation of opioid receptors in gastric-projecting nodose ganglia neurons contributes to the reduction in gastric motility and emptying associated with opioid use. In the present study, whole-cell patch-clamp recordings were performed to determine the mechanism underlying opioid receptor-mediated modulation of Ca<sup>2+</sup> currents in acutely isolated gastric vagal afferent neurons. Our results demonstrate that Ca<sub>V</sub>2.2 channels provide the majority (71% ± 16%) of Ca<sup>2+</sup> currents in gastric vagal afferent neurons. Furthermore, we found that application of oxycodone, U-50488, or deltorphin II on gastric nodose ganglia neurons inhibited Ca<sup>2+</sup> currents through a voltage-dependent mechanism by coupling to the G<i>α</i> <sub>i/o</sub> family of heterotrimeric G-proteins. Because previous studies have demonstrated that the nodose ganglia expresses low levels of <i>δ</i>-opioid receptors, we also determined the deltorphin II concentration-response relationship and assessed deltorphin-mediated Ca<sup>2+</sup> current inhibition following exposure to the <i>δ</i>-opioid receptor antagonist ICI 174,864 (0.3 µM). The peak mean Ca<sup>2+</sup> current inhibition following deltorphin II application was 47% ± 24% (EC<sub>50</sub> = 302.6 nM), and exposure to ICI 174,864 blocked deltorphin II-mediated Ca<sup>2+</sup> current inhibition (4% ± 4% versus 37% ± 20%). Together, our results suggest that analgesics targeting any opioid receptor subtype can modulate gastric vagal circuits. SIGNIFICANCE STATEMENT: This study demonstrated that in gastric nodose ganglia neurons, agonists targeting all three classical opioid receptor subtypes (<i>μ</i>, <i>δ</i>, and <i>κ</i>) inhibit voltage-gated Ca<sup>2+</sup> channels in a voltage-dependent mechanism by coupling to Gα<sub>i/o</sub>. These findings suggest that analgesics targeting any opioid receptor subtype would modulate gastric vagal circuits responsible for regulating gastric reflexes.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10877734/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139106397","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
Development of an LC-MS/MS Method to Measure Sphingolipids in CSF from Patients with Multiple Sclerosis. 开发一种 LC-MS/MS 方法,用于测量多发性硬化症患者 CSF 中的鞘磷脂。
IF 3.6 3区 医学
Molecular Pharmacology Pub Date : 2024-02-15 DOI: 10.1124/molpharm.123.000779
Yadira X Perez-Paramo, Dawn Dufield, Rathna Veeramachaneni, Emily Parkhurst, Christopher Harp, Akshaya Ramesh, Ryan C Winger, Anne H Cross, Jeffrey M Gelfand, Amit Bar-Or, W Rodney Mathews, Veronica G Anania
{"title":"Development of an LC-MS/MS Method to Measure Sphingolipids in CSF from Patients with Multiple Sclerosis.","authors":"Yadira X Perez-Paramo, Dawn Dufield, Rathna Veeramachaneni, Emily Parkhurst, Christopher Harp, Akshaya Ramesh, Ryan C Winger, Anne H Cross, Jeffrey M Gelfand, Amit Bar-Or, W Rodney Mathews, Veronica G Anania","doi":"10.1124/molpharm.123.000779","DOIUrl":"10.1124/molpharm.123.000779","url":null,"abstract":"<p><p>Multiple sclerosis is an inflammatory and degenerative disease characterized by different clinical courses including relapsing multiple sclerosis (RMS) and primary progressive multiple sclerosis (PPMS). A hallmark of patients with multiple sclerosis (pwMS) includes a putative autoimmune response, which results in demyelination and neuroaxonal damage in the central nervous system. Sphingolipids in cerebrospinal fluid (CSF) have been proposed as potential biomarkers reflective of disease activity in pwMS. Hence, sensitive methods to accurately quantify sphingolipids in CSF are needed. In this study, we report the development of a sensitive high-throughput multiplexed liquid chromatography coupled to a tandem mass spectrometry method to perform quantitation on 14 species of sphingolipids in human CSF. We applied this method to measure CSF sphingolipids in healthy controls (n = 10), PPMS (n = 27), and RMS (n = 17) patients before and after ocrelizumab treatment. The median CSF levels of the 14 sphingolipids measured herein was higher in PPMS (17.2 ng/mL) and RMS (17.6 ng/mL) when compared with the healthy controls (13.8 ng/mL). Levels of sphingolipids were decreased by 8.6% at week 52 after treatment with ocrelizumab in RMS patients but not in PPMS patients. Specifically, C16 glucosylceramide (-26%; <i>P</i> = 0.004) and C18 ceramides (-13%; <i>P</i> = 0.042) decreased from baseline in RMS patients. Additionally, in PPMS patients C16 glucosylceramide levels correlated with CSF neurofilament heavy levels at baseline (<i>Rho =</i>0.532; <i>P</i> = 0.004) and after treatment (<i>Rho =</i>0.424; <i>P</i> = 0.028). Collectively, these results indicate that CSF sphingolipid levels are altered in pwMS and treatment with ocrelizumab results in significant shifts in the sphingolipid profile that may reflect a reduction in disease activity supporting further investigation into sphingolipids as tools to monitor disease state. SIGNIFICANCE STATEMENT: This study describes the development of a new method to measure 14 sphingolipid species in CSF. These results demonstrate that sphingolipids levels are elevated in CSF from pwMS compared to healthy controls. Distinct sphingolipid signatures were observed between patients with different clinical disease courses, and these lipid signatures changed after treatment with ocrelizumab, especially in RMS patients. This method enables further investigation into the role of sphingolipids as candidate biomarkers in pwMS and other central nervous system disorders.</p>","PeriodicalId":18767,"journal":{"name":"Molecular Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139106398","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:481959085
Book学术官方微信