Joshua P Dignam, Smriti Sharma, Gregor Aitchison, Ayman Gebril, Ioannis Stasinopoulos, Sofia Laforest, Chelbi Coyle, Ruth Andrew, Natalie Z M Homer, Sébastien Bonnet, Sandra Breuils-Bonnet, Martin Wabitsch, Margaret R MacLean
Background and purpose: The contribution of obesity to pulmonary arterial hypertension (PAH) pathophysiology remains poorly understood. Adipose tissue synthesises estrogens via cytochrome P450 (CYP) 19A1 (aromatase), whereas circulating estrogens are metabolised in the lung by CYP1A1. This study investigated whether obesity predisposes to PAH through enhanced estrogen synthesis and metabolism.
Experimental approach: A normoxic, two-hit, rat model of obesity-associated pulmonary hypertension (PH) was developed, combining Sugen 5416 (Sugen, Su) with a high-fat diet (HFD). Estrogen levels in SuHFD rat plasma and epicardial adipose tissue (EAT) from PAH patients were quantified using LC-MS/MS. CYP1A1 expression was assessed in lung and cardiac adipose tissue from SuHFD rats and PAH patients. The therapeutic potential of the CYP1A1 inhibitor hesperetin was evaluated in vivo. Complementary studies used pulmonary artery smooth muscle cells (PASMCs) from PAH patients and Simpson-Golabi-Behmel syndrome (SGBS) adipocytes.
Key results: HFD-fed rats of both sexes developed mild PH, which Sugen moderately exacerbated. EAT from PAH patients exhibited up-regulated aromatase and CYP1A1 expression, along with elevated estrogen levels. Circulating estrone was increased in male SuHFD rats. Pulmonary CYP1A1 expression was elevated in SuHFD rats and PAH patients. Hesperetin attenuated obesity-associated PH, reducing CYP1A1 expression in SuHFD rat lungs and PAH PASMCs. CYP1A1 induction in female SuHFD rat pericardial adipose tissue and Sugen-treated SGBS adipocytes was also tempered.
Conclusion and implications: These findings implicate augmented estrogen production by adipose tissue and elevated pulmonary CYP1A1 expression in the pathogenesis of obesity-associated PH. CYP1A1 may represent a novel therapeutic target in obese PAH patients.
{"title":"Cytochrome P450 1A1 influences obesity-induced pulmonary hypertension.","authors":"Joshua P Dignam, Smriti Sharma, Gregor Aitchison, Ayman Gebril, Ioannis Stasinopoulos, Sofia Laforest, Chelbi Coyle, Ruth Andrew, Natalie Z M Homer, Sébastien Bonnet, Sandra Breuils-Bonnet, Martin Wabitsch, Margaret R MacLean","doi":"10.1111/bph.70244","DOIUrl":"https://doi.org/10.1111/bph.70244","url":null,"abstract":"<p><strong>Background and purpose: </strong>The contribution of obesity to pulmonary arterial hypertension (PAH) pathophysiology remains poorly understood. Adipose tissue synthesises estrogens via cytochrome P450 (CYP) 19A1 (aromatase), whereas circulating estrogens are metabolised in the lung by CYP1A1. This study investigated whether obesity predisposes to PAH through enhanced estrogen synthesis and metabolism.</p><p><strong>Experimental approach: </strong>A normoxic, two-hit, rat model of obesity-associated pulmonary hypertension (PH) was developed, combining Sugen 5416 (Sugen, Su) with a high-fat diet (HFD). Estrogen levels in SuHFD rat plasma and epicardial adipose tissue (EAT) from PAH patients were quantified using LC-MS/MS. CYP1A1 expression was assessed in lung and cardiac adipose tissue from SuHFD rats and PAH patients. The therapeutic potential of the CYP1A1 inhibitor hesperetin was evaluated in vivo. Complementary studies used pulmonary artery smooth muscle cells (PASMCs) from PAH patients and Simpson-Golabi-Behmel syndrome (SGBS) adipocytes.</p><p><strong>Key results: </strong>HFD-fed rats of both sexes developed mild PH, which Sugen moderately exacerbated. EAT from PAH patients exhibited up-regulated aromatase and CYP1A1 expression, along with elevated estrogen levels. Circulating estrone was increased in male SuHFD rats. Pulmonary CYP1A1 expression was elevated in SuHFD rats and PAH patients. Hesperetin attenuated obesity-associated PH, reducing CYP1A1 expression in SuHFD rat lungs and PAH PASMCs. CYP1A1 induction in female SuHFD rat pericardial adipose tissue and Sugen-treated SGBS adipocytes was also tempered.</p><p><strong>Conclusion and implications: </strong>These findings implicate augmented estrogen production by adipose tissue and elevated pulmonary CYP1A1 expression in the pathogenesis of obesity-associated PH. CYP1A1 may represent a novel therapeutic target in obese PAH patients.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145647397","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background and purpose: Although the impact of nicotine on the dopaminergic system is well established, its effects on neural activity in the brain regions implicated in addiction remain unclear. The major objective of the study was to assess the impact of acute nicotine on neuronal and astrocytic metabolic activity in the prefrontal cortex, cerebral cortex and hippocampus of awake mice.
