Pub Date : 2025-12-01Epub Date: 2025-10-10DOI: 10.1152/ajpcell.00197.2025
Luigi Regenburgh De La Motte, Barbara Bassani, Francesco Trepiccione, Giovambattista Capasso, Antonino Bruno, Giuseppe Ambrosio
The prevalence of heart failure (HF) and of chronic kidney disease (CKD) is continuously rising. Both diseases require significant management efforts, and more importantly, HF is often associated with CKD, aggravating the clinical scenario and leading to "cardiorenal syndrome" (CRS). Although clinical studies suggest a bidirectional interaction between HF and CKD, the pathophysiological understanding of CRS remains incomplete. Several mechanisms are involved in CRS, including changes in systemic and renal hemodynamics, endothelial dysfunction, inflammation, and activation of the renin-angiotensin-aldosterone and sympathetic nervous systems. However, the precise mechanisms are still unclear, partly because of the incomplete characterization of experimental models recapitulating CRS. In this review, we analyze recent studies using different animal models of CRS, such as primary HF, primary CKD, and the "double-hit" models that have been proposed to investigate the pathophysiology of this condition. In HF models, data on renal pathology showed renal fibrosis, inflammation, and decreased glomerular filtration rate (GFR), whereas kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) were used as markers of early kidney damage. In CKD models, data on heart pathology indicated changes in hemodynamics, increased systolic blood pressure, and the presence of fibrosis. These models provide new insights into the pathophysiological development of CRS, particularly the "double-hit" models, which may offer more information about the cross talk between the heart and kidneys. This review emphasizes the complexity of CRS and highlights the need for further research to clarify the underlying interactions and mechanisms.
{"title":"Preclinical models of cardio-renal syndrome: a systematic review.","authors":"Luigi Regenburgh De La Motte, Barbara Bassani, Francesco Trepiccione, Giovambattista Capasso, Antonino Bruno, Giuseppe Ambrosio","doi":"10.1152/ajpcell.00197.2025","DOIUrl":"10.1152/ajpcell.00197.2025","url":null,"abstract":"<p><p>The prevalence of heart failure (HF) and of chronic kidney disease (CKD) is continuously rising. Both diseases require significant management efforts, and more importantly, HF is often associated with CKD, aggravating the clinical scenario and leading to \"cardiorenal syndrome\" (CRS). Although clinical studies suggest a bidirectional interaction between HF and CKD, the pathophysiological understanding of CRS remains incomplete. Several mechanisms are involved in CRS, including changes in systemic and renal hemodynamics, endothelial dysfunction, inflammation, and activation of the renin-angiotensin-aldosterone and sympathetic nervous systems. However, the precise mechanisms are still unclear, partly because of the incomplete characterization of experimental models recapitulating CRS. In this review, we analyze recent studies using different animal models of CRS, such as primary HF, primary CKD, and the \"double-hit\" models that have been proposed to investigate the pathophysiology of this condition. In HF models, data on renal pathology showed renal fibrosis, inflammation, and decreased glomerular filtration rate (GFR), whereas kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) were used as markers of early kidney damage. In CKD models, data on heart pathology indicated changes in hemodynamics, increased systolic blood pressure, and the presence of fibrosis. These models provide new insights into the pathophysiological development of CRS, particularly the \"double-hit\" models, which may offer more information about the cross talk between the heart and kidneys. This review emphasizes the complexity of CRS and highlights the need for further research to clarify the underlying interactions and mechanisms.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C1857-C1872"},"PeriodicalIF":4.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145273565","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
At the neuromuscular junction, nicotinic acetylcholine receptor (nAChR) dynamics are regulated in a nerve- and activity-dependent manner. Correlated local alterations in myoplasmic [Ca2+]i, induced by IP3-sensitive subsynaptic Ca2+ stores, have been proposed to signal motor endplate adaptation to motor neuron stimulation. Accordingly, there is evidence for a modulatory role of Ca2+/calmodulin-dependent protein kinase IIβ (CaMKIIβ) in the sorting, targeting, and/or incorporation of nAChRs into the postsynaptic membrane. As the scaffold protein Homer 2 emerges as a key player in integrating downstream postsynaptic signaling pathways, this study investigated the possible involvement of Homer 2 in the molecular mechanism controlling nAChR dynamics. Using Homer 2-/- transgenic mice, it was found that Homer 2 ablation leads to a chronic adaptation of the endplate characterized by: 1) reduction in nAChR activity due to slower insertion of nAChRs into the endplate; 2) reduced subsynaptic IP3R1 content and IP3-releasable Ca2+; and 3) impaired colocalization of CaMKIIβ with nAChRs. Overall, the present results demonstrate that Homer 2 ablation produces a significant alteration in endplate nAChR dynamics, which is associated with impaired organization of the subsynaptic IP3-driven Ca2+ signaling mechanism.NEW & NOTEWORTHY This research sheds light on the role of Homer 2 in organizing the subsynaptic microdomain, where nAChRs, IP3R1s, and CaMKIIβ assemble to regulate nAChR dynamics. The present results point to a novel type of endplate instability, which may have implications for understanding neuromuscular junction function and related disorders.
