663 results on '"EndMT"'
Search Results
2. Salvianic acid A ameliorates atherosclerosis through metabolic-dependent anti-EndMT pathway and repression of TGF-β/ALK5 signaling
- Author
-
Gao, Yijun, Ye, Fei, Dong, Yafen, Wang, Tingfang, Xiong, Liyan, Chen, Ting, Wang, Yun, Liu, Xiaoyan, Zhang, Yunan, Qiu, Zheng, Jiang, Jianfang, Liu, Xijun, Hu, Qingxun, and Zhang, Chuan
- Published
- 2025
- Full Text
- View/download PDF
3. An integrated and multifunctional homemade cell sensor platform based on Si-d-CQDs and CRISPR-Cas12a for CD31 detection during endothelial-to-mesenchymal transition
- Author
-
Yuan, Guolin, Cheng, Dongliang, Huang, Jian, Wang, Meifang, Xia, Xianru, An, He, Xie, Fei, Li, Xiandong, Chen, Jiayi, Tang, Yijun, and Peng, Chunyan
- Published
- 2025
- Full Text
- View/download PDF
4. Mmu_circ_0001148 promotes endothlial-mesenchymal transition via regulating miR-218-5p/JMY axis and drives progression of atherosclerosis
- Author
-
Yu, Shang-Min, Liu, Jia-Qi, Zhang, Lin-Lin, Ma, Ya-Ting, Yin, Fei-Yang, and Liu, Shan
- Published
- 2025
- Full Text
- View/download PDF
5. Endothelial-to-mesenchymal transition in the tumor microenvironment: Roles of transforming growth factor-β and matrix metalloproteins
- Author
-
Du, Fei, Li, Jing, Zhong, Xiaolin, Zhang, Zhuo, and Zhao, Yueshui
- Published
- 2024
- Full Text
- View/download PDF
6. Cannabinoid 2 Receptor Activation Protects against Diabetic Cardiomyopathy through Inhibition of AGE/RAGE-Induced Oxidative Stress, Fibrosis, and Inflammasome Activation
- Author
-
Hashiesh, Hebaallah Mamdouh, Azimullah, Sheikh, Nagoor Meeran, Mohamed Fizur, Saraswathiamma, Dhanya, Arunachalam, Seenipandi, Jha, Niraj Kumar, Sadek, Bassem, Adeghate, Ernest, Sethi, Gautam, Albawardi, Alia, Al Marzooqi, Saeeda, and Ojha, Shreesh
- Published
- 2024
- Full Text
- View/download PDF
7. Empagliflozin attenuating renal interstitial fibrosis in diabetic kidney disease by inhibiting lymphangiogenesis and lymphatic endothelial-to-mesenchymal transition via the VEGF-C/VEGFR3 pathway
- Author
-
Huang, Jiaan, Liu, Yan, Shi, Mengting, Zhang, Xiaoyun, Zhong, Yan, Guo, Shuai, Ma, Yun, Pan, Limin, Yang, Fan, and Wang, Yuehua
- Published
- 2024
- Full Text
- View/download PDF
8. Bioinformatics analysis of MMP14+ myeloid cells affecting endothelial-mesenchymal transformation and immune microenvironment in glioma
- Author
-
Luo, Wei, Quan, Qi, Xu, Zihao, Lei, Jinju, and Peng, Roujun
- Published
- 2024
- Full Text
- View/download PDF
9. Triptolide attenuates cardiac remodeling by inhibiting pyroptosis and EndMT via modulating USP14/Keap1/Nrf2 pathway
- Author
-
Ba, Lina, E, Mingyao, Wang, Ruixuan, Wu, Nan, Wang, Rui, Liu, Renling, Feng, Xiang, Qi, Hanping, Sun, Hongli, and Qiao, Guofen
- Published
- 2024
- Full Text
- View/download PDF
10. circSIRT2/miR‐542‐3p/VASH1 axis regulates endothelial‐to‐mesenchymal transition (EndMT) in subretinal fibrosis in age‐related macular degeneration models.
- Author
-
Zhang, Min, Wu, Jiali, Wang, Yimin, Wu, Yidong, Wan, Xiaoling, Jiang, Mei, Bo, Qiyu, Chen, Jieqiong, and Sun, Xiaodong
- Subjects
- *
ENDOTHELIAL growth factors , *MACULAR degeneration , *UMBILICAL veins , *VISION disorders , *ENDOTHELIAL cells , *CIRCULAR RNA - Abstract
Neovascular age‐related macular degeneration (nAMD), characterized by choroidal neovascularization (CNV), is one of the leading causes of severe visual impairment and irreversible vision loss around the world. Subretinal fibrosis (SRF) contributes to the incomplete response to anti‐vascular endothelial growth factor (VEGF) treatment and is one of the main reasons for long‐term poor visual outcomes in nAMD. Reducing SRF is urgently needed in the anti‐VEGF era. The role of non‐coding RNAs has been implicated in CNV; however, their roles in SRF have not been elucidated yet. Herein, we comprehensively investigated circular RNA (circRNA) profiles in the laser‐induced mouse SRF model and the transforming growth factor‐β (TGF‐β) induced human umbilical vein endothelial cell (HUVEC) fibrosis model. A novel circRNA, circSIRT2, was identified, and its function in SRF and endothelial‐to‐mesenchymal transition (EndMT) regulation was investigated. circSIRT2 was consistently upregulated in fibrotic models in vivo and in vitro. circSIRT2 overexpression downregulated the fibrotic markers and inhibited the proliferation and migration of endothelial cells in vitro. circSIRT2 overexpression in vivo also reduced SRF area in mice. Mechanistically, circSIRT2 functioned by sponging miR‐542‐3p, which further upregulated the expression of vasohibin‐1 (VASH1) and reduced SRF lesion development. Vitreous delivery of miR‐542‐3p and VASH1 in the mouse SRF model also confirmed the pro‐fibrotic function of miR‐542‐3p and anti‐fibrotic function of VASH1, respectively. In conclusion, circSIRT2 inhibited SRF by binding miR‐542‐3p, which stimulated the VASH1 expression and subsequently suppressed EndMT. The circSIRT2/miR‐542‐3p/VASH1 axis may serve as a promising therapeutic target for SRF in nAMD. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
11. miR-23b-3p Ameliorates LPS-Induced Pulmonary Fibrosis by Inhibiting EndMT via DPP4 Inhibition.