Experimental approach: Nicotine (0.0125-2.00 mg kg-1) was administered subcutaneously to 2- to 2.5-month-old C57BL/6NCrl male mice. The neuronal and astrocytic metabolic activity was measured by infusing [1,6-13C2]glucose and [2-13C]acetate, respectively, 15 min after injection, and monitoring amino acids labelling in the 1H-[13C]-NMR spectrum of brain tissue extracts.
Key results: Nicotine perturbed glucose metabolism in a dose- and brain- region-dependent manner. At lower doses, it enhanced the rate of glucose oxidation in glutamatergic neurons in the hippocampus (0.0125 mg kg-1) and prefrontal cortex (0.025 mg kg-1), with no change in the cerebral cortex. In contrast, a higher nicotine dose (1.0 mg kg-1) suppressed glutamatergic and GABAergic neurometabolic activity in all three brain regions. Nicotine did not affect the astrocytic metabolic activity at the lower dose (0.025 mg kg-1) but suppressed it at the high dose (2.0 mg kg-1).
Conclusions and implications: Nicotine has biphasic impacts on glutamatergic activity, enhancing excitatory activity at low doses but reducing both excitatory and inhibitory activity at higher doses. Most interestingly, acute nicotine increases neuronal excitability by shifting the excitation-to-inhibition balance in the prefrontal cortex, a critical component of the mesocortical circuitry.
{"title":"Biphasic effect of nicotine on glutamatergic activity in male mouse brain.","authors":"Prajakta Pramod Biyani, Ajay Sarawagi, Anant Bahadur Patel","doi":"10.1111/bph.70242","DOIUrl":"https://doi.org/10.1111/bph.70242","url":null,"abstract":"<p><strong>Background and purpose: </strong>Although the impact of nicotine on the dopaminergic system is well established, its effects on neural activity in the brain regions implicated in addiction remain unclear. The major objective of the study was to assess the impact of acute nicotine on neuronal and astrocytic metabolic activity in the prefrontal cortex, cerebral cortex and hippocampus of awake mice.</p><p><strong>Experimental approach: </strong>Nicotine (0.0125-2.00 mg kg<sup>-1</sup>) was administered subcutaneously to 2- to 2.5-month-old C57BL/6NCrl male mice. The neuronal and astrocytic metabolic activity was measured by infusing [1,6-<sup>13</sup>C<sub>2</sub>]glucose and [2-<sup>13</sup>C]acetate, respectively, 15 min after injection, and monitoring amino acids labelling in the <sup>1</sup>H-[<sup>13</sup>C]-NMR spectrum of brain tissue extracts.</p><p><strong>Key results: </strong>Nicotine perturbed glucose metabolism in a dose- and brain- region-dependent manner. At lower doses, it enhanced the rate of glucose oxidation in glutamatergic neurons in the hippocampus (0.0125 mg kg<sup>-1</sup>) and prefrontal cortex (0.025 mg kg<sup>-1</sup>), with no change in the cerebral cortex. In contrast, a higher nicotine dose (1.0 mg kg<sup>-1</sup>) suppressed glutamatergic and GABAergic neurometabolic activity in all three brain regions. Nicotine did not affect the astrocytic metabolic activity at the lower dose (0.025 mg kg<sup>-1</sup>) but suppressed it at the high dose (2.0 mg kg<sup>-1</sup>).</p><p><strong>Conclusions and implications: </strong>Nicotine has biphasic impacts on glutamatergic activity, enhancing excitatory activity at low doses but reducing both excitatory and inhibitory activity at higher doses. Most interestingly, acute nicotine increases neuronal excitability by shifting the excitation-to-inhibition balance in the prefrontal cortex, a critical component of the mesocortical circuitry.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145647357","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Helmut Bischof, Katarina Cisarova, Sandra Burgstaller, Mia Schwerer, Markus Absenger-Novak, Philipp J Jost, Roland Malli, Wolfgang F Graier, Robert Lukowski
Background and purpose: Hexokinase 2 (HK2) is a key enzyme linked to high tumour cell proliferation. Its inhibitors such as 3-bromopyruvic acid (3-BP) induce cancer cell death, highlighting HK2 modulation as potential anti-cancer treatment. However, standard chemotherapies often cause the emergence of senescent cancer cells, which goes along with cell metabolic reprogramming and treatment failure. This study explores whether targeting HK2 can induce cancer cell senescence and whether metabolic changes in senescent cancer cells are tied to the cellular HK2 status.
Experimental approach: The expression of hexokinase 1 (HK1) and HK2 was assessed using immunoblot and immunofluorescence analysis in cell lines and in primary murine breast cancer (BC) cells. The senescence-inducing potential of HK2 inhibition and the effect of chemotherapy-induced senescence on HK1 and HK2 expression were assessed. Cell-based approaches were complemented by analysing single-cell RNA sequencing data from BC patients.
