首页 > 最新文献

American journal of physiology. Lung cellular and molecular physiology最新文献

英文 中文
Parenchymal and inflammatory responses to ozone exposure in the aging healthy and surfactant protein C mutant lung. 老化健康和表面活性蛋白C突变肺对臭氧暴露的实质和炎症反应。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-20 DOI: 10.1152/ajplung.00261.2024
Jenna R Cheminant, Cassandra E Deering-Rice, Christopher B Massa, Ujjwal Adhikari, Jessica Noll, Christopher A Reilly, Alessandro Venosa

Ozone (O3) is a ubiquitous pollutant known to produce acute, transient inflammation through oxidative injury and inflammation. These effects are exacerbated in susceptible populations, such as the elderly and those exhibiting genetic mutations in central nodes of pulmonary function. To comprehend the impact of these predisposing factors, the present study examines structural, mechanical, and immunological responses to single acute O3 exposure (0.8 ppm, 3 h) in young (8-14-wk old), middle-aged (44-52-wk old), and old (>80-wk old) mice. Furthermore, this work compares the impact of a clinically relevant mutation in the gene encoding for the alveolar epithelial type 2 specific surfactant protein C. Aging was associated with reduced lung resistance and increases in respiratory elastic properties, the latter of which was exacerbated in SP-C mutant mice. Ozone exposure produced focal injury localized at the terminal bronchiole-to-alveolar junctions and enlarged alveoli in aged SP-C mutant lungs. Flow cytometric analysis revealed increases in mononuclear myeloid abundance in aged SP-C mutant lungs, paired with a contraction in CD8+ expressing cells. Expansion of tertiary lymphoid tissues was also noted in aged groups, more evident in the mutant mice. Spatial transcriptomics of CD68+ macrophages and CD45- nonimmune parenchymal cells highlighted age-dependent shifts in inflammatory and extracellular matrix organization signaling, and enrichment in senescence and chromatin remodeling pathways. These results illustrate the structural and immunological impact of O3 in the aging wild-type and mutant lung and emphasize the significance of modeling environmental exposure in at-risk populations.NEW & NOTEWORTHY Environmental stress and genetic mutations in key functional nodes are linked to the pathogenesis and exacerbation of respiratory pathologies. These responses are exacerbated by aging, though the impact of these factors in combination is not clearly defined. Using a surfactant protein-C mutant line, our studies describe structural changes and phenotypic responses triggered by acute ozone exposure in the young/middle-aged/old lung. Spatial transcriptomics also found regionally distinct and enhanced activation in the aged lung.

臭氧(O3)是一种普遍存在的污染物,已知可通过氧化损伤和炎症产生急性、短暂性炎症。这些影响在易感人群中更为严重,如老年人和那些表现出肺功能中心淋巴结基因突变的人。为了了解这些易感因素的影响,本研究检查了年轻(8-14周龄)、中年(44-52周龄)和老年(80周龄)小鼠对单次急性臭氧暴露(0.8 ppm, 3h)的结构、机械和免疫反应。此外,本研究比较了肺泡上皮2型特异性表面活性剂蛋白c编码基因的临床相关突变的影响。衰老与肺阻力降低和呼吸弹性特性增加相关,后者在SP-C突变小鼠中加剧。臭氧暴露在老年SP-C突变肺中产生局灶性损伤,局限于终末细支气管与肺泡连接处,肺泡增大。流式细胞术分析显示,老年SP-C突变体肺部单核髓细胞丰度增加,同时CD8+表达细胞收缩。在老龄组中,三级淋巴组织的扩张也被注意到,在突变小鼠中更为明显。CD68+巨噬细胞和CD45-非免疫实质细胞的空间转录组学强调了炎症和细胞外基质组织信号的年龄依赖性变化,以及衰老和染色质重塑途径的富集。这些结果说明了O3在衰老野生型和突变型肺中的结构和免疫学影响,并强调了在高危人群中模拟环境暴露的重要性。
{"title":"Parenchymal and inflammatory responses to ozone exposure in the aging healthy and surfactant protein C mutant lung.","authors":"Jenna R Cheminant, Cassandra E Deering-Rice, Christopher B Massa, Ujjwal Adhikari, Jessica Noll, Christopher A Reilly, Alessandro Venosa","doi":"10.1152/ajplung.00261.2024","DOIUrl":"10.1152/ajplung.00261.2024","url":null,"abstract":"<p><p>Ozone (O<sub>3</sub>) is a ubiquitous pollutant known to produce acute, transient inflammation through oxidative injury and inflammation. These effects are exacerbated in susceptible populations, such as the elderly and those exhibiting genetic mutations in central nodes of pulmonary function. To comprehend the impact of these predisposing factors, the present study examines structural, mechanical, and immunological responses to single acute O<sub>3</sub> exposure (0.8 ppm, 3 h) in young (8-14-wk old), middle-aged (44-52-wk old), and old (>80-wk old) mice. Furthermore, this work compares the impact of a clinically relevant mutation in the gene encoding for the alveolar epithelial type 2 specific surfactant protein C. Aging was associated with reduced lung resistance and increases in respiratory elastic properties, the latter of which was exacerbated in SP-C mutant mice. Ozone exposure produced focal injury localized at the terminal bronchiole-to-alveolar junctions and enlarged alveoli in aged SP-C mutant lungs. Flow cytometric analysis revealed increases in mononuclear myeloid abundance in aged SP-C mutant lungs, paired with a contraction in CD8<sup>+</sup> expressing cells. Expansion of tertiary lymphoid tissues was also noted in aged groups, more evident in the mutant mice. Spatial transcriptomics of CD68<sup>+</sup> macrophages and CD45<sup>-</sup> nonimmune parenchymal cells highlighted age-dependent shifts in inflammatory and extracellular matrix organization signaling, and enrichment in senescence and chromatin remodeling pathways. These results illustrate the structural and immunological impact of O<sub>3</sub> in the aging wild-type and mutant lung and emphasize the significance of modeling environmental exposure in at-risk populations.<b>NEW & NOTEWORTHY</b> Environmental stress and genetic mutations in key functional nodes are linked to the pathogenesis and exacerbation of respiratory pathologies. These responses are exacerbated by aging, though the impact of these factors in combination is not clearly defined. Using a surfactant protein-C mutant line, our studies describe structural changes and phenotypic responses triggered by acute ozone exposure in the young/middle-aged/old lung. Spatial transcriptomics also found regionally distinct and enhanced activation in the aged lung.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":" ","pages":"L334-L349"},"PeriodicalIF":3.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998422","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}
引用次数: 0
Engineered hydrogel biomaterials facilitate lung progenitor cell differentiation from induced pluripotent stem cells.
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-30 DOI: 10.1152/ajplung.00419.2024
Alicia E Tanneberger, Rachel Blomberg, Ganna Bilousova, Amy L Ryan, Chelsea M Magin

