Pub Date : 2024-11-23DOI: 10.1007/s00395-024-01091-9
Neven Stevic, Alexandre Pinède, Nathan Mewton, Michel Ovize, Laurent Argaud, Sandrine Lecour, Clément Boiteux, Thomas Bochaton, Martin Cour
Ventricular fibrillation (VF)-induced cardiac arrest frequently complicates ST-segment elevation myocardial infarction (STEMI). Although larger infarct sizes (IS) correlate with a higher risk of VF, the influence of VF itself on IS has remained poorly investigated. To address this knowledge gap, we analyzed the effect of VF on IS in patients and two experimental models. From a prospective cohort, 30 STEMI patients with VF were matched 1:2 with STEMI patients without VF on the common determinants of IS. The primary endpoint was IS, assessed using the 48-h area under the curve (AUC) for troponin. We also compared IS in pigs with/without spontaneous VF during STEMI (n = 15/group), and in an isolated rat heart model of myocardial infarction with/without electrically induced VF (n = 7/group). After matching, the patient characteristics, including the area at risk (AR), were similar. IS was 33% lower in the VF group compared to the control group (troponin AUC 1.6 [0.5–3.3] 106 arbitrary units vs. 2.4 [0.9–4.1] 106 arbitrary units; p < 0.05), but infarct scar size (assessed using MRI and ECG) did not differ between the groups at 1 and 6 months. In both experimental models, IS, expressed as a percentage of AR, was lower (p < 0.05) in the VF group than in the control group. When common determinants of IS are comparable, VF occurring prior to myocardial infarction reperfusion appears to be associated with smaller IS. Nevertheless, this finding, observed under specific experimental conditions and in a highly selected group of patients, was not associated with reduced infarct scar size.
心室颤动(VF)引起的心脏骤停经常并发 ST 段抬高型心肌梗死(STEMI)。虽然梗死面积(IS)越大,发生室颤的风险就越高,但室颤本身对 IS 的影响却鲜有研究。为了填补这一知识空白,我们分析了患者和两种实验模型中 VF 对 IS 的影响。在一个前瞻性队列中,30 名有 VF 的 STEMI 患者与没有 VF 的 STEMI 患者就 IS 的共同决定因素进行了 1:2 匹配。主要终点是IS,使用肌钙蛋白的48小时曲线下面积(AUC)进行评估。我们还比较了 STEMI 期间有/无自发 VF 的猪(n = 15/组)和有/无电诱导 VF 的离体大鼠心肌梗死模型(n = 7/组)的 IS 情况。配对后,患者特征(包括危险区域(AR))相似。心房颤动组的 IS 比对照组低 33%(肌钙蛋白 AUC 1.6 [0.5-3.3] 106 个任意单位 vs. 2.4 [0.9-4.1] 106 个任意单位;p < 0.05),但心肌梗死瘢痕大小(通过核磁共振成像和心电图评估)在 1 个月和 6 个月时在两组之间没有差异。在两种实验模型中,以 AR 百分比表示的 IS 在 VF 组均低于对照组(p < 0.05)。当 IS 的共同决定因素具有可比性时,心肌梗死再灌注前发生的 VF 似乎与较小的 IS 有关。然而,这一发现是在特定的实验条件下、在经过严格筛选的患者群体中观察到的,与心肌梗死瘢痕缩小无关:注册(HIBISCUS-STEMI 队列):ClinicalTrials.gov NCT05794022。
{"title":"Effect of ventricular fibrillation on infarct size after myocardial infarction: a translational study","authors":"Neven Stevic, Alexandre Pinède, Nathan Mewton, Michel Ovize, Laurent Argaud, Sandrine Lecour, Clément Boiteux, Thomas Bochaton, Martin Cour","doi":"10.1007/s00395-024-01091-9","DOIUrl":"https://doi.org/10.1007/s00395-024-01091-9","url":null,"abstract":"<p>Ventricular fibrillation (VF)-induced cardiac arrest frequently complicates ST-segment elevation myocardial infarction (STEMI). Although larger infarct sizes (IS) correlate with a higher risk of VF, the influence of VF itself on IS has remained poorly investigated. To address this knowledge gap, we analyzed the effect of VF on IS in patients and two experimental models. From a prospective cohort, 30 STEMI patients with VF were matched 1:2 with STEMI patients without VF on the common determinants of IS. The primary endpoint was IS, assessed using the 48-h area under the curve (AUC) for troponin. We also compared IS in pigs with/without spontaneous VF during STEMI (<i>n</i> = 15/group), and in an isolated rat heart model of myocardial infarction with/without electrically induced VF (<i>n</i> = 7/group). After matching, the patient characteristics, including the area at risk (AR), were similar. IS was 33% lower in the VF group compared to the control group (troponin AUC 1.6 [0.5–3.3] 10<sup>6</sup> arbitrary units vs. 2.4 [0.9–4.1] 10<sup>6</sup> arbitrary units; <i>p</i> < 0.05), but infarct scar size (assessed using MRI and ECG) did not differ between the groups at 1 and 6 months. In both experimental models, IS, expressed as a percentage of AR, was lower (<i>p</i> < 0.05) in the VF group than in the control group. When common determinants of IS are comparable, VF occurring prior to myocardial infarction reperfusion appears to be associated with smaller IS. Nevertheless, this finding, observed under specific experimental conditions and in a highly selected group of patients, was not associated with reduced infarct scar size.</p><p>Registration (HIBISCUS-STEMI cohort): ClinicalTrials.gov NCT05794022.