这是用户在 2024-3-12 10:24 为 https://www.science.org/doi/10.1126/science.abn0910?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&... 保存的双语快照页面,由 沉浸式翻译 提供双语支持。了解如何保存?
Full access
Research Article 研究文章
HEART FAILURE 心力衰竭

Liver-heart cross-talk mediated by coagulation factor XI protects against heart failure
凝血因子Xi介导的肝心串扰对心力衰竭的保护作用

Yang Cao https://orcid.org/0000-0003-2086-6314, Yuchen Wang, Zhenqi Zhou https://orcid.org/0000-0001-6560-9704, Calvin Pan https://orcid.org/0000-0002-0752-833X, Ling Jiang https://orcid.org/0000-0002-1003-850X, Zhiqiang Zhou, Yonghong Meng, Sarada Charugundla, Tao Li https://orcid.org/0000-0002-7988-3090, Hooman Allayee https://orcid.org/0000-0002-2384-5239, Marcus M. Seldin, and Aldons J. Lusis https://orcid.org/0000-0001-9013-0228 jlusis@mednet.ucla.eduAuthors Info & Affiliations
曹阳HTTPS://ORCID.ORG/0000-0003-2086-6314,王宇晨,周贞琪HTTPS://ORCID.ORG/0000-0001-6560-9704,潘凯文HTTPS://ORCID.ORG/0000-0002-0752-833X,凌江HTTPS://ORCID.ORG/0000-002-1003-850X,周志强,孟永红,SARADA CHARUGUNDLA,李涛HTTPS://ORCID.ORG/0000-0002-7988-3090,[...],和ALDONS J. LUSIS HTTPS://ORCID.ORG/0000-0001-9013-0228 +2作者作者信息&附属机构
Science 科学
22 Sep 2022 2022年9月22日
Vol 377, Issue 6613 第377期,第6613期
pp. 1399-1406

A liver-to-heart chat 肝胆相照的谈话

It is thought that the liver and the heart share physiological communication, helping to explain why, for example, nonalcoholic fatty liver disease increases the risk of heart failure. By examining a large collection of mouse strains, Cao et al. identified coagulation factor XI as a mediator of such liver-heart cross-talk (see the Perspective by Tong and Hill). In mouse models of diet-induced heart failure with preserved ejection fraction, factor XI was inversely correlated with the extent of diastolic dysfunction, with greater expression of factor XI reducing cardiac fibrosis and inflammation. Factor XI itself was only expressed in the liver, but it had reproducible effects on the heart, which the authors connected to the activity of the SMAD pathway. —YN
人们认为肝脏和心脏共享生理信息,这有助于解释为什么非酒精性脂肪肝会增加心力衰竭的风险。通过研究大量小鼠品系,Cao等人鉴定出凝血因子Xi是这种肝-心相互作用的介导物(参见Tong和Hill的《透视》)。在射血分数保留的饮食诱导心力衰竭小鼠模型中,因子Xi与舒张功能障碍的程度呈负相关,因子Xi表达增加可减少心脏纤维化和炎症。因子Xi本身仅在肝脏中表达,但它对心脏具有可重复的作用,作者将其与SMAD途径的活性联系起来。-YN

Abstract 摘要

Tissue-tissue communication by endocrine factors is a vital mechanism for physiologic homeostasis. A systems genetics analysis of transcriptomic and functional data from a cohort of diverse, inbred strains of mice predicted that coagulation factor XI (FXI), a liver-derived protein, protects against diastolic dysfunction, a key trait of heart failure with preserved ejection fraction. This was confirmed using gain- and loss-of-function studies, and FXI was found to activate the bone morphogenetic protein (BMP)–SMAD1/5 pathway in the heart. The proteolytic activity of FXI is required for the cleavage and activation of extracellular matrix–associated BMP7 in the heart, thus inhibiting genes involved in inflammation and fibrosis. Our results reveal a protective role of FXI in heart injury that is distinct from its role in coagulation.
内分泌因子介导的组织间通讯是维持生理稳态的重要机制。对来自一组不同近交系小鼠的转录组和功能数据的系统遗传学分析预测,凝血因子Xi(FXi)是一种肝源性蛋白,可防止舒张功能障碍,舒张功能障碍是射血分数保留的心力衰竭的关键特征。使用功能获得和丧失研究证实了这一点,发现FXI可激活心脏中的骨形态发生蛋白(BMP)-SMAD 1/5通路。FXI的蛋白水解活性是心脏中细胞外基质相关BMP 7的切割和激活所必需的,从而抑制炎症和纤维化相关基因。我们的研究结果揭示了FXI在心脏损伤中的保护作用,这与其在凝血中的作用不同。
Tissue-tissue cross-talk by endocrine factors, including secreted proteins (1), is a vital mechanism to maintain proper physiologic homeostasis. The heart and the liver display multifaceted interactions (2), and in clinical practice it is common to observe heart diseases affecting the liver and vice versa (3). For instance, nonalcoholic fatty liver disease increases the risk for heart failure with diastolic and systolic dysfunction (4, 5). On the basis of these observations, we hypothesized that secreted proteins may mediate communication between liver and heart. We screened for such endocrine factors using a “systems genetics” approach that integrates natural variation for physiological and clinical traits with global transcriptomics data in cohorts of genetically diverse mice. In our studies, we used a resource consisting of ~100 diverse, inbred strains of mice called the Hybrid Mouse Diversity Panel (HMDP) (6). Among several candidates that were identified was coagulation factor XI (FXI), a protein produced exclusively by liver.
内分泌因子(包括分泌蛋白)引起的组织间串扰(1)是维持适当生理稳态的重要机制。心脏和肝脏表现出多方面的相互作用(2),在临床实践中,经常观察到心脏病影响肝脏,反之亦然(3)。例如,非酒精性脂肪肝会增加心力衰竭伴舒张和收缩功能障碍的风险(4,5)。基于这些观察,我们假设分泌蛋白可能介导肝脏和心脏之间的通讯。我们使用“系统遗传学”方法筛选此类内分泌因子,该方法将生理和临床特征的自然变异与遗传多样性小鼠队列中的全局转录组学数据相结合。在我们的研究中,我们使用了由约100种不同的近交系小鼠组成的资源,称为杂交小鼠多样性小组(HMDP)(6)。 在几个候选者中,确定了凝血因子Xi(FXi),一种专门由肝脏产生的蛋白质。
We validated several of these factors in a mouse model of a common form of heart failure, heart failure with preserved ejection fraction (HFpEF). We reasoned that we were more likely to see an effect if the heart was stressed. HFpEF is characterized by diastolic dysfunction and preserved ejection fraction, which is distinct from heart failure with reduced ejection fraction (HFrEF) (7). HFpEF accounts for half of all cases of heart failure and is associated with multiple comorbidities, including diabetes, hypertension, and restrictive cardiomyopathies (8, 9). In HFpEF, chronic systemic inflammation and metabolic disorders affect not only the myocardium, but also other organs such as the kidneys, lungs, and skeletal muscles. However, little is known about the molecular mechanisms underlying impaired cardiac relaxation and how other organs interact with the heart to regulate the pathophysiology of HFpEF.
我们在一种常见形式的心力衰竭,射血分数保留性心力衰竭(HFpEF)的小鼠模型中验证了这些因素中的几个。我们推断,如果心脏受到压力,我们更有可能看到效果。HFpEF的特征是舒张功能障碍和射血分数保留,这与射血分数降低的心力衰竭(HFrEF)不同(7)。HFpEF占所有心力衰竭病例的一半,并与多种合并症相关,包括糖尿病、高血压和限制性心肌病(8,9)。在HFpEF中,慢性全身性炎症和代谢紊乱不仅影响心肌,还影响其他器官,如肾、肺和骨骼肌。然而,很少有人知道受损的心脏舒张的分子机制,以及其他器官如何与心脏相互作用,以调节HFpEF的病理生理。
We chose to follow up on FXI, which was particularly interesting because not only did it perturb gene expression in the heart, but it also affected diastolic function. In the mouse model of HFpEF, mice overexpressing FXI in liver showed improved diastolic function, whereas FXI-knockout mice were sensitized for diastolic dysfunction. We identified potential pathways by which FX1 affects diastolic function by examining differential gene expression in response to changes in FXI levels. FXI overexpression activated the bone morphogenetic protein (BMP)–SMAD1/5 pathway in the heart. The action of FXI on the heart requires proteolytic activity, because point mutations in its catalytic domain eliminated the effects on BMP signaling and heart function. BMP7 is secreted as an inactive precursor that binds to the extracellular matrix, and our results indicate that it is cleaved by FXI, releasing the active growth factor from the prodomain. We also provide evidence that FXI has a similar function in humans. Our results identify FXI as an endocrine factor that influences heart function, and this is distinct from its role in coagulation.
我们选择对FXI进行随访,这特别有趣,因为它不仅干扰心脏中的基因表达,而且还影响舒张功能。在HFpEF小鼠模型中,肝脏中过表达FXI的小鼠显示舒张功能改善,而FXI敲除小鼠对舒张功能障碍敏感。我们确定了潜在的途径,FX 1影响舒张功能,通过检查差异基因表达的变化,在FXI水平。FXI过表达激活了心脏中的骨形态发生蛋白(BMP)-SMAD 1/5通路。FXI对心脏的作用需要蛋白水解活性,因为其催化结构域中的点突变消除了对BMP信号传导和心脏功能的影响。BMP 7作为结合细胞外基质的无活性前体分泌,并且我们的结果表明它被FXI切割,从前结构域释放活性生长因子。 我们还提供了FXI在人类中具有类似功能的证据。我们的研究结果确定FXI作为影响心脏功能的内分泌因子,这与其在凝血中的作用不同。

Results 结果

Systems genetics screening for potential regulators of liver-heart cross-talk
肝心串扰潜在调控因子的系统遗传学筛选

To identify endocrine circuits mediating liver-heart cross-talk (10), we took advantage of a recently developed bioinformatics approach that uses natural variation to identify correlations between tissues. For this, we used a panel of 100 diverse inbred mouse strains in the HMDP (6, 11). Global transcriptomic data from the heart and the liver were generated across all 100 inbred strains and used to detect the correlations between secreted proteins from the liver and their downstream effects on the heart (Fig. 1A). By assessing the strength of cross-tissue predictions, we generated a list of potential liver-heart mediators (Fig. 1B and table S1). The top-ranked candidates included Igfbp7, Lipc, Emilin1, Lgals9, St6gal1, Ghr, Crlf2, Lcat, and F11. This list revealed several previously described mediators with consistent functions. For instance, insulin-like growth factor-binding protein-7 (Igfbp7) has been reported to be correlated with diastolic function in HFrEF and HFpEF patients (12).
为了识别介导肝脏-心脏串扰的内分泌回路(10),我们利用了最近开发的生物信息学方法,该方法使用自然变异来识别组织之间的相关性。为此,我们在HMDP中使用了一组100种不同的近交系小鼠品系(6,11)。在所有100个近交系中生成来自心脏和肝脏的全局转录组数据,并用于检测来自肝脏的分泌蛋白与其对心脏的下游效应之间的相关性(图1A)。通过评估跨组织预测的强度,我们生成了潜在的肝-心介质列表(图1B和表S1)。排名靠前的候选基因包括Igfbp 7、Lipc、Emilin 1、Lgals 9、St 6 gal 1、Ghr、Crlf 2、Lcat和F11。该列表显示了几个先前描述的具有一致功能的介体。 例如,据报道胰岛素样生长因子结合蛋白-7(Igfbp 7)与HFrEF和HFpEF患者的舒张功能相关(12)。
Fig. 1. Systems genetics analysis of cross-tissue correlations identifies proteins mediating liver-heart cross-talk.
Fig. 1.跨组织相关性的系统遗传学分析鉴定了介导肝-心串扰的蛋白质。
(A) Schematic illustrating the identification of the liver-heart interaction using 100 inbred strains of mice (HMDP). The correlation between the secreted factors (from the liver) and cardiac gene expression (RNA-Seq) was used for liver-heart predictions. This framework identified peptides secreted by the liver and strongly associated with the cardiac gene network. n = 4 to 20 mice for each strain. (B) Distribution of significance score for all liver genes across all heart gene expression in 100 strains (left). List shows the top 20 genes potentially mediating liver-heart communication (right). (C) Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis of F11 expression across indicated tissues in C57BL/6J mice (n = 4). All data are presented as means ± SEM. (D) Pathway enrichment derived from heart genes correlated with liver F11 expression. (E) GWAS loci for indicated clinical traits in human populations. The GWAS catalog and PhenoScanner databases consist of human genotype-phenotype associations from publicly available genetic association studies.
(A)使用100只近交系小鼠(HMDP)鉴定肝-心相互作用的示意图。分泌因子(来自肝脏)和心脏基因表达(RNA-Seq)之间的相关性用于肝-心预测。该框架确定了由肝脏分泌的肽,并与心脏基因网络密切相关。每个品系n = 4至20只小鼠。(B)100个菌株中所有肝脏基因在所有心脏基因表达中的显著性评分分布(左)。列表显示了前20个可能介导肝-心通信的基因(右)。(C)C57 BL/6 J小鼠(n = 4)中指定组织中F11表达的定量逆转录聚合酶链反应(qRT-PCR)分析。所有数据均以平均值± SEM表示。(D)来自心脏基因的途径富集与肝脏F11表达相关。(E)人群中指示临床特征的GWAS基因座。 GWAS目录和PhenoScanner数据库由来自公开遗传关联研究的人类基因型-表型关联组成。
Open in viewer

FXI protects against diastolic dysfunction, fibrosis, and inflammation in a mouse model of HFpEF
FXI在HFpEF小鼠模型中保护舒张功能障碍、纤维化和炎症