{"title":"Adaptation of the endplate in skeletal muscle of Homer 2<sup>-/-</sup> mice.","authors":"Paola Lorenzon, Stefano Amoretti, Sandra Furlan, Barbara Ravara, Annalisa Bernareggi, Marina Sciancalepore, Roberta Sacchetto, Aram Megighian, Sandra Zampieri, Alessandra Nori, Pompeo Volpe","doi":"10.1152/ajpcell.00419.2025","DOIUrl":"10.1152/ajpcell.00419.2025","url":null,"abstract":"<p><p>At the neuromuscular junction, nicotinic acetylcholine receptor (nAChR) dynamics are regulated in a nerve- and activity-dependent manner. Correlated local alterations in myoplasmic [Ca<sup>2+</sup>]<sub>i</sub>, induced by IP<sub>3</sub>-sensitive subsynaptic Ca<sup>2+</sup> stores, have been proposed to signal motor endplate adaptation to motor neuron stimulation. Accordingly, there is evidence for a modulatory role of Ca<sup>2+</sup>/calmodulin-dependent protein kinase IIβ (CaMKIIβ) in the sorting, targeting, and/or incorporation of nAChRs into the postsynaptic membrane. As the scaffold protein Homer 2 emerges as a key player in integrating downstream postsynaptic signaling pathways, this study investigated the possible involvement of Homer 2 in the molecular mechanism controlling nAChR dynamics. Using Homer 2<sup>-/-</sup> transgenic mice, it was found that Homer 2 ablation leads to a chronic adaptation of the endplate characterized by: <i>1</i>) reduction in nAChR activity due to slower insertion of nAChRs into the endplate; <i>2</i>) reduced subsynaptic IP<sub>3</sub>R1 content and IP<sub>3</sub>-releasable Ca<sup>2+</sup>; and <i>3</i>) impaired colocalization of CaMKIIβ with nAChRs. Overall, the present results demonstrate that Homer 2 ablation produces a significant alteration in endplate nAChR dynamics, which is associated with impaired organization of the subsynaptic IP<sub>3</sub>-driven Ca<sup>2+</sup> signaling mechanism.<b>NEW & NOTEWORTHY</b> This research sheds light on the role of Homer 2 in organizing the subsynaptic microdomain, where nAChRs, IP<sub>3</sub>R1s, and CaMKIIβ assemble to regulate nAChR dynamics. The present results point to a novel type of endplate instability, which may have implications for understanding neuromuscular junction function and related disorders.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C2064-C2078"},"PeriodicalIF":4.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145556040","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-10-10DOI: 10.1152/ajpcell.00564.2025
Weronika Wilczak, Rieke Schleinhege, Christine M Loescher, Albrecht Schwab, Zoltán Pethő
Fibrosis plays a crucial role in a range of chronic diseases, including cancer. Emerging evidence suggests that ion channels, transporters, and pumps-the transportome-have an essential share in fibrogenesis and fibrosis by regulating fibroblast and myofibroblast activity. This review bridges current knowledge gaps by integrating insights from multiple diseases affecting the heart, lungs, pancreas, kidney, and liver, as well as cancer. Thereby, we reveal shared molecular mechanisms of how the transportome modulates fibroblast activation, extracellular matrix deposition, tissue stiffness, and remodeling. We focus on the roles of various ion transport proteins, including PIEZO1, transient receptor potential (TRP), K+, and cystic fibrosis transmembrane regulator (CFTR) channels; the Na+/H+ exchanger NHE1; and the Na+/K+-ATPase. By comparing analogous pathways across different fibrotic diseases such as Ca2+ signaling and transforming growth factor β1 (TGF-β1) and Wnt/β-catenin pathways, we highlight the druggable potential of these ion transport proteins and suggest novel concepts for therapeutic intervention.
{"title":"A tough job: ion channels, transporters, and pumps during organ fibrosis.","authors":"Weronika Wilczak, Rieke Schleinhege, Christine M Loescher, Albrecht Schwab, Zoltán Pethő","doi":"10.1152/ajpcell.00564.2025","DOIUrl":"10.1152/ajpcell.00564.2025","url":null,"abstract":"<p><p>Fibrosis plays a crucial role in a range of chronic diseases, including cancer. Emerging evidence suggests that ion channels, transporters, and pumps-the transportome-have an essential share in fibrogenesis and fibrosis by regulating fibroblast and myofibroblast activity. This review bridges current knowledge gaps by integrating insights from multiple diseases affecting the heart, lungs, pancreas, kidney, and liver, as well as cancer. Thereby, we reveal shared molecular mechanisms of how the transportome modulates fibroblast activation, extracellular matrix deposition, tissue stiffness, and remodeling. We focus on the roles of various ion transport proteins, including PIEZO1, transient receptor potential (TRP), K<sup>+</sup>, and cystic fibrosis transmembrane regulator (CFTR) channels; the Na<sup>+</sup>/H<sup>+</sup> exchanger NHE1; and the Na<sup>+</sup>/K<sup>+</sup>-ATPase. By comparing analogous pathways across different fibrotic diseases such as Ca<sup>2+</sup> signaling and transforming growth factor β1 (TGF-β1) and Wnt/β-catenin pathways, we highlight the druggable potential of these ion transport proteins and suggest novel concepts for therapeutic intervention.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C2091-C2111"},"PeriodicalIF":4.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145273584","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-10-20DOI: 10.1152/ajpcell.00441.2025
Karin Åvall, Julie M Williams
The diversity of fibroblasts across different organs, and within the same structures, means that their role in both health and disease is manifold. This review focuses on their job in the heart and kidney, specifically during the course of cardiorenal syndrome (CRS). During CRS, there is a complex bidirectional interplay between the two body systems whereby the failure of one drives the decline of the other. These effects manifest by a response that leads to the deposition of fibrotic tissue, attributable to fibroblast dysfunction. Fibroblasts in themselves provide essential functions within organs, which are determined by the specific identity of their subtype. During disease, fibroblast function is further constrained and directed by the niches that form at the sites of injury. This review delves into the origins of fibroblasts in the heart and kidney, their functions in each tissue, and the processes and stressors whereby they become activated to form myofibroblasts. We discuss tools that can be used to study the phenomenon of fibroblast activation in vitro and in human studies and, finally, what therapeutic possibilities there may be in the future.