Lung progenitor (LP) cells identified by the expression of transcription factor NK2 homeobox 1 (NKX2.1) are essential for the development of all lung epithelial cell types and hold tremendous potential for pulmonary research and translational regenerative medicine applications. Here, we present engineered hydrogels as a promising alternative to the naturally derived materials that are often used to differentiate human-induced pluripotent stem cells (iPSCs) into LP cells. Poly(ethylene glycol) norbornene (PEGNB) hydrogels with defined composition were used to systematically investigate the role of microenvironmental stiffness, cell origin, and splitting during the differentiation process. Results demonstrated that each factor impacted LP differentiation efficiency and that the soft hydrogels replicating healthy lung stiffness [elastic modulus (E) = 4.00 ± 0.25 kPa] produced the highest proportion of LP cells based on flow cytometric analysis results (54%) relative to the stiff hydrogels (48%) and Matrigel controls (32%) at the end of the nonsplit differentiation protocol. Collectively, these results showed that engineered hydrogels provide a well-defined microenvironment for iPSC-to-LP differentiation and perform as effectively as the current gold standard Matrigel-coated tissue culture plastic. Adopting engineered biomaterials in cell culture protocols may enable greater control over differentiation parameters and has the potential to enhance the clinical translation of iPSC-derived LP cells.NEW & NOTEWORTHY Standard iPSC differentiation protocols rely on Matrigel, a basement membrane extract from mouse sarcoma cells that is poorly defined and exhibits significant batch-to-batch variation. Due to these limitations, Matrigel-derived products have never been approved by the Food and Drug Administration. This study introduces a novel method for differentiating iPSCs into lung progenitor cells using well-defined hydrogel substrates. These biomaterials not only enhance differentiation efficiency but also streamline the regulatory pathway, facilitating their potential therapeutic application.

{"title":"Engineered hydrogel biomaterials facilitate lung progenitor cell differentiation from induced pluripotent stem cells.","authors":"Alicia E Tanneberger, Rachel Blomberg, Ganna Bilousova, Amy L Ryan, Chelsea M Magin","doi":"10.1152/ajplung.00419.2024","DOIUrl":"10.1152/ajplung.00419.2024","url":null,"abstract":"<p><p>Lung progenitor (LP) cells identified by the expression of transcription factor NK2 homeobox 1 (NKX2.1) are essential for the development of all lung epithelial cell types and hold tremendous potential for pulmonary research and translational regenerative medicine applications. Here, we present engineered hydrogels as a promising alternative to the naturally derived materials that are often used to differentiate human-induced pluripotent stem cells (iPSCs) into LP cells. Poly(ethylene glycol) norbornene (PEGNB) hydrogels with defined composition were used to systematically investigate the role of microenvironmental stiffness, cell origin, and splitting during the differentiation process. Results demonstrated that each factor impacted LP differentiation efficiency and that the soft hydrogels replicating healthy lung stiffness [elastic modulus (<i>E</i>) = 4.00 ± 0.25 kPa] produced the highest proportion of LP cells based on flow cytometric analysis results (54%) relative to the stiff hydrogels (48%) and Matrigel controls (32%) at the end of the nonsplit differentiation protocol. Collectively, these results showed that engineered hydrogels provide a well-defined microenvironment for iPSC-to-LP differentiation and perform as effectively as the current gold standard Matrigel-coated tissue culture plastic. Adopting engineered biomaterials in cell culture protocols may enable greater control over differentiation parameters and has the potential to enhance the clinical translation of iPSC-derived LP cells.<b>NEW & NOTEWORTHY</b> Standard iPSC differentiation protocols rely on Matrigel, a basement membrane extract from mouse sarcoma cells that is poorly defined and exhibits significant batch-to-batch variation. Due to these limitations, Matrigel-derived products have never been approved by the Food and Drug Administration. This study introduces a novel method for differentiating iPSCs into lung progenitor cells using well-defined hydrogel substrates. These biomaterials not only enhance differentiation efficiency but also streamline the regulatory pathway, facilitating their potential therapeutic application.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":" ","pages":"L379-L388"},"PeriodicalIF":3.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062986","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}
引用次数: 0
Secondhand vape exposure regulation of CFTR and immune function in cystic fibrosis. 二手电子烟暴露对囊性纤维化患者CFTR和免疫功能的调节。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-21 DOI: 10.1152/ajplung.00328.2024
Benjamin L Wisniewski, Mahesh Shrestha, Dinesh Bojja, Chandra L Shrestha, Chris S Lee, Hazel Ozuna, Rachael E Rayner, Shasha Bai, Estelle Cormet-Boyaka, Susan D Reynolds, Benjamin T Kopp

