首页 > 最新文献

American Journal of Respiratory Cell and Molecular Biology最新文献

英文 中文
CSP7 Protects Alveolar Epithelial Cells by Targeting p53-Fibrinolytic Pathways During Lung Injuries. CSP7通过靶向肺损伤过程中的p53-纤维蛋白溶解途径保护肺泡上皮细胞
IF 6.4 2区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-13 DOI: 10.1165/rcmb.2023-0453oc
Bijesh Puthusseri,Ashoka Kumar Bhagavath,Daniel Korir,Rashmi Shetty,Gretchen A Johnson,Dorota L Stankowska,Nagarjun V Konduru,Hua Tang,Sudhir Bolla,Gerald J Criner,Nathaniel Marchetti,Durgesh Nandini Das,Sreerama Shetty
Impaired alveolar epithelial regeneration in patients with idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD) is attributed to telomere dysfunction in type II alveolar epithelial cells (A2Cs). Genetic susceptibility, aging, and toxicant exposures, including tobacco smoke (TS), contribute to telomere dysfunction in A2Cs. Here we investigated whether improvement of telomere function plays a role in CSP7-mediated protection of A2Cs against ongoing senescence and apoptosis during bleomycin (BLM)-induced pulmonary fibrosis (PF) as well as alveolar injury caused by chronic TS exposure. We found a significant telomere shortening in A2Cs isolated from IPF and COPD lungs in line with other studies. These cells showed increased p53 in addition to its post-translational modification with induction of activated caspase-3 and β-galactosidase, suggesting a p53-mediated loss of A2C renewal. Further, we found increased expression of SIAH-1, a p53-inducible E3 ubiquitin ligase known to down-regulate telomere repeats binding factor 2 (TRF2). Consistent with the loss of TRF2 and upregulation of TRF1, telomerase reverse transcriptase (TERT) was downregulated in A2Cs. A2Cs from fibrotic lungs of mice either repeatedly instilled with BLM or isolated from chronic TS exposure-induced lung injury model showed reduced telomere length along with induction of p53, PAI-1, SIAH1 and TRF1 as well as loss of TRF2 and TERT, which were reversed in wild-type mice after treatment with CSP7. Interestingly, PAI-1-/- mice, or those lacking microRNA-34a expression in A2Cs, resisted telomere dysfunction, while uPA-/- mice failed to respond to CSP7 treatment, suggesting p53-microRNA-34a feed-forward induction and p53-uPA pathway contributes to telomere dysfunction.
特发性肺纤维化(IPF)和慢性阻塞性肺病(COPD)患者的肺泡上皮再生功能受损,是由于II型肺泡上皮细胞(A2Cs)的端粒功能障碍造成的。遗传易感性、衰老和毒物暴露(包括烟草烟雾(TS))导致了肺泡上皮细胞端粒功能障碍。在此,我们研究了端粒功能的改善是否在 CSP7 介导的保护 A2Cs 免受博莱霉素(BLM)诱导的肺纤维化(PF)以及慢性 TS 暴露引起的肺泡损伤过程中的衰老和凋亡中发挥作用。我们发现,从 IPF 和 COPD 肺中分离出的 A2Cs 中存在明显的端粒缩短现象,这与其他研究结果一致。这些细胞显示出 p53 的增加,以及其翻译后修饰与活化的 caspase-3 和 β-半乳糖苷酶的诱导,这表明 p53 介导的 A2C 更新损失。此外,我们还发现SIAH-1的表达增加,SIAH-1是p53诱导的E3泛素连接酶,已知可下调端粒重复结合因子2(TRF2)。与TRF2的缺失和TRF1的上调相一致,端粒酶逆转录酶(TERT)在A2Cs中也出现了下调。反复灌注 BLM 或从慢性 TS 暴露诱导的肺损伤模型中分离的小鼠纤维化肺中的 A2Cs 显示端粒长度减少,同时诱导 p53、PAI-1、SIAH1 和 TRF1 以及 TRF2 和 TERT 的缺失。有趣的是,PAI-1-/-小鼠或那些在A2Cs中缺乏microRNA-34a表达的小鼠能抵抗端粒功能障碍,而uPA-/-小鼠则对CSP7处理没有反应,这表明p53-microRNA-34a前馈诱导和p53-uPA途径导致了端粒功能障碍。
{"title":"CSP7 Protects Alveolar Epithelial Cells by Targeting p53-Fibrinolytic Pathways During Lung Injuries.","authors":"Bijesh Puthusseri,Ashoka Kumar Bhagavath,Daniel Korir,Rashmi Shetty,Gretchen A Johnson,Dorota L Stankowska,Nagarjun V Konduru,Hua Tang,Sudhir Bolla,Gerald J Criner,Nathaniel Marchetti,Durgesh Nandini Das,Sreerama Shetty","doi":"10.1165/rcmb.2023-0453oc","DOIUrl":"https://doi.org/10.1165/rcmb.2023-0453oc","url":null,"abstract":"Impaired alveolar epithelial regeneration in patients with idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD) is attributed to telomere dysfunction in type II alveolar epithelial cells (A2Cs). Genetic susceptibility, aging, and toxicant exposures, including tobacco smoke (TS), contribute to telomere dysfunction in A2Cs. Here we investigated whether improvement of telomere function plays a role in CSP7-mediated protection of A2Cs against ongoing senescence and apoptosis during bleomycin (BLM)-induced pulmonary fibrosis (PF) as well as alveolar injury caused by chronic TS exposure. We found a significant telomere shortening in A2Cs isolated from IPF and COPD lungs in line with other studies. These cells showed increased p53 in addition to its post-translational modification with induction of activated caspase-3 and β-galactosidase, suggesting a p53-mediated loss of A2C renewal. Further, we found increased expression of SIAH-1, a p53-inducible E3 ubiquitin ligase known to down-regulate telomere repeats binding factor 2 (TRF2). Consistent with the loss of TRF2 and upregulation of TRF1, telomerase reverse transcriptase (TERT) was downregulated in A2Cs. A2Cs from fibrotic lungs of mice either repeatedly instilled with BLM or isolated from chronic TS exposure-induced lung injury model showed reduced telomere length along with induction of p53, PAI-1, SIAH1 and TRF1 as well as loss of TRF2 and TERT, which were reversed in wild-type mice after treatment with CSP7. Interestingly, PAI-1-/- mice, or those lacking microRNA-34a expression in A2Cs, resisted telomere dysfunction, while uPA-/- mice failed to respond to CSP7 treatment, suggesting p53-microRNA-34a feed-forward induction and p53-uPA pathway contributes to telomere dysfunction.","PeriodicalId":7655,"journal":{"name":"American Journal of Respiratory Cell and Molecular Biology","volume":null,"pages":null},"PeriodicalIF":6.4,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142264347","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
Identification of SNHG11 as a Therapeutic Target in Pulmonary Hypertension. 确定 SNHG11 为肺动脉高压的治疗靶点
IF 6.4 2区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-12 DOI: 10.1165/rcmb.2023-0428oc
Huayang Li,Quan Liu,Chiyu Liu,Shunjun Wang,Yitao Zhang,Jinyu Pan,Kaizheng Liu,Suiqing Huang,Tongxin Chu,Liqun Shang,Qingyang Song,Kangni Feng,Zhongkai Wu