</p>","PeriodicalId":8723,"journal":{"name":"Basic Research in Cardiology","volume":"20 1","pages":""},"PeriodicalIF":9.5,"publicationDate":"2024-11-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142690692","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Immune checkpoint inhibitor (ICI)-associated myocarditis is a rare but potentially fatal immune-related adverse event. Previously, we reported a case of ICI-associated myocarditis with elevated autoantibodies to 4E-binding protein 3 (4E-BP3). Recent studies have suggested that 4E-BP3 may play an important role in tumor development. However, its role in cardiac diseases including myocarditis is unknown. We investigated the role of 4E-BP3 in an autoimmune myocarditis mouse model. Myocarditis was induced in wild-type and 4E-BP3 knockout mice by immunization with murine α-myosin peptide. 4E-BP3 gene expression was upregulated in the heart of myocarditis mouse. We found that genetic deletion of 4E-BP3 attenuated myocardial inflammation, reduced fibrosis area, and improved cardiac function in myocarditis mice. Studies in bone marrow-chimeric mice demonstrated that immune cell-derived 4E-BP3 plays a pivotal role in the pathogenesis of myocarditis. Immune cell transfer experiments revealed that 4E-BP3 deficiency in dendritic cells and CD4+ T cells decreased disease severity in recipient mice. Furthermore, dendritic cells that were deficient in 4E-BP3 exhibited a diminished capacity to produce IL-6 and IL-1β. Naive CD4+ T cells lacking 4E-BP3 had a reduced ability to differentiate into T-helper (Th)1 and Th17 cells. These findings suggest that 4E-BP3 in dendritic cells and CD4+ T cells may be critically involved in the pathogenesis of α-myosin-specific T cell-mediated myocarditis. Thus, 4E-BP3 could be a possible therapeutic target for myocarditis.
免疫检查点抑制剂(ICI)相关心肌炎是一种罕见但可能致命的免疫相关不良事件。此前,我们曾报道过一例 ICI 相关性心肌炎患者,其 4E 结合蛋白 3(4E-BP3)自身抗体升高。最近的研究表明,4E-BP3 可能在肿瘤发生发展中扮演重要角色。然而,它在包括心肌炎在内的心脏疾病中的作用尚不清楚。我们研究了 4E-BP3 在自身免疫性心肌炎小鼠模型中的作用。用鼠α肌球蛋白肽免疫诱导野生型小鼠和4E-BP3基因敲除小鼠患心肌炎。4E-BP3基因在心肌炎小鼠心脏中表达上调。我们发现,基因缺失4E-BP3可减轻心肌炎小鼠的心肌炎症,减少纤维化面积,并改善其心脏功能。对骨髓嵌合小鼠的研究表明,免疫细胞衍生的4E-BP3在心肌炎的发病机制中起着关键作用。免疫细胞转移实验显示,树突状细胞和 CD4+ T 细胞缺乏 4E-BP3 会降低受体小鼠的疾病严重程度。此外,缺乏4E-BP3的树突状细胞产生IL-6和IL-1β的能力减弱。缺乏 4E-BP3 的幼稚 CD4+ T 细胞分化成 Thelper (Th)1 和 Th17 细胞的能力降低。这些研究结果表明,树突状细胞和CD4+ T细胞中的4E-BP3可能与α-肌球蛋白特异性T细胞介导的心肌炎的发病机制密切相关。因此,4E-BP3可能是心肌炎的治疗靶点。
{"title":"4E-BP3 deficiency impairs dendritic cell activation and CD4+ T cell differentiation and attenuates α-myosin-specific T cell-mediated myocarditis in mice","authors":"Siqi Li, Kazuko Tajiri, Zixun Yuan, Yoshiko Murakata, Zonghu Song, Seiya Mizuno, Dongzhu Xu, Nobuyuki Murakoshi","doi":"10.1007/s00395-024-01089-3","DOIUrl":"https://doi.org/10.1007/s00395-024-01089-3","url":null,"abstract":"<p>Immune checkpoint inhibitor (ICI)-associated myocarditis is a rare but potentially fatal immune-related adverse event. Previously, we reported a case of ICI-associated myocarditis with elevated autoantibodies to 4E-binding protein 3 (4E-BP3). Recent studies have suggested that 4E-BP3 may play an important role in tumor development. However, its role in cardiac diseases including myocarditis is unknown. We investigated the role of 4E-BP3 in an autoimmune myocarditis mouse model. Myocarditis was induced in wild-type and 4E-BP3 knockout mice by immunization with murine α-myosin peptide. 4E-BP3 gene expression was upregulated in the heart of myocarditis mouse. We found that genetic deletion of 4E-BP3 attenuated myocardial inflammation, reduced fibrosis area, and improved cardiac function in myocarditis mice. Studies in bone marrow-chimeric mice demonstrated that immune cell-derived 4E-BP3 plays a pivotal role in the pathogenesis of myocarditis. Immune cell transfer experiments revealed that 4E-BP3 deficiency in dendritic cells and CD4<sup>+</sup> T cells decreased disease severity in recipient mice. Furthermore, dendritic cells that were deficient in 4E-BP3 exhibited a diminished capacity to produce IL-6 and IL-1β. Naive CD4<sup>+</sup> T cells lacking 4E-BP3 had a reduced ability to differentiate into T-helper (Th)1 and Th17 cells. These findings suggest that 4E-BP3 in dendritic cells and CD4<sup>+</sup> T cells may be critically involved in the pathogenesis of α-myosin-specific T cell-mediated myocarditis. Thus, 4E-BP3 could be a possible therapeutic target for myocarditis.</p>","PeriodicalId":8723,"journal":{"name":"Basic Research in Cardiology","volume":"38 1","pages":""},"PeriodicalIF":9.5,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142597907","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-08DOI: 10.1007/s00395-024-01088-4
Nabil V. Sayour, Dániel Kucsera, Ayham R. Alhaddad, Viktória É. Tóth, Tamás G. Gergely, Tamás Kovács, Zsombor I. Hegedűs, Márk E. Jakab, Péter Ferdinandy, Zoltán V. Varga