On the basis of their specific expression in the liver, data from the literature, and functional annotation, we selected Hgfac, C8g, and F11 as candidate mediators of liver-heart communication (Fig. 1B). To determine whether these factors have clinically relevant effects on the heart, we examined them in a mouse model of HFpEF, which is characterized by diastolic dysfunction. We overexpressed these genes individually in the livers of C57BL/6J male mice with an adeno-associated virus serotype 8 (AAV8) vector carrying target genes or green fluorescent protein (GFP) control and directed by the liver-specific thyroid hormone-binding globulin promoter (fig. S1A). After AAV8 injection, mice were subjected to a “two-hit” HFpEF model induced by a combination of high-fat diet (HFD) and inhibition of nitric oxide synthase using Nω-nitrol-arginine methyl ester (l-NAME) (13), followed by assessment of cardiac functions (fig. S1B). After 7 weeks of HFD + l-NAME feeding, mice developed heart failure phenotypes that recapitulated the clinical symptoms of HFpEF, including diastolic dysfunction [increased E/A ratio, E/eʹ ratio, left ventricular (LV) mass, heart weight, and lung weight], metabolic disorders (increased body weight, fat mass, plasma lipids, and glucose intolerance), exercise intolerance (reduced running distance), and preserved LV ejection fraction (LVEF) (fig. S1, C to N).
基于它们在肝脏中的特异性表达、来自文献的数据和功能注释,我们选择Hgfac、C8 g和F11作为肝-心通讯的候选介质(图1B)。为了确定这些因素是否对心脏有临床相关影响,我们在HFpEF小鼠模型中对其进行了检查,HFpEF的特征在于舒张功能障碍。我们在C57 BL/6 J雄性小鼠的肝脏中分别过表达这些基因,使用携带靶基因的腺相关病毒血清型8(AAV 8)载体或绿色荧光蛋白(GFP)对照,并由肝脏特异性甲状腺肿结合球蛋白启动子指导(图S1 A)。在AAV 8注射后,使小鼠经受通过高脂肪饮食(HFD)和使用Nω-硝基-精氨酸甲酯(I-NAME)抑制一氧化氮合酶的组合诱导的“两次打击”HFpEF模型(13),随后评估心脏功能(图S1 B)。 在HFD + l-NAME喂养7周后,小鼠发展出心力衰竭表型,其概括了HFpEF的临床症状,包括舒张功能障碍[增加的E/A比率、E/E比值、左心室(LV)质量、心脏重量和肺重量]、代谢紊乱[增加的E/A比率、E/E比值、左心室(LV)质量、心脏重量和肺重量]、代谢紊乱[增加的E/A比率、E/E比值、左心室(LV)质量、心脏重量和肺重量]、代谢紊乱[增加的E/A比率、E/E比值、左心室(LV)质量和肺重量]。(体重、脂肪量、血脂和葡萄糖耐受不良增加)、运动不耐受(减少的跑步距离)和保留的LV射血分数(LVEF)(图S1,C至N)。
We observed that when overexpressed, liver-derived hepatocyte growth factor activator (HGFAC) increased LV mass and complement C8 gamma chain (C8G) decreased heart weight in the model of HFpEF (figs. S2 and S3). However, we focused on the other top candidate, FXI, because it had additional effects on several HFpEF traits, including diastolic function. FXI acts downstream of FXII (14, 15) and triggers the middle phase of the intrinsic pathway of blood coagulation by activating FIX. Like HGFAC and C8G, FXI is also exclusively expressed in the liver (Fig. 1C and fig. S4, A and B). Furthermore, on the basis of associations with heart transcript levels in the HMDP, FXI was predicted to be strongly correlated with critical pathways in the heart and a number of clinical traits related to HFpEF (Fig. 1D and fig. S4C). In addition, human genome-wide association studies (GWAS) revealed that genetic loci encompassing the F11 gene were associated with the total cholesterol and BMP7 levels (Fig. 1E and tables S2 and S3) (16). These data suggested a potential role of FXI in heart failure.
我们观察到,在HFpEF模型中,当过表达时,肝源性肝细胞生长因子激活剂(HGFAC)增加LV质量,补体C8 γ链(C8G)降低心脏重量(图1A和1B)。S2和S3)。然而,我们专注于另一个最佳候选药物FXI,因为它对几个HFpEF特征有额外的影响,包括舒张功能。FXI作用于FXII的下游(14,15),并通过激活FIX触发凝血内源性途径的中间阶段。与HGFAC和C8 G一样,FXI也仅在肝脏中表达(图1C和图S4,A和B)。此外,基于与HMDP中心脏转录物水平的相关性,预测FXI与心脏中的关键途径和与HFpEF相关的许多临床特征强烈相关(图1D和图S4C)。此外,人类全基因组关联研究(GWAS)显示,包含F11基因的遗传基因座与总胆固醇和BMP 7水平相关(图1)。 表1E和表S2和S3)(16)。这些数据表明FXI在心力衰竭中的潜在作用。
We observed reduced plasma FXI in the mice on HFD + l-NAME relative to those given a chow diet (fig. S4, D and E). We then induced the HFpEF model in 30 genetically diverse, inbred strains of mice, a subset of HMDP, to examine the association between plasma FXI and diastolic dysfunction in the context of naturally occurring variation. We found that FXI levels were inversely correlated with diastolic dysfunction after feeding the HFpEF diet (Fig. 2A). Taken together, these results suggest that FXI may protect against diastolic dysfunction.
我们观察到相对于给予普通食物的小鼠,HFD + l-NAME的小鼠中血浆FXI减少(图S4、D和E)。然后,我们在30个遗传多样的近交系小鼠(HMDP的一个子集)中诱导HFpEF模型,以检查血浆FXI和舒张功能障碍之间的相关性。我们发现,喂食HFpEF饮食后,FXI水平与舒张功能障碍呈负相关(图2A)。综上所述,这些结果表明,FXI可预防舒张功能障碍。
Fig. 2. FXI overexpression reverses HFpEF-induced diastolic dysfunction, inflammation, and fibrosis.
图二. FXI过表达逆转HFpEF诱导的舒张功能障碍、炎症和纤维化。
(A) Thirty inbred strains of male mice were subjected to HFD + l-NAME to induce HFpEF. Plasma FXI concentrations and diastolic function (E/eʹ ratio) were assessed after 7 weeks of feeding. Plasma FXI concentrations were inversely correlated with diastolic dysfunction. (B to L) C57BL/6J male mice were injected with AAV8 containing the cDNA sequence for GFP or F11 and then fed with HFD + l-NAME for 7 weeks. Western blotting shows liver FXI protein (B), plasma FXI concentrations (C), E/A ratio (D), E/eʹ ratio (E), representative images of echocardiography (F), LVEF (G), heart weight/tibia length ratio (H), lung weight [wet/dry ratio (I)], running distance (J), thrombin-antithrombin complexes [TAT (K)], and relative mRNA expression of indicated genes in the heart (L). n = 4 for chow in (D) to (H); in other panels, n = 8 to 10. (M and N) C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 were on HFD + l-NAME for 7 weeks (n = 5). Representative images of immunohistochemistry staining (M) and quantification of positive cells (N) showing inflammatory cell infiltration in the heart tissue. (O and P) C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 were given a chow diet or HFD + l-NAME for 7 weeks (n = 5). Representative images of Masson’s trichrome staining (O) and quantification (P) show fibrosis in the heart tissue. Each point represents a mouse. All data are presented as means ± SEM. ns, not significant. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 by two-way ANOVA [(D) to (J) and (P)] or by Student’s t test [(B) and (C) and (K) to (N)]. For (A) to (C) and (K) to (N), all mice were on HFD + l-NAME. LYM, lymphocytes; MONO, monocytes; GRAN, granulocytes.
(A)对30个近交系雄性小鼠进行HFD + l-NAME诱导HFpEF。喂养7周后评估血浆FXI浓度和舒张功能(E/e β比值)。血浆FXI浓度与舒张功能障碍呈负相关。(B至L)用含有GFP或F11的cDNA序列的AAV 8注射C57 BL/6 J雄性小鼠,然后用HFD +1-NAME喂养7周。蛋白质印迹法显示肝脏FXI蛋白(B)、血浆FXI浓度(C)、E/A比(D)、E/e比值(E)、超声心动图的代表性图像(F)、LVEF(G)、心脏重量/胫骨长度比(H)、肺重量[湿/干比(I)]、跑步距离(J)、凝血酶-抗凝血酶复合物[TAT(K)]和心脏中指定基因的相对mRNA表达(L)。对于(D)至(H)中的食物,n = 4;在其他组中,n = 8至10。(M和N)注射AAV 8-GFP或AAV 8-F11的C57 BL/6 J雄性小鼠接受HFD +1-NAME 7周(n = 5)。 免疫组织化学染色(M)和阳性细胞定量(N)的代表性图像,显示心脏组织中的炎性细胞浸润。(0和P)注射AAV 8-GFP或AAV 8-F11的C57 BL/6 J雄性小鼠给予普通饮食或HFD +1-NAME 7周(n = 5)。Masson三色染色(0)和定量(P)的代表性图像显示心脏组织中的纤维化。每个点代表一只老鼠。所有数据均以平均值± SEM表示。ns,不显著。*P < 0.05,**P < 0.01,*P < 0.001,和 *P < 0.0001,通过双因素方差分析[(D)至(J)和(P)]或通过学生t检验[(B)和(C)和(K)至(N)]。对于(A)至(C)和(K)至(N),所有小鼠均接受HFD +1-NAME。LYM,淋巴细胞; MONO,单核细胞; GRAN,粒细胞。
Open in viewer
We then directly validated the function of FXI in the HFpEF model using overexpression. C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 were subjected to chow diet or HFD + l-NAME for 7 weeks (fig. S1B). After AAV8 injection, F11 expression was elevated in the liver and FXI protein was increased in the plasma (Fig. 2, B and C, and fig. S5, A and B). We injected sufficient virus to increase FXI protein levels only modestly (about 1.4-fold) to avoid nonphysiological artifacts. Plasma alanine transaminase levels were not significantly changed (P = 0.29) by FXI overexpression, suggesting no deleterious effects on the liver from overexpression (fig. S5C). FXI protein was not detected in the heart, confirming the specificity of AAV8 to target the liver and supporting the concept that FXI is an endocrine factor produced by the liver that affects the heart (fig. S5D). Mice receiving AAV8-F11 had lower body weight and fat mass after HFpEF compared with those receiving AAV8-GFP (fig. S5E). Blood pressure was not affected by FXI (fig. S5F). Consistent with our genetic results in the HMDP, FXI overexpression decreased E/A ratio, E/eʹ ratio, heart weight, and lung weight in the HFpEF model while preserving LVEF, indicating an improvement in diastolic function (Fig. 2, D to I, and fig. S5G). Running distance was also improved by FXI overexpression, indicating that FXI ameliorates exercise intolerance in HFpEF (Fig. 2J). FXI overexpression also had beneficial metabolic effects on fat mass and plasma lipid levels (fig. S6, A to D) but not on glucose tolerance (fig. S6, E to G).
然后,我们使用过表达直接验证了FXI在HFpEF模型中的功能。注射AAV 8-GFP或AAV 8-F11的C57 BL/6 J雄性小鼠经受普通饮食或HFD +1-NAME 7周(图S1 B)。AAV 8注射后,肝脏中F11表达升高,血浆中FXI蛋白增加(图2,B和C,以及图S5,A和B)。我们注射了足够的病毒,仅适度增加FXI蛋白水平(约1.4倍),以避免非生理性伪影。FXI过表达未显著改变血浆丙氨酸转氨酶水平(P = 0.29),表明过表达对肝脏无有害影响(图S5 C)。在心脏中未检测到FXI蛋白,证实了AAV 8靶向肝脏的特异性,并支持FXI是由肝脏产生的影响心脏的内分泌因子的概念(图S5 D)。与接受AAV 8-GFP的小鼠相比,接受AAV 8-F11的小鼠在HFpEF后具有较低的体重和脂肪量(图S5 E)。 血压不受FXI影响(图S5 F)。与我们在HMDP中的遗传结果一致,在HFpEF模型中,FXI过表达降低了E/A比、E/E比值、心脏重量和肺重量,同时保留了LVEF,表明舒张功能改善(图2,D至I和图S5 G)。FXI过表达也改善了跑步距离,表明FXI改善了HFpEF中的运动不耐受(图2 J)。FXI过表达还对脂肪量和血浆脂质水平具有有益的代谢作用(图S6,A至D),但对葡萄糖耐量无作用(图S6,E至G)。
To test whether FXI overexpression affects blood coagulation, we measured blood thrombin-antithrombin complexes in mice with GFP or FXI overexpression and found that they were not significantly changed (P = 0.79) in mice receiving AAV8-F11 versus AAV8-GFP (Fig. 2K), suggesting that the coagulation system was not affected by FXI overexpression. It has been found that mean platelet volume, reflecting the size and activity of platelets, is increased in decompensated heart failure patients and correlates with disease severity, serving as an independent predictor of 6-month mortality after decompensation (17). We observed a small but significant increase of mean platelet volume upon HFpEF development (P < 0.05), and FXI overexpression reversed it (P < 0.01) (fig. S6H). FXI overexpression significantly reduced circulating inflammatory cells (P < 0.05) and cytokine levels [interleukin β (IL-β) and IL-6, P < 0.05; interferon γ, P < 0.01] in the HFpEF model (fig. S6, I and J). Moreover, the expression of inflammatory genes in the heart was also reduced by FXI overexpression (Fig. 2L and fig. S6K). When mice were maintained on a chow diet, the number of blood immune cells was not changed by FXI overexpression (fig. S6L).
为了测试FXI过表达是否影响血液凝固,我们测量了GFP或FXI过表达的小鼠中的血液凝血酶-抗凝血酶复合物,发现它们在接受AAV 8-F11的小鼠中相对于AAV 8-GFP没有显著变化(P = 0.79)(图2K),表明凝血系统不受FXI过表达的影响。研究发现,反映血小板大小和活性的平均血小板体积在失代偿性心力衰竭患者中增加,并与疾病严重程度相关,可作为失代偿后6个月死亡率的独立预测因素(17)。我们观察到HFpEF发展后平均血小板体积的小但显著的增加(P < 0.05),并且FXI过表达逆转了它(P < 0.01)(图S6 H)。在HFpEF模型中,FXI过表达显著降低了循环炎性细胞(P < 0.05)和细胞因子水平[白细胞介素β(IL-β)和IL-6,P < 0.05;干扰素γ,P < 0.01](图S6,I和J)。 此外,心脏中炎性基因的表达也通过FXI过表达而降低(图2L和图S6K)。当小鼠维持食物饮食时,血液免疫细胞的数量不因FXI过表达而改变(图S6L)。
To further test whether the cardiac infiltration of inflammatory cells was attenuated by FXI, we performed multiplex immunohistochemistry using antibodies against macrophages (F4/80), T cells (CD3), monocytes (Ly6C), and granulocytes (Ly6G). We observed significantly decreased inflammatory cells (F4/80, P < 0.0001; CD3, P < 0.05; Ly6C and Ly6G, P < 0.01) in heart tissue from FXI-overexpressing mice versus GFP-overexpressing mice (Fig. 2, M and N), suggesting that FXI overexpression reduced inflammation in heart tissue in the HFpEF model. In addition, FXI overexpression also decreased fibrosis in the heart (Fig. 2, O and P).
为了进一步测试FXI是否减弱了炎性细胞的心脏浸润,我们使用抗巨噬细胞(F4/80)、T细胞(CD 3)、单核细胞(Ly 6C)和粒细胞(Ly 6 G)的抗体进行了多重免疫组织化学。我们观察到与GFP过表达小鼠相比,FXI过表达小鼠的心脏组织中的炎性细胞显著减少(F4/80,P < 0.0001; CD 3,P < 0.05; Ly 6C和Ly 6 G,P < 0.01)(图2,M和N),表明FXI过表达减少了HFpEF模型中心脏组织的炎症。此外,FXI过表达也减少了心脏中的纤维化(图2,O和P)。

FXI activates the BMP-SMAD1/5 pathway in cardiomyocytes
FXI激活心肌细胞中的BMP-SMAD 1/5通路