{"title":"The role of the fibroblast in cardiorenal syndrome.","authors":"Karin Åvall, Julie M Williams","doi":"10.1152/ajpcell.00441.2025","DOIUrl":"10.1152/ajpcell.00441.2025","url":null,"abstract":"<p><p>The diversity of fibroblasts across different organs, and within the same structures, means that their role in both health and disease is manifold. This review focuses on their job in the heart and kidney, specifically during the course of cardiorenal syndrome (CRS). During CRS, there is a complex bidirectional interplay between the two body systems whereby the failure of one drives the decline of the other. These effects manifest by a response that leads to the deposition of fibrotic tissue, attributable to fibroblast dysfunction. Fibroblasts in themselves provide essential functions within organs, which are determined by the specific identity of their subtype. During disease, fibroblast function is further constrained and directed by the niches that form at the sites of injury. This review delves into the origins of fibroblasts in the heart and kidney, their functions in each tissue, and the processes and stressors whereby they become activated to form myofibroblasts. We discuss tools that can be used to study the phenomenon of fibroblast activation in vitro and in human studies and, finally, what therapeutic possibilities there may be in the future.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C2049-C2063"},"PeriodicalIF":4.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145336255","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-17DOI: 10.1152/ajpcell.00587.2025
Himanshu Verma, Yumei Feng Earley
Astrocytes, traditionally viewed as passive support cells, have emerged as critical regulators of neuronal signaling, autonomic function, and cardiovascular homeostasis. Accumulating evidence highlights the active participation of astrocytes in maintaining neurotransmitter balance, ion homeostasis, synaptic plasticity, and cerebral metabolism. In particular, astrocytes form integral components of tripartite synapses, mediating neuronal communication through calcium-dependent release of gliotransmitters, including ATP, glutamate, d-serine, and γ-aminobutyric acid. This astrocyte-mediated signaling is essential in modulating autonomic circuits involved in blood pressure regulation and sympathetic nerve activity. Recent research underscores the role of astrocyte dysfunction-mediated inflammation, termed astrogliosis, in driving pathological states such as hypertension. Astrocyte activation within critical cardiovascular control centers, including the nucleus tractus solitarii, paraventricular nucleus, and rostral ventrolateral medulla, promotes neuroinflammation, disrupts neurotransmitter clearance, and enhances sympathetic nervous system activity. These processes contribute significantly to hypertension development, particularly under conditions of metabolic stress, such as obesity and high-fat diet consumption. Key molecular mechanisms implicated include NF-κB-mediated inflammatory pathways, impaired astrocytic glutamate transporters, overactivation of angiotensin II signaling, and abnormal gliotransmitter release. In this review, we summarize recent advances in our understanding of the physiological roles of astrocytes in autonomic and cardiovascular regulation and discuss the pathological consequences of astrocyte-driven neuroinflammation in hypertension. We further outline promising directions for future research and therapeutic interventions targeting astrocytic pathways, offering potential new strategies for preventing or reversing autonomic dysfunction and hypertension.