Despite accumulating data on underlying mechanisms, the influence of sex and prevalent cardio-metabolic co-morbidities on the manifestation and severity of immune checkpoint inhibitor (ICI)-induced cardiotoxicity has not been well defined. To elucidate whether sex and prevalent cardio-metabolic co-morbidities affect ICI-induced cardiotoxicity, we randomized 17-month-old male and female mice to receive control diet (CON) or high-fat diet (HFD) + L-NAME—a well-established mouse model of cardio-metabolic co-morbidities—for 17 weeks (n = 5–7), and evaluated markers of T-cell function in the spleen. As expected, HFD + L-NAME significantly increased body- and heart weight, and serum cholesterol levels, and caused no systolic dysfunction, however, led to diastolic dysfunction, cardiomyocyte hypertrophy, and increased fibrosis only in males compared to corresponding CON. Western blot analyses of splenic immune checkpoint protein levels showed differential expression depending on sex and prevalent cardio-metabolic co-morbidities, suggesting T-cell exhaustion in both sexes on HFD + L-NAME, but more pronounced in males. In a sub-study with a similar setup, we tested cardiotoxic manifestations of ICI by treating mice with anti-PD-1 monoclonal antibody (ICI) for the last 2 weeks of diet administration (n = 5–7). After 2 weeks of ICI treatment, cardiac systolic functions significantly decreased in CON, but not in HFD + L-NAME groups of both sexes compared to baseline (before ICI administration). In conclusion, in this exploratory study using aged mice, we describe for the first time that ICI-related systolic dysfunction is diminished in both sexes when obesity and hypercholesterolemia are present, possibly due to obesity-related T-cell exhaustion.
尽管有关潜在机制的数据不断积累,但性别和普遍存在的心脏代谢并发症对免疫检查点抑制剂(ICI)诱导的心脏毒性的表现和严重程度的影响尚未得到很好的界定。为了弄清性别和普遍存在的心血管代谢并发症是否会影响 ICI 诱导的心脏毒性,我们将 17 个月大的雄性和雌性小鼠随机分为对照饮食(CON)和高脂饮食(HFD)+ L-NAME--心血管代谢并发症的成熟小鼠模型--17 周(n = 5-7),并评估了脾脏中 T 细胞功能的标记物。不出所料,HFD + L-NAME 会显著增加体重、心脏重量和血清胆固醇水平,但不会导致收缩功能障碍,不过,与相应的 CON 相比,只有雄性小鼠会导致舒张功能障碍、心肌细胞肥大和纤维化增加。脾脏免疫检查点蛋白水平的 Western 印迹分析显示,不同性别和心血管代谢并发症的表达存在差异,这表明在高密度脂蛋白胆固醇膳食 + L-NAME 条件下,男女均会出现 T 细胞衰竭,但男性更为明显。在一项设置类似的子研究中,我们在小鼠饮食的最后2周用抗PD-1单克隆抗体(ICI)治疗小鼠(n = 5-7),以测试ICI的心脏毒性表现。ICI治疗2周后,与基线(ICI治疗前)相比,CON组的心脏收缩功能明显下降,而HFD + L-NAME组的心脏收缩功能没有明显下降。总之,在这项使用老年小鼠进行的探索性研究中,我们首次描述了当肥胖和高胆固醇血症存在时,ICI 相关的收缩功能障碍在雌雄小鼠中都会减弱,这可能是由于肥胖相关的 T 细胞衰竭所致。
{"title":"Effects of sex and obesity on immune checkpoint inhibition-related cardiac systolic dysfunction in aged mice","authors":"Nabil V. Sayour, Dániel Kucsera, Ayham R. Alhaddad, Viktória É. Tóth, Tamás G. Gergely, Tamás Kovács, Zsombor I. Hegedűs, Márk E. Jakab, Péter Ferdinandy, Zoltán V. Varga","doi":"10.1007/s00395-024-01088-4","DOIUrl":"https://doi.org/10.1007/s00395-024-01088-4","url":null,"abstract":"<p>Despite accumulating data on underlying mechanisms, the influence of sex and prevalent cardio-metabolic co-morbidities on the manifestation and severity of immune checkpoint inhibitor (ICI)-induced cardiotoxicity has not been well defined. To elucidate whether sex and prevalent cardio-metabolic co-morbidities affect ICI-induced cardiotoxicity, we randomized 17-month-old male and female mice to receive control diet (CON) or high-fat diet (HFD) + L-NAME—a well-established mouse model of cardio-metabolic co-morbidities—for 17 weeks (<i>n</i> = 5–7), and evaluated markers of T-cell function in the spleen. As expected, HFD + L-NAME significantly increased body- and heart weight, and serum cholesterol levels, and caused no systolic dysfunction, however, led to diastolic dysfunction, cardiomyocyte hypertrophy, and increased fibrosis only in males compared to corresponding CON. Western blot analyses of splenic immune checkpoint protein levels showed differential expression depending on sex and prevalent cardio-metabolic co-morbidities, suggesting T-cell exhaustion in both sexes on HFD + L-NAME, but more pronounced in males. In a sub-study with a similar setup, we tested cardiotoxic manifestations of ICI by treating mice with anti-PD-1 monoclonal antibody (ICI) for the last 2 weeks of diet administration (<i>n</i> = 5–7). After 2 weeks of ICI treatment, cardiac systolic functions significantly decreased in CON, but not in HFD + L-NAME groups of both sexes compared to baseline (before ICI administration). In conclusion, in this exploratory study using aged mice, we describe for the first time that ICI-related systolic dysfunction is diminished in both sexes when obesity and hypercholesterolemia are present, possibly due to obesity-related T-cell exhaustion.</p>","PeriodicalId":8723,"journal":{"name":"Basic Research in Cardiology","volume":"215 1","pages":""},"PeriodicalIF":9.5,"publicationDate":"2024-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142596479","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-07DOI: 10.1007/s00395-024-01087-5
Rui-Ze Niu, Hong-Yan Xu, Hui Tian, Dan Zhang, Chun-Yu He, Xiao-Lan Li, Yu-Ye Li, Juan He