Key results: BC cell sensitivity to HK2 inhibition did not correlate with HK2 expression levels. Consistently, senescence was linked to a decrease in HK2 and an increase in HK1 expression. Moreover, genetic knockdown of HK2 induced senescence, indicating that a change in the HK2/HK1 ratio drives, rather than results, from cellular senescence. This shift in HK2/HK1 ratio was confirmed in single-cell RNA sequencing data of BC biopsies.
Conclusions and implications: Expressional shifts in the HK2/HK1 ratio may serve as a novel marker for BC cell senescence. Whereas targeting HK2 shows promise in untreated cancers, senescence-inducing anti-cancer therapies may limit the effectiveness of HK2-targeted treatments in pre-treated cancer patients.
{"title":"Targeting hexokinase 2 to induce breast cancer cell senescence.","authors":"Helmut Bischof, Katarina Cisarova, Sandra Burgstaller, Mia Schwerer, Markus Absenger-Novak, Philipp J Jost, Roland Malli, Wolfgang F Graier, Robert Lukowski","doi":"10.1111/bph.70282","DOIUrl":"https://doi.org/10.1111/bph.70282","url":null,"abstract":"<p><strong>Background and purpose: </strong>Hexokinase 2 (HK2) is a key enzyme linked to high tumour cell proliferation. Its inhibitors such as 3-bromopyruvic acid (3-BP) induce cancer cell death, highlighting HK2 modulation as potential anti-cancer treatment. However, standard chemotherapies often cause the emergence of senescent cancer cells, which goes along with cell metabolic reprogramming and treatment failure. This study explores whether targeting HK2 can induce cancer cell senescence and whether metabolic changes in senescent cancer cells are tied to the cellular HK2 status.</p><p><strong>Experimental approach: </strong>The expression of hexokinase 1 (HK1) and HK2 was assessed using immunoblot and immunofluorescence analysis in cell lines and in primary murine breast cancer (BC) cells. The senescence-inducing potential of HK2 inhibition and the effect of chemotherapy-induced senescence on HK1 and HK2 expression were assessed. Cell-based approaches were complemented by analysing single-cell RNA sequencing data from BC patients.</p><p><strong>Key results: </strong>BC cell sensitivity to HK2 inhibition did not correlate with HK2 expression levels. Consistently, senescence was linked to a decrease in HK2 and an increase in HK1 expression. Moreover, genetic knockdown of HK2 induced senescence, indicating that a change in the HK2/HK1 ratio drives, rather than results, from cellular senescence. This shift in HK2/HK1 ratio was confirmed in single-cell RNA sequencing data of BC biopsies.</p><p><strong>Conclusions and implications: </strong>Expressional shifts in the HK2/HK1 ratio may serve as a novel marker for BC cell senescence. Whereas targeting HK2 shows promise in untreated cancers, senescence-inducing anti-cancer therapies may limit the effectiveness of HK2-targeted treatments in pre-treated cancer patients.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145647410","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sujin Lim, Junhyuk Woo, Sungmin Pak, Joonho Paik, Gyu-Sang Hong, Ji Hun Wi, Sanghee Lee, Cristina Fenollar-Ferrer, Kyungreem Han, Young Kee Shin, Uhtaek Oh
Background and purpose: Tentonin 3 (TTN3/TMEM150C) is a mechanosensitive ion channel that plays critical roles in mechanotransduction processes. TTN3 forms a tetramer with a predicted rectangular shape and a central pore. A conotoxin ρ-TIA and its synthetic analog, noxious mechanosensation blocker 1 (NMB-1), were initially developed to inhibit slowly adapting (SA)-type mechanically activated (MA) currents in dorsal root ganglion (DRG) neurons. Since TTN3 confers slowly-adapting MA currents in DRG neurons, both NMB-1 and ρ-TIA were hypothesized to inhibit TTN3.
Experimental approach: To record MA currents, a few micrometre step indentations were applied to HEK cells expressing TTN3. NMB-1 and ρ-TIA were applied to these cells to test their specific antagonism. Mutations of the conopeptides and TTN3 were made to identify underlying mechanisms of inhibition.
Key results: NMB-1 strongly inhibited TTN3, whereas ρ-TIA had only a weak effect, and neither peptide affected Piezo channels. Alanine-scanning mutagenesis coupled with electrophysiological assays pinpointed that a positively charged residue in NMB-1 and ρ-TIA is essential for their inhibitory action. Additionally, a glutamate residue (Glu126) near the pore entrance of TTN3 was identified as critical for the NMB-1 inhibitory action, suggesting a key electrostatic interaction between NMB-1 and TTN3. Molecular dynamics simulations further supported this electrostatic interaction between the peptide ligand and the channel protein.