Secondhand smoke exposure (SHSe) is a public health threat for people with cystic fibrosis (CF) and other lung diseases. Primary smoking reduces CF transmembrane conductance regulator (CFTR) channel function, the causative defect in CF. We reported that SHSe worsens respiratory and nutritional outcomes in CF by disrupting immune responses and metabolic signaling. Recently, electronic cigarette (e-cigs) usage by caregivers and peers has increased rapidly, causing new secondhand e-cig vape exposures. Primary vaping is associated with immunologic deficits in healthy people, but it is unknown whether e-cigs similarly impacts CF immune function or how it differs from SHSe. Human CF and non-CF blood monocyte-derived macrophages (MDMs) and bronchial epithelial cells (HBECs) were exposed to flavored and unflavored e-cigs. The effect of e-cigs on CFTR expression and function, bacterial killing, cytokine signaling, lipid mediators, and metabolism was measured during treatment with CFTR modulators. E-cigs decreased CFTR expression and function in CF and non-CF MDMs and negated CFTR functional restoration by elexacaftor/tezacaftor/ivacaftor (ETI). E-cigs also negated the restoration of anti-inflammatory PGD2 expression in CF MDMs treated with ETI compared with controls. Flavored but not unflavored e-cigs increased proinflammatory cytokine expression in CF MDMs and e-cigs promoted glycolytic metabolism. E-cigs did not impact bacterial killing. Overall, HBECs were less impacted by e-cigs compared with MDMs. E-cigs reduced macrophage CFTR expression and hindered functional CFTR restoration by CFTR modulators, promoting a glycolytic, proinflammatory state. E-cigs are an emerging public health threat that may limit the efficacy of CFTR modulators in people with CF.NEW & NOTEWORTHY New research reveals that e-cigarettes pose a serious health risk for individuals with cystic fibrosis (CF). Exposure to electronic cigarette (e-cig) vapors decreases CF transmembrane conductance regulator (CFTR) function and undermines the effectiveness of CFTR modulators, potentially worsening inflammation and metabolic responses. This highlights an urgent need for awareness around e-cig use, especially among caregivers and peers of those with CF. E-cigarettes may further complicate the management of this chronic lung disease.

背景:二手烟暴露(SHSe)是囊性纤维化(CF)和其他肺部疾病患者的公共卫生威胁。吸烟降低CFTR通道功能,这是CF的致病缺陷。我们报道了SHSe通过破坏免疫反应和代谢信号恶化CF的呼吸和营养结果。最近,护理人员和同龄人使用电子烟(e-cigs)的人数迅速增加,导致新的二手电子烟暴露。初级电子烟与健康人的免疫缺陷有关,但尚不清楚电子烟是否同样影响CF免疫功能,或者它与SHSe有何不同。方法:将人CF和非CF血液单核细胞来源的巨噬细胞(MDMs)和支气管上皮细胞(HBECs)暴露于有味和无味电子烟中。在使用CFTR调节剂治疗期间,测量电子烟对CFTR表达和功能、细菌杀灭、细胞因子信号传导、脂质介质和代谢的影响。结果:电子烟降低了CF和非CF MDMs中CFTR的表达和功能,并否定了elexextractor /tezacaftor/ivacaftor (ETI)对CFTR功能的恢复。与对照组相比,电子烟也抑制了经ETI治疗的CF MDMs中抗炎PGD2表达的恢复。加味电子烟增加了CF MDMs中促炎细胞因子的表达,并促进了糖酵解代谢。电子烟对细菌杀灭没有影响。总体而言,与mdm相比,HBECs受电子烟的影响较小。结论:电子烟降低巨噬细胞CFTR表达,抑制CFTR调节剂对CFTR功能的恢复,促进糖酵解、促炎状态。电子烟是一种新出现的公共卫生威胁,可能会限制CFTR调节剂对CF患者的疗效。
{"title":"Secondhand vape exposure regulation of CFTR and immune function in cystic fibrosis.","authors":"Benjamin L Wisniewski, Mahesh Shrestha, Dinesh Bojja, Chandra L Shrestha, Chris S Lee, Hazel Ozuna, Rachael E Rayner, Shasha Bai, Estelle Cormet-Boyaka, Susan D Reynolds, Benjamin T Kopp","doi":"10.1152/ajplung.00328.2024","DOIUrl":"10.1152/ajplung.00328.2024","url":null,"abstract":"<p><p>Secondhand smoke exposure (SHSe) is a public health threat for people with cystic fibrosis (CF) and other lung diseases. Primary smoking reduces CF transmembrane conductance regulator (CFTR) channel function, the causative defect in CF. We reported that SHSe worsens respiratory and nutritional outcomes in CF by disrupting immune responses and metabolic signaling. Recently, electronic cigarette (e-cigs) usage by caregivers and peers has increased rapidly, causing new secondhand e-cig vape exposures. Primary vaping is associated with immunologic deficits in healthy people, but it is unknown whether e-cigs similarly impacts CF immune function or how it differs from SHSe. Human CF and non-CF blood monocyte-derived macrophages (MDMs) and bronchial epithelial cells (HBECs) were exposed to flavored and unflavored e-cigs. The effect of e-cigs on CFTR expression and function, bacterial killing, cytokine signaling, lipid mediators, and metabolism was measured during treatment with CFTR modulators. E-cigs decreased CFTR expression and function in CF and non-CF MDMs and negated CFTR functional restoration by elexacaftor/tezacaftor/ivacaftor (ETI). E-cigs also negated the restoration of anti-inflammatory PGD<sub>2</sub> expression in CF MDMs treated with ETI compared with controls. Flavored but not unflavored e-cigs increased proinflammatory cytokine expression in CF MDMs and e-cigs promoted glycolytic metabolism. E-cigs did not impact bacterial killing. Overall, HBECs were less impacted by e-cigs compared with MDMs. E-cigs reduced macrophage CFTR expression and hindered functional CFTR restoration by CFTR modulators, promoting a glycolytic, proinflammatory state. E-cigs are an emerging public health threat that may limit the efficacy of CFTR modulators in people with CF.<b>NEW & NOTEWORTHY</b> New research reveals that e-cigarettes pose a serious health risk for individuals with cystic fibrosis (CF). Exposure to electronic cigarette (e-cig) vapors decreases CF transmembrane conductance regulator (CFTR) function and undermines the effectiveness of CFTR modulators, potentially worsening inflammation and metabolic responses. This highlights an urgent need for awareness around e-cig use, especially among caregivers and peers of those with CF. E-cigarettes may further complicate the management of this chronic lung disease.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":" ","pages":"L324-L333"},"PeriodicalIF":3.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998424","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}
引用次数: 0
Carotid bodies mediate glial cell activation and neuroinflammation in the NTS following long-term intermittent hypoxia: role in cardiorespiratory dysfunction. 颈动脉小体在NTS中长期间歇性缺氧后介导胶质细胞激活和神经炎症:在心肺功能障碍中的作用。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-08 DOI: 10.1152/ajplung.00280.2024
Katherin Pereyra, Esteban Diaz-Jara, Ignacio Bernal-Santander, Sinay Vicencio, Rodrigo Del Rio, Rodrigo Iturriaga