Pulmonary hypertension (PH) is a life-threatening condition characterized by pulmonary vascular remodeling and endothelial dysfunction. Current therapies primarily target vasoactive imbalances but often fail to address adverse vascular remodeling. Long non-coding RNA (lncRNA), which are key regulators of various cellular processes, remain underexplored in the context of PH. To investigate the role of lncRNA in PH, we performed a comprehensive analysis using Weighted Gene Co-expression Network Analysis (WGCNA) on the GSE113439 dataset, comprising human lung tissue samples from different PH subtypes. Our analysis identified the lncRNA SNHG11 as consistently downregulated in PH. Functional assays in human pulmonary artery endothelial cells (HPAECs) demonstrated that SNHG11 plays a critical role in modulating inflammation, cell proliferation, apoptosis, and the JAK/STAT and MAPK signaling pathways. Mechanistically, SNHG11 influences the stability of PRPF8, a crucial mRNA spliceosome component, thereby affecting multiple cellular functions beyond splicing. In vivo experiments using a hypoxic rat model showed that knockdown of SNHG11 alleviates PH development and improves right ventricular function. These findings highlight SNHG11 as a key regulator in PH pathogenesis and suggest it as a potential therapeutic target.
肺动脉高压(PH)是一种危及生命的疾病,其特点是肺血管重塑和内皮功能障碍。目前的疗法主要针对血管活性失衡,但往往无法解决不利的血管重塑问题。长非编码 RNA(lncRNA)是各种细胞过程的关键调控因子,但在 PH 方面的研究仍然不足。为了研究lncRNA在PH中的作用,我们使用加权基因共表达网络分析(WGCNA)对GSE113439数据集进行了全面分析,该数据集包括来自不同PH亚型的人类肺组织样本。我们的分析确定了lncRNA SNHG11在PH中持续下调。在人肺动脉内皮细胞(HPAECs)中进行的功能测试表明,SNHG11在调节炎症、细胞增殖、细胞凋亡以及JAK/STAT和MAPK信号通路方面起着关键作用。从机理上讲,SNHG11会影响mRNA剪接体的关键成分PRPF8的稳定性,从而影响剪接以外的多种细胞功能。利用缺氧大鼠模型进行的体内实验表明,敲除 SNHG11 可减轻 PH 的发展并改善右心室功能。这些发现凸显了SNHG11是PH发病机制中的一个关键调控因子,并建议将其作为一个潜在的治疗靶点。
{"title":"Identification of SNHG11 as a Therapeutic Target in Pulmonary Hypertension.","authors":"Huayang Li,Quan Liu,Chiyu Liu,Shunjun Wang,Yitao Zhang,Jinyu Pan,Kaizheng Liu,Suiqing Huang,Tongxin Chu,Liqun Shang,Qingyang Song,Kangni Feng,Zhongkai Wu","doi":"10.1165/rcmb.2023-0428oc","DOIUrl":"https://doi.org/10.1165/rcmb.2023-0428oc","url":null,"abstract":"Pulmonary hypertension (PH) is a life-threatening condition characterized by pulmonary vascular remodeling and endothelial dysfunction. Current therapies primarily target vasoactive imbalances but often fail to address adverse vascular remodeling. Long non-coding RNA (lncRNA), which are key regulators of various cellular processes, remain underexplored in the context of PH. To investigate the role of lncRNA in PH, we performed a comprehensive analysis using Weighted Gene Co-expression Network Analysis (WGCNA) on the GSE113439 dataset, comprising human lung tissue samples from different PH subtypes. Our analysis identified the lncRNA SNHG11 as consistently downregulated in PH. Functional assays in human pulmonary artery endothelial cells (HPAECs) demonstrated that SNHG11 plays a critical role in modulating inflammation, cell proliferation, apoptosis, and the JAK/STAT and MAPK signaling pathways. Mechanistically, SNHG11 influences the stability of PRPF8, a crucial mRNA spliceosome component, thereby affecting multiple cellular functions beyond splicing. In vivo experiments using a hypoxic rat model showed that knockdown of SNHG11 alleviates PH development and improves right ventricular function. These findings highlight SNHG11 as a key regulator in PH pathogenesis and suggest it as a potential therapeutic target.","PeriodicalId":7655,"journal":{"name":"American Journal of Respiratory Cell and Molecular Biology","volume":null,"pages":null},"PeriodicalIF":6.4,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142264350","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
Estrogen Oppositely Regulates Pulmonary Hypertension via METTL3/PFKFB3 Under Normoxia and Hypoxia. 雌激素通过 METTL3/PFKFB3 在正常缺氧和缺氧条件下对肺动脉高压进行相反调控
IF 6.4 2区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-12 DOI: 10.1165/rcmb.2024-0042oc
Xiaosa Li,Jiale Wang,Yuqin Chen,Ping Li,Hao Wen,Xingyan Xu,Jian Wang,Yiming Xu,Yingying Chen,Jiangping Song,Wenju Lu,Dongxing Zhu,Xiaodong Fu
Despite extensive investigation into estrogen's role in pulmonary hypertension (PH) development, its effects-whether beneficial or detrimental-remains contentious. This study aimed to elucidate estrogen's potential role in PH under normoxic and hypoxic conditions. Utilizing norfenfluramine- and hypoxia-induced rat models of PH, the study evaluated the impact of 17β-estradiol (E2) on PH progression. E2 promoted PH development under normoxia while providing protection under hypoxia. Mechanistically, under normoxia, E2 upregulated methyltransferase-like 3 (METTL3) gene transcription and protein via an estrogen response element-dependent pathway, which in turn elevated the m6A methylation and translational efficiency of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3 (PFKFB3) mRNA, leading to increased PFKFB3 protein levels and enhanced proliferation and migration of pulmonary artery smooth muscle cells (PASMCs). Conversely, under hypoxia, E2 downregulated METTL3 transcription through a hypoxia response element-dependent mechanism, driven by elevated hypoxia-induced factor 1α (HIF-1α) levels, resulting in reduced PFKFB3 protein expression and diminished PASMCs proliferation and migration. Both METTL3 and PFKFB3 proteins are upregulated in the pulmonary arteries of patients with PAH. Collectively, these findings suggest that E2 exerts differential effects on PH progression via dual regulation of the METTL3/PFKFB3 protein under normoxic and hypoxic conditions, positioning the METTL3/PFKFB3 protein as a potential therapeutic target for PH treatment.