{"title":"Emerging roles of astrocytes in autonomic control of blood pressure and hypertension.","authors":"Himanshu Verma, Yumei Feng Earley","doi":"10.1152/ajpcell.00587.2025","DOIUrl":"10.1152/ajpcell.00587.2025","url":null,"abstract":"<p><p>Astrocytes, traditionally viewed as passive support cells, have emerged as critical regulators of neuronal signaling, autonomic function, and cardiovascular homeostasis. Accumulating evidence highlights the active participation of astrocytes in maintaining neurotransmitter balance, ion homeostasis, synaptic plasticity, and cerebral metabolism. In particular, astrocytes form integral components of tripartite synapses, mediating neuronal communication through calcium-dependent release of gliotransmitters, including ATP, glutamate, d-serine, and γ-aminobutyric acid. This astrocyte-mediated signaling is essential in modulating autonomic circuits involved in blood pressure regulation and sympathetic nerve activity. Recent research underscores the role of astrocyte dysfunction-mediated inflammation, termed astrogliosis, in driving pathological states such as hypertension. Astrocyte activation within critical cardiovascular control centers, including the nucleus tractus solitarii, paraventricular nucleus, and rostral ventrolateral medulla, promotes neuroinflammation, disrupts neurotransmitter clearance, and enhances sympathetic nervous system activity. These processes contribute significantly to hypertension development, particularly under conditions of metabolic stress, such as obesity and high-fat diet consumption. Key molecular mechanisms implicated include NF-κB-mediated inflammatory pathways, impaired astrocytic glutamate transporters, overactivation of angiotensin II signaling, and abnormal gliotransmitter release. In this review, we summarize recent advances in our understanding of the physiological roles of astrocytes in autonomic and cardiovascular regulation and discuss the pathological consequences of astrocyte-driven neuroinflammation in hypertension. We further outline promising directions for future research and therapeutic interventions targeting astrocytic pathways, offering potential new strategies for preventing or reversing autonomic dysfunction and hypertension.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C2013-C2021"},"PeriodicalIF":4.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145538683","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-10-31DOI: 10.1152/ajpcell.00716.2025
Dachuan Dong, Wen-Jun Shen, Stefanie Bittner, Jia Chen, Xiao Hao, Mark Donowitz, Fredric B Kraemer, Salman Azhar
Dysregulation of cholesterol metabolism can lead to obesity and increase the risk of developing many diseases, including type 2 diabetes and cardiovascular diseases. Our previous studies have identified postsynaptic density-95, disc large, and zonula occludens-1 (PDZ) adaptor proteins Na+/H+ exchange regulatory factor Nherf1 (encoded by Nherf1) and Nherf2 (encoded by Nherf2) to be potential regulators of cholesterol metabolism in vitro. In this study, we explored their physiological regulatory function in vivo by using Nherf1- and Nherf2-deficient (Nherf1-/- and Nherf2-/-), and wild-type (C57BL/6) mice. All mice were fed either a chow diet or a cholesterol-enriched Western diet (42% fat, 0.2% cholesterol) for 8 wk starting at 8-wk-old. Our results demonstrate that Nherf2-/-, but not Nherf1-/-, mice are resistant to diet-induced obesity. In Nherf2-/- mice, serum high-density lipoprotein and low-density lipoprotein/very low-density lipoprotein decreased substantially without affecting lipolysis or steroid hormone levels. In addition, distended gallbladders were observed in Nherf2-/- mice, with reduced bile acid output into the intestine and feces, which correlated with decreased cholesterol reabsorption. This led to attenuated Fxr/Shp signaling in the liver and derepressing Cyp7a1 transcription in the absence of Nherf2. These findings suggest a potential role of in regulating gallbladder emptying and lipid homeostasis, offering new insights into potential therapeutic targets for treating diet-induced obesity.NEW & NOTEWORTHYNherf2-/- but not Nherf1-/- mice demonstrate a resistance to diet-induced obesity. Notably, male Nherf2-/- mice exhibit impaired glucose tolerance and insulin responsiveness, yet neither sex shows further worsening with diet challenge. In addition, elevated hepatic Cyp7a1 levels were observed in Nherf2-/- mice, but there was reduced cholesterol absorption in the ileum, along with enlarged gallbladders and diminished ileal bile acid content, highlighting significant metabolic alterations linked to Nherf2 deficiency.
{"title":"Nherf2 is a major determinant of bile acid pool dynamics and contributes to regulation of Western diet-induced obesity.","authors":"Dachuan Dong, Wen-Jun Shen, Stefanie Bittner, Jia Chen, Xiao Hao, Mark Donowitz, Fredric B Kraemer, Salman Azhar","doi":"10.1152/ajpcell.00716.2025","DOIUrl":"10.1152/ajpcell.00716.2025","url":null,"abstract":"<p><p>Dysregulation of cholesterol metabolism can lead to obesity and increase the risk of developing many diseases, including type 2 diabetes and cardiovascular diseases. Our previous studies have identified postsynaptic density-95, disc large, and zonula occludens-1 (PDZ) adaptor proteins Na<sup>+</sup>/H<sup>+</sup> exchange regulatory factor Nherf1 (encoded by <i>Nherf1</i>) and Nherf2 (encoded by <i>Nherf2</i>) to be potential regulators of cholesterol metabolism in vitro. In this study, we explored their physiological regulatory function in vivo by using Nherf1- and Nherf2-deficient (<i>Nherf1</i><sup>-/-</sup> and <i>Nherf2</i><sup>-/-</sup>), and wild-type (C57BL/6) mice. All mice were fed either a chow diet or a cholesterol-enriched Western diet (42% fat, 0.2% cholesterol) for 8 wk starting at 8-wk-old. Our results demonstrate that <i>Nherf2</i><sup>-/-</sup>, but not <i>Nherf1</i><sup>-/-</sup>, mice are resistant to diet-induced obesity. In <i>Nherf2</i><sup>-/-</sup> mice, serum high-density lipoprotein and low-density lipoprotein/very low-density lipoprotein decreased substantially without affecting lipolysis or steroid hormone levels. In addition, distended gallbladders were observed in <i>Nherf2</i><sup>-/-</sup> mice, with reduced bile acid output into the intestine and feces, which correlated with decreased cholesterol reabsorption. This led to attenuated Fxr/Shp signaling in the liver and derepressing <i>Cyp7a1</i> transcription in the absence of Nherf2. These findings suggest a potential role of in regulating gallbladder emptying and lipid homeostasis, offering new insights into potential therapeutic targets for treating diet-induced obesity.