Conclusions and implications: NMB-1 specifically blocks a mechanosensitive channel, TTN3, via electrostatic interaction. These findings offer mechanistic insights into the selective inhibition of TTN3 by NMB-1 and provide a foundation for developing therapeutic agents targeting TTN3-related channelopathies.
{"title":"Molecular basis for a pore block of Tentonin 3 expressed in HEK293 cells by a conopeptide, NMB-1.","authors":"Sujin Lim, Junhyuk Woo, Sungmin Pak, Joonho Paik, Gyu-Sang Hong, Ji Hun Wi, Sanghee Lee, Cristina Fenollar-Ferrer, Kyungreem Han, Young Kee Shin, Uhtaek Oh","doi":"10.1111/bph.70278","DOIUrl":"https://doi.org/10.1111/bph.70278","url":null,"abstract":"<p><strong>Background and purpose: </strong>Tentonin 3 (TTN3/TMEM150C) is a mechanosensitive ion channel that plays critical roles in mechanotransduction processes. TTN3 forms a tetramer with a predicted rectangular shape and a central pore. A conotoxin ρ-TIA and its synthetic analog, noxious mechanosensation blocker 1 (NMB-1), were initially developed to inhibit slowly adapting (SA)-type mechanically activated (MA) currents in dorsal root ganglion (DRG) neurons. Since TTN3 confers slowly-adapting MA currents in DRG neurons, both NMB-1 and ρ-TIA were hypothesized to inhibit TTN3.</p><p><strong>Experimental approach: </strong>To record MA currents, a few micrometre step indentations were applied to HEK cells expressing TTN3. NMB-1 and ρ-TIA were applied to these cells to test their specific antagonism. Mutations of the conopeptides and TTN3 were made to identify underlying mechanisms of inhibition.</p><p><strong>Key results: </strong>NMB-1 strongly inhibited TTN3, whereas ρ-TIA had only a weak effect, and neither peptide affected Piezo channels. Alanine-scanning mutagenesis coupled with electrophysiological assays pinpointed that a positively charged residue in NMB-1 and ρ-TIA is essential for their inhibitory action. Additionally, a glutamate residue (Glu126) near the pore entrance of TTN3 was identified as critical for the NMB-1 inhibitory action, suggesting a key electrostatic interaction between NMB-1 and TTN3. Molecular dynamics simulations further supported this electrostatic interaction between the peptide ligand and the channel protein.</p><p><strong>Conclusions and implications: </strong>NMB-1 specifically blocks a mechanosensitive channel, TTN3, via electrostatic interaction. These findings offer mechanistic insights into the selective inhibition of TTN3 by NMB-1 and provide a foundation for developing therapeutic agents targeting TTN3-related channelopathies.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145630327","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background and purpose: d-cycloserine, N-methyl-d-aspartate/glutamate (NMDA) receptor co-agonist (GluN2A/GluN2B partial and GluN2C super-agonist), improves negative symptom of schizophrenia with narrow therapeutic window, but the mechanisms remains unclear.
Experimental approach: Effects of chronic d-cycloserine administration (2-5 mg·kg-1) on sucrose preference of adult male rats were determined. Dose/concentration-dependent effects of acute/chronic administrations of d-cycloserine (25-300 μM and 2-25 mg·kg-1) on expression of GluN2 subunits and associated transmission of l-glutamate/d-serine/GABA were determined using microdialysis in adult male rats, primary cultured astrocytes (male/female neonatal rats) and capillary immunoblotting.
Key results: d-cycloserine dose-dependently increased release of astroglial l-glutamate/d-serine and neuronal GABA in the thalamus and medial prefrontal cortex (mPFC) (d-serine > GABA > l-glutamate), according to intrinsic activities (GluN2C > GluN2A [but unaffected GluN2B]). Chronic d-cycloserine dose-dependently down-regulated GluN2C > GluN2B > GluN2A (according to affinity) and attenuated d-cycloserine-induced astroglial release of l-glutamate/d-serine, dose dependently (GluN2C > GluN2A). Chronic exposure (25 μM) d-cycloserine down-regulated GluN2C but increased GluN2C-related astroglial l-glutamate/d-serine release. Chronic exposure to >60μM d-cycloserine diminished GluN2C-related astroglial release but activated GluN2A-related release. Chronic administration of 2 but not 5-mg·kg-1 d-cycloserine restored MK-801-induced decrease sucrose preference.
Conclusion and implications: Chronic d-cycloserine (25 μM and 2 mg·kg-1) down-regulated GluN2C without affecting GluN2A/GluN2B but increased GluN2-related astroglial l-glutamate/d-serine release. Higher d-cycloserine dose (>60 μM; >5 mg·kg-1) inactivated GluN2C, but increased GluN2A related astroglial release. These results indicate that dose-dependent activation and inactivation of GluN2C by d-cycloserine is possibly involved in its efficacy on negative symptom of schizophrenia, but with a narrow therapeutic window.