尽管对雌激素在肺动脉高压(PH)发展中的作用进行了广泛的研究,但其作用--无论是有益还是有害--仍存在争议。本研究旨在阐明雌激素在常氧和缺氧条件下对肺动脉高压的潜在作用。该研究利用去甲芬氟拉明和缺氧诱导的大鼠 PH 模型,评估了 17β-estradiol (E2) 对 PH 进展的影响。在常氧条件下,E2促进了PH的发展,而在低氧条件下则提供了保护。从机理上讲,在常氧条件下,E2通过雌激素反应元件依赖途径上调甲基转移酶样3(METTL3)基因转录和蛋白,进而提高6-磷酸果糖-2-激酶/果糖-2,6-二磷酸酶同工酶3(PFKFB3)mRNA的m6A甲基化和翻译效率,导致PFKFB3蛋白水平升高,增强肺动脉平滑肌细胞(PASMCs)的增殖和迁移。相反,在低氧条件下,E2 通过低氧反应元件依赖性机制下调 METTL3 的转录,而低氧诱导因子 1α(HIF-1α)水平的升高会导致 PFKFB3 蛋白表达减少,并降低 PASMCs 的增殖和迁移。在 PAH 患者的肺动脉中,METTL3 和 PFKFB3 蛋白均上调。总之,这些研究结果表明,E2 在常氧和缺氧条件下通过对 METTL3/PFKFB3 蛋白的双重调控对 PH 的进展产生不同的影响,从而将 METTL3/PFKFB3 蛋白定位为治疗 PH 的潜在治疗靶点。
{"title":"Estrogen Oppositely Regulates Pulmonary Hypertension via METTL3/PFKFB3 Under Normoxia and Hypoxia.","authors":"Xiaosa Li,Jiale Wang,Yuqin Chen,Ping Li,Hao Wen,Xingyan Xu,Jian Wang,Yiming Xu,Yingying Chen,Jiangping Song,Wenju Lu,Dongxing Zhu,Xiaodong Fu","doi":"10.1165/rcmb.2024-0042oc","DOIUrl":"https://doi.org/10.1165/rcmb.2024-0042oc","url":null,"abstract":"Despite extensive investigation into estrogen's role in pulmonary hypertension (PH) development, its effects-whether beneficial or detrimental-remains contentious. This study aimed to elucidate estrogen's potential role in PH under normoxic and hypoxic conditions. Utilizing norfenfluramine- and hypoxia-induced rat models of PH, the study evaluated the impact of 17β-estradiol (E2) on PH progression. E2 promoted PH development under normoxia while providing protection under hypoxia. Mechanistically, under normoxia, E2 upregulated methyltransferase-like 3 (METTL3) gene transcription and protein via an estrogen response element-dependent pathway, which in turn elevated the m6A methylation and translational efficiency of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3 (PFKFB3) mRNA, leading to increased PFKFB3 protein levels and enhanced proliferation and migration of pulmonary artery smooth muscle cells (PASMCs). Conversely, under hypoxia, E2 downregulated METTL3 transcription through a hypoxia response element-dependent mechanism, driven by elevated hypoxia-induced factor 1α (HIF-1α) levels, resulting in reduced PFKFB3 protein expression and diminished PASMCs proliferation and migration. Both METTL3 and PFKFB3 proteins are upregulated in the pulmonary arteries of patients with PAH. Collectively, these findings suggest that E2 exerts differential effects on PH progression via dual regulation of the METTL3/PFKFB3 protein under normoxic and hypoxic conditions, positioning the METTL3/PFKFB3 protein as a potential therapeutic target for PH treatment.","PeriodicalId":7655,"journal":{"name":"American Journal of Respiratory Cell and Molecular Biology","volume":null,"pages":null},"PeriodicalIF":6.4,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142264349","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
Cell-Specific Contribution of IL4 Receptor α Signaling Shapes the Overall Manifestation of Allergic Airway Disease. IL4受体α信号的细胞特异性贡献决定了过敏性气道疾病的整体表现。
IF 6.4 2区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-10 DOI: 10.1165/rcmb.2024-0208oc
Ishita Choudhary,Richa Lamichhane,Dhruthi Singamsetty,Thao Vo,Frank Brombacher,Sonika Patial,Yogesh Saini
IL-4 and IL-13 play a critical role in allergic asthma pathogenesis via their common receptor, i.e., IL4Rα. However, the cell-specific role of IL4Rα in mixed allergens (MA)-induced allergic asthma has remained unclear. Therefore, we aimed to identify the cell-specific contribution of IL4Rα signaling in the manifestation of various pathological outcomes in mice with allergic airway disease. We compared MA-induced pathological outcomes between hematopoietic progenitor cells (HPCs)- or non-HPCs-specific IL4Rα-deficient chimera, myeloid cell-specific IL4Rα-deficient (LysMcre+/+/IL4Rαfl/fl), and airway epithelial cell-specific IL4Rα-deficient (CCSP-Cre+ /IL4Rαfl/fl) mice. Chimeric mice with systemic IL4Rα sufficiency displayed hallmark features of allergic asthma, including eosinophilic and lymphocytic infiltration, type 2 (Th2) cytokine/chemokine production, IgE production, and lung pathology. These features were markedly reduced in chimeric mice with systemic IL4Rα deficiency. Non-HPCs-specific IL4Rα-deficient mice displayed typical inflammatory features of allergic asthma but with markedly reduced mucous cell metaplasia (MCM). Deletion of IL4Rα signaling on airway epithelial cells, a subpopulation within the non-HPC lineage, resulted in almost complete absence of MCM. In contrast, all features of allergic asthma except for MCM and mucin production were mitigated in HPCs-specific IL4Rα-deficient chimeric mice. Deleting IL4Rα signaling in myeloid cells, a subpopulation within the HPC lineage, significantly alleviated MA-induced allergic airway inflammatory responses, but similar to the HPCs-specific IL4Rα-deficient chimeric mice, these mice showed significant MCM and mucin production. Our findings demonstrate that the differential allergen responsiveness seen in mice with HPCs-specific and non-HPCs-specific IL4Rα deficiency is predominantly driven by the absence of IL4Rα in myeloid cells and airway epithelial cells, respectively. Our findings also highlight distinct and mutually exclusive roles of IL4Rα signaling in mediating pathological outcomes within the myeloid and airway epithelial cell compartments.