Chronic intermittent hypoxia (CIH), the main feature of obstructive sleep apnea, heightened chemosensory discharges of the carotid body (CB), which contributes to potentiate the ventilatory hypoxic response and elicits hypertension. We aimed to determine 1) whether the persistence of cardiorespiratory alterations found in long-term CIH depends on the inputs from the CB and 2) in what extension the activation of glial cells and neuroinflammation in the caudal region of the nucleus of the solitary tract (NTS) require functional CB chemosensory activity. To evaluate these hypotheses, we exposed male mice to CIH for 60 days. At 50 days of CIH, CBs were denervated and animals were kept in CIH for 10 additional days. At the end of the experiments, we measured arterial blood pressure, breathing regularity, and hypoxic ventilatory response (HVR) and assessed astrocyte and microglia cell activation. Compared to sham treatment, CIH induced hypertension [mean arterial blood pressure (MABP): 83.47 ± 1.39 vs. 95.00 ± 2.18 mmHg] and disordered breathing [irregularity score (IS): 7.77 ± 0.49 vs. 12.56 ± 1.66], increased the HVR [1.69 ± 0.17 vs. 4.31 ± 0.87 change in minute ventilation (ΔV̇e)/min], and produced an early transient activation of astrocytes followed by a late and persistent activation of microglia in the NTS. In addition, CIH increased IL-1β, IL-6, and TNF-α levels in the NTS. Bilateral CB denervation after 50 days of CIH results in the restoration of normal glial cell activation in the NTS, lower levels of IL-6 and TNF-α, and reductions in arterial blood pressure (83.47 ± 1.38 mmHg) and HVR (1.63 ± 0.43 ΔV̇e/min). The present results suggest that CB inputs to the NTS during long-term CIH contribute to maintain the cardiorespiratory alterations and the formation of a neuroinflammatory niche at the NTS by modifying glial cell activity.NEW & NOTEWORTHY Chronic intermittent hypoxia (CIH), a feature of obstructive sleep apnea, causes cardiorespiratory alterations (i.e. hypertension) linked to oxidative stress, inflammation, and sympathoexcitation. In the present study, we highlight the role of enhanced carotid body (CB) chemosensory afferent discharges to the nucleus of the solitary tract (NTS) in long-term CIH-induced cardiorespiratory disorders. Indeed, we provide evidence that supports the notion that increased CB afferent activity contributes to persistent CIH-induced hypertension, likely triggering neuroinflammation in the NTS.

慢性间歇性缺氧(CIH)是阻塞性睡眠呼吸暂停的主要特征,颈动脉体(CB)的化学感觉放电增加,这有助于增强通气性缺氧反应并引起高血压。我们的目的是确定:1)长期CIH中发现的心肺改变的持久性是否依赖于CB的输入;2)孤立束核(NTS)尾侧区域的胶质细胞激活和神经炎症在多大程度上需要功能性CB化学感觉活动。为了评估这些假设,我们将雄性小鼠暴露于CIH 60天。在体外培养50天时,将CBs去神经,动物再在体外培养10天。在实验结束时,我们测量了动脉血压、呼吸规律和缺氧通气反应(HVR),并评估了星形胶质细胞和小胶质细胞的激活情况。与Sham相比,CIH诱导高血压(MABP: 83.47±1.39 vs. 95.00±2.18 mmHg),呼吸障碍(IS: 7.77±0.49 vs. 12.56±1.66),HVR增加(1.69±0.17 vs. 4.31±0.87 ΔVE/min),并在NTS中产生早期短暂性星形胶质细胞激活,随后是晚期和持久的小胶质细胞激活。此外,CIH增加了NTS中IL-1β、IL-6和TNF-α的水平。CIH 50天后,双侧CB去神经支配导致NTS中正常胶质细胞活化恢复,IL-6和TNF-α水平降低,动脉血压(83.47±1.38 mmHg)和HVR(1.63±0.43 ΔVE/min)降低。目前的研究结果表明,在长期体外循环过程中,脑脊液输入NTS有助于通过改变神经胶质细胞活性来维持NTS的心肺改变和神经炎症生态位的形成。
{"title":"Carotid bodies mediate glial cell activation and neuroinflammation in the NTS following long-term intermittent hypoxia: role in cardiorespiratory dysfunction.","authors":"Katherin Pereyra, Esteban Diaz-Jara, Ignacio Bernal-Santander, Sinay Vicencio, Rodrigo Del Rio, Rodrigo Iturriaga","doi":"10.1152/ajplung.00280.2024","DOIUrl":"10.1152/ajplung.00280.2024","url":null,"abstract":"<p><p>Chronic intermittent hypoxia (CIH), the main feature of obstructive sleep apnea, heightened chemosensory discharges of the carotid body (CB), which contributes to potentiate the ventilatory hypoxic response and elicits hypertension. We aimed to determine <i>1</i>) whether the persistence of cardiorespiratory alterations found in long-term CIH depends on the inputs from the CB and <i>2</i>) in what extension the activation of glial cells and neuroinflammation in the caudal region of the nucleus of the solitary tract (NTS) require functional CB chemosensory activity. To evaluate these hypotheses, we exposed male mice to CIH for 60 days. At 50 days of CIH, CBs were denervated and animals were kept in CIH for 10 additional days. At the end of the experiments, we measured arterial blood pressure, breathing regularity, and hypoxic ventilatory response (HVR) and assessed astrocyte and microglia cell activation. Compared to sham treatment, CIH induced hypertension [mean arterial blood pressure (MABP): 83.47 ± 1.39 vs. 95.00 ± 2.18 mmHg] and disordered breathing [irregularity score (IS): 7.77 ± 0.49 vs. 12.56 ± 1.66], increased the HVR [1.69 ± 0.17 vs. 4.31 ± 0.87 change in minute ventilation (ΔV̇e)/min], and produced an early transient activation of astrocytes followed by a late and persistent activation of microglia in the NTS. In addition, CIH increased IL-1β, IL-6, and TNF-α levels in the NTS. Bilateral CB denervation after 50 days of CIH results in the restoration of normal glial cell activation in the NTS, lower levels of IL-6 and TNF-α, and reductions in arterial blood pressure (83.47 ± 1.38 mmHg) and HVR (1.63 ± 0.43 ΔV̇e/min). The present results suggest that CB inputs to the NTS during long-term CIH contribute to maintain the cardiorespiratory alterations and the formation of a neuroinflammatory niche at the NTS by modifying glial cell activity.<b>NEW & NOTEWORTHY</b> Chronic intermittent hypoxia (CIH), a feature of obstructive sleep apnea, causes cardiorespiratory alterations (i.e. hypertension) linked to oxidative stress, inflammation, and sympathoexcitation. In the present study, we highlight the role of enhanced carotid body (CB) chemosensory afferent discharges to the nucleus of the solitary tract (NTS) in long-term CIH-induced cardiorespiratory disorders. Indeed, we provide evidence that supports the notion that increased CB afferent activity contributes to persistent CIH-induced hypertension, likely triggering neuroinflammation in the NTS.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":" ","pages":"L357-L371"},"PeriodicalIF":3.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142942647","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}
引用次数: 0
Distinct single-cell transcriptional profile in CD4+ T-lymphocytes among obese children with asthma.
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-27 DOI: 10.1152/ajplung.00270.2024
Vickram Tejwani, Rulin Wang, Andres Villabona-Rueda, Karthik Suresh, Tianshi David Wu, Ian M Adcock, Nazanin Z Kermani, Joe Zein, Nadia N Hansel, Srinivasan Yegnasubramanian, Meredith C McCormack, Franco R D'Alessio