To investigate the molecular mechanism underlying the impact of FXI on the heart, we performed RNA sequencing (RNA-Seq) of the mice with FXI versus GFP overexpression in heart and adipose. Compared with GFP controls, 124 genes in the heart were significantly changed (adjusted P < 0.05) by FXI overexpression (fig. S7, A to C). Differentially expressed genes were enriched in pathways related to circadian rhythm, cardiac muscle contraction, inflammation, focal adhesion, the phosphatidylinositol 3-kinase (PI3K)–Akt pathway, and insulin signaling (fig. S7D). In contrast to the heart, only six genes in white adipose tissue were significantly changed (adjusted P < 0.05) by FXI overexpression (fig. S7E). Tcap (Titin-cap), and Lrrc10 (Leucine-rich repeat-containing 10), two genes involved in cardiac myofibril assembly and cardiac muscle tissue morphogenesis (2), were increased in the heart tissue of FXI-overexpressing mice (fig. S7, F and G).
为了研究FXI对心脏影响的分子机制,我们对心脏和脂肪中FXI与GFP过表达的小鼠进行了RNA测序(RNA-Seq)。与GFP对照相比,心脏中的124个基因被FXI过表达显著改变(调整的P < 0.05)(图S7,A至C)。差异表达的基因在与昼夜节律、心肌收缩、炎症、粘着斑、磷脂酰肌醇3-激酶(PI 3 K)-Akt途径和胰岛素信号传导相关的途径中富集(图S7 D)。与心脏相反,白色脂肪组织中只有6个基因被FXI过表达显著改变(调整的P < 0.05)(图S7 E)。Tcap(Titin-cap)和Lrrc 10(富含亮氨酸的重复序列10),这两个基因参与心脏肌原纤维组装和心肌组织形态发生(2),在FXI过表达小鼠的心脏组织中增加(图S7、F和G)。
To identify pathways perturbed by FXI, we again turned to the HMDP. Because we had performed global transcriptomics in the heart as well as the liver in all 100 strains, we could identify heart genes in which expression was correlated with the expression of FXI in the liver. On the basis of this, we examined the protein or RNA levels of the predicted pathways (Fig. 1E) and RNA-Seq (fig. S7), including the PI3K-Akt, nuclear factor κB, SMAD, and tumor necrosis factor-α (TNF-α) pathways (Fig. 3A and fig. S8A). We observed that members of the BMP pathway were correlated with FXI expression, and the link to BMP was supported by data from human GWAS (discussed below). Consistent with this, our overexpression studies showed that FXI induced an increase in SMAD1/5 phosphorylation and a decrease in TNF-α in the heart but not in other tissues (Fig. 3A and fig. S8, A to H), suggesting activation of the BMP-SMAD1/5 pathway and a decrease of inflammation in the heart. To test whether nuclear p-SMAD1/5 was also increased, we isolated the nuclear fraction from the same heart tissue and observed that it was significantly induced in the FXI overexpression group relative to GFP controls (P < 0.0001) (fig. S8I). We injected C57BL/6J male mice with saline control or mouse FXI protein and, after 2 hours, observed the phosphorylation of SMAD1/5 in the heart but not in other tissues, supporting the tissue-specific activation of the BMP-SMAD1/5 pathway by FXI (fig. S8, J to M). Plasminogen activator inhibitor-1 (PAI-1) was comparable in the hearts receiving AAV8-F11 relative to those receiving AAV8-GFP (fig. S8A). However, FXI overexpression reversed the expression of the fibrotic and inflammatory genes Col5a1, Col5a3, Adam19, IL1β, IL6, and Tnf in the heart but not in other tissues examined, consistent with the observed decrease in fibrosis and inflammation in the heart (Fig. 3B and fig. S8, N and O).
为了确定FXI干扰的途径,我们再次转向HMDP。因为我们已经在所有100个菌株的心脏和肝脏中进行了全局转录组学,所以我们可以鉴定心脏基因,其中表达与肝脏中FXI的表达相关。在此基础上,我们检查了预测途径(图1 E)和RNA-Seq(图S7)的蛋白质或RNA水平,包括PI 3 K-Akt、核因子κB、SMAD和肿瘤坏死因子-α(TNF-α)途径(图3A和图S8 A)。我们观察到BMP途径的成员与FXI表达相关,并且与BMP的联系得到了来自人GWAS的数据的支持(下文讨论)。与此一致,我们的过表达研究表明,FXI诱导心脏中SMAD 1/5磷酸化增加和TNF-α降低,但在其他组织中未诱导(图3A和图S8,A至H),表明BMP-SMAD 1/5通路激活和心脏炎症减少。 为了测试核p-SMAD 1/5是否也增加,我们从相同的心脏组织中分离核级分,并观察到相对于GFP对照,它在FXI过表达组中被显著诱导(P < 0.0001)(图S8 I)。我们向C57 BL/6 J雄性小鼠注射生理盐水对照或小鼠FXI蛋白,2小时后,观察到心脏中SMAD 1/5的磷酸化,但其他组织中没有,支持FXI对BMP-SMAD 1/5途径的组织特异性激活(图S8,J至M)。相对于接受AAV 8-GFP的心脏,接受AAV 8-F11的心脏中的纤溶酶原激活物抑制剂-1(派-1)相当(图S8 A)。然而,FXI过表达逆转了心脏中纤维化和炎症基因Col 5a 1、Col 5a 3、Adam 19、IL 1 β、IL 6和Tnf的表达,但未逆转检查的其他组织中的表达,这与观察到的心脏纤维化和炎症减少一致(图3B和图S8,N和O)。
Fig. 3. FXI activates BMP-SMAD1/5 pathway in the heart.
图三. FXI激活心脏中的BMP-SMAD 1/5通路。
(A) Western blotting and quantification showing protein levels in heart tissue from C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 and fed with 7 weeks of HFD + l-NAME. Actin served as the loading control. n = 5. (B) qRT-PCR analysis showing the mRNA levels of Col5a3 in the indicated tissue from C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 and fed HFD + l-NAME for 7 weeks. Heart, P < 0.001; others, not significant. n = 8. (C and D) NRVMs were treated with control or human FXIa protein (1 μg/ml) with medium containing control or phenylephrine (PE, 100 μM) for 24 hours. p-SMAD1/5 (C) and the indicated genes (D) were examined. Actin served as the loading control. n = 6. (E to H) C57BL/6J male mice were injected with AAV8-GFP or AAV8-F11 with DMH1 and fed with HFD + l-NAME for 7 weeks. Heart p-SMAD1/5 level (E), heart weight/tibia length ratio (F), E/eʹ ratio (G), and LVEF (H) were determined. n = 3 for (E) and n = 8 for (F) to (H). (I to M) C57BL/6J male mice were injected with AAV8-GFP, AAV8-F11, or AAV8-F11-Mut (mF11-Mut2) and fed with HFD + l-NAME for 7 weeks. Plasma FXI levels (I), heart p-SMAD1/5 protein level (J), heart weight/tibia length ratio (K), E/eʹ ratio (L), and LVEF (M) were measured. n = 5 for (I), n = 6 for (J), and n = 10 to 20 for (K) to (M). (N) C57BL/6J male mice were injected with either AAV8-GFP or AAV8-F11 and then fed with HFD + l-NAME for 7 weeks. BMP7 proteins in unprocessed monomer, growth factor dimer, and monomer under nonreducing condition were determined. (O) NRVMs were treated with control or human FXIa protein (1 μg/ml) plus negative control or Bmp7 siRNA with medium containing PE (100 μM) for 24 hours. p-SMAD1/5 and Tcap proteins were examined. Actin served as the loading control. n = 3. (P) NRVMs were treated with control or human FXIa protein (1 μg/ml), BMP7 antibody (no antibody control, 1:100 and 1:50), with medium containing PE (100 μM) for 2 hours, and the p-SMAD1/5 level was determined. Actin served as the loading control. n = 4. Each point represents a mouse. All data are presented as means ± SEM. ns, not significant. *P < 0.05, **P < 0.01, and ***P < 0.001 by two-way ANOVA [(D) to (H)], one-way ANOVA [(I) to (M)], or Student’s t test [(A) to (C)].
(A)蛋白质印迹和定量显示来自注射AAV 8-GFP或AAV 8-F11并喂食7周HFD +1-NAME的C57 BL/6 J雄性小鼠的心脏组织中的蛋白质水平。肌动蛋白作为上样对照。n = 5。(B)qRT-PCR分析显示来自注射AAV 8-GFP或AAV 8-F11并喂食HFD + l-NAME 7周的C57 BL/6 J雄性小鼠的指定组织中Col 5a 3的mRNA水平。心脏,P < 0.001;其他,不显著。n = 8。(C和D)NRVM用对照或人FXIa蛋白(1 μg/ml)和含有对照或苯丙氨酸(PE,100 μM)的培养基处理24小时。检测p-SMAD 1/5(C)和所示基因(D)。肌动蛋白作为上样对照。n = 6。(E至H)C57 BL/6 J雄性小鼠注射AAV 8-GFP或具有DMH 1的AAV 8-F11,并用HFD +1-NAME喂养7周。测定心脏p-SMAD 1/5水平(E)、心脏重量/胫骨长度比值(F)、E/e比值(G)和LVEF(H)。对于(E),n = 3,对于(F)至(H),n = 8。 (I至M)C57 BL/6 J雄性小鼠注射AAV 8-GFP、AAV 8-F11或AAV 8-F11-Mut(mF 11-Mut 2),并用HFD +1-NAME喂养7周。测量血浆FXI水平(I)、心脏p-SMAD 1/5蛋白水平(J)、心脏重量/胫骨长度比(K)、E/e比值(L)和LVEF(M)。对于(I)n = 5,对于(J)n = 6,对于(K)至(M)n = 10至20。(N)用AAV 8-GFP或AAV 8-F11注射C57 BL/6 J雄性小鼠,然后用HFD +1-NAME喂养7周。在未加工的单体,生长因子二聚体和单体在非还原条件下的BMP 7蛋白进行了测定。(O)NRVM用对照或人FXIa蛋白(1 μg/ml)加阴性对照或Bmp 7 siRNA与含有PE(100 μM)的培养基处理24小时。检测p-SMAD 1/5和Tcap蛋白。肌动蛋白作为上样对照。n = 3。 (P)用对照或人FXIa蛋白(1 μg/ml)、BMP 7抗体(无抗体对照,1:100和1:50)和含PE(100 μM)的培养基处理NRVM 2小时,并测定p-SMAD 1/5水平。肌动蛋白作为上样对照。n = 4。每个点代表一只老鼠。所有数据均以平均值± SEM表示。ns,不显著。通过双因素方差分析[(D)至(H)]、单因素方差分析[(I)至(M)]或学生t检验[(A)至(C)],*P < 0.05、**P < 0.01和 *P < 0.001。
Open in viewer
To determine the localization of p-SMAD1/5, we stained p-SMAD1/5 and the markers of cardiomyocytes (troponin I), fibroblasts (vimentin), macrophages (CD68), and endothelial cells (CD31) in the heart after FXI overexpression. p-SMAD1/5 was colocalized with troponin I, but not with other markers (fig. S9), suggesting that p-SMAD1/5 was mainly activated in cardiomyocytes. To directly test whether FXI protein activates the BMP-SMAD1/5 pathway in cardiomyocytes, we incubated neonatal rat ventricular myocytes (NRVMs), human embryonic stem cell–induced cardiomyocytes, and other cell lines with control medium or medium containing human activated FXI protein (FXIa) in the presence of phenylephrine for 24 hours. We observed that FXIa increased the phosphorylation of SMAD1/5 and decreased the expression of Nppa, Nppb, Col5a3, and Adam19 in NRVMs and human embryonic stem cell–induced cardiomyocytes but not in other cell types (Fig. 3, C and D, and fig. S10).
为了确定p-SMAD 1/5的定位,我们对FXI过表达后心脏中的p-SMAD 1/5和心肌细胞(肌钙蛋白I)、成纤维细胞(波形蛋白)、巨噬细胞(CD 68)和内皮细胞(CD 31)的标志物进行染色。p-SMAD 1/5与肌钙蛋白I共定位,但不与其他标志物共定位(图S9),表明p-SMAD 1/5主要在心肌细胞中活化。为了直接测试FXI蛋白是否激活心肌细胞中的BMP-SMAD 1/5通路,我们在苯丙氨酸存在下将新生大鼠心室肌细胞(NRVM)、人胚胎干细胞诱导的心肌细胞和其他细胞系与对照培养基或含有人活化FXI蛋白(FXIa)的培养基孵育24小时。我们观察到FXIa增加了SMAD 1/5的磷酸化,并降低了NRVM和人胚胎干细胞诱导的心肌细胞中Nppa、Nppb、Col 5a 3和Adam 19的表达,但在其他细胞类型中未观察到(图3,C和D,以及图S10)。
The above experiments were performed with male mice and we were interested in determining whether the results were similar with females. C57BL/6J female mice injected with AAV8-GFP or AAV8-F11 were subjected to HFD + l-NAME for 7 weeks (fig. S11A). After AAV8 injection, F11 expression was increased in the liver (fig. S11B). We observed similar effects of FXI in female mice, including decreased body mass, decreased inflammatory cells in the blood, reduced plasma lipids, increased SMAD1/5 phosphorylation, improved diastolic function, and reduced heart weight and lung weight (fig. S11, C to N). By contrast, blood pressure was comparable between the FXI and GFP groups (fig. S11O).
上述实验是用雄性小鼠进行的,我们有兴趣确定结果是否与雌性小鼠相似。注射AAV 8-GFP或AAV 8-F11的C57 BL/6 J雌性小鼠经受HFD +1-NAME 7周(图S11 A)。AAV 8注射后,肝脏中F11表达增加(图S11 B)。我们在雌性小鼠中观察到FXI的相似作用,包括体重减轻、血液中炎性细胞减少、血脂降低、SMAD 1/5磷酸化增加、舒张功能改善以及心脏重量和肺重量减轻(图S11,C至N)。相比之下,FXI和GFP组之间的血压相当(图S11 O)。
To determine whether the effects of FXI that we observed were specific to the HFpEF model, we examined a “multi-hit” HFpEF model induced by the combination of aging, HFD, and angiotensin II (18). Aged C57BL/6J male mice were injected with AAV8-GFP or AAV8-F11 and then fed a HFD for 12 weeks. After 8 weeks of HFD, mice were infused with angiotensin II for 4 weeks (fig. S12A). F11 mRNA was increased in the liver by AAV8-F11 compared with AAV8-GFP controls (fig. S12B). Similar to the beneficial effects in the “two-hit” HFpEF model, we observed significant improvement in diastolic function and related traits in FXI-overexpressing mice relative to GFP-overexpressing mice, including reduced body mass (P < 0.05) (fig. S12C) and improved diastolic function as measured by lower E/A ratio (P < 0.05), lower E/eʹ ratio (P < 0.001), and lower LV mass (P < 0.05) (fig. S12, D to H). In addition, FXI-overexpressing mice exhibited reduced heart weight and plasma total cholesterol, as well as increased p-SMAD1/5 relative to GFP controls (fig. S12, I to K).
为了确定我们观察到的FXI效应是否对HFpEF模型具有特异性,我们检查了由衰老、HFD和血管紧张素II联合诱导的“多次打击”HFpEF模型(18)。用AAV 8-GFP或AAV 8-F11注射老年C57 BL/6 J雄性小鼠,然后喂食HFD 12周。HFD 8周后,向小鼠输注血管紧张素II 4周(图S12 A)。与AAV 8-GFP对照相比,通过AAV 8-F11在肝脏中增加F11 mRNA(图S12 B)。与“两次打击”HFpEF模型中的有益作用相似,我们观察到相对于GFP过表达小鼠,FXI过表达小鼠的舒张功能和相关性状显著改善,包括体重减轻(P < 0.05)。(图S12 C)和通过较低的E/A比测量的改善的舒张功能(P < 0.05),较低的E/E比值(P < 0.001),和较低的LV质量(P < 0.05)(图S12,D至H)。 此外,相对于GFP对照,过表达FXI的小鼠表现出心脏重量和血浆总胆固醇降低,以及p-SMAD 1/5增加(图S12,I至K)。
To confirm that FXI overexpression activates BMP signaling to protect against diastolic dysfunction, we blocked the BMP receptor with the dorsomorphin homolog 1 (DMH1) (19). DMH1 is a selective inhibitor of activin receptor-like kinase 3, a type 1 BMP receptor. In NRVMs, DMH1 treatment suppressed SMAD1/5 phosphorylation induction by FXIa (fig. S13A). We also examined the effect of DMH1 in vivo. C57BL/6J mice were injected with AAV8-F11 and fed with HFD + l-NAME for 7 weeks. Injection of DMH1 every other day to block SMAD1/5 phosphorylation (20) suppressed the change of body mass induced by FXI (fig. S13B), as well as the effect of FXI on p-SMAD1/5 levels, diastolic function, adipose weight, blood cell numbers, and plasma cholesterol (Fig. 3, E to H, and fig. S13, C to G). These results confirmed that FXI protects against HFpEF by activating the BMP pathway.
为了证实FXI过表达激活BMP信号传导以防止舒张功能障碍,我们用dorsomorphin同系物1(DMH 1)阻断BMP受体(19)。DMH 1是激活素受体样激酶3(一种1型BMP受体)的选择性抑制剂。在NRVM中,DMH 1处理抑制了FXIa对SMAD 1/5磷酸化的诱导(图S13 A)。我们还研究了DMH 1在体内的作用。C57 BL/6 J小鼠注射AAV 8-F11,并用HFD + l-NAME喂养7周。每隔一天注射DMH 1以阻断SMAD 1/5磷酸化(20)可抑制FXI诱导的体重变化(图S13 B),以及FXI对p-SMAD 1/5水平、舒张功能、脂肪重量、血细胞数量和血浆胆固醇的影响(图3,E至H,图S13,C至G)。这些结果证实,FXI通过激活BMP通路保护免受HFpEF。