{"title":"Exploring effects of chronic d-cycloserine administration on expression of GluN2 subunits and tripartite synaptic transmission in thalamocortical pathway.","authors":"Motohiro Okada, Ruri Okubo, Nobutomo Yamamoto, Eishi Motomura","doi":"10.1111/bph.70262","DOIUrl":"https://doi.org/10.1111/bph.70262","url":null,"abstract":"<p><strong>Background and purpose: </strong>d-cycloserine, N-methyl-d-aspartate/glutamate (NMDA) receptor co-agonist (GluN2A/GluN2B partial and GluN2C super-agonist), improves negative symptom of schizophrenia with narrow therapeutic window, but the mechanisms remains unclear.</p><p><strong>Experimental approach: </strong>Effects of chronic d-cycloserine administration (2-5 mg·kg<sup>-1</sup>) on sucrose preference of adult male rats were determined. Dose/concentration-dependent effects of acute/chronic administrations of d-cycloserine (25-300 μM and 2-25 mg·kg<sup>-1</sup>) on expression of GluN2 subunits and associated transmission of l-glutamate/d-serine/GABA were determined using microdialysis in adult male rats, primary cultured astrocytes (male/female neonatal rats) and capillary immunoblotting.</p><p><strong>Key results: </strong>d-cycloserine dose-dependently increased release of astroglial l-glutamate/d-serine and neuronal GABA in the thalamus and medial prefrontal cortex (mPFC) (d-serine > GABA > l-glutamate), according to intrinsic activities (GluN2C > GluN2A [but unaffected GluN2B]). Chronic d-cycloserine dose-dependently down-regulated GluN2C > GluN2B > GluN2A (according to affinity) and attenuated d-cycloserine-induced astroglial release of l-glutamate/d-serine, dose dependently (GluN2C > GluN2A). Chronic exposure (25 μM) d-cycloserine down-regulated GluN2C but increased GluN2C-related astroglial l-glutamate/d-serine release. Chronic exposure to >60μM d-cycloserine diminished GluN2C-related astroglial release but activated GluN2A-related release. Chronic administration of 2 but not 5-mg·kg<sup>-1</sup> d-cycloserine restored MK-801-induced decrease sucrose preference.</p><p><strong>Conclusion and implications: </strong>Chronic d-cycloserine (25 μM and 2 mg·kg<sup>-1</sup>) down-regulated GluN2C without affecting GluN2A/GluN2B but increased GluN2-related astroglial l-glutamate/d-serine release. Higher d-cycloserine dose (>60 μM; >5 mg·kg<sup>-1</sup>) inactivated GluN2C, but increased GluN2A related astroglial release. These results indicate that dose-dependent activation and inactivation of GluN2C by d-cycloserine is possibly involved in its efficacy on negative symptom of schizophrenia, but with a narrow therapeutic window.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145630330","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mohamed Zamzamy, Sophie Post, Wing Hang Ip, Elin Hahlin, Sebastian Kühn, Andrea Pirosu, Christian Conze, Martin Baumdick, Madeleine J Bunders, Britta F Zecher, Angelique Hoelzemer, Stephan Linder, Niklas Arnberg, Thomas Dobner, Marcus Altfeld, Sebastian Schloer
Background and purpose: Human adenovirus (HAdV) causes respiratory or gastrointestinal tract infections depending on the virus subtype. While HAdV infections are generally self-limiting in immunocompetent people, they can result in significant morbidity and mortality in immunocompromised adults and children. Due to the limited availability of effective therapeutic options, there is an urgent need for novel therapeutics to combat HAdV infection and mitigate its severity.
Experimental approach: Here, we have repurposed the clinically well-used antifungal, itraconazole, to control HAdV infection. We tested the antiviral potential of the itraconazole and the mTOR inhibitor Ku-63794 on the production of infectious HAdV in A549 and Caco-2 cells as well as human intestinal organoids (HIOs). Additionally, we evaluated the benefit of a combination of these host-directed drugs with the direct-acting antiviral brincidofovir.
Key results: Pharmacological treatment with itraconazole significantly reduced virus titres in different in vitro models, including HIOs. Treatment with itraconazole impairs HAdV entry by entrapping incoming virus particles in endolysosomes and by promoting autophagy in HAdV-infected cells. Moreover, combining itraconazole with brincidofovir, a cidofovir derivative currently under clinical evaluation for anti-HAdV applications, demonstrated a synergistic effect in reducing HAdV titres.
Conclusion and implications: Given the gastrointestinal toxicity associated with brincidofovir, its combination with the host-directed drug itraconazole allowed lower brincidofovir doses to be used to decrease HAdV titres, thereby minimizing adverse drug effects while maintaining antiviral efficacy.