- Author
-
Yue, Linlin, Chen, Feng, Liu, Xin, Wu, Chaoyu, Wang, Jie, Lai, Jiying, and Zhu, Hongquan
- Abstract
Acute respiratory distress syndrome is a disease triggered by severe pulmonary and systemic inflammation that may lead to fibrosis and the decline of lung function. Lung capillary endothelial-to-mesenchymal transition (EndMT) is one of the primary sources of fibroblasts in pulmonary fibrosis. The role of miRNAs as molecular markers of pulmonary fibrosis, and miRNAs as nucleic acid drugs has attracted increasing attention. To mock EndMT process, Human pulmonary microvascular endothelial cells (HPMECs) were induced with lipopolysaccharide (LPS). Similarly, LPS treatment was used to generate a mouse model of LPS-induced EndMT and pulmonary fibrosis. LPS-induced EndMT in HPMECs resulted in a significant reduction of miR-23b-3p. miR-23b-3p inhibited the interstitial transition of HPMECs, and miR-23b-3p could mediate this process via inhibiting dipeptidyl peptidase-4 (DPP4). Dual-luciferase assays confirmed the regulatory mechanism of miR-23b-3p. In our mouse model of LPS-induced pulmonary fibrosis, miR-23b-3p and a DPP4 inhibitor (sitagliptin) individually alleviated LPS-induced EndMT progression and pulmonary fibrosis, and their combined use achieved the strongest remission effect. To sum up, miR-23b-3p alleviates EndMT in pulmonary fibrosis by inhibiting the expression of DPP4. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
12. Mural Cells Initiate Endothelial-to-Mesenchymal Transition in Adjacent Endothelial Cells in Extracranial AVMs.
- Author
-
Mehdi, Syed J., Zhang, Haihong, Sun, Ravi W., Richter, Gresham T., and Strub, Graham M.
- Subjects
- *
VASCULAR endothelial cells , *ENDOTHELIAL cells , *ARTERIOVENOUS malformation , *UMBILICAL veins , *CD34 antigen , *CEREBRAL arteriovenous malformations - Abstract
Extracranial arteriovenous malformations (eAVMs) are complex vascular lesions characterized by anomalous arteriovenous connections, vascular instability, and disruptions in endothelial cell (EC)-to-mural cell (MC) interactions. This study sought to determine whether eAVM-MCs could induce endothelial-to-mesenchymal transition (EndMT), a process known to disrupt vascular integrity, in the eAVM microenvironment. eAVM and paired control tissues were analyzed using RT-PCR for EC (CD31, CD34, and CDH5) and EndMT-specific markers (SNAI1, SNAI2, ACTA2/α-SMA, N-cadherin/CDH2, VIM). Immunohistochemistry (IHC) was also performed to analyze MC- (PDGFR-β and α-SMA), EC (CD31, CD34, and CDH5), and EndMT-specific markers (CDH2 and SNAI1) in sequential paraffin-embedded sections of eAVM patient biopsies and in adjacent normal tissue biopsies from the same patients. Furthermore, eAVM-MCs and MCs from normal paired tissues (NMCs) were then isolated from fresh human surgical samples using flow cytometry and co-cultured with normal human umbilical vein vascular endothelial cells (HUVECs), followed by analysis of CD31 by immunofluorescence. RT-PCR analysis did not show a significant difference in the expression of EC markers between eAVM tissues and controls, whereas expression of EndMT-specific markers was upregulated in eAVM tissues compared to controls. IHC of eAVMs and paired control tissues demonstrated regions of significant perivascular eAVM-MC expansion (PDGFR-β+, and α-SMA+) surrounding dilated, morphologically abnormal vessels. These regions contained endothelium undergoing EndMT as evidenced by loss of CD31, CD34, and CDH5 expression and upregulation of the EndMT-associated genes CDH2 and SNAI1. Isolated eAVM-MCs induced loss of CD31 in HUVECs when grown in co-culture, while NMCs did not. This study suggests that the eAVM endothelium surrounded by expanded eAVM-MCs undergoes EndMT, potentially leading to the formation of dilated and fragile vessels, and implicates the eAVM-MCs in EndMT initiation and eAVM pathology. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. The novel anthraquinone compound Kanglexin prevents endothelial-to-mesenchymal transition in atherosclerosis by activating FGFR1 and suppressing integrin β1/TGFβ signaling.