IL-4和IL-13通过它们的共同受体(即IL4Rα)在过敏性哮喘发病机制中发挥关键作用。然而,IL4Rα在混合过敏原(MA)诱导的过敏性哮喘中的细胞特异性作用仍不清楚。因此,我们旨在确定IL4Rα信号在过敏性气道疾病小鼠各种病理结果的表现中的细胞特异性贡献。我们比较了造血祖细胞(HPCs)或非HPCs特异性IL4Rα缺陷嵌合体、骨髓细胞特异性IL4Rα缺陷(LysMcre+/+/IL4Rαfl/fl)和气道上皮细胞特异性IL4Rα缺陷(CCSP-Cre+ /IL4Rαfl/fl)小鼠的MA诱导病理结果。全身 IL4Rα 充分的嵌合小鼠显示出过敏性哮喘的标志性特征,包括嗜酸性粒细胞和淋巴细胞浸润、2 型(Th2)细胞因子/趋化因子产生、IgE 产生和肺部病理变化。全身性 IL4Rα 缺乏的嵌合小鼠的这些特征明显减少。非 HPCs 特异性 IL4Rα 缺乏的小鼠表现出典型的过敏性哮喘炎症特征,但粘液细胞增生(MCM)明显减少。删除气道上皮细胞(非 HPC 系的一个亚群)上的 IL4Rα 信号导致 MCM 几乎完全消失。相反,在 HPC 特异性 IL4Rα 缺失的嵌合小鼠中,除 MCM 和粘蛋白产生外,过敏性哮喘的所有特征都得到了缓解。删除髓系细胞(HPC 系中的一个亚群)中的 IL4Rα 信号可显著减轻 MA 诱导的过敏性气道炎症反应,但与 HPCs 特异性 IL4Rα 缺失嵌合小鼠类似,这些小鼠表现出明显的 MCM 和粘蛋白生成。我们的研究结果表明,HPCs 特异性和非 HPCs 特异性 IL4Rα 缺陷小鼠对过敏原的不同反应主要是由于髓系细胞和气道上皮细胞中分别缺乏 IL4Rα 所导致的。我们的研究结果还突显了IL4Rα信号在介导髓系细胞和气道上皮细胞病理结果中的不同且相互排斥的作用。
{"title":"Cell-Specific Contribution of IL4 Receptor α Signaling Shapes the Overall Manifestation of Allergic Airway Disease.","authors":"Ishita Choudhary,Richa Lamichhane,Dhruthi Singamsetty,Thao Vo,Frank Brombacher,Sonika Patial,Yogesh Saini","doi":"10.1165/rcmb.2024-0208oc","DOIUrl":"https://doi.org/10.1165/rcmb.2024-0208oc","url":null,"abstract":"IL-4 and IL-13 play a critical role in allergic asthma pathogenesis via their common receptor, i.e., IL4Rα. However, the cell-specific role of IL4Rα in mixed allergens (MA)-induced allergic asthma has remained unclear. Therefore, we aimed to identify the cell-specific contribution of IL4Rα signaling in the manifestation of various pathological outcomes in mice with allergic airway disease. We compared MA-induced pathological outcomes between hematopoietic progenitor cells (HPCs)- or non-HPCs-specific IL4Rα-deficient chimera, myeloid cell-specific IL4Rα-deficient (LysMcre+/+/IL4Rαfl/fl), and airway epithelial cell-specific IL4Rα-deficient (CCSP-Cre+ /IL4Rαfl/fl) mice. Chimeric mice with systemic IL4Rα sufficiency displayed hallmark features of allergic asthma, including eosinophilic and lymphocytic infiltration, type 2 (Th2) cytokine/chemokine production, IgE production, and lung pathology. These features were markedly reduced in chimeric mice with systemic IL4Rα deficiency. Non-HPCs-specific IL4Rα-deficient mice displayed typical inflammatory features of allergic asthma but with markedly reduced mucous cell metaplasia (MCM). Deletion of IL4Rα signaling on airway epithelial cells, a subpopulation within the non-HPC lineage, resulted in almost complete absence of MCM. In contrast, all features of allergic asthma except for MCM and mucin production were mitigated in HPCs-specific IL4Rα-deficient chimeric mice. Deleting IL4Rα signaling in myeloid cells, a subpopulation within the HPC lineage, significantly alleviated MA-induced allergic airway inflammatory responses, but similar to the HPCs-specific IL4Rα-deficient chimeric mice, these mice showed significant MCM and mucin production. Our findings demonstrate that the differential allergen responsiveness seen in mice with HPCs-specific and non-HPCs-specific IL4Rα deficiency is predominantly driven by the absence of IL4Rα in myeloid cells and airway epithelial cells, respectively. Our findings also highlight distinct and mutually exclusive roles of IL4Rα signaling in mediating pathological outcomes within the myeloid and airway epithelial cell compartments.","PeriodicalId":7655,"journal":{"name":"American Journal of Respiratory Cell and Molecular Biology","volume":null,"pages":null},"PeriodicalIF":6.4,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142219606","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
Nicotine-induced ER Stress and ASM Cell Proliferation is Mediated by α7nAChR and Chaperones-RIC-3 and TMEM35. 尼古丁诱导的ER应激和ASM细胞增殖由α7nAChR和伴侣蛋白-RIC-3及TMEM35介导
IF 5.9 2区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-05 DOI: 10.1165/rcmb.2024-0194OC
Niyati A Borkar, Michael A Thompson, Brian Kelley, Barnabas T Shiferaw, Samantha K Hamrick, Sathish Venkatachalem, Y S Prakash, Christina M Pabelick

Nicotine exposure in the context of smoking or vaping worsens airway function. Although commonly thought to exert effects through the peripheral nervous system, we previously showed airway smooth muscle (ASM) expresses nicotinic acetylcholine receptors (nAChRs), particularly alpha7 subtype (α7nAChR) with functional effects on contractility and metabolism. However, the mechanisms of nAChR regulation and downstream effects in ASM are not fully understood. Using human ASM cells from non-asthmatics vs. mild-moderate asthmatics, we tested the hypothesis that nAChR-specific ER chaperones RIC-3 and TMEM35 promote cell surface localization of α7nAChR with downstream influence on its functionality: effects exacerbated by inflammation. We found that mild-moderate asthma and exposure to pro-inflammatory cytokines relevant to asthma promote chaperone and α7nAChR expression in ASM. Downstream, ER stress was linked to nicotine/α7nAChR signaling, where RIC-3 and TMEM35 regulate nicotine-induced ER stress, Ca2+ regulation and ASM cell proliferation. Overall, our data highlights the importance α7nAChR chaperones in mediating and modulating nicotine effects in ASM towards airway contractility and remodeling.

