Ceramides in peripheral arterial plaque lead to endothelial cell dysfunction

Q3 Medicine
Rodrigo Meade BS , Yang Chao MD, PhD , Nikolai Harroun MD , Chenglong Li MD , Shahab Hafezi MD , Fong-Fu Hsu PhD , Clay F. Semenkovich MD , Mohamed A. Zayed MD, PhD, MBA
{"title":"Ceramides in peripheral arterial plaque lead to endothelial cell dysfunction","authors":"Rodrigo Meade BS ,&nbsp;Yang Chao MD, PhD ,&nbsp;Nikolai Harroun MD ,&nbsp;Chenglong Li MD ,&nbsp;Shahab Hafezi MD ,&nbsp;Fong-Fu Hsu PhD ,&nbsp;Clay F. Semenkovich MD ,&nbsp;Mohamed A. Zayed MD, PhD, MBA","doi":"10.1016/j.jvssci.2023.100181","DOIUrl":null,"url":null,"abstract":"<div><h3>Background</h3><p>Peripheral arterial atheroprogression is increasingly prevalent, and is a risk factor for major limb amputations in individuals with risk factors such as diabetes. We previously demonstrated that bioactive lipids are significantly altered in arterial tissue of individuals with diabetes and advanced peripheral arterial disease.</p></div><div><h3>Methods</h3><p>Here we evaluated whether sphingolipid ceramide 18:1/16:0 (C16) is a cellular regulator in endothelial cells and peripheral tibial arterial tissue in individuals with diabetes.</p></div><div><h3>Results</h3><p>We observed that C16 is the single most elevated ceramide in peripheral arterial tissue from below the knee in individuals with diabetes (11% increase, <em>P</em> &lt; .05). C16 content in tibial arterial tissue positively correlates with sphingomyelin (SPM) content in patients with and without diabetes (r<sup>2</sup> = 0.5, <em>P</em> &lt; .005; r<sup>2</sup> = 0.17, <em>P</em> &lt; .05; respectively). Tibial arteries of individuals with diabetes demonstrated no difference in <em>CERS6</em> expression (encoding ceramide synthase 6; the predominate ceramide synthesis enzyme), but higher <em>SMPD</em> expression (encoding sphingomyelin phosphodiesterase that catalyzes ceramide synthesis from sphingomyelins; <em>P</em> &lt; .05). <em>SMPD4,</em> but not <em>SMPD2</em>, was particularly elevated in maximally diseased (Max) tibial arterial segments (<em>P</em> &lt; .05). In vitro, exogenous C16 caused endothelial cells (HUVECs) to have decreased proliferation (<em>P</em> &lt; .03), increased apoptosis (<em>P</em> &lt; .003), and decreased autophagy (<em>P</em> &lt; .008). Selective knockdown of <em>SMPD2</em> and <em>SMPD4</em> decreased native production of C16 (<em>P</em> &lt; .01 and <em>P</em> &lt; .001, respectively), but only knockdown of SMPD4 rescued cellular proliferation (<em>P</em> &lt; .005) following exogenous supplementation with C16.</p></div><div><h3>Conclusions</h3><p>Our findings suggest that C16 is a tissue biomarker for peripheral arterial disease severity in the setting of diabetes, and can impact endothelial cell viability and function.</p></div><div><h3>Clinical relevance</h3><p>Peripheral arterial disease and its end-stage manifestation known as chronic limb-threatening ischemia (CLTI) represent ongoing prevalent and intricate medical challenges. Individuals with diabetes have a heightened risk of developing CLTI and experiencing its complications, including wounds, ulcers, and major amputations. In the present study, we conducted a comprehensive examination of the molecular lipid composition within arterial segments from individuals with CLTI, and with and without diabetes. Our investigations unveiled a striking revelation: the sphingolipid ceramide 18:1/16:0 emerged as the predominant ceramide species that was significantly elevated in the peripheral arterial intima below the knee in patients with diabetes. Moreover, this heightened ceramide presence is associated with a marked impairment of endothelial cell function and viability. Additionally, our study revealed a concurrent elevation in the expression of sphingomyelin phosphodiesterases, enzymes responsible for catalyzing ceramide synthesis from sphingomyelins, within maximally diseased arterial segments. These findings underscore the pivotal role of ceramides and their biosynthesis enzymes in the context of CLTI, offering new insights into potential therapeutic avenues for managing this challenging disease process.</p></div>","PeriodicalId":74035,"journal":{"name":"JVS-vascular science","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2666350323000858/pdfft?md5=3d3bd95adba87869a0086af3cb8d6207&pid=1-s2.0-S2666350323000858-main.pdf","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"JVS-vascular science","FirstCategoryId":"1085","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S2666350323000858","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"Medicine","Score":null,"Total":0}
引用次数: 0

Abstract

Background

Peripheral arterial atheroprogression is increasingly prevalent, and is a risk factor for major limb amputations in individuals with risk factors such as diabetes. We previously demonstrated that bioactive lipids are significantly altered in arterial tissue of individuals with diabetes and advanced peripheral arterial disease.

Methods

Here we evaluated whether sphingolipid ceramide 18:1/16:0 (C16) is a cellular regulator in endothelial cells and peripheral tibial arterial tissue in individuals with diabetes.