- Author
-
Zhao, Yixiu, Wang, Zhiqi, Ren, Jing, Chen, Huan, Zhu, Jia, Zhang, Yue, Zheng, Jiangfei, Cao, Shifeng, Li, Yanxi, Liu, Xue, An, Na, Ban, Tao, Yang, Baofeng, and Zhang, Yan
- Abstract
Endothelial-mesenchymal transition (EndMT) disrupts vascular endothelial integrity and induces atherosclerosis. Active integrin β1 plays a pivotal role in promoting EndMT by facilitating TGFβ/Smad signaling in endothelial cells. Here, we report a novel anthraquinone compound, Kanglexin (KLX), which prevented EndMT and atherosclerosis by activating MAP4K4 and suppressing integrin β1/TGFβ signaling. First, KLX effectively counteracted the EndMT phenotype and mitigated the dysregulation of endothelial and mesenchymal markers induced by TGFβ1. Second, KLX suppressed TGFβ/Smad signaling by inactivating integrin β1 and inhibiting the polymerization of TGFβR1/2. The underlying mechanism involved the activation of FGFR1 by KLX, resulting in the phosphorylation of MAP4K4 and Moesin, which led to integrin β1 inactivation by displacing Talin from its β-tail. Oral administration of KLX effectively stimulated endothelial FGFR1 and inhibited integrin β1, thereby preventing vascular EndMT and attenuating plaque formation and progression in the aorta of atherosclerotic Apoe
−/− mice. Notably, KLX (20 mg/kg) exhibited superior efficacy compared with atorvastatin, a clinically approved lipid-regulating drug. In conclusion, KLX exhibited potential in ameliorating EndMT and retarding the formation and progression of atherosclerosis through direct activation of FGFR1. Therefore, KLX is a promising candidate for the treatment of atherosclerosis to mitigate vascular endothelial injury. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
14. Severe and post-COVID-19 are associated with high expression of vimentin and reduced expression of N-cadherin
- Author
-
Esra Yilmaz, Dilek Yilmaz, and Ercan Cacan
- Subjects
COVID-19 ,Vimentin ,N-cadherin ,EndMT ,Endothelial dysfunction ,Medicine ,Science - Abstract
Abstract SARS-CoV-2 penetrates human cells via its spike protein, which mainly interacts with ACE2 receptors, triggering viral replication and an exacerbated immune response characterized by a cytokine storm. Vimentin III, an intermediate filament protein predominantly found in mesenchymal cells, has garnered considerable attention in recent research due to its multifaceted biological roles and significance in the endothelial-mesenchymal transition (EndMT) during various fibrotic processes. However, the pathophysiological mechanisms linking vimentin to SARS-CoV-2 remain incompletely elucidated. In this study, we determined the expression profiles of vimentin in three cohorts: patients admitted to the intensive care unit with SARS-CoV-2 infection, individuals in the 6–12 month convalescent phase post-infection and COVID-19 negative controls. Our objective was to assess the association between peripheral blood biomarkers implicated in endothelial dysfunction and genes related to fibrosis. Serum levels of vimentin and N-cadherin were determined by ELISA, while vimentin gene expression was determined by qRT-PCR. In addition, we examined the correlation between clinical parameters and serum levels of vimentin and N-cadherin in severe COVID-19 patients and healthy counterparts. Our findings revealed elevated serum vimentin levels and increased gene expression in severe COVID-19 patients compared to healthy controls. Conversely, serum N-cadherin levels were diminished in both acute and convalescent stages of severe COVID-19 relative to healthy individuals. Notably, associations were observed between C-reactive protein, lactate dehydrogenase, lymphocyte count and vimentin levels in severe COVID-19 patients, indicative of endothelial dysfunction. Furthermore, our study identified vimentin and N-cadherin as potential diagnostic markers via ROC analysis. Overall, delineating the dysregulation of vimentin and N-cadherin due to SARS-CoV-2 infection in disease pathogenesis and tissue homeostasis offers novel insights for clinical management and targeted therapeutic interventions.
- Published
- 2024
- Full Text
- View/download PDF
15. Severe and post-COVID-19 are associated with high expression of vimentin and reduced expression of N-cadherin.
- Author
-
Yilmaz, Esra, Yilmaz, Dilek, and Cacan, Ercan
- Subjects
INTERMEDIATE filament proteins ,INTENSIVE care patients ,COVID-19 ,LYMPHOCYTE count ,VIMENTIN - Abstract
SARS-CoV-2 penetrates human cells via its spike protein, which mainly interacts with ACE2 receptors, triggering viral replication and an exacerbated immune response characterized by a cytokine storm. Vimentin III, an intermediate filament protein predominantly found in mesenchymal cells, has garnered considerable attention in recent research due to its multifaceted biological roles and significance in the endothelial-mesenchymal transition (EndMT) during various fibrotic processes. However, the pathophysiological mechanisms linking vimentin to SARS-CoV-2 remain incompletely elucidated. In this study, we determined the expression profiles of vimentin in three cohorts: patients admitted to the intensive care unit with SARS-CoV-2 infection, individuals in the 6–12 month convalescent phase post-infection and COVID-19 negative controls. Our objective was to assess the association between peripheral blood biomarkers implicated in endothelial dysfunction and genes related to fibrosis. Serum levels of vimentin and N-cadherin were determined by ELISA, while vimentin gene expression was determined by qRT-PCR. In addition, we examined the correlation between clinical parameters and serum levels of vimentin and N-cadherin in severe COVID-19 patients and healthy counterparts. Our findings revealed elevated serum vimentin levels and increased gene expression in severe COVID-19 patients compared to healthy controls. Conversely, serum N-cadherin levels were diminished in both acute and convalescent stages of severe COVID-19 relative to healthy individuals. Notably, associations were observed between C-reactive protein, lactate dehydrogenase, lymphocyte count and vimentin levels in severe COVID-19 patients, indicative of endothelial dysfunction. Furthermore, our study identified vimentin and N-cadherin as potential diagnostic markers via ROC analysis. Overall, delineating the dysregulation of vimentin and N-cadherin due to SARS-CoV-2 infection in disease pathogenesis and tissue homeostasis offers novel insights for clinical management and targeted therapeutic interventions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. THBS1 mediates hypoxia driven EndMT in pulmonary hypertension.
- Author
-
Peng, Bingming, Zhou, Yingzhen, Fu, Xingmeng, Chen, Li, Pan, Zhengxia, Yi, Qijian, Zhao, Tengteng, Fu, Zhou, and Wang, Ting
- Subjects
- *
VASCULAR remodeling , *PULMONARY hypertension , *CONGENITAL heart disease , *CHILD patients , *HYPERTENSION - Abstract
Long‐term hypoxia is one of the main causes of pulmonary vascular remodeling in pulmonary hypertension (PH) associated with congenital heart disease (CHD) children. Endothelial to mesenchymal transition (EndMT) is an important pathological basis of pulmonary vascular remodeling in PH. We observed that Fibronectin 1 (FN1) had strong protein–protein interactions with both Thrombospondin 1 (THBS1) and Transglutaminase 2 (TGM2) in PH with venous peripheral bloods samples from pediatric patients and healthy children. LungMAP CellCards and heatmaps of human PAEC in PH patients and lung tissues in hypoxia induced PH mice model were used to show that THBS1 and FN1 were significantly elevated. We studied the relationship between THBS1 and FN1 in vivo, by using SUHX‐induced PH mice model, and in vitro, by using hypoxia‐induced human PAEC. The results showed that hypoxia could result in EndMT and inhibiting THBS1 could reverse EndMT in vivo and in vitro, verifying our transcriptome results. Taken together, our research demonstrated that THBS1 could mediate hypoxia driven EndMT of PH, providing a new insight of research in the pathophysiology of PH. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Melatonin Attenuates Diabetic Retinopathy by Regulating EndMT of Retinal Vascular Endothelial Cells via Inhibiting the HDAC7/FOXO1/ZEB1 Axis.