FXI protease activity is required for the activation of BMP signaling
FXI蛋白酶活性是激活BMP信号传导所必需的

The FXI protein is conserved in human, mouse, rat, and other species and consists of four apple domains and one catalytic domain (fig. S14A). It is present in that plasma as a zymogen, which exists as a homodimer consisting of two identical polypeptide chains linked by disulfide bonds (fig. S14B) (21). During FXI activation, an internal peptide bond is cleaved by FXIIa (or XII) in each of the two chains, resulting in activated FXIa, a serine protease composed of two heavy and two light chains held together by disulfide bonds (fig. S14B). To test whether the catalytic domain is required for the function of FXI on the heart, we introduced two missense mutations in human and mouse FXI catalytic domains (fig. S14, A to E). These mutations were predicted to be exposed at the surface of the FXI molecule and to cause functional defects (type II mutation) (22). Next, we tested their function in vitro using a co-culture system. Huh7 human liver cells and AML12 mouse liver cells were transfected with respective human or mouse plasmids containing GFP control, wild-type (WT) F11, or F11 with mutations. Then, cells were placed in co-cultures with NRVMs or 3T3-L1 adipocytes (fig. S15A). Twenty-four hours after transfection, FXI was highly induced in both Huh7 cells and AML12 cells (fig. S15B). In NRVMs, phosphorylation of SMAD1/5 was induced by WT FXI overexpression from both human and mouse liver cells, whereas mutant FXI did not exhibit a comparable effect (fig. S15C). By contrast, SMAD1/5 phosphorylation was not significantly (P = 0.84) induced by FXI in 3T3-L1 adipocytes, suggesting a heart-specific effect (fig. S15D). Consistent with phosphorylated SMAD1/5, Col5a3 was decreased by WT FXI but not mutant FXI in NRVMs, indicating that the catalytic activity is required for its effect (fig. S15E).
FXI蛋白在人、小鼠、大鼠和其他物种中是保守的,由四个苹果结构域和一个催化结构域组成(图S14 A)。它作为酶原存在于血浆中,作为由通过二硫键连接的两条相同多肽链组成的同源二聚体存在(图S14 B)(21)。在FXI活化过程中,两条链中的每条链的内部肽键被FXIIa(或XII)切割,产生活化的FXIa,这是一种丝氨酸蛋白酶,由两条重链和两条轻链通过二硫键连接在一起(图S14 B)。为了测试催化结构域是否是心脏上FXI功能所必需的,我们在人和小鼠FXI催化结构域中引入了两个错义突变(图S14,A至E)。预测这些突变暴露于FXI分子表面并导致功能缺陷(II型突变)(22)。接下来,我们使用共培养系统在体外测试它们的功能。 Huh 7人肝细胞和AML 12小鼠肝细胞用含有GFP对照、野生型(WT)F11或具有突变的F11的相应人或小鼠质粒转染。然后,将细胞置于与NRVM或3 T3-L1脂肪细胞的共培养物中(图S15 A)。转染后24小时,FXI在Huh 7细胞和AML 12细胞中均被高度诱导(图S15 B)。在NRVM中,SMAD 15的磷酸化由来自人和小鼠肝细胞的WT FXI过表达诱导,而突变体FXI未表现出相当的作用(图S15 C)。相比之下,FXI在3 T3-L1脂肪细胞中未显著诱导SMAD 15磷酸化(P = 0.84),表明心脏特异性效应(图S15 D)。与磷酸化SMAD 15一致,在NRVM中WT FXI降低了Col 5a 3,但突变体FXI没有降低,表明其作用需要催化活性(图S15 E)。
To test the effect of missense mutation in vivo, we produced AAV8 with the mouse WT and mutant F11 coding sequences. AAV8 containing GFP control, WT F11, and mutant F11 (mF11-Mut2) was injected into C57BL/6J male mice followed by HFD + l-NAME for 7 weeks, after which plasma FXI was increased in FXI group and was comparable to the mutant FXI group (Fig. 3I and fig. S15F). Body weight and fat mass were decreased by WT FXI overexpression, but there was no significant difference (P > 0.05) between groups of mutant FXI and GFP controls (fig. S15, G to I), suggesting functional defects of mutant FXI. Consistently, the effects of FXI on p-SMAD1/5, heart weight, E/A ratio, E/eʹ ratio, adipose weight, plasma cholesterol, and blood immune cells were not observed in mice carrying mutant FXI, demonstrating that catalytic activity is essential for the function of FXI in protecting against deleterious phenotypes in HFpEF (Fig. 3, J to M, and fig. S15, J to P).
为了测试体内错义突变的效果,我们用小鼠WT和突变体F11编码序列产生AAV 8。将含有GFP对照、WT F11和突变体F11(mF 11-Mut 2)的AAV 8注射到C57 BL/6 J雄性小鼠中,随后注射HFD + l-NAME 7周,之后FXI组中血浆FXI增加并且与突变体FXI组相当(图3 I和图S15 F)。WT FXI过表达降低了体重和脂肪量,但在突变体FXI组和GFP对照组之间没有显著差异(P > 0.05)(图S15,G至I),表明突变体FXI的功能缺陷。同样,在携带突变FXI的小鼠中未观察到FXI对p-SMAD 15、心脏重量、E/A比、E/E比值、脂肪重量、血浆胆固醇和血液免疫细胞的影响,表明催化活性对于FXI在保护HFpEF免受有害表型影响方面的功能至关重要(图3,J至M和图S15,J至P)。

FXI cleaves the BMP7 proprotein, activating the resulting growth factor fragment
FXI切割BMP 7前蛋白,激活生成的生长因子片段

As a serine protease, FXIa catalyzes the proteolysis of its substrates. BMP7 is synthesized as a large precursor molecule (inactive) that is cleaved to growth factor dimer or monomer (active) by proteolytic enzymes (23). We found that the cleavage site of the full-length BMP7 protein, at an arginine, is a common FXIa cleavage site (fig. S16, A and B) (24). We therefore hypothesized that BMP7 is a substrate of FXIa that mediates SMAD1/5 activation. FXI overexpression increased BMP7 growth factor dimer and monomer in the heart (Fig. 3N). Moreover, incubation of FXIa with BMP7 protein resulted in the cleavage of BMP7 (fig. S16C). Knocking down BMP7 or treatment with BMP7 antibody in NRVMs greatly reduced the activation of SMAD1/5 by FXIa (Fig. 3, O and P). The BMP7 protein is considerably enriched in heart tissue and cardiomyocytes (fig. S17), which may explain the preferential effect of FXI on the heart. The prodomain of BMP7 appears to bind to the extracellular matrix (23), suggesting that FXI cleaves the precursor BMP7 bound to the extracellular matrix in the heart; this then releases the dimer and monomer growth factors from the matrix to bind to the BMP receptor and activate SMAD1/5.
作为丝氨酸蛋白酶,FXIa催化其底物的蛋白水解。BMP 7作为大的前体分子(无活性)合成,其被蛋白水解酶切割成生长因子二聚体或单体(活性)(23)。我们发现,全长BMP 7蛋白在精氨酸处的切割位点是常见的FXIa切割位点(图S16,A和B)(24)。因此,我们假设BMP 7是FXIa介导SMAD 1/5激活的底物。FXI过表达增加了心脏中的BMP 7生长因子二聚体和单体(图3 N)。此外,FXIa与BMP 7蛋白孵育导致BMP 7裂解(图S16 C)。在NRVM中敲低BMP 7或用BMP 7抗体处理大大降低了FXIa对SMAD 1/5的激活(图3,O和P)。BMP 7蛋白在心脏组织和心肌细胞中显著富集(图S17),这可以解释FXI对心脏的优先作用。 BMP 7的前结构域似乎与细胞外基质结合(23),表明FXI切割与心脏细胞外基质结合的前体BMP 7;然后从基质中释放二聚体和单体生长因子,与BMP受体结合并激活SMAD 1/5。

FXI knockout mice have reduced p-SMAD1/5 levels and increased diastolic dysfunction
FXI基因敲除小鼠p-SMAD 1/5水平降低,舒张功能障碍增加

We sought to further examine the cardioprotective effect of FXI using FXI knockout male mice in which the F11 gene was disrupted by a PGK-neo cassette (25). F11 transcripts in the liver of heterozygous null mice (F11-Het) were reduced by ~50% compared with WT littermates (Fig. 4A). FXI was either absent or barely detectable in other tissues (Fig. 4A and fig. S4, A and B). Adult WT and F11-Het mice were then subjected to HFD + l-NAME for 7 weeks to induce HFpEF phenotypes. Compared with WT littermates on the HFpEF diet, p-SMAD1/5 was reduced in the hearts of F11-Het mice (Fig. 4B). Consistent with reduced p-SMAD1/5, F11-Het mice exhibited more severe diastolic dysfunction, as evidenced by the increased E/A ratio, E/eʹ ratio, and LV mass but preserved ejection fraction (Fig. 4, C to G). Moreover, heart weight and lung weight were higher in F11-Het mice relative to WT controls, suggesting cardiac hypertrophy and lung congestion in FXI-deficient mice (Fig. 4, H and I). Exercise tolerance was also decreased in F11-Het mice compared with WT mice (Fig. 4J). By contrast, blood pressure was not significantly changed (P > 0.05) by FXI deficiency (fig. S18), indicating that FXI does not influence heart function through effects on blood pressure. These results collectively demonstrated the increased severity of diastolic dysfunction in FXI-deficient mice. We observed consistent effects of FXI in female mice with FXI heterozygous knockout (fig. S19).
我们试图使用FXI基因敲除雄性小鼠进一步检查FXI的心脏保护作用,其中F11基因被PGK-neo盒破坏(25)。与WT同窝出生小鼠相比,杂合无效小鼠(F11-Het)肝脏中的F11转录物减少约50%(图4A)。在其他组织中,FXI不存在或几乎检测不到(图4A和图S4,A和B)。然后使成年WT和F11-Het小鼠经受HFD +1-NAME 7周以诱导HFpEF表型。与使用HFpEF饮食的WT同窝仔相比,F11-Het小鼠心脏中的p-SMAD 1/5减少(图4 B)。与p-SMAD 1/5降低一致,F11-Het小鼠表现出更严重的舒张功能障碍,如E/A比、E/E比值和LV质量增加但射血分数保持所证明的(图4,C至G)。此外,相对于WT对照,F11-Het小鼠的心脏重量和肺重量更高,表明FXI缺陷小鼠的心脏肥大和肺充血(图4,H和I)。 与WT小鼠相比,F11-Het小鼠的运动耐量也降低(图4J)。相比之下,FXI缺乏没有显著改变血压(P > 0.05)(图S18),表明FXI不会通过影响血压影响心脏功能。这些结果共同证明了FXI缺陷小鼠中舒张功能障碍的严重程度增加。我们在FXI杂合敲除雌性小鼠中观察到FXI的一致效应(图S19)。
Fig. 4. Reduced FXI concentrations are associated with diastolic dysfunction in mice and humans.
见图4。FXI浓度降低与小鼠和人的舒张功能障碍相关。
Heterozygous B6.129X1-F11tm1Gjb/J (F11-Het) mice and WT littermates at 8 weeks of age were subjected to HFD + l-NAME for 7 weeks. n = 8 for WT and n = 6 for F11-Het. (A) qRT-PCR showing F11 mRNA in the indicated tissues from WT and F11-Het mice. n = 4. (B) Western blotting showing p-SMAD1/5 in the hearts of WT mice fed with chow diet (Chow), WT, and F11-Het mice fed with HFD + l-NAME for 7 weeks. n = 5. (C to G) Representative images of echocardiography (C), E/A ratio (D), E/eʹ ratio (E), LV mass (F), and LVEF (G) were examined at baseline (BSL) and after 7 weeks of HFD + l-NAME feeding (HFpEF). n = 8 for WT and n = 6 for F11-Het. (H to J) Heart weight/tibia length ratio (H), lung weight [wet/dry ratio (I)], and running distance (J) were examined after 7 weeks of HFD + l-NAME feeding (HFpEF). n = 8 for WT and n = 6 for F11-Het. (K) Plasma FXI protein in non-HFpEF controls (NHF, n = 20) and HFpEF patients (n = 21). (L and M) Plasma FXI protein was inversely correlated with E/eʹ ratio in all participants (L), including HFpEF patients (M). (N) Illustration summarizing FXI-mediated liver-heart cross-talk in protecting against heart failure. Using a bioinformatic framework that integrates global liver-heart transcriptome and cardiometabolic trait data from the HMDP, we found that coagulation FXI, secreted by the liver, exhibits cardioprotective effects on the progression of HFpEF. FXI overexpression in the liver mitigates the diastolic dysfunction, inflammation, and fibrosis induced by HFpEF. FXIa cleaves the BMP7 precursor and activates the BMP7-SMAD1/5 pathway in the heart to mediate the anti-inflammatory and anti-fibrotic effects. Each point represents a mouse. All data are presented as means ± SEM. ns, not significant. *P < 0.05, **P < 0.01, and ***P < 0.001 by two-way ANOVA [(D) to (G)], one-way ANOVA (B), or Student’s t test [(A) and (H) to (K)].
使8周龄的杂合B6.129X1-F11 tm1Gjb /J(F11-Het)小鼠和WT同窝出生的小鼠经受HFD +1-NAME 7周。对于WT,n = 8,对于F11-Het,n = 6。(A)qRT-PCR显示WT和F11-Het小鼠指定组织中的F11 mRNA。n = 4。(B)蛋白质印迹显示喂食普通食物(Chow)的WT小鼠、喂食HFD + l-NAME 7周的WT和F11-Het小鼠的心脏中的p-SMAD 1/5。n = 5。(C至G)在基线(BSL)和HFD + l-NAME喂养7周后(HFpEF)检查超声心动图(C)、E/A比(D)、E/E比值(E)、LV质量(F)和LVEF(G)的代表性图像。对于WT,n = 8,对于F11-Het,n = 6。(H至J)在HFD + l-NAME喂养(HFpEF)7周后检查心脏重量/胫骨长度比(H)、肺重量[湿/干比(I)]和跑步距离(J)。对于WT,n = 8,对于F11-Het,n = 6。(K)非HFpEF对照(NHF,n = 20)和HFpEF患者(n = 21)的血浆FXI蛋白。 (L和M)所有参与者(L)(包括HFpEF患者(M))的血浆FXI蛋白与E/E比值呈负相关。(N)总结FXI介导的肝-心串扰在预防心力衰竭中的示意图。使用整合来自HMDP的全局肝-心转录组和心脏代谢性状数据的生物信息学框架,我们发现由肝脏分泌的凝血FXI对HFpEF的进展具有心脏保护作用。肝脏中的FXI过表达减轻了HFpEF诱导的舒张功能障碍、炎症和纤维化。FXIa切割BMP 7前体并激活心脏中的BMP 7-SMAD 1/5通路,以介导抗炎和抗纤维化作用。每个点代表一只老鼠。所有数据均以平均值± SEM表示。ns,不显著。通过双因素方差分析[(D)至(G)]、单因素方差分析(B)或学生t检验[(A)和(H)至(K)],*P < 0.05、**P < 0.01和 *P < 0.001。
Open in viewer