<b>NEW & NOTEWORTHY</b> <i>Nherf2</i><sup>-/-</sup> but not <i>Nherf1</i><sup>-/-</sup> mice demonstrate a resistance to diet-induced obesity. Notably, male <i>Nherf2</i><sup>-/-</sup> mice exhibit impaired glucose tolerance and insulin responsiveness, yet neither sex shows further worsening with diet challenge. In addition, elevated hepatic <i>Cyp7a1</i> levels were observed in <i>Nherf2</i><sup>-/-</sup> mice, but there was reduced cholesterol absorption in the ileum, along with enlarged gallbladders and diminished ileal bile acid content, highlighting significant metabolic alterations linked to Nherf2 deficiency.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C1765-C1774"},"PeriodicalIF":4.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12716856/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145420948","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-10-21DOI: 10.1152/ajpcell.00254.2025
Connor A Stead, Stuart J Hesketh, Aaron C Q Thomas, Mark R Viggars, Hazel Sutherland, Jonathan C Jarvis, Jatin G Burniston
Resistance training promotes muscle protein accretion and myofiber hypertrophy, driven by dynamic processes of protein synthesis and degradation. Muscle adaptations to ongoing resistance training occur over weeks, but most molecular knowledge on the process of adaptation is derived from static measurements at specific time points, which do not capture the dynamics of the adaptation process. To address this, we utilized deuterium oxide labeling and peptide mass spectrometry to quantify absolute protein content (grams) and synthesis rates (grams/day) in skeletal muscle during a time series experimental design. A daily programmed resistance training regimen was applied to male rat tibialis anterior via electrical stimulation of the left hindlimb for 10, 20, and 30 days (5 sets of 10 repetitions daily). Muscle samples from stimulated and contralateral control limbs were analyzed, quantifying 658 protein abundances and 215 protein synthesis rates. Unsupervised temporal clustering of protein responses revealed distinct phases of muscle adaptation. The early (0-10 days) response was driven by greater rates of ribosomal protein accretion and the mid (10-20 days) response by expansion of mitochondrial networks. These findings highlight that subsets of proteins exhibit distinct adaptation timelines due to variations in translation and/or degradation rates. The new understanding of temporal patterns highlighted by our dynamic proteomic data helps interpret static data from studies at isolated time points and could improve the development of strategies for optimizing muscle growth and functional adaptation to resistance training.NEW & NOTEWORTHY We used stable isotope labeling and proteomic analyses to quantify absolute changes in the synthesis and abundance of muscle proteins during programmed resistance training in rat in vivo. This novel time-resolved approach revealed distinct phases of adaptation, characterized by early ribosomal and later mitochondrial protein accretion. Strikingly, we observed substantial "oversynthesis" of muscle proteins; that is, the net gain in muscle protein content was much less than the amount of newly synthesized protein.
{"title":"Dynamic time course of muscle proteome adaptation to programmed resistance training in rats.","authors":"Connor A Stead, Stuart J Hesketh, Aaron C Q Thomas, Mark R Viggars, Hazel Sutherland, Jonathan C Jarvis, Jatin G Burniston","doi":"10.1152/ajpcell.00254.2025","DOIUrl":"10.1152/ajpcell.00254.2025","url":null,"abstract":"<p><p>Resistance training promotes muscle protein accretion and myofiber hypertrophy, driven by dynamic processes of protein synthesis and degradation. Muscle adaptations to ongoing resistance training occur over weeks, but most molecular knowledge on the process of adaptation is derived from static measurements at specific time points, which do not capture the dynamics of the adaptation process. To address this, we utilized deuterium oxide labeling and peptide mass spectrometry to quantify absolute protein content (grams) and synthesis rates (grams/day) in skeletal muscle during a time series experimental design. A daily programmed resistance training regimen was applied to male rat tibialis anterior via electrical stimulation of the left hindlimb for 10, 20, and 30 days (5 sets of 10 repetitions daily). Muscle samples from stimulated and contralateral control limbs were analyzed, quantifying 658 protein abundances and 215 protein synthesis rates. Unsupervised temporal clustering of protein responses revealed distinct phases of muscle adaptation. The early (0-10 days) response was driven by greater rates of ribosomal protein accretion and the mid (10-20 days) response by expansion of mitochondrial networks. These findings highlight that subsets of proteins exhibit distinct adaptation timelines due to variations in translation and/or degradation rates. The new understanding of temporal patterns highlighted by our dynamic proteomic data helps interpret static data from studies at isolated time points and could improve the development of strategies for optimizing muscle growth and functional adaptation to resistance training.<b>NEW & NOTEWORTHY</b> We used stable isotope labeling and proteomic analyses to quantify absolute changes in the synthesis and abundance of muscle proteins during programmed resistance training in rat in vivo. This novel time-resolved approach revealed distinct phases of adaptation, characterized by early ribosomal and later mitochondrial protein accretion. Strikingly, we observed substantial \"oversynthesis\" of muscle proteins; that is, the net gain in muscle protein content was much less than the amount of newly synthesized protein.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C1792-C1809"},"PeriodicalIF":4.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145336284","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-10-25DOI: 10.1152/ajpcell.00060.2025