- Author
-
Ning, Jiayi, Pan, Minghong, Yang, Hanyi, Wang, Zhaoyang, Wang, Xiaolan, Guo, Kai, Feng, Yingtong, Xie, Tingke, Chen, Yixuan, Chen, Chengming, Liu, Sida, Zhang, Yimeng, Wang, Yuanyong, Yan, Xiaolong, and Han, Jing
- Subjects
- *
VASCULAR endothelial cells , *DIABETIC retinopathy , *BASAL lamina , *PINEAL gland , *NUCLEAR proteins - Abstract
Diabetic retinopathy (DR) is characterized as a microvascular disease. Nonproliferative diabetic retinopathy (NPDR) presents with alterations in retinal blood flow and vascular permeability, thickening of the basement membrane, loss of pericytes, and formation of acellular capillaries. Endothelial–mesenchymal transition (EndMT) of retinal microvessels may play a critical role in advancing NPDR. Melatonin, a hormone primarily secreted by the pineal gland, is a promising therapeutic for DR. This study explored the EndMT in retinal microvessels of NPDR and its related mechanisms. The effect of melatonin on the retina of diabetic rats was evaluated by electroretinogram (ERG) and histopathologic slide staining. Furthermore, the effect of melatonin on human retinal microvascular endothelial cells (HRMECs) was detected by EdU incorporation assay, scratch assay, transwell assay, and tube formation test. Techniques such as RNA‐sequencing, overexpression or knockdown of target genes, extraction of cytoplasmic and nuclear protein, co‐immunoprecipitation (co‐IP), and multiplex immunofluorescence facilitated the exploration of the mechanisms involved. Our findings reveal, for the first time, that melatonin attenuates diabetic retinopathy by regulating EndMT of retinal vascular endothelial cells via inhibiting the HDAC7/FOXO1/ZEB1 axis. Collectively, these results suggest that melatonin holds potential as a therapeutic strategy to reduce retinal vascular damage and protect vision in NPDR. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. The role of the purinergic P2X7 receptor in renal haemodynamic physiology and hypertensive renovascular injury
- Author
-
Nespoux, Josselin Georges, Bailey, Matthew, and Baker, Andrew
- Subjects
P2X7 ,Renal haemodynamics ,Kidney function ,CRISPR/Cas9 ,Hypertension ,Kidney injury ,Purinergic ,Pressure natriuresis ,Rat ,EndMT ,endothelial-to-mesenchymal transition ,ATP - Abstract
Purinergic signaling regulates numerous intrarenal mechanisms contributing to the long-term control of blood pressure. The P2X7 receptor is an ionotropic receptor activated by extra-cellular ATP that participates in inflammation and activation of immune cells. P2X7 is also expressed in the vascular endothelium, including in the kidney where its function remains poorly understood. Previous research identified the gene P2rx7, which encodes the P2X7 receptor, as a candidate gene for susceptibility to hypertensive renal vascular injury in Fischer (F344) rats. Higher expression of P2X7 was reported in several rat models of hypertensive kidney injury and both pharmacological blockade and P2X7 deletion were found renoprotective. However, studies raised concerns regarding the specificity of the broadly used P2X7 antagonists which have generated conflicting results. Here, I hypothesized that global genetic deletion of P2X7 in F344 rats will prevent the development of renal inflammation and injury in a model of chronic angiotensin II (Ang II)-induced hypertension. In this study, I sought to characterize the contribution of P2X7 to basal renal vascular and tubular functions in male and female rats. I also aimed to determine whether absence of P2X7 exerts renoprotective effects during chronic Ang II infusion, and to provide cellular and molecular mechanistic evidence for such protection using a combination of in vivo, ex vivo and in vitro studies. To this end, I used F344 rats to generate a novel CRISPR-Cas9-designed P2X7 knockout (KO) and showed that it is a true global KO with no functional P2X7 protein expressed. Using wire myography, I found that P2X7 KO impaired endothelial-dependent vasodilation in the renal artery of male, but not female, rats that may reflect diminished nitric oxide (NO) production in response to acetylcholine. NO is a key paracrine factor regulating the renal pressure natriuresis mechanism and blood pressure (BP). Thus, I assessed kidney function in anesthetized male and female P2X7 KO and wild-type (WT) rats at baseline and following acute stepwise increases in renal perfusion pressure. Overall, results showed no significant genotype effect on renal haemodynamics, BP and NO production. Moreover, my data do not support a major role of P2X7 in the modulation of tubular sodium reabsorption. I next attempted to generate an experimental model of chronic Ang II infusion-induced hypertensive kidney and vascular injury in F344 WT and P2X7 KO rats. After 5-6 weeks of infusion, rats developed modest renal damage consisting of perivascular fibrosis and tubular injury. P2X7 did not contribute to the development of renal perivascular fibrosis and tubular injury during chronic Ang II infusion. Finally, I investigated the implication of P2X7 to endothelial-to-mesenchymal transition (EndMT) using the potent P2X7 antagonist A438079 in vitro. I found that P2X7 blockade did not affect EndMT. In summary, I propose that P2X7 receptors modestly promote endothelial NO production in healthy rat renal arteries and inhibit tubular sodium reabsorption in a sex-specific manner. The role of P2X7 in renal vascular functions may become more relevant in a pathological context and requires further investigations with better disease models.