FXI levels correlate with diastolic function in human cohorts
FXI水平与人类队列中的舒张功能相关

To determine the clinical relevance of FXI, we quantified plasma FXI in human patients with HFpEF and in normal participants. Plasma FXI protein was not significantly different (P = 0.91) between non–heart failure controls and HFpEF patients (Fig. 4K). However, plasma FXI was inversely correlated with E/eʹ ratio in all participants (Fig. 4L), including HFpEF patients (Fig. 4M), supporting the conclusion that FXI protects against diastolic dysfunction in HFpEF.
为了确定FXI的临床相关性,我们定量了HFpEF患者和正常受试者的血浆FXI。血浆FXI蛋白在非心力衰竭对照和HFpEF患者之间没有显著差异(P = 0.91)(图4K)。然而,在所有参与者(图4L)中,包括HFpEF患者(图4 M),血浆FXI与E/E比值呈负相关,支持FXI可预防HFpEF舒张功能障碍的结论。

Discussion 讨论

Our results indicate that liver-derived FXI specifically regulates cardiomyocytes through the BMP-SMAD1/5 pathway, resulting in attenuation of fibrosis, inflammation, and diastolic dysfunction in the context of an HFpEF model (Fig. 4N). Our analysis of diastolic function in a cohort of heart failure patients indicates the relevance of the pathway in humans and in mouse models, and the human GWAS results are consistent with that conclusion.
我们的结果表明,肝源性FXI通过BMP-SMAD 1/5途径特异性调节心肌细胞,导致HFpEF模型背景下纤维化、炎症和舒张功能障碍减弱(图4 N)。我们对一组心力衰竭患者舒张功能的分析表明了该途径在人类和小鼠模型中的相关性,人类GWAS结果与该结论一致。
Prior studies have implicated the BMP and SMAD pathways in traits relevant to heart failure. It has been reported that the BMP pathway is enriched for HFpEF but not HFrEF (26). BMP2 has been found to alleviate heart failure with type 2 diabetes by inhibiting inflammasome formation (27), and is inversely correlated with the concentrations of atrial natriuretic peptide and brain natriuretic peptide in chronic heart failure patients with diabetes. Another study observed increased BMP6 in chronic heart failure patients, suggesting that BMP6 may be involved in the pathophysiology of systolic heart failure (28). In addition, SMAD1 protein was differentially expressed in a high-salt diet-induced HFpEF model (29).
先前的研究表明BMP和SMAD通路与心力衰竭相关。据报道,BMP途径富含HFpEF,但不富含HFrEF(26)。已发现BMP 2通过抑制炎性小体形成来缓解2型糖尿病心力衰竭(27),并且与糖尿病慢性心力衰竭患者的心房利钠肽和脑利钠肽浓度呈负相关。另一项研究观察到慢性心力衰竭患者的BMP6增加,表明BMP6可能参与收缩性心力衰竭的病理生理学(28)。此外,SMAD1蛋白在高盐饮食诱导的HFpEF模型中差异表达(29)。
FXI is a component of the intrinsic pathway of blood coagulation, acting downstream of FXII and functioning as a protease to activate FIX (14, 15, 21, 30, 31). Our data indicate that FXI overexpression also influences various systemic aspects of metabolism, and we cannot rule out the possibility that it may also affect organs other than the heart. FXI-deficient patients generally do not have spontaneous bleeding, because FXI is not required for the initial thrombin generation step (32), consistent with the possibility that it exhibits other previously unknown functions. Inactivating mutations of F11 are relatively common among Ashkenazi Jews (33). A number of studies investigated the relationship between FXI and incident coronary heart disease, stroke, and ischemic cardiomyopathy (34, 35). FXI was reported to improve or protect against the inflammatory responses and cytokine responses to infections independently of its intrinsic coagulation activity (3640).
FXI是凝血内源性途径的一种组分,在FXII下游发挥作用,并作为蛋白酶激活FIX(14、15、21、30、31)。我们的数据表明,FXI过表达也影响代谢的各个系统方面,我们不能排除它也可能影响心脏以外的器官的可能性。FXI缺乏的患者通常不会发生自发性出血,因为FXI不是最初凝血酶生成步骤所必需的(32),这与其表现出其他先前未知功能的可能性一致。F11的失活突变在德系犹太人中相对常见(33)。许多研究调查了FXI与冠心病、卒中和缺血性心肌病之间的关系(34,35)。据报告,FXI可改善或预防感染的炎症反应和细胞因子反应,与其内在凝血活性无关(36-40)。
The fact that FXI is a direct mediator of liver-heart communication suggests the possibility of using it in therapeutic applications for heart failure. It is important to note that elevated FXI is associated with various thromboses and ischemic stroke (4143), so elevating FXI would be problematic as a therapeutic goal. However, the downstream BMP pathway could provide potential therapeutic targets.
事实上,FXI是一个直接调解人的肝心通信表明,可能性使用它在治疗应用心力衰竭。值得注意的是,FXI升高与各种血栓形成和缺血性卒中相关(41-43),因此升高FXI作为治疗目标存在问题。然而,下游BMP通路可以提供潜在的治疗靶点。

Acknowledgments 致谢

Funding: This work was supported by the National Institutes of Health (grants DK120342 and HL147883; grants HL138193, DK130640, and DK097771 to M.M.S.; grants R01HL133169 and R01HL148110 to H.A.; and grant DK125354 to Z.Z.).
资金来源:这项工作得到了美国国立卫生研究院的支持(赠款DK 120342和HL 147883;拨款HL 138193,DK 130640和DK 097771给M.M.S.;赠款R 01 HL 133169和R 01 HL 148110授予H.A.;并将DK 125354授予Z.Z.)。
Author contributions: Y.C., M.M.S., and A.J.L. designed the experiments. Y.C., Y.W., Z.Z., L.J., Z.Z., Y.M., S.C., and T.L. performed the experiments. Y.C., L.J., M.M.S., and C.P. analyzed raw data. Z.Z., H.A., M.M.S., and A.J.L. reviewed the data and made substantial contributions to improving the studies. Y.C. and A.J.L. wrote the manuscript, which was reviewed by all authors.
作者:Y.C.,多发性硬化症,和A.J.L.设计了实验。Y.C.,Y.W.,Z.Z.,L.J.,Z.Z.,Y.M.,南卡罗来纳州,和T.L.之间。进行了实验。Y.C.,L.J.,多发性硬化症,C. P分析了原始数据Z.Z.,H.A.,多发性硬化症,和A.J.L.审查了数据,并为改进研究作出了重大贡献。Y.C.和A.J.L.他写了手稿,所有作者都审阅了。
Competing interests: The authors declare no competing interests.
竞争利益:作者声明没有竞争利益。
Data and materials availability: All data supporting the conclusions in this manuscript can be found in the main text or the supplementary materials. RNA-Seq data were deposited to the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/) under accession number GSE200496. The R script used to perform the volcano plots and PCA plot are available at Zenodo (44).
数据和材料的可获得性:所有支持本文结论的数据可在正文或补充材料中找到。将RNA-Seq数据以登录号GSE 200496保藏于基因表达综合数据库(GEO)(https://www.ncbi.nlm.nih.gov/geo/)。用于执行火山图和PCA图的R脚本可在Zenodo(44)获得。
License information: Copyright © 2022 the authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original US government works. https://www.science.org/about/science-licenses-journal-article-reuse
版权所有© 2022作者,保留部分权利;美国科学促进会独家许可。没有要求美国政府的原始作品。https://www.science.org/about/science-licenses-journal-article-reuse

Supplementary Materials 补充材料

This PDF file includes: 此PDF文件包括:

Materials and Methods 材料和方法
Figs. S1 to S20 图S1至S20
Tables S1 to S8 表S1至S8
References (4550) 参考文献(45-50)

Other Supplementary Material for this manuscript includes the following:
本手稿的其他补充材料包括:

MDAR Reproducibility Checklist
MDAR再现性检查表

References and Notes 参考和注释

1
J. M. Friedman, J. L. Halaas, Leptin and the regulation of body weight in mammals. Nature 395, 763–770 (1998).
J. M.弗里德曼Halaas、Leptin与哺乳动物体重调节。Nature 395,763-770(1998)。
2
S. Møller, M. Bernardi, Interactions of the heart and the liver. Eur. Heart J. 34, 2804–2811 (2013).
S. Møller,M. Bernardi,心脏和肝脏的相互作用。EUR. Heart J.34,2804-2811(2013).
3
K. K. Baskin, A. L. Bookout, E. N. Olson, The heart-liver metabolic axis: Defective communication exacerbates disease. EMBO Mol. Med. 6, 436–438 (2014).
K. K.巴斯金,A. L. Bookout,E. N.奥尔森,心脏-肝脏代谢轴:有缺陷的沟通加剧疾病。EMBO分子量6,436-438(2014)。
4
M. Packer, Atrial fibrillation and heart failure with preserved ejection fraction in patients with nonalcoholic fatty liver disease. Am. J. Med. 133, 170–177 (2020).
M.非酒精性脂肪肝患者射血分数正常的房颤和心力衰竭。Am. 133,170-177(2020)。
5
L. Pacifico, M. Di Martino, A. De Merulis, M. Bezzi, J. F. Osborn, C. Catalano, C. Chiesa, Left ventricular dysfunction in obese children and adolescents with nonalcoholic fatty liver disease. Hepatology 59, 461–470 (2014).
6
Y. Cao, C. Pan, Y.-C. Wang, Z. Zhou, V. Jedian, Y. Meng, G. Campbell, K. Guardino, C. Li, J. Wang, A. J. Lusis, Identification of DNA damage repair enzyme Ascc2 as causal for heart failure with preserved ejection fraction. Circulation 145, 1102–1104 (2022).
7
S. M. Dunlay, V. L. Roger, M. M. Redfield, Epidemiology of heart failure with preserved ejection fraction. Nat. Rev. Cardiol. 14, 591–602 (2017).
8
K. Hogg, K. Swedberg, J. McMurray, Heart failure with preserved left ventricular systolic function; epidemiology, clinical characteristics, and prognosis. J. Am. Coll. Cardiol. 43, 317–327 (2004).
9
T. E. Owan, D. O. Hodge, R. M. Herges, S. J. Jacobsen, V. L. Roger, M. M. Redfield, Trends in prevalence and outcome of heart failure with preserved ejection fraction. N. Engl. J. Med. 355, 251–259 (2006).
10
M. M. Seldin, S. Koplev, P. Rajbhandari, L. Vergnes, G. M. Rosenberg, Y. Meng, C. Pan, T. M. N. Phuong, R. Gharakhanian, N. Che, S. Mäkinen, D. M. Shih, M. Civelek, B. W. Parks, E. D. Kim, F. Norheim, K. Chella Krishnan, Y. Hasin-Brumshtein, M. Mehrabian, M. Laakso, C. A. Drevon, H. A. Koistinen, P. Tontonoz, K. Reue, R. M. Cantor, J. L. M. Björkegren, A. J. Lusis, A strategy for discovery of endocrine interactions with application to whole-body metabolism. Cell Metab. 27, 1138–1155.e6 (2018).
11
A. J. Lusis, M. M. Seldin, H. Allayee, B. J. Bennett, M. Civelek, R. C. Davis, E. Eskin, C. R. Farber, S. Hui, M. Mehrabian, F. Norheim, C. Pan, B. Parks, C. D. Rau, D. J. Smith, T. Vallim, Y. Wang, J. Wang, The Hybrid Mouse Diversity Panel: A resource for systems genetics analyses of metabolic and cardiovascular traits. J. Lipid Res. 57, 925–942 (2016).
12
P. U. Gandhi, H. K. Gaggin, M. M. Redfield, H. H. Chen, S. R. Stevens, K. J. Anstrom, M. J. Semigran, P. Liu, J. L. Januzzi Jr., Insulin-like growth factor-binding protein-7 as a biomarker of diastolic dysfunction and functional capacity in heart failure with preserved ejection fraction: Results From the RELAX trial. JACC Heart Fail. 4, 860–869 (2016).
13
G. G. Schiattarella, F. Altamirano, D. Tong, K. M. French, E. Villalobos, S. Y. Kim, X. Luo, N. Jiang, H. I. May, Z. V. Wang, T. M. Hill, P. P. A. Mammen, J. Huang, D. I. Lee, V. S. Hahn, K. Sharma, D. A. Kass, S. Lavandero, T. G. Gillette, J. A. Hill, Nitrosative stress drives heart failure with preserved ejection fraction. Nature 568, 351–356 (2019).
14
P. N. Walsh, Roles of platelets and factor XI in the initiation of blood coagulation by thrombin. Thromb. Haemost. 86, 75–82 (2001).
15
J. Emsley, P. A. McEwan, D. Gailani, Structure and function of factor XI. Blood 115, 2569–2577 (2010).
16
E. Ferkingstad, P. Sulem, B. A. Atlason, G. Sveinbjornsson, M. I. Magnusson, E. L. Styrmisdottir, K. Gunnarsdottir, A. Helgason, A. Oddsson, B. V. Halldorsson, B. O. Jensson, F. Zink, G. H. Halldorsson, G. Masson, G. A. Arnadottir, H. Katrinardottir, K. Juliusson, M. K. Magnusson, O. T. Magnusson, R. Fridriksdottir, S. Saevarsdottir, S. A. Gudjonsson, S. N. Stacey, S. Rognvaldsson, T. Eiriksdottir, T. A. Olafsdottir, V. Steinthorsdottir, V. Tragante, M. O. Ulfarsson, H. Stefansson, I. Jonsdottir, H. Holm, T. Rafnar, P. Melsted, J. Saemundsdottir, G. L. Norddahl, S. H. Lund, D. F. Gudbjartsson, U. Thorsteinsdottir, K. Stefansson, Large-scale integration of the plasma proteome with genetics and disease. Nat. Genet. 53, 1712–1721 (2021).
17
H. Kandis, H. Ozhan, S. Ordu, I. Erden, O. Caglar, C. Basar, S. Yalcin, R. Alemdar, M. Aydin, The prognostic value of mean platelet volume in decompensated heart failure. Emerg. Med. J. 28, 575–578 (2011).
18
C. Withaar, L. M. G. Meems, G. Markousis-Mavrogenis, C. J. Boogerd, H. H. W. Silljé, E. M. Schouten, M. M. Dokter, A. A. Voors, B. D. Westenbrink, C. S. P. Lam, R. A. de Boer, The effects of liraglutide and dapagliflozin on cardiac function and structure in a multi-hit mouse model of heart failure with preserved ejection fraction. Cardiovasc. Res. 117, 2108–2124 (2021).
19
M. A. Petersen, J. K. Ryu, K.-J. Chang, A. Etxeberria, S. Bardehle, A. S. Mendiola, W. Kamau-Devers, S. P. J. Fancy, A. Thor, E. A. Bushong, B. Baeza-Raja, C. A. Syme, M. D. Wu, P. E. Rios Coronado, A. Meyer-Franke, S. Yahn, L. Pous, J. K. Lee, C. Schachtrup, H. Lassmann, E. J. Huang, M. H. Han, M. Absinta, D. S. Reich, M. H. Ellisman, D. H. Rowitch, J. R. Chan, K. Akassoglou, Fibrinogen activates BMP signaling in oligodendrocyte progenitor cells and inhibits remyelination after vascular damage. Neuron 96, 1003–1012.e7 (2017).
20
Y. Shan, F. Yang, Z. Tang, C. Bi, S. Sun, Y. Zhang, H. Liu, Dexmedetomidine ameliorates the neurotoxicity of sevoflurane on the immature brain through the BMP/SMAD signaling pathway. Front. Neurosci. 12, 964 (2018).
21
K. Fujikawa, D. W. Chung, L. E. Hendrickson, E. W. Davie, Amino acid sequence of human factor XI, a blood coagulation factor with four tandem repeats that are highly homologous with plasma prekallikrein. Biochemistry 25, 2417–2424 (1986).
22
R. E. Saunders, N. Shiltagh, K. Gomez, G. Mellars, C. Cooper, D. J. Perry, E. G. Tuddenham, S. J. Perkins, Structural analysis of eight novel and 112 previously reported missense mutations in the interactive FXI mutation database reveals new insight on FXI deficiency. Thromb. Haemost. 102, 287–301 (2009).
23
K. E. Gregory, R. N. Ono, N. L. Charbonneau, C.-L. Kuo, D. R. Keene, H. P. Bächinger, L. Y. Sakai, The prodomain of BMP-7 targets the BMP-7 complex to the extracellular matrix. J. Biol. Chem. 280, 27970–27980 (2005).
24
X. Ge, Y. Yamaguchi, L. Zhao, L. Bury, P. Gresele, C. Berube, L. L. Leung, J. Morser, Prochemerin cleavage by factor XIa links coagulation and inflammation. Blood 131, 353–364 (2018).
25
D. Gailani, N. M. Lasky, G. J. Broze Jr., A murine model of factor XI deficiency. Blood Coagul. Fibrinolysis 8, 134–144 (1997).
26
L. Adamo, J. Yu, C. Rocha-Resende, A. Javaheri, R. D. Head, D. L. Mann, Proteomic Signatures of Heart Failure in Relation to Left Ventricular Ejection Fraction. J. Am. Coll. Cardiol. 76, 1982–1994 (2020).
27
J. M. Zhang, R.-Q. Yu, F.-Z. Wu, L. Qiao, X.-R. Wu, Y.-J. Fu, Y.-F. Liang, Y. Pang, C.-Y. Xie, BMP-2 alleviates heart failure with type 2 diabetes mellitus and doxorubicin-induced AC16 cell injury by inhibiting NLRP3 inflammasome-mediated pyroptosis. Exp. Ther. Med. 22, 897 (2021).
28
J. Banach, W. Gilewski, A. Słomka, K. Buszko, J. Błażejewski, D. Karasek, D. Rogowicz, E. Żekanowska, W. Sinkiewicz, Bone morphogenetic protein 6-a possible new player in pathophysiology of heart failure. Clin. Exp. Pharmacol. Physiol. 43, 1247–1250 (2016).
29
G. Zhou, J. Chen, C. Wu, P. Jiang, Y. Wang, Y. Zhang, Y. Jiang, X. Li, Deciphering the protein, modular connections and precision medicine for heart failure with preserved ejection fraction and hypertension based on TMT quantitative proteomics and molecular docking. Front. Physiol. 12, 607089 (2021).
30
W. Wu, D. Sinha, S. Shikov, C. K. Yip, T. Walz, P. C. Billings, J. D. Lear, P. N. Walsh, Factor XI homodimer structure is essential for normal proteolytic activation by factor XIIa, thrombin, and factor XIa. J. Biol. Chem. 283, 18655–18664 (2008).
31
P. H. Bolton-Maggs, Factor XI deficiency. Baillieres Clin. Haematol. 9, 355–368 (1996).
32
A. P. Wheeler, D. Gailani, Why factor XI deficiency is a clinical concern. Expert Rev. Hematol. 9, 629–637 (2016).
33
R. Asakai, D. W. Chung, E. W. Davie, U. Seligsohn, Factor XI deficiency in Ashkenazi Jews in Israel. N. Engl. J. Med. 325, 153–158 (1991).
34
D. Appiah, O. E. Fashanu, S. R. Heckbert, M. Cushman, B. M. Psaty, A. R. Folsom, Relation of coagulation factor XI with incident coronary heart disease and stroke: The Cardiovascular Health Study. Blood Coagul. Fibrinolysis 28, 389–392 (2017).
35
M. Zabczyk, S. Butenas, I. Palka, J. Nessler, A. Undas, Active tissue factor and activated factor XI in circulating blood of patients with systolic heart failure due to ischemic cardiomyopathy. Pol. Arch. Med. Wewn. 120, 334–340 (2010).
36
I. Stroo, S. Zeerleder, C. Ding, B. M. Luken, J. J. T. H. Roelofs, O. J. de Boer, J. C. M. Meijers, F. J. Castellino, C. van ’t Veer, T. van der Poll, Coagulation factor XI improves host defence during murine pneumonia-derived sepsis independent of factor XII activation. Thromb. Haemost. 117, 1601–1614 (2017).
37
A. T. P. Ngo, K. R. Jordan, P. A. Mueller, M. W. Hagen, S. E. Reitsma, C. Puy, A. S. Revenko, C. U. Lorentz, E. I. Tucker, Q. Cheng, M. T. Hinds, S. Fazio, B. P. Monia, D. Gailani, A. Gruber, H. Tavori, O. J. T. McCarty, Pharmacological targeting of coagulation factor XI mitigates the development of experimental atherosclerosis in low-density lipoprotein receptor-deficient mice. J. Thromb. Haemost. 19, 1001–1017 (2021).
38
C. Puy, J. Pang, S. E. Reitsma, C. U. Lorentz, E. I. Tucker, D. Gailani, A. Gruber, F. Lupu, O. J. T. McCarty, Cross-talk between the complement pathway and the contact activation system of coagulation: Activated factor XI neutralizes complement factor H. J. Immunol. 206, 1784–1792 (2021).
39
E. I. Tucker, N. G. Verbout, P. Y. Leung, S. Hurst, O. J. T. McCarty, D. Gailani, A. Gruber, Inhibition of factor XI activation attenuates inflammation and coagulopathy while improving the survival of mouse polymicrobial sepsis. Blood 119, 4762–4768 (2012).
40
C. E. Bane Jr., I. Ivanov, A. Matafonov, K. L. Boyd, Q. Cheng, E. R. Sherwood, E. I. Tucker, S. T. Smiley, O. J. T. McCarty, A. Gruber, D. Gailani, Factor XI deficiency alters the cytokine response and activation of contact proteases during polymicrobial sepsis in mice. PLOS ONE 11, e0152968 (2016).
41
V. Rovite, U. Maurins, K. Megnis, I. Vaivade, R. Pečulis, J. Rits, S. Prave, J. Klovins, Association of F11 polymorphism rs2289252 with deep vein thrombosis and related phenotypes in population of Latvia. Thromb. Res. 134, 659–663 (2014).
42
Y. Li, I. D. Bezemer, C. M. Rowland, C. H. Tong, A. R. Arellano, J. J. Catanese, J. J. Devlin, P. H. Reitsma, L. A. Bare, F. R. Rosendaal, Genetic variants associated with deep vein thrombosis: The F11 locus. J. Thromb. Haemost. 7, 1802–1808 (2009).
43
M. Chong, J. Sjaarda, M. Pigeyre, P. Mohammadi-Shemirani, R. Lali, A. Shoamanesh, H. C. Gerstein, G. Paré, Novel drug targets for ischemic stroke identified through mendelian randomization analysis of the blood proteome. Circulation 140, 819–830 (2019).
44
C. Pan, R script used to perform the volcano plots and PCA plot (F11_SF7) for: Y. Cao, Y. Wang, Z. Zhou, C. Pan, L. Jiang, Z. Zhou, Y. Meng, S. Charugundla, T. Li, H. Allayee, M. M. Seldin, A. J. Lusis, Liver-heart cross-talk mediated by coagulation factor XI protects against heart failure, Zenodo (2022); https://doi.org/10.5281/zenodo.6961041.
45
B. J. Bennett, C. R. Farber, L. Orozco, H. M. Kang, A. Ghazalpour, N. Siemers, M. Neubauer, I. Neuhaus, R. Yordanova, B. Guan, A. Truong, W. P. Yang, A. He, P. Kayne, P. Gargalovic, T. Kirchgessner, C. Pan, L. W. Castellani, E. Kostem, N. Furlotte, T. A. Drake, E. Eskin, A. J. Lusis, A high-resolution association mapping panel for the dissection of complex traits in mice. Genome Res. 20, 281–290 (2010).
46
Y. Wang, S. Huang, V. P. Sah, J. Ross Jr., J. H. Brown, J. Han, K. R. Chien, Cardiac muscle cell hypertrophy and apoptosis induced by distinct members of the p38 mitogen-activated protein kinase family. J. Biol. Chem. 273, 2161–2168 (1998).
47
A. Sharma, G. Li, K. Rajarajan, R. Hamaguchi, P. W. Burridge, S. M. Wu, Derivation of highly purified cardiomyocytes from human induced pluripotent stem cells using small molecule-modulated differentiation and subsequent glucose starvation. J. Vis. Exp. ( 97): (2015).
48
F. Norheim, Y. Hasin-Brumshtein, L. Vergnes, K. Chella Krishnan, C. Pan, M. M. Seldin, S. T. Hui, M. Mehrabian, Z. Zhou, S. Gupta, B. W. Parks, A. Walch, K. Reue, S. M. Hofmann, A. P. Arnold, A. J. Lusis, Gene-by-sex interactions in mitochondrial functions and cardio-metabolic traits. Cell Metab. 29, 932–949.e4 (2019).
49
P. Langfelder, S. Horvath, WGCNA: An R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).
50
C. Lippert, J. Listgarten, Y. Liu, C. M. Kadie, R. I. Davidson, D. Heckerman, FaST linear mixed models for genome-wide association studies. Nat. Methods 8, 833–835 (2011).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Embedded figures cannot be submitted, and we discourage the use of figures within eLetters in general. If a figure is essential, please include a link to the figure within the text of the eLetter. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science
Volume 377 | Issue 6613
23 September 2022

Submission history

Received: 2 November 2021
Accepted: 5 August 2022
Published in print: 23 September 2022

Permissions

Request permissions for this article.

Acknowledgments

Funding: This work was supported by the National Institutes of Health (grants DK120342 and HL147883; grants HL138193, DK130640, and DK097771 to M.M.S.; grants R01HL133169 and R01HL148110 to H.A.; and grant DK125354 to Z.Z.).
Author contributions: Y.C., M.M.S., and A.J.L. designed the experiments. Y.C., Y.W., Z.Z., L.J., Z.Z., Y.M., S.C., and T.L. performed the experiments. Y.C., L.J., M.M.S., and C.P. analyzed raw data. Z.Z., H.A., M.M.S., and A.J.L. reviewed the data and made substantial contributions to improving the studies. Y.C. and A.J.L. wrote the manuscript, which was reviewed by all authors.
Competing interests: The authors declare no competing interests.
Data and materials availability: All data supporting the conclusions in this manuscript can be found in the main text or the supplementary materials. RNA-Seq data were deposited to the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/) under accession number GSE200496. The R script used to perform the volcano plots and PCA plot are available at Zenodo (44).
License information: Copyright © 2022 the authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original US government works. https://www.science.org/about/science-licenses-journal-article-reuse

Authors

Affiliations

Funding Information

FUNDING

Notes

*
Corresponding author. Email: jlusis@mednet.ucla.edu

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. FNDC5/Irisin Inhibits the Inflammatory Response and Mediates the Aerobic Exercise-Induced Improvement of Liver Injury after Myocardial Infarction, International Journal of Molecular Sciences, 24, 4, (4159), (2023).https://doi.org/10.3390/ijms24044159
    Crossref
  2. Subtyping based on immune cell fractions reveal heterogeneity of cardiac fibrosis in end-stage heart failure, Frontiers in Immunology, 14, (2023).https://doi.org/10.3389/fimmu.2023.1053793
    Crossref
  3. Pharmacology and Clinical Development of Factor XI Inhibitors, Circulation, 147, 11, (897-913), (2023).https://doi.org/10.1161/CIRCULATIONAHA.122.062353
    Crossref
  4. An update on hypertension, Acta Physiologica, 237, 3, (2023).https://doi.org/10.1111/apha.13942
    Crossref
  5. Substrates, Cofactors, and Cellular Targets of Coagulation Factor XIa, Seminars in Thrombosis and Hemostasis, (2023).https://doi.org/10.1055/s-0043-1764469
    Crossref
  6. Factor XI inhibitors in patients with cardiovascular disease and a high risk of bleeding: a cautionary tale, Nature Reviews Cardiology, (2023).https://doi.org/10.1038/s41569-023-00872-4
    Crossref
  7. Factor XIa Inhibitors in Anticoagulation Therapy: Recent Advances and Perspectives, Journal of Medicinal Chemistry, (2023).https://doi.org/10.1021/acs.jmedchem.2c02130
    Crossref
  8. Dysregulated systemic metabolism in a Down syndrome mouse model, Molecular Metabolism, 68, (101666), (2023).https://doi.org/10.1016/j.molmet.2022.101666
    Crossref
  9. A coagulation factor moonlights in the heart, Science, 377, 6613, (1382-1383), (2022)./doi/10.1126/science.ade2538
    Abstract
  10. Liver-derived factor XI protects against HFpEF, Nature Reviews Cardiology, 19, 12, (781-781), (2022).https://doi.org/10.1038/s41569-022-00800-y
    Crossref
  11. See more
Loading...