Obesity is a risk factor for asthma morbidity, associated with less responsiveness to inhaled corticosteroids. CD4+ T cells are central to the immunology of asthma and may contribute to the unique obese asthma phenotype. We sought to characterize the single-cell CD4+ transcriptional profile differences in obese children with asthma compared with normal-weight children with asthma. Eight normal-weight and obese participants with asthma were clinically phenotyped and matched based on asthma control. Peripheral blood (PB) CD4+ T cells were sorted, and single-cell RNA sequencing was conducted. Cell clusters were identified by canonical gene expression and differential gene expression and reactome pathway analysis was applied. The obese PB bulk transcriptomic signature from the U-BIOPRED pediatric cohort was assessed in our cohort as well. Obese children with asthma have a distinct CD4+ transcriptional profile with differential gene expression. There were more activated protein tyrosine phosphate receptor type C (PTPRC)high cells and less PTPRClow in children with obesity. Children with obesity had higher enrichment of the neutrophil degranulation, interleukin-7 (IL-7) receptor, and IL-7-related janus kinase-signal transducer and activator of transcription signaling pathways. Genes previously associated with more severe asthma, IL-32, FKBP5 gene expression, IL-6, and Rho transcriptional signaling, were also enriched in obese children with asthma. Our findings shed insight into the molecular mechanisms underpinning more severe and steroid-resistant asthma among children with obesity. Further investigation is needed to identify potential new therapeutic targets for this group.NEW & NOTEWORTHY This study identified unique contributors to asthma in children with obesity and found novel pathways. Increased expression of IL-7R, IL-32, PARP-1, FKBP5 gene, IL-6, and Rho transcriptional signaling were observed in obese individuals with asthma.

{"title":"Distinct single-cell transcriptional profile in CD4+ T-lymphocytes among obese children with asthma.","authors":"Vickram Tejwani, Rulin Wang, Andres Villabona-Rueda, Karthik Suresh, Tianshi David Wu, Ian M Adcock, Nazanin Z Kermani, Joe Zein, Nadia N Hansel, Srinivasan Yegnasubramanian, Meredith C McCormack, Franco R D'Alessio","doi":"10.1152/ajplung.00270.2024","DOIUrl":"10.1152/ajplung.00270.2024","url":null,"abstract":"<p><p>Obesity is a risk factor for asthma morbidity, associated with less responsiveness to inhaled corticosteroids. CD4+ T cells are central to the immunology of asthma and may contribute to the unique obese asthma phenotype. We sought to characterize the single-cell CD4+ transcriptional profile differences in obese children with asthma compared with normal-weight children with asthma. Eight normal-weight and obese participants with asthma were clinically phenotyped and matched based on asthma control. Peripheral blood (PB) CD4+ T cells were sorted, and single-cell RNA sequencing was conducted. Cell clusters were identified by canonical gene expression and differential gene expression and reactome pathway analysis was applied. The obese PB bulk transcriptomic signature from the U-BIOPRED pediatric cohort was assessed in our cohort as well. Obese children with asthma have a distinct CD4+ transcriptional profile with differential gene expression. There were more activated protein tyrosine phosphate receptor type C (PTPRC)<sup>high</sup> cells and less PTPRC<sup>low</sup> in children with obesity. Children with obesity had higher enrichment of the neutrophil degranulation, interleukin-7 (IL-7) receptor, and IL-7-related janus kinase-signal transducer and activator of transcription signaling pathways. Genes previously associated with more severe asthma, <i>IL-32</i>, <i>FKBP5</i> gene expression, <i>IL-6</i>, and Rho transcriptional signaling, were also enriched in obese children with asthma. Our findings shed insight into the molecular mechanisms underpinning more severe and steroid-resistant asthma among children with obesity. Further investigation is needed to identify potential new therapeutic targets for this group.<b>NEW & NOTEWORTHY</b> This study identified unique contributors to asthma in children with obesity and found novel pathways. Increased expression of IL-7R, IL-32, PARP-1, <i>FKBP5</i> gene, IL-6, and Rho transcriptional signaling were observed in obese individuals with asthma.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":" ","pages":"L372-L378"},"PeriodicalIF":3.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143045562","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}
引用次数: 0
Every breath you take: exploring macrophages and environmental exposures in the lung-a tribute to Dr. Joseph Brain's legacy. 你所做的每一次呼吸:探索巨噬细胞和肺部环境暴露-致敬约瑟夫·布莱恩博士的遗产。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-17 DOI: 10.1152/ajplung.00407.2024
Larissa A Shimoda, Julie A Bastarache, Rodney D Britt, Wolfgang M Kuebler
{"title":"Every breath you take: exploring macrophages and environmental exposures in the lung-a tribute to Dr. Joseph Brain's legacy.","authors":"Larissa A Shimoda, Julie A Bastarache, Rodney D Britt, Wolfgang M Kuebler","doi":"10.1152/ajplung.00407.2024","DOIUrl":"10.1152/ajplung.00407.2024","url":null,"abstract":"","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":" ","pages":"L321-L323"},"PeriodicalIF":3.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998421","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}
引用次数: 0
Shear stress-induced restoration of pulmonary microvascular endothelial barrier function following ischemia reperfusion injury requires VEGFR2 signaling. 缺血再灌注损伤后剪切应力诱导的肺微血管内皮屏障功能恢复需要 VEGFR2 信号。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2025-03-01 Epub Date: 2024-12-19 DOI: 10.1152/ajplung.00200.2024
Don Walsh, Daria S Kostyunina, Aoife Blake, John Boylan, Paul McLoughlin