- Published
- 2023
- Full Text
- View/download PDF
19. Furin knockdown inhibited EndMT and abnormal proliferation and migration of endothelial cells.
- Author
-
Zeng, Rui, Wang, Yimin, Chen, Jun, and Liu, Qiang
- Subjects
- *
REVERSE transcriptase polymerase chain reaction , *ENDOTHELIAL cells , *GENE expression , *CELL migration , *VASCULAR endothelium - Abstract
BACKGROUND: In the pathogenesis of atherosclerotic cardiovascular disorders, vascular endothelium is crucial. A critical step in the development of atherosclerosis is endothelial dysfunction. Furin may play a factor in vascular remodeling, inflammatory cell infiltration, regulation of plaque stability, and atherosclerosis by affecting the adhesion and migration of endothelial cells. It is yet unknown, though, how furin contributes to endothelial dysfunction. METHODS: We stimulated endothelial cells with oxidized modified lipoprotein (ox-LDL). Endothelial-to-mesenchymal transition (EndMT) was found using immunofluorescence (IF) and western blot (WB). Furin expression level and Hippo/YAP signal activation were found using reverse transcription-quantitative PCR (RT-qPCR) and WB, respectively. To achieve the goal of furin knockdown, we transfected siRNA using the RNA transmate reagent. Following furin knockdown, cell proliferation, and migration were assessed by the CCK-8, scratch assay, and transwell gold assay, respectively. WB and IF both picked up on EndMT. WB and RT-qPCR, respectively, were used to find furin's expression level. We chose the important micrornas that can regulate furin and we then confirmed them using RT-qPCR. RESULTS: EndMT was created by ox-LDL, evidenced by the up-regulation of mesenchymal cell markers and the down-regulation of endothelial cell markers. Furin expression levels in both protein and mRNA were increased, and the Hippo/YAP signaling pathway was turned on. Furin knockdown dramatically reduced the aberrant migration and proliferation of endothelial cells by ox-LDL stimulation. Furin knockdown can also suppress ox-LDL-induced EndMT, up-regulate indicators of endothelial cells, and down-regulate markers of mesenchymal cells. After ox-LDL stimulation and siRNA transfection, furin's expression level was up-regulated and down-regulated. CONCLUSION: Our study demonstrated that furin knockdown could affect ox-LDL-induced abnormal endothelial cell proliferation, migration, and EndMT. This implies that furin plays an important role in endothelial dysfunction. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease.
- Author
-
Larionov, Alexey, Hammer, Christian Manfred, Fiedler, Klaus, and Filgueira, Luis
- Subjects
- *
CYTOLOGY , *PATHOGENESIS , *HUMAN body , *CARCINOGENESIS , *CYTOARCHITECTONICS , *ENDOTHELIAL cells - Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Diverse roles of SARS-CoV-2 Spike and Nucleocapsid proteins in EndMT stimulation through the TGF-β-MRTF axis inhibited by aspirin.
- Author
-
Ciszewski, Wojciech M., Woźniak, Lucyna A., and Sobierajska, Katarzyna
- Subjects
- *
SARS-CoV-2 , *VIRAL proteins , *ASPIRIN , *PROTEINS , *WOUND healing , *HEART block - Abstract
Background: The SARS-CoV-2 virus causes severe COVID-19 in one-fifth of patients. In addition to high mortality, infection may induce respiratory failure and cardiovascular complications associated with inflammation. Acute or prolonged inflammation results in organ fibrosis, the cause of which might be endothelial disorders arising during the endothelial-mesenchymal transition (EndMT). Methods: HUVECs and HMEC-1 cells were stimulated with SARS-CoV-2 S (Spike) and N (Nucleocapsid) proteins, and EndMT induction was evaluated by studying specific protein markers via Western blotting. Wound healing and tube formation assays were employed to assess the potential of SARS-CoV-2 to stimulate changes in cell behaviour. MRTF nuclear translocation, ROS generation, TLR4 inhibitors, TGF-β-neutralizing antibodies, and inhibitors of the TGF-β-dependent pathway were used to investigate the role of the TGF-β-MRTF signalling axis in SARS-CoV-2-dependent EndMT stimulation. Results: Both viral proteins stimulate myofibroblast trans-differentiation. However, the N protein is more effective at EndMT induction. The TGF-β-MRTF pathway plays a critical role in this process. The N protein preferentially favours action through TGF-β2, whose secretion is induced through TLR4-ROS action. TGF-β2 stimulates MRTF-A and MRTF-B nuclear translocation and strongly regulates EndMT. In contrast, the Spike protein stimulates TGF-β1 secretion as a result of ACE2 downregulation. TGF-β1 induces only MRTF-B, which, in turn, weakly regulates EndMT. Furthermore, aspirin, a common nonsteroidal anti-inflammatory drug, might prevent and reverse SARS-CoV-2-dependent EndMT induction through TGF-β-MRTF pathway deregulation. Conclusion: The reported study revealed that SARS-CoV-2 infection induces EndMT. Moreover, it was demonstrated for the first time at the molecular level that the intensity of the EndMT triggered by SARS-CoV-2 infection may vary and depend on the viral protein involved. The N protein acts through TLR4-ROS-TGF-β2-MRTF-A/B, whereas the S protein acts through ACE2-TGF-β1-MRTF-B. Furthermore, we identified aspirin as a potential anti-fibrotic drug for treating patients with SARS-CoV-2 infection. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Endothelial-to-Mesenchymal Transition in Cardiovascular Pathophysiology.
- Author
-
Singh, Aman, Bhatt, Kriti S., Nguyen, Hien C., Frisbee, Jefferson C., and Singh, Krishna K.