View Options

View options

PDF format

Download this article as a PDF file

Download PDF

Media

Figures

Fig. 1. Systems genetics analysis of cross-tissue correlations identifies proteins mediating liver-heart cross-talk.
(A) Schematic illustrating the identification of the liver-heart interaction using 100 inbred strains of mice (HMDP). The correlation between the secreted factors (from the liver) and cardiac gene expression (RNA-Seq) was used for liver-heart predictions. This framework identified peptides secreted by the liver and strongly associated with the cardiac gene network. n = 4 to 20 mice for each strain. (B) Distribution of significance score for all liver genes across all heart gene expression in 100 strains (left). List shows the top 20 genes potentially mediating liver-heart communication (right). (C) Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis of F11 expression across indicated tissues in C57BL/6J mice (n = 4). All data are presented as means ± SEM. (D) Pathway enrichment derived from heart genes correlated with liver F11 expression. (E) GWAS loci for indicated clinical traits in human populations. The GWAS catalog and PhenoScanner databases consist of human genotype-phenotype associations from publicly available genetic association studies.
Fig. 2. FXI overexpression reverses HFpEF-induced diastolic dysfunction, inflammation, and fibrosis.
(A) Thirty inbred strains of male mice were subjected to HFD + l-NAME to induce HFpEF. Plasma FXI concentrations and diastolic function (E/eʹ ratio) were assessed after 7 weeks of feeding. Plasma FXI concentrations were inversely correlated with diastolic dysfunction. (B to L) C57BL/6J male mice were injected with AAV8 containing the cDNA sequence for GFP or F11 and then fed with HFD + l-NAME for 7 weeks. Western blotting shows liver FXI protein (B), plasma FXI concentrations (C), E/A ratio (D), E/eʹ ratio (E), representative images of echocardiography (F), LVEF (G), heart weight/tibia length ratio (H), lung weight [wet/dry ratio (I)], running distance (J), thrombin-antithrombin complexes [TAT (K)], and relative mRNA expression of indicated genes in the heart (L). n = 4 for chow in (D) to (H); in other panels, n = 8 to 10. (M and N) C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 were on HFD + l-NAME for 7 weeks (n = 5). Representative images of immunohistochemistry staining (M) and quantification of positive cells (N) showing inflammatory cell infiltration in the heart tissue. (O and P) C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 were given a chow diet or HFD + l-NAME for 7 weeks (n = 5). Representative images of Masson’s trichrome staining (O) and quantification (P) show fibrosis in the heart tissue. Each point represents a mouse. All data are presented as means ± SEM. ns, not significant. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 by two-way ANOVA [(D) to (J) and (P)] or by Student’s t test [(B) and (C) and (K) to (N)]. For (A) to (C) and (K) to (N), all mice were on HFD + l-NAME. LYM, lymphocytes; MONO, monocytes; GRAN, granulocytes.
Fig. 3. FXI activates BMP-SMAD1/5 pathway in the heart.
(A) Western blotting and quantification showing protein levels in heart tissue from C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 and fed with 7 weeks of HFD + l-NAME. Actin served as the loading control. n = 5. (B) qRT-PCR analysis showing the mRNA levels of Col5a3 in the indicated tissue from C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 and fed HFD + l-NAME for 7 weeks. Heart, P < 0.001; others, not significant. n = 8. (C and D) NRVMs were treated with control or human FXIa protein (1 μg/ml) with medium containing control or phenylephrine (PE, 100 μM) for 24 hours. p-SMAD1/5 (C) and the indicated genes (D) were examined. Actin served as the loading control. n = 6. (E to H) C57BL/6J male mice were injected with AAV8-GFP or AAV8-F11 with DMH1 and fed with HFD + l-NAME for 7 weeks. Heart p-SMAD1/5 level (E), heart weight/tibia length ratio (F), E/eʹ ratio (G), and LVEF (H) were determined. n = 3 for (E) and n = 8 for (F) to (H). (I to M) C57BL/6J male mice were injected with AAV8-GFP, AAV8-F11, or AAV8-F11-Mut (mF11-Mut2) and fed with HFD + l-NAME for 7 weeks. Plasma FXI levels (I), heart p-SMAD1/5 protein level (J), heart weight/tibia length ratio (K), E/eʹ ratio (L), and LVEF (M) were measured. n = 5 for (I), n = 6 for (J), and n = 10 to 20 for (K) to (M). (N) C57BL/6J male mice were injected with either AAV8-GFP or AAV8-F11 and then fed with HFD + l-NAME for 7 weeks. BMP7 proteins in unprocessed monomer, growth factor dimer, and monomer under nonreducing condition were determined. (O) NRVMs were treated with control or human FXIa protein (1 μg/ml) plus negative control or Bmp7 siRNA with medium containing PE (100 μM) for 24 hours. p-SMAD1/5 and Tcap proteins were examined. Actin served as the loading control. n = 3. (P) NRVMs were treated with control or human FXIa protein (1 μg/ml), BMP7 antibody (no antibody control, 1:100 and 1:50), with medium containing PE (100 μM) for 2 hours, and the p-SMAD1/5 level was determined. Actin served as the loading control. n = 4. Each point represents a mouse. All data are presented as means ± SEM. ns, not significant. *P < 0.05, **P < 0.01, and ***P < 0.001 by two-way ANOVA [(D) to (H)], one-way ANOVA [(I) to (M)], or Student’s t test [(A) to (C)].
Fig. 4. Reduced FXI concentrations are associated with diastolic dysfunction in mice and humans.
Heterozygous B6.129X1-F11tm1Gjb/J (F11-Het) mice and WT littermates at 8 weeks of age were subjected to HFD + l-NAME for 7 weeks. n = 8 for WT and n = 6 for F11-Het. (A) qRT-PCR showing F11 mRNA in the indicated tissues from WT and F11-Het mice. n = 4. (B) Western blotting showing p-SMAD1/5 in the hearts of WT mice fed with chow diet (Chow), WT, and F11-Het mice fed with HFD + l-NAME for 7 weeks. n = 5. (C to G) Representative images of echocardiography (C), E/A ratio (D), E/eʹ ratio (E), LV mass (F), and LVEF (G) were examined at baseline (BSL) and after 7 weeks of HFD + l-NAME feeding (HFpEF). n = 8 for WT and n = 6 for F11-Het. (H to J) Heart weight/tibia length ratio (H), lung weight [wet/dry ratio (I)], and running distance (J) were examined after 7 weeks of HFD + l-NAME feeding (HFpEF). n = 8 for WT and n = 6 for F11-Het. (K) Plasma FXI protein in non-HFpEF controls (NHF, n = 20) and HFpEF patients (n = 21). (L and M) Plasma FXI protein was inversely correlated with E/eʹ ratio in all participants (L), including HFpEF patients (M). (N) Illustration summarizing FXI-mediated liver-heart cross-talk in protecting against heart failure. Using a bioinformatic framework that integrates global liver-heart transcriptome and cardiometabolic trait data from the HMDP, we found that coagulation FXI, secreted by the liver, exhibits cardioprotective effects on the progression of HFpEF. FXI overexpression in the liver mitigates the diastolic dysfunction, inflammation, and fibrosis induced by HFpEF. FXIa cleaves the BMP7 precursor and activates the BMP7-SMAD1/5 pathway in the heart to mediate the anti-inflammatory and anti-fibrotic effects. Each point represents a mouse. All data are presented as means ± SEM. ns, not significant. *P < 0.05, **P < 0.01, and ***P < 0.001 by two-way ANOVA [(D) to (G)], one-way ANOVA (B), or Student’s t test [(A) and (H) to (K)].