The heterogeneity of endothelial cells (ECs) across human tissues remains incompletely inventoried. We constructed an atlas of > 210,000 ECs derived from 38 regions across 24 human tissues. Our analysis reveals significant differences in transcriptome, phenotype, metabolism and transcriptional regulation among ECs from various tissues. Notably, arterial, venous, and lymphatic ECs shared more common markers in multiple tissues than capillary ECs, which exhibited higher heterogeneity. This diversity in capillary ECs suggests their greater potential as targets for drug development. ECs from different tissues and vascular beds were found to be associated with specific diseases. Importantly, tissue specificity of EC senescence is more determined by somatic site than by tissue type (e.g. subcutaneus adipose tissue and visceral adipose tissue). Additionally, sex-specific differences in brain EC senescence were observed. Our EC atlas offers valuble resoursce for identifying EC subclusters in single-cell datasets from body tissues or organoids, facilitating the screen of tissue-specific targeted therapies, and serving as a powerful tool for future discoveries.
{"title":"Single-cell transcriptome unveils unique transcriptomic signatures of human organ-specific endothelial cells","authors":"Rui-Ze Niu, Hong-Yan Xu, Hui Tian, Dan Zhang, Chun-Yu He, Xiao-Lan Li, Yu-Ye Li, Juan He","doi":"10.1007/s00395-024-01087-5","DOIUrl":"https://doi.org/10.1007/s00395-024-01087-5","url":null,"abstract":"<p>The heterogeneity of endothelial cells (ECs) across human tissues remains incompletely inventoried. We constructed an atlas of > 210,000 ECs derived from 38 regions across 24 human tissues. Our analysis reveals significant differences in transcriptome, phenotype, metabolism and transcriptional regulation among ECs from various tissues. Notably, arterial, venous, and lymphatic ECs shared more common markers in multiple tissues than capillary ECs, which exhibited higher heterogeneity. This diversity in capillary ECs suggests their greater potential as targets for drug development. ECs from different tissues and vascular beds were found to be associated with specific diseases. Importantly, tissue specificity of EC senescence is more determined by somatic site than by tissue type (e.g. subcutaneus adipose tissue and visceral adipose tissue). Additionally, sex-specific differences in brain EC senescence were observed. Our EC atlas offers valuble resoursce for identifying EC subclusters in single-cell datasets from body tissues or organoids, facilitating the screen of tissue-specific targeted therapies, and serving as a powerful tool for future discoveries.</p>","PeriodicalId":8723,"journal":{"name":"Basic Research in Cardiology","volume":"80 1","pages":""},"PeriodicalIF":9.5,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142594909","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-30DOI: 10.1007/s00395-024-01086-6
Siavash Beikoghli Kalkhoran, Maryna Basalay, Zhenhe He, Pelin Golforoush, Tayeba Roper, Ben Caplin, Alan D. Salama, Sean M. Davidson, Derek M. Yellon
Myocardial ischemia–reperfusion (IR) injury is a major cause of morbidity and mortality in patients with chronic kidney disease (CKD). The most frequently used and representative experimental model is the rat dietary adenine-induced CKD, which leads to CKD-associated CVD. However, the continued intake of adenine is a potential confounding factor. This study investigated cardiovascular dysfunction following brief adenine exposure, CKD development and return to a normal diet. Male Wistar rats received a 0.3% adenine diet for 10 weeks and normal chow for an additional 8 weeks. Kidney function was assessed by urinalysis and histology. Heart function was assessed by echocardiography. Sensitivity to myocardial IR injury was assessed using the isolated perfused rat heart (Langendorff) model. The inflammation profile of rats with CKD was assessed via cytokine ELISA, tissue histology and RNA sequencing. Induction of CKD was confirmed by a significant increase in plasma creatinine and albuminuria. Histology revealed extensive glomerular and tubular damage. Diastolic dysfunction, measured by the reduction of the E/A ratio, was apparent in rats with CKD even following a normal diet. Hearts from rats with CKD had significantly larger infarcts after IR injury. The CKD rats also had statistically higher levels of markers of inflammation including myeloperoxidase, KIM-1 and interleukin-33. RNA sequencing revealed several changes including an increase in inflammatory signaling pathways. In addition, we noted that CKD induced significant cardiac capillary rarefaction. We have established a modified model of adenine-induced CKD, which leads to cardiovascular dysfunction in the absence of adenine. Our observations of capillary rarefaction and inflammation suggest that these may contribute to detrimental cardiovascular outcomes.