吸烟或吸食电子烟时接触尼古丁会使气道功能恶化。虽然人们通常认为尼古丁是通过外周神经系统产生作用的,但我们之前发现气道平滑肌(ASM)表达尼古丁乙酰胆碱受体(nAChR),尤其是α7亚型(α7nAChR),对收缩力和新陈代谢具有功能性影响。然而,nAChR 在 ASM 中的调节机制和下游效应尚未完全明了。我们利用来自非哮喘患者和轻中度哮喘患者的人类 ASM 细胞,测试了 nAChR 特异性 ER 合子 RIC-3 和 TMEM35 促进 α7nAChR 细胞表面定位并对其功能产生下游影响的假设:炎症会加剧这种影响。我们发现,轻度-中度哮喘和暴露于与哮喘相关的促炎细胞因子会促进 ASM 中伴侣和 α7nAChR 的表达。在下游,ER 压力与烟碱/α7nAChR 信号传导有关,其中 RIC-3 和 TMEM35 可调节烟碱诱导的 ER 压力、Ca2+ 调节和 ASM 细胞增殖。总之,我们的数据强调了α7nAChR伴侣在介导和调节尼古丁在ASM中对气道收缩性和重塑的影响方面的重要性。
{"title":"Nicotine-induced ER Stress and ASM Cell Proliferation is Mediated by α7nAChR and Chaperones-RIC-3 and TMEM35.","authors":"Niyati A Borkar, Michael A Thompson, Brian Kelley, Barnabas T Shiferaw, Samantha K Hamrick, Sathish Venkatachalem, Y S Prakash, Christina M Pabelick","doi":"10.1165/rcmb.2024-0194OC","DOIUrl":"https://doi.org/10.1165/rcmb.2024-0194OC","url":null,"abstract":"<p><p>Nicotine exposure in the context of smoking or vaping worsens airway function. Although commonly thought to exert effects through the peripheral nervous system, we previously showed airway smooth muscle (ASM) expresses nicotinic acetylcholine receptors (nAChRs), particularly alpha7 subtype (α7nAChR) with functional effects on contractility and metabolism. However, the mechanisms of nAChR regulation and downstream effects in ASM are not fully understood. Using human ASM cells from non-asthmatics vs. mild-moderate asthmatics, we tested the hypothesis that nAChR-specific ER chaperones RIC-3 and TMEM35 promote cell surface localization of α7nAChR with downstream influence on its functionality: effects exacerbated by inflammation. We found that mild-moderate asthma and exposure to pro-inflammatory cytokines relevant to asthma promote chaperone and α7nAChR expression in ASM. Downstream, ER stress was linked to nicotine/α7nAChR signaling, where RIC-3 and TMEM35 regulate nicotine-induced ER stress, Ca<sup>2+</sup> regulation and ASM cell proliferation. Overall, our data highlights the importance α7nAChR chaperones in mediating and modulating nicotine effects in ASM towards airway contractility and remodeling.</p>","PeriodicalId":7655,"journal":{"name":"American Journal of Respiratory Cell and Molecular Biology","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142139032","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
Smooth Muscle Cell-Specific LKB1 Protects Against Sugen5416/Hypoxia-Induced Pulmonary Hypertension through Inhibition of BMP4. 平滑肌细胞特异性 LKB1 通过抑制 BMP4 防止 Sugen5416/缺氧诱导的肺动脉高压
IF 5.9 2区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-05 DOI: 10.1165/rcmb.2023-0430OC
Xiaoping Ma, Yan Liu, Lingli Lei, Lin Wang, Qiming Deng, Hanlin Lu, Hongxuan Li, Shuhui Tian, Xiaoteng Qin, Wencheng Zhang, Yuanyuan Sun

Pulmonary hypertension (PH) is a life-threatening syndrome associated with hyperproliferation of pulmonary artery smooth muscle cells (PASMCs), which exhibit similar features to cancer cells. Currently, there is no curative treatment for PH. LKB1 is known as a tumor suppressor gene with an anti-proliferative effect on cancer cells. However, its role and mechanism in the development of PH remain unclear. Gain-and loss-of-function strategies were used to elucidate the mechanisms of LKB1 in regulating the occurrence and progression of PH. Sugen5416/Hypoxia (SuHx) PH model was utilized for in vivo study. We observed not only a decreased expression of LKB1 in the lung vessels of the SuHx mouse model, but also in human pulmonary artery smooth muscle cells (HPASMCs) exposed to hypoxia. Smooth muscle-specific LKB1 knockout significantly aggravated SuHx-induced PH in mice. RNA sequencing analysis revealed a substantial increase in bone morphogenetic protein-4 (BMP4) in the aortas of LKB1SMKO mice compared with controls, identifying BMP4 as a novel target of LKB1. LKB1 knockdown in HPASMCs cultured under hypoxic conditions increased BMP4 protein level and HPASMC proliferation and migration. The co-immunoprecipitation analysis revealed that LKB1 directly modulates BMP4 protein degradation through phosphorylation. Therapeutically, suppressing BMP4 expression in SMCs alleviates PH in LKB1SMKO mice. Our findings demonstrate that LKB1 attenuates PH by enhancing the lysosomal degradation of BMP4, thus suppressing the proliferation and migration of HPASMCs. Modulating LKB1-BMP4 axis in SMC could be a promising therapeutic strategy of PH.