Results

We observed that C16 is the single most elevated ceramide in peripheral arterial tissue from below the knee in individuals with diabetes (11% increase, P < .05). C16 content in tibial arterial tissue positively correlates with sphingomyelin (SPM) content in patients with and without diabetes (r2 = 0.5, P < .005; r2 = 0.17, P < .05; respectively). Tibial arteries of individuals with diabetes demonstrated no difference in CERS6 expression (encoding ceramide synthase 6; the predominate ceramide synthesis enzyme), but higher SMPD expression (encoding sphingomyelin phosphodiesterase that catalyzes ceramide synthesis from sphingomyelins; P < .05). SMPD4, but not SMPD2, was particularly elevated in maximally diseased (Max) tibial arterial segments (P < .05). In vitro, exogenous C16 caused endothelial cells (HUVECs) to have decreased proliferation (P < .03), increased apoptosis (P < .003), and decreased autophagy (P < .008). Selective knockdown of SMPD2 and SMPD4 decreased native production of C16 (P < .01 and P < .001, respectively), but only knockdown of SMPD4 rescued cellular proliferation (P < .005) following exogenous supplementation with C16.

Conclusions

Our findings suggest that C16 is a tissue biomarker for peripheral arterial disease severity in the setting of diabetes, and can impact endothelial cell viability and function.

Clinical relevance

Peripheral arterial disease and its end-stage manifestation known as chronic limb-threatening ischemia (CLTI) represent ongoing prevalent and intricate medical challenges. Individuals with diabetes have a heightened risk of developing CLTI and experiencing its complications, including wounds, ulcers, and major amputations. In the present study, we conducted a comprehensive examination of the molecular lipid composition within arterial segments from individuals with CLTI, and with and without diabetes. Our investigations unveiled a striking revelation: the sphingolipid ceramide 18:1/16:0 emerged as the predominant ceramide species that was significantly elevated in the peripheral arterial intima below the knee in patients with diabetes. Moreover, this heightened ceramide presence is associated with a marked impairment of endothelial cell function and viability. Additionally, our study revealed a concurrent elevation in the expression of sphingomyelin phosphodiesterases, enzymes responsible for catalyzing ceramide synthesis from sphingomyelins, within maximally diseased arterial segments. These findings underscore the pivotal role of ceramides and their biosynthesis enzymes in the context of CLTI, offering new insights into potential therapeutic avenues for managing this challenging disease process.

外周动脉斑块中的神经酰胺导致内皮细胞功能障碍
背景:外周动脉粥样硬化进展越来越普遍,是糖尿病等危险因素导致截肢的危险因素。我们之前证明,生物活性脂质在糖尿病和晚期外周动脉疾病患者的动脉组织中显著改变。方法研究鞘脂神经酰胺18:1/16:0 (C16)是否在糖尿病患者的内皮细胞和胫外周动脉组织中起细胞调节作用。结果我们观察到C16是糖尿病患者膝以下外周动脉组织中升高最多的神经酰胺(增加11%,P <. 05)。糖尿病患者和非糖尿病患者胫骨动脉组织C16含量与鞘磷脂(SPM)含量呈正相关(r2 = 0.5, P <.005;r2 = 0.17, P <. 05;分别)。糖尿病患者胫骨动脉中CERS6的表达无差异(编码神经酰胺合成酶6;主要的神经酰胺合成酶),但较高的SMPD表达(编码鞘磷脂磷酸二酯酶,催化鞘磷脂合成神经酰胺;P & lt;. 05)。SMPD4,而不是SMPD2,在最大病变(Max)胫骨动脉段特别升高(P <. 05)。体外,外源性C16引起内皮细胞(HUVECs)增殖降低(P <.03),细胞凋亡增加(P <.003),自噬减少(P <.008)。选择性敲除SMPD2和SMPD4可降低C16的天然产量(P <.01和P <.001),但只有敲低SMPD4才能挽救细胞增殖(P <.005),外源性添加C16后。结论C16是糖尿病患者外周动脉疾病严重程度的组织生物标志物,可以影响内皮细胞的活力和功能。外周动脉疾病及其终末期表现为慢性肢体威胁性缺血(CLTI),是持续流行和复杂的医学挑战。糖尿病患者发生CLTI及其并发症的风险更高,包括伤口、溃疡和主要截肢。在本研究中,我们对CLTI患者、糖尿病患者和非糖尿病患者动脉段内的分子脂质组成进行了全面检查。我们的研究揭示了一个惊人的发现:鞘脂神经酰胺18:1/16:0成为糖尿病患者膝盖以下外周动脉内膜中显著升高的主要神经酰胺种类。此外,这种神经酰胺的存在与内皮细胞功能和活力的显著损害有关。此外,我们的研究还发现,在病变最严重的动脉段内,鞘磷脂磷酸二酯酶(负责催化鞘磷脂合成神经酰胺的酶)的表达同时升高。这些发现强调了神经酰胺及其生物合成酶在CLTI中的关键作用,为管理这一具有挑战性的疾病过程的潜在治疗途径提供了新的见解。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
CiteScore
4.20
自引率
0.00%
发文量
0
审稿时长
28 weeks
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信