Normal shear stress produced by blood flow is sensed by the vascular endothelium and required for maintenance of the homeostatic functions of the endothelium in systemic conduit and resistance vessels. Many critical illnesses are characterized by periods of abnormally reduced or absent shear stress in the lung (e.g., hemorrhagic shock, embolism, ischemia reperfusion injury, and lung transplantation) and are complicated by pulmonary edema following reperfusion due to microvascular leak. The role of shear stress in regulating the pulmonary microvascular endothelial barrier in the intact vascular bed has not been previously examined. We tested the hypothesis that, in lungs injured by a period of ischemia and reperfusion (IRI), reduced shear stress contributes to increased pulmonary microvascular endothelial barrier permeability and edema formation. Furthermore, we examined the role of vascular endothelial-derived growth factor receptor 2 (VEGFR2) as a mechanosensor mediating the endothelial response to this altered shear stress. Following IRI, we perfused isolated ventilated mouse lungs with a low viscosity solution (LVS) or a higher, physiological viscosity solution (PVS) at constant flow to produce differing endothelial shear stresses in the intact microcirculation. Lungs perfused with LVS developed pulmonary edema due to increased endothelial permeability whereas those perfused with PVS were protected from edema formation by reduced endothelial permeability. This effect of PVS required normal VEGFR2 mechanoreceptor function. These data show for the first time that shear stress has an important role in restoring endothelial barrier function in the intact pulmonary microcirculation following injury and have important implications for the treatment of pulmonary edema in critically ill patients.NEW & NOTEWORTHY Critical illnesses are frequently complicated by noncardiogenic pulmonary edema. Many such illnesses include periods of reduced blood flow, often accompanied by hemodilution, which together reduce endothelial shear stress. We report that in ischemia-reperfusion injury reduced shear stress contributes to increased permeability of the in situ pulmonary microvascular endothelium and worsens alveolar edema. Restoring shear stress toward normal reduces endothelial permeability and edema formation, an effect that requires the normal mechanoreceptor function of VEGFR2.

血流产生的正常剪切应力被血管内皮感知,这是维持全身导管和阻力血管内皮稳态功能所必需的。许多危重疾病的特征是肺内剪切应力异常减少或不存在(如出血性休克、栓塞、缺血再灌注损伤和肺移植),并伴有微血管泄漏引起的再灌注后肺水肿。在完整的血管床中,剪切应力在调节肺微血管内皮屏障中的作用尚未被研究过。我们验证了一个假设,即在一段时间的缺血再灌注(IRI)损伤的肺中,剪切应力的减少有助于增加肺微血管内皮屏障的通透性和水肿的形成。此外,我们研究了VEGFR2作为机械传感器介导内皮对这种改变的剪切应力的反应的作用。IRI后,我们用低粘度溶液(LVS)或更高的生理粘度溶液(PVS)以恒定流量灌注离体通气小鼠肺,以在完整微循环中产生不同的内皮剪切应力。LVS灌注的肺由于内皮通透性增加而发生肺水肿,而PVS灌注的肺由于内皮通透性降低而免于水肿的形成。这种PVS效应需要正常的VEGFR2机械受体功能。这些数据首次表明,剪切应力在损伤后完整肺微循环中恢复内皮屏障功能具有重要作用,对危重患者肺水肿的治疗具有重要意义。
{"title":"Shear stress-induced restoration of pulmonary microvascular endothelial barrier function following ischemia reperfusion injury requires VEGFR2 signaling.","authors":"Don Walsh, Daria S Kostyunina, Aoife Blake, John Boylan, Paul McLoughlin","doi":"10.1152/ajplung.00200.2024","DOIUrl":"10.1152/ajplung.00200.2024","url":null,"abstract":"<p><p>Normal shear stress produced by blood flow is sensed by the vascular endothelium and required for maintenance of the homeostatic functions of the endothelium in systemic conduit and resistance vessels. Many critical illnesses are characterized by periods of abnormally reduced or absent shear stress in the lung (e.g., hemorrhagic shock, embolism, ischemia reperfusion injury, and lung transplantation) and are complicated by pulmonary edema following reperfusion due to microvascular leak. The role of shear stress in regulating the pulmonary microvascular endothelial barrier in the intact vascular bed has not been previously examined. We tested the hypothesis that, in lungs injured by a period of ischemia and reperfusion (IRI), reduced shear stress contributes to increased pulmonary microvascular endothelial barrier permeability and edema formation. Furthermore, we examined the role of vascular endothelial-derived growth factor receptor 2 (VEGFR2) as a mechanosensor mediating the endothelial response to this altered shear stress. Following IRI, we perfused isolated ventilated mouse lungs with a low viscosity solution (LVS) or a higher, physiological viscosity solution (PVS) at constant flow to produce differing endothelial shear stresses in the intact microcirculation. Lungs perfused with LVS developed pulmonary edema due to increased endothelial permeability whereas those perfused with PVS were protected from edema formation by reduced endothelial permeability. This effect of PVS required normal VEGFR2 mechanoreceptor function. These data show for the first time that shear stress has an important role in restoring endothelial barrier function in the intact pulmonary microcirculation following injury and have important implications for the treatment of pulmonary edema in critically ill patients.<b>NEW & NOTEWORTHY</b> Critical illnesses are frequently complicated by noncardiogenic pulmonary edema. Many such illnesses include periods of reduced blood flow, often accompanied by hemodilution, which together reduce endothelial shear stress. We report that in ischemia-reperfusion injury reduced shear stress contributes to increased permeability of the in situ pulmonary microvascular endothelium and worsens alveolar edema. Restoring shear stress toward normal reduces endothelial permeability and edema formation, an effect that requires the normal mechanoreceptor function of VEGFR2.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":" ","pages":"L389-L404"},"PeriodicalIF":3.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142863000","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}
引用次数: 0
TRAPping the effects of tobacco smoking: the regulation and function of Acp5 expression in lung macrophages.
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2025-02-24 DOI: 10.1152/ajplung.00157.2024
Suzanne H Willems, Shilei Qian, Pernilla Lång, Bjarne E Overtoom, Sina Alimostafazadeh, Rocío Fuentes-Mateos, Gwenda F Vasse, T Anienke van der Veen, Jelmer Vlasma, Marina H De Jager, Victor Guryev, Gyorgy Fejer, Göran Andersson, Barbro N Melgert