- Subjects
- *
HYPERTROPHIC scars , *CELL adhesion , *CELL adhesion molecules , *VON Willebrand factor , *ENDOTHELIAL cells , *PATHOLOGICAL physiology , *EMBRYOLOGY , *HEART fibrosis - Abstract
Under different pathophysiological conditions, endothelial cells lose endothelial phenotype and gain mesenchymal cell-like phenotype via a process known as endothelial-to-mesenchymal transition (EndMT). At the molecular level, endothelial cells lose the expression of endothelial cell-specific markers such as CD31/platelet-endothelial cell adhesion molecule, von Willebrand factor, and vascular-endothelial cadherin and gain the expression of mesenchymal cell markers such as α-smooth muscle actin, N-cadherin, vimentin, fibroblast specific protein-1, and collagens. EndMT is induced by numerous different pathways triggered and modulated by multiple different and often redundant mechanisms in a context-dependent manner depending on the pathophysiological status of the cell. EndMT plays an essential role in embryonic development, particularly in atrioventricular valve development; however, EndMT is also implicated in the pathogenesis of several genetically determined and acquired diseases, including malignant, cardiovascular, inflammatory, and fibrotic disorders. Among cardiovascular diseases, aberrant EndMT is reported in atherosclerosis, pulmonary hypertension, valvular disease, fibroelastosis, and cardiac fibrosis. Accordingly, understanding the mechanisms behind the cause and/or effect of EndMT to eventually target EndMT appears to be a promising strategy for treating aberrant EndMT-associated diseases. However, this approach is limited by a lack of precise functional and molecular pathways, causes and/or effects, and a lack of robust animal models and human data about EndMT in different diseases. Here, we review different mechanisms in EndMT and the role of EndMT in various cardiovascular diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Single-Cell RNA-Sequencing Analyses Identify APLNR, INS-IGF2, RGCC Genes May Be Involved in the Pathogenesis of Systemic Sclerosis Skin
- Author
-
Zhu P and Deng W
- Subjects
systemic sclerosis skin ,single cell rna-sequencing analysis ,the endothelial to mesencchymal transition ,endmt ,aplnr ,ins-igf2 ,rgcc ,Dermatology ,RL1-803 - Abstract
Peiqiu Zhu,1 Weiwei Deng2 1Department of Dermatology and Venereology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, People’s Republic of China; 2Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, People’s Republic of ChinaCorrespondence: Weiwei Deng, Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, People’s Republic of China, Tel +86 15622165048, Email 1601178683@qq.comBackground: Systemic sclerosis represents a persistent autoimmune disorder marked with fibrosis affecting both skin and other organs, which leads to a diminished quality of life and increased mortality. The affected skin provides a valuable opportunity to explore the pathogenesis of systemic sclerosis. Nevertheless, the roles of various cell populations within scleroderma remain intricate.Methods: We conducted a comprehensive reanalysis of recently published single-cell RNA-sequencing data from skin tissue cells in scleroderma. Through the utilization of Seurat, irGSEA, AUCell packages, and WGCNA analysis, we aimed to unveil crucial genes associated with the disease’s etiological factors. Our investigation involved the characterization of heterogeneous pathway activities in both healthy and SSc-affected skin. Furthermore, we employed immunofluorescence techniques to validate the expression patterns of hub genes and differentially expressed genes.Results: The Endothelial-to-Mesenchymal Transition (EndMT) pathway was upregulated in SSc skin. Notably, the M4 module within Endothelial cell subpopulation 1 exhibited a strong association with EndMT. Furthermore, we identified three overexpressed genes (APLNR, INS-IGF2, RGCC) that demonstrated a significant correlation with EndMT. Importantly, their expression levels were markedly higher in skin of individuals with SSc when compared to healthy controls.Conclusion: APLNR, INS-IGF2 and RGCC serve as potential key players in the pathogenesis of SSc skin through EndMT-dependent mechanisms.Keywords: systemic sclerosis skin, single cell RNA-sequencing analysis, the Endothelial to Mesencchymal Transition, EndMT, APLNR, INS-IGF2, RGCC
- Published
- 2024
24. Study on the mechanism of OSM participating in myocardial fibrosis by inhibiting TGFβ-induced EndMT of cardiac microvascular endothelial cells through SPARC/SMAD signaling
- Author
-
Zhu, Ying, Xu, Zhuo, and Chen, Min
- Published
- 2024
- Full Text
- View/download PDF
25. USF1 transcriptionally activates USP14 to drive atherosclerosis by promoting EndMT through NLRC5/Smad2/3 axis
- Author
-
Zhang, Zhiwen, Guo, Quan, Ma, Chao, Zhao, Zhenzhou, Shi, Qingbo, Yu, Haosen, Rao, Lixin, and Li, Muwei
- Published
- 2024
- Full Text
- View/download PDF
26. Endothelial Reprogramming in Atherosclerosis.
- Author
-
Zhang, Lu, Wu, Xin, and Hong, Liang
- Subjects
- *
ATHEROSCLEROSIS , *ATHEROSCLEROTIC plaque , *ENDOTHELIUM diseases , *ENDOTHELIAL cells , *MUSCLE cells - Abstract
Atherosclerosis (AS) is a severe vascular disease that results in millions of cases of mortality each year. The development of atherosclerosis is associated with vascular structural lesions, characterized by the accumulation of immune cells, mesenchymal cells, lipids, and an extracellular matrix at the intimal resulting in the formation of an atheromatous plaque. AS involves complex interactions among various cell types, including macrophages, endothelial cells (ECs), and smooth muscle cells (SMCs). Endothelial dysfunction plays an essential role in the initiation and progression of AS. Endothelial dysfunction can encompass a constellation of various non-adaptive dynamic alterations of biology and function, termed "endothelial reprogramming". This phenomenon involves transitioning from a quiescent, anti-inflammatory state to a pro-inflammatory and proatherogenic state and alterations in endothelial cell identity, such as endothelial to mesenchymal transition (EndMT) and endothelial-to-immune cell-like transition (EndIT). Targeting these processes to restore endothelial balance and prevent cell identity shifts, alongside modulating epigenetic factors, can attenuate atherosclerosis progression. In the present review, we discuss the role of endothelial cells in AS and summarize studies in endothelial reprogramming associated with the pathogenesis of AS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Multiscale computational modeling of aortic valve calcification.