Kemly Philip, Hannah P Thompson, Scott D Collum, Isabella Lefebvre, Bindu Akkanti, Bihong Zhao, Rahat Hussain, Manish Patel, Michael R Blackburn, Tingting W Mills, Harry Karmouty-Quintana
A complication of viral lung infections is the development of pulmonary fibrosis. This phenomenon is most evident in patients with COVID-19, where in its most aggressive form patients developed nonresolvable (NR) COVID-19 requiring lung transplantation. NR-COVID-19 was characterized by the presentation of a fulminant fibrotic lung injury that progressed rapidly, even in patients with limited comorbidities. However, the mechanisms that led to this rapidly progressing form of fibrosis are not fully understood. A common clinical manifestation in the most severe cases of COVID-19 was the presence of "silent" hypoxemia. Thus, we hypothesized that a dysfunctional hypoxic response may result in exacerbated lung injury seen in patients with severe forms of COVID-19. Our results demonstrate that despite increased expression of hypoxia-inducible factor 1A (HIF1A) and its downstream mediator adenosine A2B receptor (ADORA2B), reduced macrophage HIF1A was observed in patients with severe COVID-19, including NR-COVID-19. Utilizing mice lacking HIF1A in myeloid cells using the lysozyme M Cre promoter, we demonstrate that these mice present with increased lung inflammation and pulmonary fibrosis following chronic low-dose bleomycin treatment. The augmented lung injury was associated with reduced markers for alternatively activated macrophages also observed in NR-COVID-19 lungs. These results point to reduced myeloid HIF1A as a mechanism that can lead to exacerbated lung injury in mice, which parallels the rapid fibrotic response observed in NR-COVID-19. Collectively, our results point to using HIF1A stabilizers as a potential avenue to prevent the development of rapidly progressing postviral lung fibrosis. However, special care is necessary since chronic HIF1A activation is also linked to fibrotic outcomes.NEW & NOTEWORTHY Postviral-induced lung fibrosis represents a severe and potentially fatal outcome. This was most evident during the COVID-19 pandemic, where a subset of individuals presented with fulminant lung fibrosis requiring lung transplantation. The mechanisms that promote this exacerbated lung injury are not fully known. Herein, we demonstrate that mice lacking myeloid hypoxia-inducible factor 1A (HIF1A) develop an exacerbated lung injury response to bleomycin that was consistent with reduced macrophage HIF1A expression in nonresolvable (NR)-COVID-19, characterized by extensive lung fibrosis.
{"title":"Decoding fibrosis in nonresolvable COVID-19: a role for myeloid-specific HIF1A deletion.","authors":"Kemly Philip, Hannah P Thompson, Scott D Collum, Isabella Lefebvre, Bindu Akkanti, Bihong Zhao, Rahat Hussain, Manish Patel, Michael R Blackburn, Tingting W Mills, Harry Karmouty-Quintana","doi":"10.1152/ajpcell.00060.2025","DOIUrl":"10.1152/ajpcell.00060.2025","url":null,"abstract":"<p><p>A complication of viral lung infections is the development of pulmonary fibrosis. This phenomenon is most evident in patients with COVID-19, where in its most aggressive form patients developed nonresolvable (NR) COVID-19 requiring lung transplantation. NR-COVID-19 was characterized by the presentation of a fulminant fibrotic lung injury that progressed rapidly, even in patients with limited comorbidities. However, the mechanisms that led to this rapidly progressing form of fibrosis are not fully understood. A common clinical manifestation in the most severe cases of COVID-19 was the presence of \"silent\" hypoxemia. Thus, we hypothesized that a dysfunctional hypoxic response may result in exacerbated lung injury seen in patients with severe forms of COVID-19. Our results demonstrate that despite increased expression of hypoxia-inducible factor 1A (HIF1A) and its downstream mediator adenosine A<sub>2B</sub> receptor (ADORA2B), reduced macrophage HIF1A was observed in patients with severe COVID-19, including NR-COVID-19. Utilizing mice lacking HIF1A in myeloid cells using the lysozyme M Cre promoter, we demonstrate that these mice present with increased lung inflammation and pulmonary fibrosis following chronic low-dose bleomycin treatment. The augmented lung injury was associated with reduced markers for alternatively activated macrophages also observed in NR-COVID-19 lungs. These results point to reduced myeloid HIF1A as a mechanism that can lead to exacerbated lung injury in mice, which parallels the rapid fibrotic response observed in NR-COVID-19. Collectively, our results point to using HIF1A stabilizers as a potential avenue to prevent the development of rapidly progressing postviral lung fibrosis. However, special care is necessary since chronic HIF1A activation is also linked to fibrotic outcomes.<b>NEW & NOTEWORTHY</b> Postviral-induced lung fibrosis represents a severe and potentially fatal outcome. This was most evident during the COVID-19 pandemic, where a subset of individuals presented with fulminant lung fibrosis requiring lung transplantation. The mechanisms that promote this exacerbated lung injury are not fully known. Herein, we demonstrate that mice lacking myeloid hypoxia-inducible factor 1A (HIF1A) develop an exacerbated lung injury response to bleomycin that was consistent with reduced macrophage HIF1A expression in nonresolvable (NR)-COVID-19, characterized by extensive lung fibrosis.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C1752-C1764"},"PeriodicalIF":4.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145370132","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-11DOI: 10.1152/ajpcell.00567.2025