{"title":"Investigating the cause of cardiovascular dysfunction in chronic kidney disease: capillary rarefaction and inflammation may contribute to detrimental cardiovascular outcomes","authors":"Siavash Beikoghli Kalkhoran, Maryna Basalay, Zhenhe He, Pelin Golforoush, Tayeba Roper, Ben Caplin, Alan D. Salama, Sean M. Davidson, Derek M. Yellon","doi":"10.1007/s00395-024-01086-6","DOIUrl":"https://doi.org/10.1007/s00395-024-01086-6","url":null,"abstract":"<p>Myocardial ischemia–reperfusion (IR) injury is a major cause of morbidity and mortality in patients with chronic kidney disease (CKD). The most frequently used and representative experimental model is the rat dietary adenine-induced CKD, which leads to CKD-associated CVD. However, the continued intake of adenine is a potential confounding factor. This study investigated cardiovascular dysfunction following brief adenine exposure, CKD development and return to a normal diet. Male Wistar rats received a 0.3% adenine diet for 10 weeks and normal chow for an additional 8 weeks. Kidney function was assessed by urinalysis and histology. Heart function was assessed by echocardiography. Sensitivity to myocardial IR injury was assessed using the isolated perfused rat heart (Langendorff) model. The inflammation profile of rats with CKD was assessed via cytokine ELISA, tissue histology and RNA sequencing. Induction of CKD was confirmed by a significant increase in plasma creatinine and albuminuria. Histology revealed extensive glomerular and tubular damage. Diastolic dysfunction, measured by the reduction of the E/A ratio, was apparent in rats with CKD even following a normal diet. Hearts from rats with CKD had significantly larger infarcts after IR injury. The CKD rats also had statistically higher levels of markers of inflammation including myeloperoxidase, KIM-1 and interleukin-33. RNA sequencing revealed several changes including an increase in inflammatory signaling pathways. In addition, we noted that CKD induced significant cardiac capillary rarefaction. We have established a modified model of adenine-induced CKD, which leads to cardiovascular dysfunction in the absence of adenine. Our observations of capillary rarefaction and inflammation suggest that these may contribute to detrimental cardiovascular outcomes.</p>","PeriodicalId":8723,"journal":{"name":"Basic Research in Cardiology","volume":"15 1","pages":""},"PeriodicalIF":9.5,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142541628","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-18DOI: 10.1007/s00395-024-01083-9
Petra Kleinbongard, Carlos Galán Arriola, Lina Badimon, Veronica Crisostomo, Zoltán Giricz, Mariann Gyöngyösi, Gerd Heusch, Borja Ibanez, Attila Kiss, Dominique P. V. de Kleijn, Bruno K. Podesser, Rafael Ramírez Carracedo, Antonio Rodríguez-Sinovas, Marisol Ruiz-Meana, Francisco M. Sanchez Margallo, Gemma Vilahur, José Luis Zamorano, Carlos Zaragoza, Peter Ferdinandy, Derek J. Hausenloy
Numerous cardioprotective interventions have been reported to reduce myocardial infarct size (IS) in pre-clinical studies. However, their translation for the benefit of patients with acute myocardial infarction (AMI) has been largely disappointing. One reason for the lack of translation is the lack of rigor and reproducibility in pre-clinical studies. To address this, we have established the European IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT) pig AMI network with centralized randomization and blinded core laboratory IS analysis and validated the network with ischemic preconditioning (IPC) as a positive control. Ten sites in the COST Innovators Grant (IG16225) network participated in the IMPACT network. Three sites were excluded from the final analysis through quality control of infarct images and use of pre-defined exclusion criteria. Using a centrally generated randomization list, pigs were allocated to myocardial ischemia/reperfusion (I/R, N = 5/site) or IPC + I/R (N = 5/site). The primary endpoint was IS [% area-at-risk (AAR)], as quantified by triphenyl-tetrazolium-chloride (TTC) staining in a centralized, blinded core laboratory (5 sites), or IS [% left-ventricular mass (LV)], as quantified by a centralized, blinded cardiac magnetic resonance (CMR) core laboratory (2 sites). In pooled analyses, IPC significantly reduced IS when compared to I/R (57 ± 14 versus 32 ± 19 [%AAR] N = 25 pigs/group; p < 0.001; 25 ± 13 versus 14 ± 8 [%LV]; N = 10 pigs/group; p = 0.021). In site-specific analyses, in 4 of the 5 sites, IS was significantly reduced by IPC when compared to I/R when quantified by TTC and in 1 of 2 sites when quantified by CMR. A pig AMI multicenter European network with centralized randomization and core blinded IS analysis was established and validated with the aim to improve the reproducibility of cardioprotective interventions in pre-clinical studies and the translation of cardioprotection for patient benefit.