Tartrate-resistant acid phosphatase (TRAP, gene Acp5) is highly expressed in alveolar macrophages with proposed roles in lung inflammation and lung fibrosis development. We previously showed that its expression and activity are higher in lung macrophages of smokers and COPD patients, suggesting involvement in smoke-induced lung damage. In this study we explored the function of TRAP and regulation of its different mRNA transcripts (Acp5 201-206) in lung tissue exposed to cigarette smoke to elucidate its function in alveolar macrophages. In mice exposed to cigarette smoke or air for 4-6 weeks, higher Acp5 mRNA expression in lung tissue after smoking was mainly driven by transcript Acp5-202, which originates from macrophages. Expression of Acp5-202 correlated with transcription factors previously found to drive proliferation of macrophages. Treating fetal liver progenitors-derived alveolar-like macrophages (MPI macrophages) with cigarette smoke extract resulted in more proliferation compared to nontreated cells. In contrast, Acp5-deficient MPI macrophages and MPI macrophages treated with a TRAP inhibitor proliferated significantly less than control macrophages. Mechanistically this lack of proliferation after TRAP inhibition was associated with higher presence of phosphorylated β-catenin compared to nontreated controls. Phosphorylation of β-catenin is known to mark it for ubiquitination and degradation by the proteasome, preventing its activity in promoting cell proliferation. In conclusion, our findings provide strong evidence for TRAP stimulating alveolar macrophage proliferation by dephosphorylating β-catenin. By driving proliferation, TRAP likely helps sustain alveolar macrophage populations during smoke exposure, either compensating for their loss due to smoking or increasing their numbers to better manage smoke-induced damage.

{"title":"TRAPping the effects of tobacco smoking: the regulation and function of <i>Acp5</i> expression in lung macrophages.","authors":"Suzanne H Willems, Shilei Qian, Pernilla Lång, Bjarne E Overtoom, Sina Alimostafazadeh, Rocío Fuentes-Mateos, Gwenda F Vasse, T Anienke van der Veen, Jelmer Vlasma, Marina H De Jager, Victor Guryev, Gyorgy Fejer, Göran Andersson, Barbro N Melgert","doi":"10.1152/ajplung.00157.2024","DOIUrl":"https://doi.org/10.1152/ajplung.00157.2024","url":null,"abstract":"<p><p>Tartrate-resistant acid phosphatase (TRAP, gene <i>Acp5</i>) is highly expressed in alveolar macrophages with proposed roles in lung inflammation and lung fibrosis development. We previously showed that its expression and activity are higher in lung macrophages of smokers and COPD patients, suggesting involvement in smoke-induced lung damage. In this study we explored the function of TRAP and regulation of its different mRNA transcripts (Acp5 201-206) in lung tissue exposed to cigarette smoke to elucidate its function in alveolar macrophages. In mice exposed to cigarette smoke or air for 4-6 weeks, higher Acp5 mRNA expression in lung tissue after smoking was mainly driven by transcript Acp5-202, which originates from macrophages. Expression of Acp5-202 correlated with transcription factors previously found to drive proliferation of macrophages. Treating fetal liver progenitors-derived alveolar-like macrophages (MPI macrophages) with cigarette smoke extract resulted in more proliferation compared to nontreated cells. In contrast, <i>Acp5</i>-deficient MPI macrophages and MPI macrophages treated with a TRAP inhibitor proliferated significantly less than control macrophages. Mechanistically this lack of proliferation after TRAP inhibition was associated with higher presence of phosphorylated β-catenin compared to nontreated controls. Phosphorylation of β-catenin is known to mark it for ubiquitination and degradation by the proteasome, preventing its activity in promoting cell proliferation. In conclusion, our findings provide strong evidence for TRAP stimulating alveolar macrophage proliferation by dephosphorylating β-catenin. By driving proliferation, TRAP likely helps sustain alveolar macrophage populations during smoke exposure, either compensating for their loss due to smoking or increasing their numbers to better manage smoke-induced damage.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":" ","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-02-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490531","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}
引用次数: 0
Culture Conditions Differentially Regulate the Inflammatory Niche and Cellular Phenotype of Tracheo-Bronchial Basal Stem Cells.
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2025-02-21 DOI: 10.1152/ajplung.00293.2024
Shubha Murthy, Denise A Seabold, Lalit K Gautam, Adrian M Caceres, Rosemary Sease, Ben A Calvert, Shana M Busch, Aaron Neely, Crystal N Marconett, Amy L Ryan

Bronchial epithelial cells derived from the tracheo-bronchial regions of human airways (HBECs) provide a valuable in vitro model for studying pathological mechanisms and evaluating therapeutics. This cell population comprises a mixed population of basal cells (BCs), the predominant stem cell in airways capable of both self-renewal and functional differentiation. Despite their potential for regenerative medicine, BCs exhibit significant phenotypic variability in culture. To investigate how culture conditions influence BC phenotype and function, we expanded three independent BC isolates in three media: airway epithelial cell growth medium (AECGM), dual-SMAD inhibitor (DSI)-enriched AECGM, and Pneumacult Ex plus (PEx+). Analysis through RNA sequencing, immune assays and impedance measurements revealed that PEx+ media significantly drove cell proliferation and a broad pro-inflammatory phenotype in BCs. In contrast, BCs expanded in AECGM, displayed increased expression of structural and extracellular matrix components at higher passage. AECGM increased expression of some cytokines at high passage, while DSI suppressed inflammation implicating the involvement TGF-β in BC inflammatory processes. Differentiation capacity of BCs declined with time in culture irrespective of expansion media. This was associated with an increase in PLUNC expressing secretory cells in AECGM and PEx+ media consistent with the known immune modulatory role of PLUNC in the airways. These findings highlight the profound impact of media conditions on inflammatory niche established by, and function of, in vitro expanded BCs. The broad pro-inflammatory phenotype driven by PEx+ media, in particular, should be considered in the development of cell-based models for airway diseases and therapeutic application.