- Author
-
Azimi-Boulali, Javid, Mahler, Gretchen J., Murray, Bruce T., and Huang, Peter
- Subjects
- *
AORTIC valve , *MULTISCALE modeling , *TRANSFORMING growth factors-beta , *SYSTEMS biology , *CALCIFICATION , *AORTIC valve diseases - Abstract
Calcific aortic valve disease (CAVD) is a common cardiovascular disease that affects millions of people worldwide. The disease is characterized by the formation of calcium nodules on the aortic valve leaflets, which can lead to stenosis and heart failure if left untreated. The pathogenesis of CAVD is still not well understood, but involves several signaling pathways, including the transforming growth factor beta (TGF β ) pathway. In this study, we developed a multiscale computational model for TGF β -stimulated CAVD. The model framework comprises cellular behavior dynamics, subcellular signaling pathways, and tissue-level diffusion fields of pertinent chemical species, where information is shared among different scales. Processes such as endothelial to mesenchymal transition (EndMT), fibrosis, and calcification are incorporated. The results indicate that the majority of myofibroblasts and osteoblast-like cells ultimately die due to lack of nutrients as they become trapped in areas with higher levels of fibrosis or calcification, and they subsequently act as sources for calcium nodules, which contribute to a polydispersed nodule size distribution. Additionally, fibrosis and calcification processes occur more frequently in regions closer to the endothelial layer where the cell activity is higher. Our results provide insights into the mechanisms of CAVD and TGF β signaling and could aid in the development of novel therapeutic approaches for CAVD and other related diseases such as cancer. More broadly, this type of modeling framework can pave the way for unraveling the complexity of biological systems by incorporating several signaling pathways in subcellular models to simulate tissue remodeling in diseases involving cellular mechanobiology. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. H3K27Me3 abundance increases fibrogenesis during endothelial-to-mesenchymal transition via the silencing of microRNA-29c
- Author
-
Jolien Fledderus, Linda Brouwer, Timara Kuiper, Martin C. Harmsen, and Guido Krenning
- Subjects
EZH2 ,H3K27Me3 ,EndMT ,miR-29c ,fibrogenesis ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
ObjectiveEndothelial-to-mesenchymal transition (EndMT) is a transdifferentiation process in which endothelial cells (ECs) adopt a mesenchymal-like phenotype. Over the past few years, it became clear that EndMT can contribute to several cardiovascular pathologies. However, the molecular pathways underlying the development of EndMT remain incompletely understood. Since the epigenetic enzyme Enhancer of Zeste Homolog 2 (EZH2) and its concomitant mark H3K27Me3 have been shown to be elevated in many cardiovascular diseases that associate with EndMT, we hypothesized that H3K27Me3 is a determinant for the susceptibility of EndMT.MethodsTo study the association between H3K27Me3 and EndMT, a knockdown model of EZH2 in human endothelial cells (HUVEC) was utilized to reduce H3K27Me3 abundance, followed by induction of EndMT using TGFβ1. The expression of molecular markers of EndMT and fibrogenesis were analysed.ResultsIn cultured HUVECs, a reduction of H3K27Me3 abundance facilitates EndMT but mitigates fibrogenesis as shown by a decreased expression of collagen I and III. In HUVEC, H3K27Me3 abundance directly affects the expression of miR29c, a collagen-targeting miRNA. Additionally, knockdown of miR-29c in HUVEC with low H3K27Me3 abundance partly restored the expression of collagen I and III. Expectedly, in rats with perivascular fibrosis an increased abundance of H3K27Me3 associated with a decreased expression of miR-29c.Conclusionour data shows that endothelial fibrogenesis underlies an epigenetic regulatory pathway and we demonstrate that a decreased abundance of H3K27Me3 in ECs blunts fibrogenesis in part in a miR-29c dependent manner. Therefore, a reduction of H3K27Me3 could serve as a novel therapeutical strategy to mitigate fibrogenesis and may prove to be beneficial in fibrogenic diseases including atherosclerosis, cardiac fibrosis, and PAH.
- Published
- 2024
- Full Text
- View/download PDF
29. Epidermal Growth Factor‐Like Repeats and Discoidin I‐Like Domains 3 Deficiency Attenuates Dilated Cardiomyopathy by Inhibiting Ubiquitin Specific Peptidase 10 Dependent Smad4 Deubiquitination
- Author
-
Mengmeng Zhao, Zihui Zheng, Shanshan Peng, Yao Xu, Jishou Zhang, Jianfang Liu, Wei Pan, Zheng Yin, Shuwan Xu, Cheng Wei, Menglong Wang, Jun Wan, and Juan‐Juan Qin
- Subjects
dilated cardiomyopathy ,EDIL3 ,EndMT ,Smad4 deubiquitination ,USP10 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background Dilated cardiomyopathy (DCM) is the leading cause of heart failure with a poor prognosis. Recent studies suggest that endothelial to mesenchymal transition (EndMT) may be involved in the pathogenesis and cardiac remodeling during DCM development. EDIL3 (epidermal growth factor‐like repeats and discoidin I‐like domains 3) is an extracellular matrix glycoprotein that has been reported to promote EndMT in various diseases. However, the roles of EDIL3 in DCM still remain unclear. Methods and Results A mouse model of DCM and human umbilical vein endothelial cells were used to explore the roles and mechanisms of EDIL3 in DCM. The results indicated that EndMT and EDIL3 were activated in DCM mice. EDIL3 deficiency attenuated cardiac dysfunction and remodeling in DCM mice. EDIL3 knockdown alleviated EndMT by inhibiting USP10 (ubiquitin specific peptidase 10) dependent Smad4 deubiquitination in vivo and in vitro. Recombinant human EDIL3 promoted EndMT via reinforcing deubiquitination of Smad4 in human umbilical vein endothelial cells treated with IL‐1β (interleukin 1β) and TGF‐β (transforming growth factor beta). Inhibiting USP10 abolished EndMT exacerbated by EDIL3. In addition, recombinant EDIL3 also aggravates doxorubicin‐induced EndMT by promoting Smad4 deubiquitination in HUVECs. Conclusions Taken together, these results indicate that EDIL3 deficiency attenuated EndMT by inhibiting USP10 dependent Smad4 deubiquitination in DCM mice.
- Published
- 2024
- Full Text
- View/download PDF
30. An application of pirfenidone-loaded, lung-targeted nanoliposomes for treating inflammation and early pulmonary fibrosis in ARDS
- Author
-
Sheng Li, Wanshi Chen, Yuhua Zhong, Di Qi, Yiwen Tan, Renzi Zhang, and Daoxin Wang
- Subjects
ARDS ,EndMT ,Lung-targeting peptide ,Lipid nanoparticles ,PFD ,Inflammation ,Materials of engineering and construction. Mechanics of materials ,TA401-492 - Abstract
Acute respiratory distress syndrome (ARDS) is a common critical respiratory disease with a high mortality rate that has been sustained for many years and is caused by a variety of intrapulmonary and extrapulmonary pathogenic factors. However, the effective clinical interventions for ARDS currently in use mainly involve respiratory and organ support therapy, and no effective targeted drug intervention is beneficial for patients with ARDS. In the present study, GALA-PFD-Lip were constructed to reduce lung inflammation, reduce lung oxidative stress, inhibit endothelial-to-mesenchymal transition (EndMT), and reduce early pulmonary fibrosis. We found that GALA-PFD-Lip have efficacious lung targeting activity and biosafety, and the anti-inflammatory and antifibrotic effects of GALA-PFD-Lip are superior to those of pure PFD. These results suggest that GALA-PFD-Lip has good clinical translation potential for the treatment of ARDS-induced pulmonary infections. This study provides new ideas for the treatment of inflammation and for the prevention of early progressive fibrosis that is characteristic of ARDS.
- Published
- 2024
- Full Text
- View/download PDF
31. miRNA-146a-5p Inhibits Hypoxia-Induced Myocardial Fibrosis Through EndMT.