{"title":"Synergistic antiviral activity against human adenovirus through combination of itraconazole and brincidofovir.","authors":"Mohamed Zamzamy, Sophie Post, Wing Hang Ip, Elin Hahlin, Sebastian Kühn, Andrea Pirosu, Christian Conze, Martin Baumdick, Madeleine J Bunders, Britta F Zecher, Angelique Hoelzemer, Stephan Linder, Niklas Arnberg, Thomas Dobner, Marcus Altfeld, Sebastian Schloer","doi":"10.1111/bph.70258","DOIUrl":"https://doi.org/10.1111/bph.70258","url":null,"abstract":"<p><strong>Background and purpose: </strong>Human adenovirus (HAdV) causes respiratory or gastrointestinal tract infections depending on the virus subtype. While HAdV infections are generally self-limiting in immunocompetent people, they can result in significant morbidity and mortality in immunocompromised adults and children. Due to the limited availability of effective therapeutic options, there is an urgent need for novel therapeutics to combat HAdV infection and mitigate its severity.</p><p><strong>Experimental approach: </strong>Here, we have repurposed the clinically well-used antifungal, itraconazole, to control HAdV infection. We tested the antiviral potential of the itraconazole and the mTOR inhibitor Ku-63794 on the production of infectious HAdV in A549 and Caco-2 cells as well as human intestinal organoids (HIOs). Additionally, we evaluated the benefit of a combination of these host-directed drugs with the direct-acting antiviral brincidofovir.</p><p><strong>Key results: </strong>Pharmacological treatment with itraconazole significantly reduced virus titres in different in vitro models, including HIOs. Treatment with itraconazole impairs HAdV entry by entrapping incoming virus particles in endolysosomes and by promoting autophagy in HAdV-infected cells. Moreover, combining itraconazole with brincidofovir, a cidofovir derivative currently under clinical evaluation for anti-HAdV applications, demonstrated a synergistic effect in reducing HAdV titres.</p><p><strong>Conclusion and implications: </strong>Given the gastrointestinal toxicity associated with brincidofovir, its combination with the host-directed drug itraconazole allowed lower brincidofovir doses to be used to decrease HAdV titres, thereby minimizing adverse drug effects while maintaining antiviral efficacy.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145630289","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p>Among the molecular targets for treating disorders of neuronal excitability, such as epilepsy, chronic pain and conditions involving excitotoxicity (e.g. stroke or traumatic brain injury), the K<sub>v</sub>7 (KCNQ) family of voltage-gated potassium channels stands out as particularly promising. Several biophysical properties make K<sub>v</sub>7 channels well-suited for this role. They have activation voltage threshold near the resting membrane potential of many neurons, they do not inactivate and have slow activation and deactivation kinetics (Jones et al., <span>2021</span>). Owing to these characteristics, a fraction of K<sub>v</sub>7 channels remains conductive in a neuron at rest, controlling firing threshold and rheobase. Gradual increase of K<sub>v</sub>7 channel activity during sustained depolarization or continuous firing introduces self-tuning or ‘accommodation’ in firing patterns.