肺动脉高压(PH)是一种危及生命的综合征,与肺动脉平滑肌细胞(PASMC)的过度增殖有关,其特征与癌细胞相似。目前,肺动脉高压尚无根治性治疗方法。众所周知,LKB1 是一种肿瘤抑制基因,具有抗癌细胞增殖的作用。然而,它在PH发病过程中的作用和机制仍不清楚。研究人员采用功能增益和功能缺失策略来阐明 LKB1 在 PH 发生和发展过程中的调控机制。我们利用 Sugen5416/Hypoxia (SuHx) PH 模型进行了体内研究。我们不仅观察到 LKB1 在 SuHx 小鼠模型肺血管中的表达减少,还观察到 LKB1 在暴露于低氧环境的人肺动脉平滑肌细胞(HPASMCs)中的表达减少。平滑肌特异性 LKB1 基因敲除明显加重了 SuHx 诱导的小鼠 PH。RNA 测序分析显示,与对照组相比,LKB1SMKO 小鼠主动脉中的骨形态发生蛋白-4(BMP4)含量大幅增加,这表明 BMP4 是 LKB1 的一个新靶点。在缺氧条件下培养的 HPASMC 中敲除 LKB1 会增加 BMP4 蛋白水平以及 HPASMC 的增殖和迁移。共免疫沉淀分析显示,LKB1通过磷酸化直接调节BMP4蛋白的降解。在治疗上,抑制 SMC 中 BMP4 的表达可缓解 LKB1SMKO 小鼠的 PH。我们的研究结果表明,LKB1 通过增强 BMP4 的溶酶体降解来减轻 PH,从而抑制 HPASMC 的增殖和迁移。调节SMC中的LKB1-BMP4轴可能是治疗PH的一种有前景的策略。
{"title":"Smooth Muscle Cell-Specific LKB1 Protects Against Sugen5416/Hypoxia-Induced Pulmonary Hypertension through Inhibition of BMP4.","authors":"Xiaoping Ma, Yan Liu, Lingli Lei, Lin Wang, Qiming Deng, Hanlin Lu, Hongxuan Li, Shuhui Tian, Xiaoteng Qin, Wencheng Zhang, Yuanyuan Sun","doi":"10.1165/rcmb.2023-0430OC","DOIUrl":"https://doi.org/10.1165/rcmb.2023-0430OC","url":null,"abstract":"<p><p>Pulmonary hypertension (PH) is a life-threatening syndrome associated with hyperproliferation of pulmonary artery smooth muscle cells (PASMCs), which exhibit similar features to cancer cells. Currently, there is no curative treatment for PH. LKB1 is known as a tumor suppressor gene with an anti-proliferative effect on cancer cells. However, its role and mechanism in the development of PH remain unclear. Gain-and loss-of-function strategies were used to elucidate the mechanisms of LKB1 in regulating the occurrence and progression of PH. Sugen5416/Hypoxia (SuHx) PH model was utilized for <i>in vivo</i> study. We observed not only a decreased expression of LKB1 in the lung vessels of the SuHx mouse model, but also in human pulmonary artery smooth muscle cells (HPASMCs) exposed to hypoxia. Smooth muscle-specific LKB1 knockout significantly aggravated SuHx-induced PH in mice. RNA sequencing analysis revealed a substantial increase in bone morphogenetic protein-4 (BMP4) in the aortas of LKB1<sup>SMKO</sup> mice compared with controls, identifying BMP4 as a novel target of LKB1. LKB1 knockdown in HPASMCs cultured under hypoxic conditions increased BMP4 protein level and HPASMC proliferation and migration. The co-immunoprecipitation analysis revealed that LKB1 directly modulates BMP4 protein degradation through phosphorylation. Therapeutically, suppressing BMP4 expression in SMCs alleviates PH in LKB1<sup>SMKO</sup> mice. Our findings demonstrate that LKB1 attenuates PH by enhancing the lysosomal degradation of BMP4, thus suppressing the proliferation and migration of HPASMCs. Modulating LKB1-BMP4 axis in SMC could be a promising therapeutic strategy of PH.</p>","PeriodicalId":7655,"journal":{"name":"American Journal of Respiratory Cell and Molecular Biology","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142139033","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
CircRNA-Cacna1d Plays a Critical Role in Sepsis-induced Lung Injury by Sponging miRNA-185-5p. CircRNA-Cacna1d在脓毒症诱发的肺损伤中通过海绵miRNA-185-5p发挥关键作用
IF 5.9 2区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-05 DOI: 10.1165/rcmb.2024-0067OC
Jiajia Wang, Jinhui Gao, Ling Ding, Xuanzhe Yang, Dong Zheng, Yuanyuan Zeng, Jianjie Zhu, Wei Lei, Cheng Chen, Zeyi Liu, Jian-An Huang

The role of circRNAs in sepsis-induced lung injury is not clear. This study investigated the role and molecular mechanism of a novel circRNA in sepsis-induced lung injury and explored its prognostic value in sepsis patients. In this study, aberrant circRNA expression profiling in lung tissues from mice with sepsis-induced lung injury was analyzed using high-throughput sequencing. CircRNA-Cacna1d was verified by quantitative real-time polymerase chain reaction, and its biological function in sepsis-induced lung injury was validated in vitro and in vivo. The interactions among circRNA-Cacna1d, miRNAs, and their downstream genes were verified. Furthermore, the clinical value of circRNA-Cacna1d in peripheral blood from sepsis patients was also evaluated. We found that circRNA-Cacna1d expression was significantly increased in lung tissues of sepsis mice and microvascular endothelial cells after lipopolysaccharide (LPS) challenge. CircRNA-Cacna1d knockdown alleviated inflammatory response and ameliorated the permeability of vascular endothelium, thereby mitigating sepsis-induced lung injury and significantly improving the survival rate of sepsis mice. Mechanistically, circRNA-Cacna1d directly interacted with miRNA-185-5p and functioned as a miRNA sponge to regulate the RhoA/ROCK1 signaling pathway. The expression level of circRNA-Cacna1d in patients with early sepsis was significantly higher than that in the healthy controls. Higher levels of circRNA-Cacna1d in sepsis patients were associated with increased disease severity and poorer outcomes. In conclusions, circRNA-Cacna1d may play a role in sepsis-induced lung injury by regulating the RhoA/ROCK1 axis by acting as miRNA-185-5p sponge. CircRNA-Cacna1d is a potential therapeutic target for sepsis-induced lung injury and a prognostic biomarker in sepsis.