Daniel Gonsales Spindola, Amber Bahr, Samantha Clark, Gabriel Pin de Jesus, Nina Martino, Anthony Lowery, Shuhan Lyu, Andrew Seeman, Grace Martino, Giesse Albeche Duarte, Elijah Crosbourne, Peter Vincent, Guangchun Bai, Alejandro P Adam, Katherine C MacNamara, Ramon Bossardi Ramos
Sepsis survivors face a heightened risk of secondary infections following discharge, yet the underlying mechanisms remain poorly defined. Our study identifies a novel mechanism of endothelial inflammatory memory, wherein inflammatory exposure induces durable chromatin remodeling in endothelial cells (ECs), priming them for exaggerated responses to a subsequent infection. Utilizing a clinically relevant two-hit mouse model, cecal ligation and puncture (CLP) followed by mild Streptococcus pneumoniae (Sp) infection in CLP survivors, we reveal transcriptional activation in endothelial cells (ECs) following secondary infection, marked by significantly elevated expression of proinflammatory cytokines, adhesion molecules, complement factors, and interferon-stimulated genes. Genome-wide ATAC-seq revealed that a subset of inflammatory gene loci retained increased chromatin accessibility even after cytokine withdrawal, demonstrating stable epigenetic remodeling consistent with transcriptional priming and inflammatory memory. In vitro, we uncovered a critical role for the activator protein-1 transcription factor JunB in mediating this epigenetic remodeling. JunB knockdown attenuated chromatin accessibility after an initial IL-6 challenge and subsequent transcriptional amplification upon a secondary LPS challenge, pinpointing JunB-driven chromatin modifications as central to endothelial reprogramming. Our findings offer mechanistic insights into how transient inflammation creates lasting epigenetic states within the endothelium, highlighting JunB as a potential therapeutic target to mitigate chronic endothelial dysfunction and increased susceptibility to secondary infections postsepsis.NEW & NOTEWORTHY We uncover that endothelial cells retain a form of inflammatory memory, driven by chromatin remodeling and sustained JunB activity. Using two-hit models in mice and human endothelial cells, we show that an initial inflammatory exposure primes the endothelium for exaggerated responses to future inflammation. This discovery reveals a new mechanism of chronic endothelial dysfunction and identifies JunB as a potential therapeutic target in postsepsis care.
{"title":"Endothelial cells retain inflammatory memory through chromatin remodeling in a two-hit model of infection-induced inflammation.","authors":"Daniel Gonsales Spindola, Amber Bahr, Samantha Clark, Gabriel Pin de Jesus, Nina Martino, Anthony Lowery, Shuhan Lyu, Andrew Seeman, Grace Martino, Giesse Albeche Duarte, Elijah Crosbourne, Peter Vincent, Guangchun Bai, Alejandro P Adam, Katherine C MacNamara, Ramon Bossardi Ramos","doi":"10.1152/ajpcell.00567.2025","DOIUrl":"10.1152/ajpcell.00567.2025","url":null,"abstract":"<p><p>Sepsis survivors face a heightened risk of secondary infections following discharge, yet the underlying mechanisms remain poorly defined. Our study identifies a novel mechanism of endothelial inflammatory memory, wherein inflammatory exposure induces durable chromatin remodeling in endothelial cells (ECs), priming them for exaggerated responses to a subsequent infection. Utilizing a clinically relevant two-hit mouse model, cecal ligation and puncture (CLP) followed by mild <i>Streptococcus pneumoniae</i> (<i>Sp</i>) infection in CLP survivors, we reveal transcriptional activation in endothelial cells (ECs) following secondary infection, marked by significantly elevated expression of proinflammatory cytokines, adhesion molecules, complement factors, and interferon-stimulated genes. Genome-wide ATAC-seq revealed that a subset of inflammatory gene loci retained increased chromatin accessibility even after cytokine withdrawal, demonstrating stable epigenetic remodeling consistent with transcriptional priming and inflammatory memory. In vitro, we uncovered a critical role for the activator protein-1 transcription factor JunB in mediating this epigenetic remodeling. JunB knockdown attenuated chromatin accessibility after an initial IL-6 challenge and subsequent transcriptional amplification upon a secondary LPS challenge, pinpointing JunB-driven chromatin modifications as central to endothelial reprogramming. Our findings offer mechanistic insights into how transient inflammation creates lasting epigenetic states within the endothelium, highlighting JunB as a potential therapeutic target to mitigate chronic endothelial dysfunction and increased susceptibility to secondary infections postsepsis.<b>NEW & NOTEWORTHY</b> We uncover that endothelial cells retain a form of inflammatory memory, driven by chromatin remodeling and sustained JunB activity. Using two-hit models in mice and human endothelial cells, we show that an initial inflammatory exposure primes the endothelium for exaggerated responses to future inflammation. This discovery reveals a new mechanism of chronic endothelial dysfunction and identifies JunB as a potential therapeutic target in postsepsis care.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C1994-C2012"},"PeriodicalIF":4.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12704011/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145487518","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-10-06DOI: 10.1152/ajpcell.00533.2025