</p><p>There are five K<sub>v</sub>7 subunits in mammals, K<sub>v</sub>7.1–K<sub>v</sub>7.5, and these are encoded by <i>KCNQ1</i>-<i>KCNQ5</i> genes. K<sub>v</sub>7.1 is mainly expressed in the cardiovascular system and epithelia, while K<sub>v</sub>7.2–K<sub>v</sub>7.5 are mostly neuronal, responsible for so-called M-type K<sup>+</sup> current in these cells (Jones et al., <span>2021</span>). Generic mutations causing loss of K<sub>v</sub>7 function often result in pathological hyperexcitability (epilepsies, cardiac arrhythmias and pain) (Jones et al., <span>2021</span>). Conversely, pharmacological activation or enhancement of K<sub>v</sub>7 activity is widely recognised therapeutic strategy for management of hyperexcitability symptoms. Two K<sub>v</sub>7 activators have been clinically used as a painkiller (flupirtine) and anticonvulsant (retigabine), and a number of other clinically used drugs may have K<sub>v</sub>7 activation among their mechanisms of action. These include painkillers, celecoxib (Du et al., <span>2011</span>) and paracetamol (Ray et al., <span>2019</span>) and a vasodilator, fasudil (Zhang et al., <span>2016</span>).</p><p>Although flupirtine and retigabine initially demonstrated clinical promise, both compounds were ultimately withdrawn from the market, mostly due to adverse effects. Retigabine was associated with blue skin discolouration, retinal pigmentation and urinary retention, while flupirtine carried a risk of hepatotoxicity. Current efforts in drug development are focused on identifying next-generation K<sub>v</sub>7 channel activators with superior pharmacological selectivity and safety. Yet, development of a new drug from scratch is a costly and time-consuming undertaking. In this context, screening libraries of already approved (or close to approval) drugs for potential repurposing represents a promising alternative strategy, offering the advantages of accelerated market entry and reduced development costs.</p><p>A new study by Lidia Carotenuto and co-authors, published in the British Journal of Pharmacology (Carotenuto et
在治疗神经元兴奋性紊乱的分子靶点中,如癫痫、慢性疼痛和涉及兴奋性毒性的疾病(如中风或创伤性脑损伤),Kv7 (KCNQ)电压门控钾通道家族尤其有前景。一些生物物理性质使得Kv7通道非常适合这个角色。它们在许多神经元的静息膜电位附近具有激活电压阈值,它们不会失活,并且具有缓慢的激活和失活动力学(Jones et al., 2021)。由于这些特性,一小部分Kv7通道在静止的神经元中保持导电,控制放电阈值和流变酶。在持续去极化或连续放电过程中,Kv7通道活动的逐渐增加引入了放电模式的自调谐或“调节”。哺乳动物中有5个Kv7亚基,Kv7.1-Kv7.5,这些亚基由KCNQ1-KCNQ5基因编码。Kv7.1主要表达于心血管系统和上皮细胞,而Kv7.2-Kv7.5主要是神经元细胞,负责这些细胞中所谓的m型K+电流(Jones et al., 2021)。导致Kv7功能丧失的基因突变通常会导致病理性高兴奋性(癫痫、心律失常和疼痛)(Jones et al., 2021)。相反,药物激活或增强Kv7活性被广泛认为是治疗高兴奋性症状的治疗策略。两种Kv7激活剂已在临床上用作止痛药(氟吡汀)和抗惊厥药(瑞加滨),许多其他临床使用的药物可能在其作用机制中具有Kv7激活作用。这些药物包括止痛药塞来昔布(Du等人,2011年)、扑热息痛(Ray等人,2019年)和血管扩张剂法舒地尔(Zhang等人,2016年)。虽然氟吡汀和瑞加滨最初表现出临床前景,但这两种化合物最终都退出了市场,主要是由于副作用。雷沙滨与蓝色皮肤变色、视网膜色素沉着和尿潴留有关,而氟吡汀则有肝毒性风险。目前药物开发的重点是鉴定具有优越药理选择性和安全性的下一代Kv7通道激活剂。然而,从头开始开发一种新药是一项昂贵且耗时的工作。在这种情况下,筛选已经批准(或接近批准)的药物库以进行潜在的重新利用是一种有希望的替代策略,具有加速市场进入和降低开发成本的优势。Lidia Carotenuto及其合著者发表在《英国药理学杂志》上的一项新研究(Carotenuto等人,2025)报告了通过再利用管道识别新的Kv7激活剂的努力。作者对来自Fraunhofer repurpose Library和EU-Openscreen Pilot Bioactive Library的约8000种化合物进行了高通量筛选,希望能找到安全有效的治疗癫痫的Kv7激活剂。筛选结果显示,JNJ-37822681(图1)是一种快速解离的D2受体拮抗剂,最初由强生公司开发用于治疗精神分裂症(Langlois et al., 2012)。该分子具有良好的生物利用度,良好的一般毒理学和安全性,目前正处于临床开发的后期阶段。作者首先使用荧光铊法筛选了针对中国仓鼠卵巢(CHO)细胞的化合物文库,这些细胞稳定过表达增强宏观电流突变的Kv7.3通道异构体(Kv7.3 A315T)。利加滨作为阳性对照。筛选确定了大约12种候选药物,其中包括瑞加滨和另一种已知的Kv7活化剂ML213。大多数新的候选药物在进一步的膜片钳实验中因其低效力和/或功效而被取消资格。然而,JNJ-37822681通过了验证,对Kv7.2、Kv7.4、Kv7.5同型异构体和Kv7.2/Kv7.3异构体的效价和疗效与瑞gabine相当,对Kv7.3的作用略弱。例如,瑞gabine和JNJ-37822681诱导Kv7.2/Kv7.3半电压的超极化位移分别为- 40和-37 mV, EC50分别为2.5和1.2 μM。JNJ-37822681与瑞加滨相似,对Kv7.1通道影响较小。利用Kv7.2与瑞gabine (PDB: 7CR2)配合物的低温电镜结构和分子对接模拟,作者能够将JNJ-37822681放入Kv7.2的瑞gabine结合口袋中。此外,对雷沙滨结合至关重要的Kv7.2 (W236L)中236位色氨酸的突变消除了JNJ-37822681的Kv7激活作用,证实了模型预测。然后,作者利用诱导多能干细胞(iPSC)技术测试了JNJ-37822681对人类iPSC衍生的皮质谷氨酸能神经元兴奋性的影响。 他们使用的皮质样谷氨酸能神经元分化自两名因KCNQ2功能缺失突变而患有遗传性癫痫的患者(KCNQ2发育性和癫痫性脑病;KCNQ2- dee),以及CRISPR/ cas9校正的等基因细胞系作为对照。在这些实验中,JNJ-37822681和瑞gabine在降低对照和DEE神经元的兴奋性和放电频率(使用膜片钳记录和多电极阵列系统的细胞外记录测量)方面同样有效。这两种化合物也可显著增强等基因对照神经元的M电流振幅。最后,作者使用了两种小鼠癫痫发作模型:戊四唑(PTZ)模型的全身性强直-阵挛性癫痫发作和遗传性癫痫易感DBA/2小鼠的听原反射性癫痫发作。在两种模型中,雷吉滨和JNJ-37822681显著减少了阵挛性和强直性癫痫发作,JNJ-37822681的效价与雷吉滨相似(PTZ)或略高(DBA/2)。综上所述,该研究确定并表征了具有良好安全性的新型Kv7激活剂,目前正作为抗精神病药物推向临床应用。该分子在Kv7通道中与瑞gabine结合袋结合,并具有相当的选择性。缺乏Kv7.1的活性是有利的,因为该亚基主要在心脏和血管组织中表达。因此,表现出显著Kv7.1相互作用的调节剂可能存在心血管风险。JNJ-37822681降低体外神经元兴奋性和体内癫痫发作严重程度,其效力和疗效与雷加滨相当。据推测,JNJ-37822681不会有皮肤变色的蓝色问题,这是雷沙滨所特有的,因为形成了色素二聚体。然而,JNJ-37822681可能仍有雷沙滨的其他非靶向副作用,如头晕、精神错乱、嗜睡和尿潴留。此外,由于JNJ-37822681是一种D2受体拮抗剂,它可能引发与这类药物相关的副作用,包括锥体外系症状和代谢问题。同样重要的是要注意,根据英国。葛兰素史克(GlaxoSmithKline)将有限的临床使用列为2017年停止使用瑞加滨(Trobalt/Potiga)的主要原因,而不是与疗效或安全性有关的担忧。因此,类似的化合物是否能获得更大的市场生存能力还有待确定。尽管如此,胡萝卜素及其合作者的研究证明了一种彻底而全面的策略,用于识别和描述治疗兴奋性障碍的新的临床相关Kv7通道打开剂。写了手稿。作者声明无利益冲突。