{"title":"The IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT): multicenter pig study on the effect of ischemic preconditioning","authors":"Petra Kleinbongard, Carlos Galán Arriola, Lina Badimon, Veronica Crisostomo, Zoltán Giricz, Mariann Gyöngyösi, Gerd Heusch, Borja Ibanez, Attila Kiss, Dominique P. V. de Kleijn, Bruno K. Podesser, Rafael Ramírez Carracedo, Antonio Rodríguez-Sinovas, Marisol Ruiz-Meana, Francisco M. Sanchez Margallo, Gemma Vilahur, José Luis Zamorano, Carlos Zaragoza, Peter Ferdinandy, Derek J. Hausenloy","doi":"10.1007/s00395-024-01083-9","DOIUrl":"https://doi.org/10.1007/s00395-024-01083-9","url":null,"abstract":"<p>Numerous cardioprotective interventions have been reported to reduce myocardial infarct size (IS) in pre-clinical studies. However, their translation for the benefit of patients with acute myocardial infarction (AMI) has been largely disappointing. One reason for the lack of translation is the lack of rigor and reproducibility in pre-clinical studies. To address this, we have established the European IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT) pig AMI network with centralized randomization and blinded core laboratory IS analysis and validated the network with ischemic preconditioning (IPC) as a positive control. Ten sites in the COST Innovators Grant (IG16225) network participated in the IMPACT network. Three sites were excluded from the final analysis through quality control of infarct images and use of pre-defined exclusion criteria. Using a centrally generated randomization list, pigs were allocated to myocardial ischemia/reperfusion (I/R, <i>N</i> = 5/site) or IPC + I/R (<i>N</i> = 5/site). The primary endpoint was IS [% area-at-risk (AAR)], as quantified by triphenyl-tetrazolium-chloride (TTC) staining in a centralized, blinded core laboratory (5 sites), or IS [% left-ventricular mass (LV)], as quantified by a centralized, blinded cardiac magnetic resonance (CMR) core laboratory (2 sites). In pooled analyses, IPC significantly reduced IS when compared to I/R (57 ± 14 versus 32 ± 19 [%AAR] <i>N</i> = 25 pigs/group; <i>p</i> < 0.001; 25 ± 13 versus 14 ± 8 [%LV]; <i>N</i> = 10 pigs/group; <i>p</i> = 0.021). In site-specific analyses, in 4 of the 5 sites, IS was significantly reduced by IPC when compared to I/R when quantified by TTC and in 1 of 2 sites when quantified by CMR. A pig AMI multicenter European network with centralized randomization and core blinded IS analysis was established and validated with the aim to improve the reproducibility of cardioprotective interventions in pre-clinical studies and the translation of cardioprotection for patient benefit.</p>","PeriodicalId":8723,"journal":{"name":"Basic Research in Cardiology","volume":"24 1","pages":""},"PeriodicalIF":9.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142448934","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-18DOI: 10.1007/s00395-024-01082-w
Roberto Bolli,Xian-Liang Tang
{"title":"CAESAR lives on with IMPACT: bringing rigor and relevance to cardioprotection research.","authors":"Roberto Bolli,Xian-Liang Tang","doi":"10.1007/s00395-024-01082-w","DOIUrl":"https://doi.org/10.1007/s00395-024-01082-w","url":null,"abstract":"","PeriodicalId":8723,"journal":{"name":"Basic Research in Cardiology","volume":"14 1","pages":""},"PeriodicalIF":9.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142449314","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-15DOI: 10.1007/s00395-024-01085-7
Payel Sen,Jules Hamers,Theresa Sittig,Bachuki Shashikadze,Laura d'Ambrosio,Jan B Stöckl,Susanne Bierschenk,Hengliang Zhang,Chiara d'Alessio,Lotte M Zandbergen,Valerie Pauly,Sebastian Clauss,Eckhard Wolf,Andreas Dendorfer,Thomas Fröhlich,Daphne Merkus
Chronic kidney disease (CKD) predisposes to cardiac remodeling and coronary microvascular dysfunction. Studies in swine identified changes in microvascular structure and function, as well as changes in mitochondrial structure and oxidative stress. However, CKD was combined with metabolic derangement, thereby obscuring the contribution of CKD alone. Therefore, we studied the impact of CKD on the heart and combined proteome studies with measurement of cardiac function and perfusion to identify processes involved in cardiac remodeling in CKD. CKD was induced in swine at 10-12 weeks of age while sham-operated swine served as controls. 5-6 months later, left ventricular (LV) function and coronary flow reserve were measured. LC-MS-MS-based proteomic analysis of LV tissue was performed. LV myocardium and kidneys were histologically examined for interstitial fibrosis and oxidative stress. Renal embolization resulted in mild chronic kidney injury (increased fibrosis and urinary NGAL). PV loops showed LV dilation and increased wall stress, while preload recruitable stroke work was impaired in CKD. Quantitative proteomic analysis of LV myocardium and STRING pre-ranked functional analysis showed enrichments in pathways related to contractile function, reactive oxygen species, and extracellular matrix (ECM) remodeling, which were confirmed histologically and associated with impaired total anti-oxidant capacity. H2O2 exposure of myocardial slices from CKD, but not normal swine, impaired contractile function. Furthermore, in CKD, mitochondrial proteins were downregulated suggesting mitochondrial dysfunction which was associated with higher basal coronary blood flow. Thus, mild CKD induces alterations in mitochondrial proteins along with contractile proteins, oxidative stress and ECM remodeling, that were associated with changes in cardiac function and perfusion.