Britt van de Haterd, Michèlle Hendriks, Bert Houben, Michelle E G Weijzen, Frank Vandenabeele, Kenneth Verboven, Anouk Agten
Cancer cachexia causes skeletal muscle wasting and metabolic dysfunction, worsening clinical outcomes in colorectal cancer (CRC). This study examines microscopic and macroscopic skeletal muscle fiber characteristics, and muscle volume in patients with CRC-associated cachexia and without cachexia compared with healthy controls (HCs), and explores how these factors relate to physical performance. In total, 12 patients with CRC-associated cachexia, 25 CRC patients without cachexia, and 25 HCs were included. Cachexia was determined by weight loss and Cachexia Staging Score. Biopsies from the vastus lateralis and erector spinae muscles were analyzed using immunohistochemistry for muscle fiber type cross-sectional area (CSA) and distribution, myonuclear content, and capillary density. Muscle volume was assessed using three-dimensional ultrasound, and CSA and density by computerized tomography scans. Physical function was evaluated with the Short Physical Performance Battery test, handgrip strength, and the Physical Activity Scale for Individuals with Physical Disabilities. Quality of life was assessed using the 36-item Short Form Survey. Patients with CRC-associated cachexia showed reduced type II muscle fiber CSA in the vastus lateralis compared with HCs and CRC patients without cachexia. CRC Patients without cachexia exhibited a slow-to-fast muscle fiber shift compared with HCs. Myonuclear content was lower in both cancer groups. Muscle volume and density were reduced in patients with CRC-associated cachexia. Positive correlations were found between microscopic and macroscopic skeletal muscle characteristics, muscle strength, physical performance, and quality of life, respectively. CRC Patients, especially those with cachexia, showed type II muscle fiber atrophy, reduced myonuclear content, and impaired physical function, emphasizing the need for targeted prehabilitation interventions.NEW & NOTEWORTHY This study reveals skeletal muscle alterations in colorectal cancer patients with cachexia, at microscopic (fiber-type specific atrophy, myonuclear content, and capillarization) and macroscopic levels (muscle volume and quality). These alterations were associated with clinically important measures of physical functioning and quality of life. Collectively, these findings establish clinically relevant links between structural muscle alterations and physical outcomes, highlighting the potential value of targeted (p)rehabilitation interventions in these patient populations.
{"title":"Skeletal muscle alterations and functional repercussions in patients with colorectal cancer-associated cachexia.","authors":"Britt van de Haterd, Michèlle Hendriks, Bert Houben, Michelle E G Weijzen, Frank Vandenabeele, Kenneth Verboven, Anouk Agten","doi":"10.1152/ajpcell.00533.2025","DOIUrl":"10.1152/ajpcell.00533.2025","url":null,"abstract":"<p><p>Cancer cachexia causes skeletal muscle wasting and metabolic dysfunction, worsening clinical outcomes in colorectal cancer (CRC). This study examines microscopic and macroscopic skeletal muscle fiber characteristics, and muscle volume in patients with CRC-associated cachexia and without cachexia compared with healthy controls (HCs), and explores how these factors relate to physical performance. In total, 12 patients with CRC-associated cachexia, 25 CRC patients without cachexia, and 25 HCs were included. Cachexia was determined by weight loss and Cachexia Staging Score. Biopsies from the vastus lateralis and erector spinae muscles were analyzed using immunohistochemistry for muscle fiber type cross-sectional area (CSA) and distribution, myonuclear content, and capillary density. Muscle volume was assessed using three-dimensional ultrasound, and CSA and density by computerized tomography scans. Physical function was evaluated with the Short Physical Performance Battery test, handgrip strength, and the Physical Activity Scale for Individuals with Physical Disabilities. Quality of life was assessed using the 36-item Short Form Survey. Patients with CRC-associated cachexia showed reduced type II muscle fiber CSA in the vastus lateralis compared with HCs and CRC patients without cachexia. CRC Patients without cachexia exhibited a slow-to-fast muscle fiber shift compared with HCs. Myonuclear content was lower in both cancer groups. Muscle volume and density were reduced in patients with CRC-associated cachexia. Positive correlations were found between microscopic and macroscopic skeletal muscle characteristics, muscle strength, physical performance, and quality of life, respectively. CRC Patients, especially those with cachexia, showed type II muscle fiber atrophy, reduced myonuclear content, and impaired physical function, emphasizing the need for targeted prehabilitation interventions.<b>NEW & NOTEWORTHY</b> This study reveals skeletal muscle alterations in colorectal cancer patients with cachexia, at microscopic (fiber-type specific atrophy, myonuclear content, and capillarization) and macroscopic levels (muscle volume and quality). These alterations were associated with clinically important measures of physical functioning and quality of life. Collectively, these findings establish clinically relevant links between structural muscle alterations and physical outcomes, highlighting the potential value of targeted (p)rehabilitation interventions in these patient populations.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C1825-C1841"},"PeriodicalIF":4.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145237736","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}