循环RNA在脓毒症诱发的肺损伤中的作用尚不明确。本研究探讨了一种新型 circRNA 在脓毒症诱导的肺损伤中的作用和分子机制,并探讨了其在脓毒症患者中的预后价值。本研究利用高通量测序技术分析了脓毒症诱发肺损伤小鼠肺组织中异常 circRNA 的表达谱。通过实时定量聚合酶链反应验证了循环RNA-Cacna1d,并在体外和体内验证了其在脓毒症诱导的肺损伤中的生物学功能。研究还验证了 circRNA-Cacna1d、miRNA 及其下游基因之间的相互作用。此外,还评估了脓毒症患者外周血中 circRNA-Cacna1d 的临床价值。我们发现,脂多糖(LPS)挑战后,脓毒症小鼠肺组织和微血管内皮细胞中的 circRNA-Cacna1d 表达明显增加。circRNA-Cacna1d的敲除减轻了炎症反应,改善了血管内皮的通透性,从而减轻了脓毒症诱发的肺损伤,显著提高了脓毒症小鼠的存活率。从机制上看,circRNA-Cacna1d直接与miRNA-185-5p相互作用,并作为miRNA海绵调控RhoA/ROCK1信号通路。早期败血症患者的 circRNA-Cacna1d 表达水平明显高于健康对照组。脓毒症患者体内较高水平的 circRNA-Cacna1d 与疾病严重程度增加和较差的预后有关。总之,circRNA-Cacna1d可能通过作为miRNA-185-5p海绵调节RhoA/ROCK1轴,在脓毒症诱发的肺损伤中发挥作用。循环RNA-Cacna1d是脓毒症诱发肺损伤的潜在治疗靶点,也是脓毒症的预后生物标志物。
{"title":"CircRNA-Cacna1d Plays a Critical Role in Sepsis-induced Lung Injury by Sponging miRNA-185-5p.","authors":"Jiajia Wang, Jinhui Gao, Ling Ding, Xuanzhe Yang, Dong Zheng, Yuanyuan Zeng, Jianjie Zhu, Wei Lei, Cheng Chen, Zeyi Liu, Jian-An Huang","doi":"10.1165/rcmb.2024-0067OC","DOIUrl":"https://doi.org/10.1165/rcmb.2024-0067OC","url":null,"abstract":"<p><p>The role of circRNAs in sepsis-induced lung injury is not clear. This study investigated the role and molecular mechanism of a novel circRNA in sepsis-induced lung injury and explored its prognostic value in sepsis patients. In this study, aberrant circRNA expression profiling in lung tissues from mice with sepsis-induced lung injury was analyzed using high-throughput sequencing. CircRNA-Cacna1d was verified by quantitative real-time polymerase chain reaction, and its biological function in sepsis-induced lung injury was validated <i>in vitro</i> and <i>in vivo</i>. The interactions among circRNA-Cacna1d, miRNAs, and their downstream genes were verified. Furthermore, the clinical value of circRNA-Cacna1d in peripheral blood from sepsis patients was also evaluated. We found that circRNA-Cacna1d expression was significantly increased in lung tissues of sepsis mice and microvascular endothelial cells after lipopolysaccharide (LPS) challenge. CircRNA-Cacna1d knockdown alleviated inflammatory response and ameliorated the permeability of vascular endothelium, thereby mitigating sepsis-induced lung injury and significantly improving the survival rate of sepsis mice. Mechanistically, circRNA-Cacna1d directly interacted with miRNA-185-5p and functioned as a miRNA sponge to regulate the RhoA/ROCK1 signaling pathway. The expression level of circRNA-Cacna1d in patients with early sepsis was significantly higher than that in the healthy controls. Higher levels of circRNA-Cacna1d in sepsis patients were associated with increased disease severity and poorer outcomes. In conclusions, circRNA-Cacna1d may play a role in sepsis-induced lung injury by regulating the RhoA/ROCK1 axis by acting as miRNA-185-5p sponge. CircRNA-Cacna1d is a potential therapeutic target for sepsis-induced lung injury and a prognostic biomarker in sepsis.</p>","PeriodicalId":7655,"journal":{"name":"American Journal of Respiratory Cell and Molecular Biology","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142139021","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
Reply to Fujino et al.: Human Lung Cell Separation Strategies for Translational Research. 回复藤野等人:转化研究中的人类肺细胞分离策略。
IF 5.9 2区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-03 DOI: 10.1165/rcmb.2024-0369LE
Allen Duong, Stephen Juvet, Tereza Martinu
{"title":"Reply to Fujino <i>et al.</i>: Human Lung Cell Separation Strategies for Translational Research.","authors":"Allen Duong, Stephen Juvet, Tereza Martinu","doi":"10.1165/rcmb.2024-0369LE","DOIUrl":"https://doi.org/10.1165/rcmb.2024-0369LE","url":null,"abstract":"","PeriodicalId":7655,"journal":{"name":"American Journal of Respiratory Cell and Molecular Biology","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124573","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
Human Lung Cell Separation Strategies for Translational Research. 转化研究中的人类肺细胞分离策略。
IF 5.9 2区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-03 DOI: 10.1165/rcmb.2024-0338LE
Naoya Fujino, Mitsuhiro Yamada, Takuya Saito, Shuichi Konno, Hisatoshi Sugiura
{"title":"Human Lung Cell Separation Strategies for Translational Research.","authors":"Naoya Fujino, Mitsuhiro Yamada, Takuya Saito, Shuichi Konno, Hisatoshi Sugiura","doi":"10.1165/rcmb.2024-0338LE","DOIUrl":"https://doi.org/10.1165/rcmb.2024-0338LE","url":null,"abstract":"","PeriodicalId":7655,"journal":{"name":"American Journal of Respiratory Cell and Molecular Biology","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124572","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
Endothelialized Bronchioalveolar Lung Organoids Model Endothelial Cell Responses to Injury. 内皮化支气管肺泡有机体模拟内皮细胞对损伤的反应
IF 5.9 2区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-03 DOI: 10.1165/rcmb.2023-0373MA