Multimedia

Tables

Share

Share

Share article link

Share on social media

References

References

1
J. M. Friedman, J. L. Halaas, Leptin and the regulation of body weight in mammals. Nature 395, 763–770 (1998).
2
S. Møller, M. Bernardi, Interactions of the heart and the liver. Eur. Heart J. 34, 2804–2811 (2013).
3
K. K. Baskin, A. L. Bookout, E. N. Olson, The heart-liver metabolic axis: Defective communication exacerbates disease. EMBO Mol. Med. 6, 436–438 (2014).
4
M. Packer, Atrial fibrillation and heart failure with preserved ejection fraction in patients with nonalcoholic fatty liver disease. Am. J. Med. 133, 170–177 (2020).
5
L. Pacifico, M. Di Martino, A. De Merulis, M. Bezzi, J. F. Osborn, C. Catalano, C. Chiesa, Left ventricular dysfunction in obese children and adolescents with nonalcoholic fatty liver disease. Hepatology 59, 461–470 (2014).
6
Y. Cao, C. Pan, Y.-C. Wang, Z. Zhou, V. Jedian, Y. Meng, G. Campbell, K. Guardino, C. Li, J. Wang, A. J. Lusis, Identification of DNA damage repair enzyme Ascc2 as causal for heart failure with preserved ejection fraction. Circulation 145, 1102–1104 (2022).
7
S. M. Dunlay, V. L. Roger, M. M. Redfield, Epidemiology of heart failure with preserved ejection fraction. Nat. Rev. Cardiol. 14, 591–602 (2017).
8
K. Hogg, K. Swedberg, J. McMurray, Heart failure with preserved left ventricular systolic function; epidemiology, clinical characteristics, and prognosis. J. Am. Coll. Cardiol. 43, 317–327 (2004).
9
T. E. Owan, D. O. Hodge, R. M. Herges, S. J. Jacobsen, V. L. Roger, M. M. Redfield, Trends in prevalence and outcome of heart failure with preserved ejection fraction. N. Engl. J. Med. 355, 251–259 (2006).
10
M. M. Seldin, S. Koplev, P. Rajbhandari, L. Vergnes, G. M. Rosenberg, Y. Meng, C. Pan, T. M. N. Phuong, R. Gharakhanian, N. Che, S. Mäkinen, D. M. Shih, M. Civelek, B. W. Parks, E. D. Kim, F. Norheim, K. Chella Krishnan, Y. Hasin-Brumshtein, M. Mehrabian, M. Laakso, C. A. Drevon, H. A. Koistinen, P. Tontonoz, K. Reue, R. M. Cantor, J. L. M. Björkegren, A. J. Lusis, A strategy for discovery of endocrine interactions with application to whole-body metabolism. Cell Metab. 27, 1138–1155.e6 (2018).
11
A. J. Lusis, M. M. Seldin, H. Allayee, B. J. Bennett, M. Civelek, R. C. Davis, E. Eskin, C. R. Farber, S. Hui, M. Mehrabian, F. Norheim, C. Pan, B. Parks, C. D. Rau, D. J. Smith, T. Vallim, Y. Wang, J. Wang, The Hybrid Mouse Diversity Panel: A resource for systems genetics analyses of metabolic and cardiovascular traits. J. Lipid Res. 57, 925–942 (2016).
12
P. U. Gandhi, H. K. Gaggin, M. M. Redfield, H. H. Chen, S. R. Stevens, K. J. Anstrom, M. J. Semigran, P. Liu, J. L. Januzzi Jr., Insulin-like growth factor-binding protein-7 as a biomarker of diastolic dysfunction and functional capacity in heart failure with preserved ejection fraction: Results From the RELAX trial. JACC Heart Fail. 4, 860–869 (2016).
13
G. G. Schiattarella, F. Altamirano, D. Tong, K. M. French, E. Villalobos, S. Y. Kim, X. Luo, N. Jiang, H. I. May, Z. V. Wang, T. M. Hill, P. P. A. Mammen, J. Huang, D. I. Lee, V. S. Hahn, K. Sharma, D. A. Kass, S. Lavandero, T. G. Gillette, J. A. Hill, Nitrosative stress drives heart failure with preserved ejection fraction. Nature 568, 351–356 (2019).
14
P. N. Walsh, Roles of platelets and factor XI in the initiation of blood coagulation by thrombin. Thromb. Haemost. 86, 75–82 (2001).
15
J. Emsley, P. A. McEwan, D. Gailani, Structure and function of factor XI. Blood 115, 2569–2577 (2010).
16
E. Ferkingstad, P. Sulem, B. A. Atlason, G. Sveinbjornsson, M. I. Magnusson, E. L. Styrmisdottir, K. Gunnarsdottir, A. Helgason, A. Oddsson, B. V. Halldorsson, B. O. Jensson, F. Zink, G. H. Halldorsson, G. Masson, G. A. Arnadottir, H. Katrinardottir, K. Juliusson, M. K. Magnusson, O. T. Magnusson, R. Fridriksdottir, S. Saevarsdottir, S. A. Gudjonsson, S. N. Stacey, S. Rognvaldsson, T. Eiriksdottir, T. A. Olafsdottir, V. Steinthorsdottir, V. Tragante, M. O. Ulfarsson, H. Stefansson, I. Jonsdottir, H. Holm, T. Rafnar, P. Melsted, J. Saemundsdottir, G. L. Norddahl, S. H. Lund, D. F. Gudbjartsson, U. Thorsteinsdottir, K. Stefansson, Large-scale integration of the plasma proteome with genetics and disease. Nat. Genet. 53, 1712–1721 (2021).
17
H. Kandis, H. Ozhan, S. Ordu, I. Erden, O. Caglar, C. Basar, S. Yalcin, R. Alemdar, M. Aydin, The prognostic value of mean platelet volume in decompensated heart failure. Emerg. Med. J. 28, 575–578 (2011).
18
C. Withaar, L. M. G. Meems, G. Markousis-Mavrogenis, C. J. Boogerd, H. H. W. Silljé, E. M. Schouten, M. M. Dokter, A. A. Voors, B. D. Westenbrink, C. S. P. Lam, R. A. de Boer, The effects of liraglutide and dapagliflozin on cardiac function and structure in a multi-hit mouse model of heart failure with preserved ejection fraction. Cardiovasc. Res. 117, 2108–2124 (2021).
19
M. A. Petersen, J. K. Ryu, K.-J. Chang, A. Etxeberria, S. Bardehle, A. S. Mendiola, W. Kamau-Devers, S. P. J. Fancy, A. Thor, E. A. Bushong, B. Baeza-Raja, C. A. Syme, M. D. Wu, P. E. Rios Coronado, A. Meyer-Franke, S. Yahn, L. Pous, J. K. Lee, C. Schachtrup, H. Lassmann, E. J. Huang, M. H. Han, M. Absinta, D. S. Reich, M. H. Ellisman, D. H. Rowitch, J. R. Chan, K. Akassoglou, Fibrinogen activates BMP signaling in oligodendrocyte progenitor cells and inhibits remyelination after vascular damage. Neuron 96, 1003–1012.e7 (2017).
20
Y. Shan, F. Yang, Z. Tang, C. Bi, S. Sun, Y. Zhang, H. Liu, Dexmedetomidine ameliorates the neurotoxicity of sevoflurane on the immature brain through the BMP/SMAD signaling pathway. Front. Neurosci. 12, 964 (2018).
21
K. Fujikawa, D. W. Chung, L. E. Hendrickson, E. W. Davie, Amino acid sequence of human factor XI, a blood coagulation factor with four tandem repeats that are highly homologous with plasma prekallikrein. Biochemistry 25, 2417–2424 (1986).
22
R. E. Saunders, N. Shiltagh, K. Gomez, G. Mellars, C. Cooper, D. J. Perry, E. G. Tuddenham, S. J. Perkins, Structural analysis of eight novel and 112 previously reported missense mutations in the interactive FXI mutation database reveals new insight on FXI deficiency. Thromb. Haemost. 102, 287–301 (2009).
23
K. E. Gregory, R. N. Ono, N. L. Charbonneau, C.-L. Kuo, D. R. Keene, H. P. Bächinger, L. Y. Sakai, The prodomain of BMP-7 targets the BMP-7 complex to the extracellular matrix. J. Biol. Chem. 280, 27970–27980 (2005).
24
X. Ge, Y. Yamaguchi, L. Zhao, L. Bury, P. Gresele, C. Berube, L. L. Leung, J. Morser, Prochemerin cleavage by factor XIa links coagulation and inflammation. Blood 131, 353–364 (2018).
25
D. Gailani, N. M. Lasky, G. J. Broze Jr., A murine model of factor XI deficiency. Blood Coagul. Fibrinolysis 8, 134–144 (1997).
26
L. Adamo, J. Yu, C. Rocha-Resende, A. Javaheri, R. D. Head, D. L. Mann, Proteomic Signatures of Heart Failure in Relation to Left Ventricular Ejection Fraction. J. Am. Coll. Cardiol. 76, 1982–1994 (2020).
27
J. M. Zhang, R.-Q. Yu, F.-Z. Wu, L. Qiao, X.-R. Wu, Y.-J. Fu, Y.-F. Liang, Y. Pang, C.-Y. Xie, BMP-2 alleviates heart failure with type 2 diabetes mellitus and doxorubicin-induced AC16 cell injury by inhibiting NLRP3 inflammasome-mediated pyroptosis. Exp. Ther. Med. 22, 897 (2021).
28
J. Banach, W. Gilewski, A. Słomka, K. Buszko, J. Błażejewski, D. Karasek, D. Rogowicz, E. Żekanowska, W. Sinkiewicz, Bone morphogenetic protein 6-a possible new player in pathophysiology of heart failure. Clin. Exp. Pharmacol. Physiol. 43, 1247–1250 (2016).
29
G. Zhou, J. Chen, C. Wu, P. Jiang, Y. Wang, Y. Zhang, Y. Jiang, X. Li, Deciphering the protein, modular connections and precision medicine for heart failure with preserved ejection fraction and hypertension based on TMT quantitative proteomics and molecular docking. Front. Physiol. 12, 607089 (2021).
30
W. Wu, D. Sinha, S. Shikov, C. K. Yip, T. Walz, P. C. Billings, J. D. Lear, P. N. Walsh, Factor XI homodimer structure is essential for normal proteolytic activation by factor XIIa, thrombin, and factor XIa. J. Biol. Chem. 283, 18655–18664 (2008).
31
P. H. Bolton-Maggs, Factor XI deficiency. Baillieres Clin. Haematol. 9, 355–368 (1996).
32
A. P. Wheeler, D. Gailani, Why factor XI deficiency is a clinical concern. Expert Rev. Hematol. 9, 629–637 (2016).
33
R. Asakai, D. W. Chung, E. W. Davie, U. Seligsohn, Factor XI deficiency in Ashkenazi Jews in Israel. N. Engl. J. Med. 325, 153–158 (1991).
34
D. Appiah, O. E. Fashanu, S. R. Heckbert, M. Cushman, B. M. Psaty, A. R. Folsom, Relation of coagulation factor XI with incident coronary heart disease and stroke: The Cardiovascular Health Study. Blood Coagul. Fibrinolysis 28, 389–392 (2017).
35
M. Zabczyk, S. Butenas, I. Palka, J. Nessler, A. Undas, Active tissue factor and activated factor XI in circulating blood of patients with systolic heart failure due to ischemic cardiomyopathy. Pol. Arch. Med. Wewn. 120, 334–340 (2010).
36
I. Stroo, S. Zeerleder, C. Ding, B. M. Luken, J. J. T. H. Roelofs, O. J. de Boer, J. C. M. Meijers, F. J. Castellino, C. van ’t Veer, T. van der Poll, Coagulation factor XI improves host defence during murine pneumonia-derived sepsis independent of factor XII activation. Thromb. Haemost. 117, 1601–1614 (2017).
37
A. T. P. Ngo, K. R. Jordan, P. A. Mueller, M. W. Hagen, S. E. Reitsma, C. Puy, A. S. Revenko, C. U. Lorentz, E. I. Tucker, Q. Cheng, M. T. Hinds, S. Fazio, B. P. Monia, D. Gailani, A. Gruber, H. Tavori, O. J. T. McCarty, Pharmacological targeting of coagulation factor XI mitigates the development of experimental atherosclerosis in low-density lipoprotein receptor-deficient mice. J. Thromb. Haemost. 19, 1001–1017 (2021).
38
C. Puy, J. Pang, S. E. Reitsma, C. U. Lorentz, E. I. Tucker, D. Gailani, A. Gruber, F. Lupu, O. J. T. McCarty, Cross-talk between the complement pathway and the contact activation system of coagulation: Activated factor XI neutralizes complement factor H. J. Immunol. 206, 1784–1792 (2021).
39
E. I. Tucker, N. G. Verbout, P. Y. Leung, S. Hurst, O. J. T. McCarty, D. Gailani, A. Gruber, Inhibition of factor XI activation attenuates inflammation and coagulopathy while improving the survival of mouse polymicrobial sepsis. Blood 119, 4762–4768 (2012).
40
C. E. Bane Jr., I. Ivanov, A. Matafonov, K. L. Boyd, Q. Cheng, E. R. Sherwood, E. I. Tucker, S. T. Smiley, O. J. T. McCarty, A. Gruber, D. Gailani, Factor XI deficiency alters the cytokine response and activation of contact proteases during polymicrobial sepsis in mice. PLOS ONE 11, e0152968 (2016).
41
V. Rovite, U. Maurins, K. Megnis, I. Vaivade, R. Pečulis, J. Rits, S. Prave, J. Klovins, Association of F11 polymorphism rs2289252 with deep vein thrombosis and related phenotypes in population of Latvia. Thromb. Res. 134, 659–663 (2014).
42
Y. Li, I. D. Bezemer, C. M. Rowland, C. H. Tong, A. R. Arellano, J. J. Catanese, J. J. Devlin, P. H. Reitsma, L. A. Bare, F. R. Rosendaal, Genetic variants associated with deep vein thrombosis: The F11 locus. J. Thromb. Haemost. 7, 1802–1808 (2009).
43
M. Chong, J. Sjaarda, M. Pigeyre, P. Mohammadi-Shemirani, R. Lali, A. Shoamanesh, H. C. Gerstein, G. Paré, Novel drug targets for ischemic stroke identified through mendelian randomization analysis of the blood proteome. Circulation 140, 819–830 (2019).
44
C. Pan, R script used to perform the volcano plots and PCA plot (F11_SF7) for: Y. Cao, Y. Wang, Z. Zhou, C. Pan, L. Jiang, Z. Zhou, Y. Meng, S. Charugundla, T. Li, H. Allayee, M. M. Seldin, A. J. Lusis, Liver-heart cross-talk mediated by coagulation factor XI protects against heart failure, Zenodo (2022); https://doi.org/10.5281/zenodo.6961041.
45
B. J. Bennett, C. R. Farber, L. Orozco, H. M. Kang, A. Ghazalpour, N. Siemers, M. Neubauer, I. Neuhaus, R. Yordanova, B. Guan, A. Truong, W. P. Yang, A. He, P. Kayne, P. Gargalovic, T. Kirchgessner, C. Pan, L. W. Castellani, E. Kostem, N. Furlotte, T. A. Drake, E. Eskin, A. J. Lusis, A high-resolution association mapping panel for the dissection of complex traits in mice. Genome Res. 20, 281–290 (2010).
46
Y. Wang, S. Huang, V. P. Sah, J. Ross Jr., J. H. Brown, J. Han, K. R. Chien, Cardiac muscle cell hypertrophy and apoptosis induced by distinct members of the p38 mitogen-activated protein kinase family. J. Biol. Chem. 273, 2161–2168 (1998).
47
A. Sharma, G. Li, K. Rajarajan, R. Hamaguchi, P. W. Burridge, S. M. Wu, Derivation of highly purified cardiomyocytes from human induced pluripotent stem cells using small molecule-modulated differentiation and subsequent glucose starvation. J. Vis. Exp. ( 97): (2015).
48
F. Norheim, Y. Hasin-Brumshtein, L. Vergnes, K. Chella Krishnan, C. Pan, M. M. Seldin, S. T. Hui, M. Mehrabian, Z. Zhou, S. Gupta, B. W. Parks, A. Walch, K. Reue, S. M. Hofmann, A. P. Arnold, A. J. Lusis, Gene-by-sex interactions in mitochondrial functions and cardio-metabolic traits. Cell Metab. 29, 932–949.e4 (2019).
49
P. Langfelder, S. Horvath, WGCNA: An R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).
50
C. Lippert, J. Listgarten, Y. Liu, C. M. Kadie, R. I. Davidson, D. Heckerman, FaST linear mixed models for genome-wide association studies. Nat. Methods 8, 833–835 (2011).
1800
1801
1802
1803
1804
View figure
Fig. 1
Fig. 1. Systems genetics analysis of cross-tissue correlations identifies proteins mediating liver-heart cross-talk.
(A) Schematic illustrating the identification of the liver-heart interaction using 100 inbred strains of mice (HMDP). The correlation between the secreted factors (from the liver) and cardiac gene expression (RNA-Seq) was used for liver-heart predictions. This framework identified peptides secreted by the liver and strongly associated with the cardiac gene network. n = 4 to 20 mice for each strain. (B) Distribution of significance score for all liver genes across all heart gene expression in 100 strains (left). List shows the top 20 genes potentially mediating liver-heart communication (right). (C) Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis of F11 expression across indicated tissues in C57BL/6J mice (n = 4). All data are presented as means ± SEM. (D) Pathway enrichment derived from heart genes correlated with liver F11 expression. (E) GWAS loci for indicated clinical traits in human populations. The GWAS catalog and PhenoScanner databases consist of human genotype-phenotype associations from publicly available genetic association studies.
View figure
Fig. 2
Fig. 2. FXI overexpression reverses HFpEF-induced diastolic dysfunction, inflammation, and fibrosis.
(A) Thirty inbred strains of male mice were subjected to HFD + l-NAME to induce HFpEF. Plasma FXI concentrations and diastolic function (E/eʹ ratio) were assessed after 7 weeks of feeding. Plasma FXI concentrations were inversely correlated with diastolic dysfunction. (B to L) C57BL/6J male mice were injected with AAV8 containing the cDNA sequence for GFP or F11 and then fed with HFD + l-NAME for 7 weeks. Western blotting shows liver FXI protein (B), plasma FXI concentrations (C), E/A ratio (D), E/eʹ ratio (E), representative images of echocardiography (F), LVEF (G), heart weight/tibia length ratio (H), lung weight [wet/dry ratio (I)], running distance (J), thrombin-antithrombin complexes [TAT (K)], and relative mRNA expression of indicated genes in the heart (L). n = 4 for chow in (D) to (H); in other panels, n = 8 to 10. (M and N) C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 were on HFD + l-NAME for 7 weeks (n = 5). Representative images of immunohistochemistry staining (M) and quantification of positive cells (N) showing inflammatory cell infiltration in the heart tissue. (O and P) C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 were given a chow diet or HFD + l-NAME for 7 weeks (n = 5). Representative images of Masson’s trichrome staining (O) and quantification (P) show fibrosis in the heart tissue. Each point represents a mouse. All data are presented as means ± SEM. ns, not significant. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 by two-way ANOVA [(D) to (J) and (P)] or by Student’s t test [(B) and (C) and (K) to (N)]. For (A) to (C) and (K) to (N), all mice were on HFD + l-NAME. LYM, lymphocytes; MONO, monocytes; GRAN, granulocytes.
View figure
Fig. 3
Fig. 3. FXI activates BMP-SMAD1/5 pathway in the heart.
(A) Western blotting and quantification showing protein levels in heart tissue from C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 and fed with 7 weeks of HFD + l-NAME. Actin served as the loading control. n = 5. (B) qRT-PCR analysis showing the mRNA levels of Col5a3 in the indicated tissue from C57BL/6J male mice injected with AAV8-GFP or AAV8-F11 and fed HFD + l-NAME for 7 weeks. Heart, P < 0.001; others, not significant. n = 8. (C and D) NRVMs were treated with control or human FXIa protein (1 μg/ml) with medium containing control or phenylephrine (PE, 100 μM) for 24 hours. p-SMAD1/5 (C) and the indicated genes (D) were examined. Actin served as the loading control. n = 6. (E to H) C57BL/6J male mice were injected with AAV8-GFP or AAV8-F11 with DMH1 and fed with HFD + l-NAME for 7 weeks. Heart p-SMAD1/5 level (E), heart weight/tibia length ratio (F), E/eʹ ratio (G), and LVEF (H) were determined. n = 3 for (E) and n = 8 for (F) to (H). (I to M) C57BL/6J male mice were injected with AAV8-GFP, AAV8-F11, or AAV8-F11-Mut (mF11-Mut2) and fed with HFD + l-NAME for 7 weeks. Plasma FXI levels (I), heart p-SMAD1/5 protein level (J), heart weight/tibia length ratio (K), E/eʹ ratio (L), and LVEF (M) were measured. n = 5 for (I), n = 6 for (J), and n = 10 to 20 for (K) to (M). (N) C57BL/6J male mice were injected with either AAV8-GFP or AAV8-F11 and then fed with HFD + l-NAME for 7 weeks. BMP7 proteins in unprocessed monomer, growth factor dimer, and monomer under nonreducing condition were determined. (O) NRVMs were treated with control or human FXIa protein (1 μg/ml) plus negative control or Bmp7 siRNA with medium containing PE (100 μM) for 24 hours. p-SMAD1/5 and Tcap proteins were examined. Actin served as the loading control. n = 3. (P) NRVMs were treated with control or human FXIa protein (1 μg/ml), BMP7 antibody (no antibody control, 1:100 and 1:50), with medium containing PE (100 μM) for 2 hours, and the p-SMAD1/5 level was determined. Actin served as the loading control. n = 4. Each point represents a mouse. All data are presented as means ± SEM. ns, not significant. *P < 0.05, **P < 0.01, and ***P < 0.001 by two-way ANOVA [(D) to (H)], one-way ANOVA [(I) to (M)], or Student’s t test [(A) to (C)].
View figure
Fig. 4
Fig. 4. Reduced FXI concentrations are associated with diastolic dysfunction in mice and humans.
Heterozygous B6.129X1-F11tm1Gjb/J (F11-Het) mice and WT littermates at 8 weeks of age were subjected to HFD + l-NAME for 7 weeks. n = 8 for WT and n = 6 for F11-Het. (A) qRT-PCR showing F11 mRNA in the indicated tissues from WT and F11-Het mice. n = 4. (B) Western blotting showing p-SMAD1/5 in the hearts of WT mice fed with chow diet (Chow), WT, and F11-Het mice fed with HFD + l-NAME for 7 weeks. n = 5. (C to G) Representative images of echocardiography (C), E/A ratio (D), E/eʹ ratio (E), LV mass (F), and LVEF (G) were examined at baseline (BSL) and after 7 weeks of HFD + l-NAME feeding (HFpEF). n = 8 for WT and n = 6 for F11-Het. (H to J) Heart weight/tibia length ratio (H), lung weight [wet/dry ratio (I)], and running distance (J) were examined after 7 weeks of HFD + l-NAME feeding (HFpEF). n = 8 for WT and n = 6 for F11-Het. (K) Plasma FXI protein in non-HFpEF controls (NHF, n = 20) and HFpEF patients (n = 21). (L and M) Plasma FXI protein was inversely correlated with E/eʹ ratio in all participants (L), including HFpEF patients (M). (N) Illustration summarizing FXI-mediated liver-heart cross-talk in protecting against heart failure. Using a bioinformatic framework that integrates global liver-heart transcriptome and cardiometabolic trait data from the HMDP, we found that coagulation FXI, secreted by the liver, exhibits cardioprotective effects on the progression of HFpEF. FXI overexpression in the liver mitigates the diastolic dysfunction, inflammation, and fibrosis induced by HFpEF. FXIa cleaves the BMP7 precursor and activates the BMP7-SMAD1/5 pathway in the heart to mediate the anti-inflammatory and anti-fibrotic effects. Each point represents a mouse. All data are presented as means ± SEM. ns, not significant. *P < 0.05, **P < 0.01, and ***P < 0.001 by two-way ANOVA [(D) to (G)], one-way ANOVA (B), or Student’s t test [(A) and (H) to (K)].
RIS DOI Dorad © 2024
RIS DOI Dorad版权所有© 2024