{"title":"Improving Kv7 targeting anticonvulsants - will repurposing save the day?","authors":"Nikita Gamper","doi":"10.1111/bph.70290","DOIUrl":"10.1111/bph.70290","url":null,"abstract":"<p>Among the molecular targets for treating disorders of neuronal excitability, such as epilepsy, chronic pain and conditions involving excitotoxicity (e.g. stroke or traumatic brain injury), the K<sub>v</sub>7 (KCNQ) family of voltage-gated potassium channels stands out as particularly promising. Several biophysical properties make K<sub>v</sub>7 channels well-suited for this role. They have activation voltage threshold near the resting membrane potential of many neurons, they do not inactivate and have slow activation and deactivation kinetics (Jones et al., <span>2021</span>). Owing to these characteristics, a fraction of K<sub>v</sub>7 channels remains conductive in a neuron at rest, controlling firing threshold and rheobase. Gradual increase of K<sub>v</sub>7 channel activity during sustained depolarization or continuous firing introduces self-tuning or ‘accommodation’ in firing patterns.</p><p>There are five K<sub>v</sub>7 subunits in mammals, K<sub>v</sub>7.1–K<sub>v</sub>7.5, and these are encoded by <i>KCNQ1</i>-<i>KCNQ5</i> genes. K<sub>v</sub>7.1 is mainly expressed in the cardiovascular system and epithelia, while K<sub>v</sub>7.2–K<sub>v</sub>7.5 are mostly neuronal, responsible for so-called M-type K<sup>+</sup> current in these cells (Jones et al., <span>2021</span>). Generic mutations causing loss of K<sub>v</sub>7 function often result in pathological hyperexcitability (epilepsies, cardiac arrhythmias and pain) (Jones et al., <span>2021</span>). Conversely, pharmacological activation or enhancement of K<sub>v</sub>7 activity is widely recognised therapeutic strategy for management of hyperexcitability symptoms. Two K<sub>v</sub>7 activators have been clinically used as a painkiller (flupirtine) and anticonvulsant (retigabine), and a number of other clinically used drugs may have K<sub>v</sub>7 activation among their mechanisms of action. These include painkillers, celecoxib (Du et al., <span>2011</span>) and paracetamol (Ray et al., <span>2019</span>) and a vasodilator, fasudil (Zhang et al., <span>2016</span>).</p><p>Although flupirtine and retigabine initially demonstrated clinical promise, both compounds were ultimately withdrawn from the market, mostly due to adverse effects. Retigabine was associated with blue skin discolouration, retinal pigmentation and urinary retention, while flupirtine carried a risk of hepatotoxicity. Current efforts in drug development are focused on identifying next-generation K<sub>v</sub>7 channel activators with superior pharmacological selectivity and safety. Yet, development of a new drug from scratch is a costly and time-consuming undertaking. In this context, screening libraries of already approved (or close to approval) drugs for potential repurposing represents a promising alternative strategy, offering the advantages of accelerated market entry and reduced development costs.</p><p>A new study by Lidia Carotenuto and co-authors, published in the British Journal of Pharmacology (Carotenuto et","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":"183 3","pages":"435-437"},"PeriodicalIF":7.7,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://bpspubs.onlinelibrary.wiley.com/doi/epdf/10.1111/bph.70290","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145630316","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}