Anna-Lena Ament, Monika Heiner, Marie Christin Hessler, Ioannis Alexopoulos, Katharina Steeg, Ulrich Gärtner, Ana Ivonne Vazquez-Armendariz, Susanne Herold

Organoid 3D systems are powerful platforms to study development and disease. Recently, the complexity of lung organoid models derived from adult mouse and human stem cells has increased substantially in terms of cellular composition and structural complexity. However, a murine lung organoid system with a clear integrated endothelial compartment is still missing. Here, we describe a novel method that adds another level of intricacy to our published bronchioalveolar lung organoid (BALO) model by microinjection of FACS-sorted lung endothelial cells (ECs) into differentiated organoid cultures. Before microinjection, ECs obtained from the lung homogenate (LH) of young mice expressed typical ECs markers such as CD31 and vascular endothelial (VE)-Cadherin and showed tube formation capacity. Following microinjection, ECs surrounded BALO´s alveolar-like compartment aligning with both alveolar epithelial cells type I (AECI) and type II (AECII), as demonstrated by confocal and electron microscopy. Notably, expression of Car4 and Aplnr was as well detected, suggesting presence of EC microvascular phenotypes in the cultured ECs. Moreover, upon epithelial cell injury by lipopolysaccharides (LPS) and influenza A virus (IV), endothelialized BALO (eBALO) released proinflammatory cytokines leading to the upregulation of the intercellular adhesion molecule 1 (ICAM-1) in ECs. In summary, we characterized for the first time a organoid model that incorporates ECs into the alveolar structures of lung organoids, not only increasing our previous model ́s cellular and structural complexity but also providing a suitable niche to model lung endothelium responses to injury ex vivo.

类器官三维系统是研究发育和疾病的强大平台。最近,由成年小鼠和人类干细胞衍生的肺器官模型在细胞组成和结构复杂性方面都有了大幅提高。然而,具有明确整合内皮区室的小鼠肺器质体系统仍然缺失。在这里,我们描述了一种新方法,通过将FACS分选的肺内皮细胞(ECs)显微注射到已分化的类器官培养物中,为我们已发表的支气管肺泡肺类器官(BALO)模型增加了另一层次的复杂性。显微注射前,从幼年小鼠肺匀浆(LH)中获得的EC表达典型的EC标志物,如CD31和血管内皮(VE)-Cadherin,并显示出管形成能力。共聚焦显微镜和电子显微镜显示,显微注射后,ECs包围了BALO的肺泡样区,与肺泡上皮细胞I型(AECI)和II型(AECII)对齐。值得注意的是,还检测到了 Car4 和 Aplnr 的表达,这表明培养的心血管细胞中存在心血管微血管表型。此外,当上皮细胞受到脂多糖(LPS)和甲型流感病毒(IV)损伤时,内皮化的 BALO(eBALO)会释放促炎细胞因子,导致 ECs 中的细胞间粘附分子 1(ICAM-1)上调。总之,我们首次鉴定了一种将肺内皮细胞纳入肺泡结构的类器官模型,这不仅增加了我们以前模型的细胞和结构复杂性,还为模拟肺内皮细胞对体内损伤的反应提供了一个合适的位置。
{"title":"Endothelialized Bronchioalveolar Lung Organoids Model Endothelial Cell Responses to Injury.","authors":"Anna-Lena Ament, Monika Heiner, Marie Christin Hessler, Ioannis Alexopoulos, Katharina Steeg, Ulrich Gärtner, Ana Ivonne Vazquez-Armendariz, Susanne Herold","doi":"10.1165/rcmb.2023-0373MA","DOIUrl":"https://doi.org/10.1165/rcmb.2023-0373MA","url":null,"abstract":"<p><p>Organoid 3D systems are powerful platforms to study development and disease. Recently, the complexity of lung organoid models derived from adult mouse and human stem cells has increased substantially in terms of cellular composition and structural complexity. However, a murine lung organoid system with a clear integrated endothelial compartment is still missing. Here, we describe a novel method that adds another level of intricacy to our published bronchioalveolar lung organoid (BALO) model by microinjection of FACS-sorted lung endothelial cells (ECs) into differentiated organoid cultures. Before microinjection, ECs obtained from the lung homogenate (LH) of young mice expressed typical ECs markers such as CD31 and vascular endothelial (VE)-Cadherin and showed tube formation capacity. Following microinjection, ECs surrounded BALO´s alveolar-like compartment aligning with both alveolar epithelial cells type I (AECI) and type II (AECII), as demonstrated by confocal and electron microscopy. Notably, expression of Car4 and Aplnr was as well detected, suggesting presence of EC microvascular phenotypes in the cultured ECs. Moreover, upon epithelial cell injury by lipopolysaccharides (LPS) and influenza A virus (IV), endothelialized BALO (eBALO) released proinflammatory cytokines leading to the upregulation of the intercellular adhesion molecule 1 (ICAM-1) in ECs. In summary, we characterized for the first time a organoid model that incorporates ECs into the alveolar structures of lung organoids, not only increasing our previous model ́s cellular and structural complexity but also providing a suitable niche to model lung endothelium responses to injury ex vivo.</p>","PeriodicalId":7655,"journal":{"name":"American Journal of Respiratory Cell and Molecular Biology","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124571","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 Respiratory Cell and Molecular Biology
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