{"title":"Oxidative stress initiates hemodynamic change in CKD-induced heart disease.","authors":"Payel Sen,Jules Hamers,Theresa Sittig,Bachuki Shashikadze,Laura d'Ambrosio,Jan B Stöckl,Susanne Bierschenk,Hengliang Zhang,Chiara d'Alessio,Lotte M Zandbergen,Valerie Pauly,Sebastian Clauss,Eckhard Wolf,Andreas Dendorfer,Thomas Fröhlich,Daphne Merkus","doi":"10.1007/s00395-024-01085-7","DOIUrl":"https://doi.org/10.1007/s00395-024-01085-7","url":null,"abstract":"Chronic kidney disease (CKD) predisposes to cardiac remodeling and coronary microvascular dysfunction. Studies in swine identified changes in microvascular structure and function, as well as changes in mitochondrial structure and oxidative stress. However, CKD was combined with metabolic derangement, thereby obscuring the contribution of CKD alone. Therefore, we studied the impact of CKD on the heart and combined proteome studies with measurement of cardiac function and perfusion to identify processes involved in cardiac remodeling in CKD. CKD was induced in swine at 10-12 weeks of age while sham-operated swine served as controls. 5-6 months later, left ventricular (LV) function and coronary flow reserve were measured. LC-MS-MS-based proteomic analysis of LV tissue was performed. LV myocardium and kidneys were histologically examined for interstitial fibrosis and oxidative stress. Renal embolization resulted in mild chronic kidney injury (increased fibrosis and urinary NGAL). PV loops showed LV dilation and increased wall stress, while preload recruitable stroke work was impaired in CKD. Quantitative proteomic analysis of LV myocardium and STRING pre-ranked functional analysis showed enrichments in pathways related to contractile function, reactive oxygen species, and extracellular matrix (ECM) remodeling, which were confirmed histologically and associated with impaired total anti-oxidant capacity. H2O2 exposure of myocardial slices from CKD, but not normal swine, impaired contractile function. Furthermore, in CKD, mitochondrial proteins were downregulated suggesting mitochondrial dysfunction which was associated with higher basal coronary blood flow. Thus, mild CKD induces alterations in mitochondrial proteins along with contractile proteins, oxidative stress and ECM remodeling, that were associated with changes in cardiac function and perfusion.","PeriodicalId":8723,"journal":{"name":"Basic Research in Cardiology","volume":"59 1","pages":""},"PeriodicalIF":9.5,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142439657","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-09DOI: 10.1007/s00395-024-01084-8
N. V. Bogert, M. Therre, S. Din, J. Furkel, X. Zhou, I. El-Battrawy, J. Heineke, P. A. Schweizer, I. Akin, H. A. Katus, N. Frey, F. Leuschner, M. H. Konstandin
Cardiac macrophages facilitate electrical conduction through the atrioventricular-node (AV) in mice. A possible role for cardiomyocyte-macrophage coupling on the effect of antiarrhythmic therapy has not been investigated yet. Holter monitoring was conducted in LysMCrexCsf1rLsL−DTR mice (MMDTR) under baseline conditions and after an elctrophysiological stress test by flecainide. In vivo effects were recapitulated in vitro by patch-clamp experiments. The underlying mechanism was characterized by expression and localization analysis of connexin43 (Cx43) and voltage-gated-sodium-channel-5 (Nav1.5). ECG monitoring in MMDTR mice did not show any significant conduction abnormalities but a significantly attenuated flecainide-induced extension of RR- and PP-intervals. Patch-clamp analysis revealed that the application of flecainide to neonatal rat ventricular cardiomyocytes (CMs) changed their resting-membrane-potential (RMP) to more negative potentials and decreased action-potential-duration (APD50). Coupling of macrophages to CMs significantly enhances the effects of flecainide, with a further reduction of the RMP and APD50, mediated by an upregulation of Cx43 and Nav1.5 surface expression. Macrophage depletion in mice does not correlate with cardiac electric conduction delay. Cardiac macrophages amplify the effects of flecainide on electrophysiological properties of cardiomyocytes in vivo and in vitro. Mechanistically, formation of macrophage-cardiomyocyte cell–cell-contacts via Cx43 facilitates the recruitment of Nav1.5 to the cell membrane increasing flecainide effects.
{"title":"Macrophages enhance sodium channel expression in cardiomyocytes","authors":"N. V. Bogert, M. Therre, S. Din, J. Furkel, X. Zhou, I. El-Battrawy, J. Heineke, P. A. Schweizer, I. Akin, H. A. Katus, N. Frey, F. Leuschner, M. H. Konstandin","doi":"10.1007/s00395-024-01084-8","DOIUrl":"https://doi.org/10.1007/s00395-024-01084-8","url":null,"abstract":"<p>Cardiac macrophages facilitate electrical conduction through the atrioventricular-node (AV) in mice. A possible role for cardiomyocyte-macrophage coupling on the effect of antiarrhythmic therapy has not been investigated yet. Holter monitoring was conducted in LysM<sup>Cre</sup>xCsf1r<sup>LsL−DTR</sup> mice (MM<sup>DTR</sup>) under baseline conditions and after an elctrophysiological stress test by flecainide. In vivo effects were recapitulated in vitro by patch-clamp experiments. The underlying mechanism was characterized by expression and localization analysis of connexin43 (Cx43) and voltage-gated-sodium-channel-5 (Na<sub>v</sub>1.5). ECG monitoring in MM<sup>DTR</sup> mice did not show any significant conduction abnormalities but a significantly attenuated flecainide-induced extension of RR- and PP-intervals. Patch-clamp analysis revealed that the application of flecainide to neonatal rat ventricular cardiomyocytes (CMs) changed their resting-membrane-potential (RMP) to more negative potentials and decreased action-potential-duration (APD50). Coupling of macrophages to CMs significantly enhances the effects of flecainide, with a further reduction of the RMP and APD50, mediated by an upregulation of Cx43 and Na<sub>v</sub>1.5 surface expression. Macrophage depletion in mice does not correlate with cardiac electric conduction delay. Cardiac macrophages amplify the effects of flecainide on electrophysiological properties of cardiomyocytes in vivo and in vitro<i>.</i> Mechanistically, formation of macrophage-cardiomyocyte cell–cell-contacts via Cx43 facilitates the recruitment of Na<sub>v</sub>1.5 to the cell membrane increasing flecainide effects.</p>","PeriodicalId":8723,"journal":{"name":"Basic Research in Cardiology","volume":"8 1","pages":""},"PeriodicalIF":9.5,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142385579","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2024-08-07DOI: 10.1007/s00395-024-01073-x