{"title":"Culture Conditions Differentially Regulate the Inflammatory Niche and Cellular Phenotype of Tracheo-Bronchial Basal Stem Cells.","authors":"Shubha Murthy, Denise A Seabold, Lalit K Gautam, Adrian M Caceres, Rosemary Sease, Ben A Calvert, Shana M Busch, Aaron Neely, Crystal N Marconett, Amy L Ryan","doi":"10.1152/ajplung.00293.2024","DOIUrl":"10.1152/ajplung.00293.2024","url":null,"abstract":"<p><p>Bronchial epithelial cells derived from the tracheo-bronchial regions of human airways (HBECs) provide a valuable <i>in vitro</i> model for studying pathological mechanisms and evaluating therapeutics. This cell population comprises a mixed population of basal cells (BCs), the predominant stem cell in airways capable of both self-renewal and functional differentiation. Despite their potential for regenerative medicine, BCs exhibit significant phenotypic variability in culture. To investigate how culture conditions influence BC phenotype and function, we expanded three independent BC isolates in three media: airway epithelial cell growth medium (AECGM), dual-SMAD inhibitor (DSI)-enriched AECGM, and Pneumacult Ex plus (PEx+). Analysis through RNA sequencing, immune assays and impedance measurements revealed that PEx+ media significantly drove cell proliferation and a broad pro-inflammatory phenotype in BCs. In contrast, BCs expanded in AECGM, displayed increased expression of structural and extracellular matrix components at higher passage. AECGM increased expression of some cytokines at high passage, while DSI suppressed inflammation implicating the involvement TGF-β in BC inflammatory processes. Differentiation capacity of BCs declined with time in culture irrespective of expansion media. This was associated with an increase in PLUNC expressing secretory cells in AECGM and PEx+ media consistent with the known immune modulatory role of PLUNC in the airways. These findings highlight the profound impact of media conditions on inflammatory niche established by, and function of, <i>in vitro</i> expanded BCs. The broad pro-inflammatory phenotype driven by PEx+ media, in particular, should be considered in the development of cell-based models for airway diseases and therapeutic application.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":" ","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-02-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143472233","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}
引用次数: 0
Epigenetic Age Acceleration in Idiopathic Pulmonary Fibrosis Revealed by DNA Methylation Clocks.
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2025-02-19 DOI: 10.1152/ajplung.00171.2024
Daniel B Kurbanov, Farida Ahangari, Taylor Adams, Ruben De Man, Jessica Tang, Marianne Carlon, Nebal Abu Hussein, Emmanuela Cortesi, Marta Zapata, Laurens De Sadeleer, Wim Wuyts, Bart Vanaudenaerde, Naftali Kaminski, John E McDonough

In this research, we delve into the association between epigenetic aging and idiopathic pulmonary fibrosis (IPF), a debilitating lung disease that progresses over time. Utilizing the Illumina MethylationEPIC array, we assessed DNA methylation levels in donated human lung tissue from IPF patients, categorizing the disease into mild, moderate, and severe stages based on clinical assessments. We employed seven epigenetic clocks to determine age acceleration, which is the discrepancy between biological (epigenetic) and chronological age. Our findings revealed a notable acceleration of biological aging in IPF tissues compared to healthy controls, with four clocks-Horvath's, Hannum's, PhenoAge, and DunedinPACE-showing significant correlations. DunedinPACE, in particular, indicated a more rapid aging process in the more severe regions within the lungs of IPF cases. These results suggest that the biological aging process in IPF is expedited and closely tied to the severity of the disease. The study underscores the potential of DNA methylation as a biomarker for IPF, providing valuable insights into the underlying methylation patterns and the dynamics of epigenetic aging in affected lung tissue. This research supports the broader application of epigenetic clocks in clinical prognosis and highlights the critical role of biological age in the context of medical research and healthcare.

{"title":"Epigenetic Age Acceleration in Idiopathic Pulmonary Fibrosis Revealed by DNA Methylation Clocks.","authors":"Daniel B Kurbanov, Farida Ahangari, Taylor Adams, Ruben De Man, Jessica Tang, Marianne Carlon, Nebal Abu Hussein, Emmanuela Cortesi, Marta Zapata, Laurens De Sadeleer, Wim Wuyts, Bart Vanaudenaerde, Naftali Kaminski, John E McDonough","doi":"10.1152/ajplung.00171.2024","DOIUrl":"https://doi.org/10.1152/ajplung.00171.2024","url":null,"abstract":"<p><p>In this research, we delve into the association between epigenetic aging and idiopathic pulmonary fibrosis (IPF), a debilitating lung disease that progresses over time. Utilizing the Illumina MethylationEPIC array, we assessed DNA methylation levels in donated human lung tissue from IPF patients, categorizing the disease into mild, moderate, and severe stages based on clinical assessments. We employed seven epigenetic clocks to determine age acceleration, which is the discrepancy between biological (epigenetic) and chronological age. Our findings revealed a notable acceleration of biological aging in IPF tissues compared to healthy controls, with four clocks-Horvath's, Hannum's, PhenoAge, and DunedinPACE-showing significant correlations. DunedinPACE, in particular, indicated a more rapid aging process in the more severe regions within the lungs of IPF cases. These results suggest that the biological aging process in IPF is expedited and closely tied to the severity of the disease. The study underscores the potential of DNA methylation as a biomarker for IPF, providing valuable insights into the underlying methylation patterns and the dynamics of epigenetic aging in affected lung tissue. This research supports the broader application of epigenetic clocks in clinical prognosis and highlights the critical role of biological age in the context of medical research and healthcare.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":" ","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-02-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143456823","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}
引用次数: 0
期刊
American journal of physiology. Lung cellular and molecular physiology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1