Dragos Andrei Duse, Nathalie Hannelore Schröder, Tanu Srivastava, Marcel Benkhoff, Jens Vogt, Melissa Kim Nowak, Florian Funk, Nina Semleit, Philipp Wollnitzke, Ralf Erkens, Sebastian Kötter, Sven Günther Meuth, Petra Keul, Webster Santos, Amin Polzin, Malte Kelm, Martina Krüger, Joachim Schmitt, Bodo Levkau
The erythrocyte S1P transporter Mfsd2b is also expressed in the heart. We hypothesized that S1P transport by Mfsd2b is involved in cardiac function. Hypertension-induced cardiac remodeling was induced by 4-weeks Angiotensin II (AngII) administration and assessed by echocardiography. Ca2+ transients and sarcomere shortening were examined in adult cardiomyocytes (ACM) from Mfsd2b+/+ and Mfsd2b-/- mice. Tension and force development were measured in skinned cardiac fibers. Myocardial gene expression was determined by real-time PCR, Protein Phosphatase 2A (PP2A) by enzymatic assay, and S1P by LC/MS, respectively. Msfd2b was expressed in the murine and human heart, and its deficiency led to higher cardiac S1P. Mfsd2b-/- mice had regular basal cardiac function but were protected against AngII-induced deterioration of left-ventricular function as evidenced by ~ 30% better stroke volume and cardiac index, and preserved ejection fraction despite similar increases in blood pressure. Mfsd2b-/- ACM exhibited attenuated Ca2+ mobilization in response to isoprenaline whereas contractility was unchanged. Mfsd2b-/- ACM showed no changes in proteins responsible for Ca2+ homeostasis, and skinned cardiac fibers exhibited reduced passive tension generation with preserved contractility. Verapamil abolished the differences in Ca2+ mobilization between Mfsd2b+/+ and Mfsd2b-/- ACM suggesting that S1P inhibits L-type-Ca2+ channels (LTCC). In agreement, intracellular S1P activated the inhibitory LTCC phosphatase PP2A in ACM and PP2A activity was increased in Mfsd2b-/- hearts. We suggest that myocardial S1P protects from hypertension-induced left-ventricular remodeling by inhibiting LTCC through PP2A activation. Pharmacologic inhibition of Mfsd2b may thus offer a novel approach to heart failure.
{"title":"Deficiency of the sphingosine-1-phosphate (S1P) transporter Mfsd2b protects the heart against hypertension-induced cardiac remodeling by suppressing the L-type-Ca<sup>2+</sup> channel.","authors":"Dragos Andrei Duse, Nathalie Hannelore Schröder, Tanu Srivastava, Marcel Benkhoff, Jens Vogt, Melissa Kim Nowak, Florian Funk, Nina Semleit, Philipp Wollnitzke, Ralf Erkens, Sebastian Kötter, Sven Günther Meuth, Petra Keul, Webster Santos, Amin Polzin, Malte Kelm, Martina Krüger, Joachim Schmitt, Bodo Levkau","doi":"10.1007/s00395-024-01073-x","DOIUrl":"10.1007/s00395-024-01073-x","url":null,"abstract":"<p><p>The erythrocyte S1P transporter Mfsd2b is also expressed in the heart. We hypothesized that S1P transport by Mfsd2b is involved in cardiac function. Hypertension-induced cardiac remodeling was induced by 4-weeks Angiotensin II (AngII) administration and assessed by echocardiography. Ca<sup>2+</sup> transients and sarcomere shortening were examined in adult cardiomyocytes (ACM) from Mfsd2b<sup>+/+</sup> and Mfsd2b<sup>-/-</sup> mice. Tension and force development were measured in skinned cardiac fibers. Myocardial gene expression was determined by real-time PCR, Protein Phosphatase 2A (PP2A) by enzymatic assay, and S1P by LC/MS, respectively. Msfd2b was expressed in the murine and human heart, and its deficiency led to higher cardiac S1P. Mfsd2b<sup>-/-</sup> mice had regular basal cardiac function but were protected against AngII-induced deterioration of left-ventricular function as evidenced by ~ 30% better stroke volume and cardiac index, and preserved ejection fraction despite similar increases in blood pressure. Mfsd2b<sup>-/-</sup> ACM exhibited attenuated Ca<sup>2+</sup> mobilization in response to isoprenaline whereas contractility was unchanged. Mfsd2b<sup>-/-</sup> ACM showed no changes in proteins responsible for Ca<sup>2+</sup> homeostasis, and skinned cardiac fibers exhibited reduced passive tension generation with preserved contractility. Verapamil abolished the differences in Ca<sup>2+</sup> mobilization between Mfsd2b<sup>+/+</sup> and Mfsd2b<sup>-/-</sup> ACM suggesting that S1P inhibits L-type-Ca<sup>2+</sup> channels (LTCC). In agreement, intracellular S1P activated the inhibitory LTCC phosphatase PP2A in ACM and PP2A activity was increased in Mfsd2b<sup>-/-</sup> hearts. We suggest that myocardial S1P protects from hypertension-induced left-ventricular remodeling by inhibiting LTCC through PP2A activation. Pharmacologic inhibition of Mfsd2b may thus offer a novel approach to heart failure.</p>","PeriodicalId":8723,"journal":{"name":"Basic Research in Cardiology","volume":" ","pages":"853-868"},"PeriodicalIF":7.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11461684/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141896648","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}