- Author
-
Wang, Yan, Yu, Jie, Ou, Chunxia, Zhao, Yue, Chen, Lixing, Cai, Wenke, Wang, Huawei, Huang, Shiying, Hu, Jie, Sun, Guihu, and Li, Longjun
- Subjects
HEART fibrosis ,FIBROSIS ,THIOREDOXIN-interacting protein ,MYOCARDIAL infarction ,VASCULAR diseases ,CARDIOMYOPATHIES - Abstract
Cardiac Vascular disease particularly myocardial infarction (MI) is a threat to health worldwide. microRNAs (miRNAs) have been shown to regulate myocardial fibrosis. Therefore, it is potential to investigate the mechanism of miRNA and fibrosis following myocardial infarction. Hypoxia human cardiac microvascular endothelial cells (HCMECs) were selected for the vitro experimental model. The miR-146a-5p expression was tested via RT-qPCR. The level of endothelial-to-mesenchymal transition (EndMT) and fibrosis markers were detected by Western blotting and immunofluorescence. Then, the inflammation, cell viability and apoptosis were investigated. The target was predicted by an online database and verified by a dual-luciferase activity assay. An MI mouse model was created to validate that miR-146a-5p regulates cardiac fibrosis in vivo. MI mouse was transfected with miR-146a-5p lentivirus. Subsequently, its effect on cardiac fibrosis of infarcted hearts was assessed by In situ hybridization (ISH), Immunohistochemistry (IHC), Triphenylterazolium chloride (TTC) staining and Masson staining. Herein, we confirmed that miR-146a-5p was down-regulated in hypoxia HCMECs. Overexpression of miR-146a-5p inhibited hypoxia-induced cardiac fibrosis following myocardial infarction by inhibiting EndMT in HCMECs. Thioredoxin-interacting protein (TXNIP) was a target that was negatively regulated by miR-146a-5p. Up-regulation of miR-146a-5p inhibited cardiac fibrosis via regulating EndMT by targeting TXNIP, and it also regulated EndMT to inhibit cardiac fibrosis in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Hydrogen sulfide attenuates atherosclerosis induced by low shear stress by sulfhydrylating endothelium NFIL3 to restrain MEST mediated endothelial mesenchymal transformation.
- Author
-
Zhou, Kun, Luo, Wen, Gui, Dan-Dan, Ren, Zhong, Wei, Dang-Heng, Liu, Lu-Shan, Li, Guo-Hua, Tang, Zhi-Han, Xiong, Wen-Hao, Hu, Heng-Jing, and Jiang, Zhi-Sheng
- Subjects
- *
SHEARING force , *HYDROGEN sulfide , *ATHEROSCLEROSIS , *ATHEROSCLEROTIC plaque , *GENETIC overexpression , *ENDOTHELIUM , *ENDOTHELIAL cells - Abstract
Endothelial-mesenchymal transition (EndMT) induced by low shear stress plays an important role in the development of atherosclerosis. However, little is known about the correlation between hydrogen sulfide (H 2 S), a protective gaseous mediator in atherosclerosis and the process of EndMT. We constructed a stable low-shear-stress-induced(2 dyn/cm2) EndMT model, acombined with the pretreatment method of hydrogen sulfide slow release agent(GYY4137). The level of MEST was detected in the common carotid artery of ApoE−/− mice with local carotid artery ligation. The effect of MEST on atherosclerosis development in vivo was verified using ApoE−/− mice were given tail-vein injection of endothelial-specific overexpressed and knock-down MEST adeno-associated virus (AAV). These findings confirmed that MEST is up-regulated in low-shear-stress-induced EndMT and atherosclerosis. In vivo experiments showed that MEST gene overexpression significantly promoted EndMT and aggravated the development of atherosclerotic plaques and MEST gene knockdown significantly inhibited EndMT and delayed the process of atherosclerosis. In vitro, H 2 S inhibits the expression of MEST and EndMT induced by low shear stress and inhibits EndMT induced by MEST overexpression. Knockdown of NFIL3 inhibit the up regulation of MEST and EndMT induced by low shear stress in HUVECs. CHIP-qPCR assay and Luciferase Reporter assay confirmed that NFIL3 binds to MEST DNA, increases its transcription and H 2 S inhibits the binding of NFIL3 and MEST DNA, weakening NFIL3's transcriptional promotion of MEST. Mechanistically, H 2 S increased the sulfhydrylation level of NFIL3, an important upstream transcription factors of MEST. In part, transcription factor NFIL3 restrain its binding to MEST DNA by sulfhydration. H 2 S negatively regulate the expression of MEST by sulfhydrylation of NFIL3, thereby inhibiting low-shear-stress-induced EndMT and atherosclerosis. • Endothelium Mest participates in the development of atherosclerosis in vivo. • Mest promotes EndMT in mice blood vessels. • H 2 S inhibits MEST and its mediated EndMT induced by low shear stress. • H 2 S inhibiting NFIL3 binding to MEST DNA via NFIL3 S-sulfhydration at Cys175 and Cys208. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. PECAM-1 drives β-catenin-mediated EndMT via internalization in colon cancer with diabetes mellitus
- Author
-
Qing Wu, Xingxing Du, Jianing Cheng, Xiuying Qi, Huan Liu, Xiaohong Lv, Xieyang Gong, Changxin Shao, Muhong Wang, Luxiao Yue, Xin Yang, Shiyu Li, Yafang Zhang, Xuemei Li, and Huike Yang
- Subjects
Colon cancer ,Diabetes mellitus ,EndMT ,PECAM-1 ,β-catenin ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background Diabetes mellitus (DM) is considered to be a risk factor in carcinogenesis and progression, although the biological mechanisms are not well understood. Here we demonstrate that platelet-endothelial cell adhesion molecule 1 (PECAM-1) internalization drives β-catenin-mediated endothelial-mesenchymal transition (EndMT) to link DM to cancer. Methods The tumor microenvironment (TME) was investigated for differences between colon cancer with and without DM by mRNA-microarray analysis. The effect of DM on colon cancer was determined in clinical patients and animal models. Furthermore, EndMT, PECAM-1 and Akt/GSK-3β/β-catenin signaling were analyzed under high glucose (HG) and human colon cancer cell (HCCC) supernatant (SN) or coculture conditions by western and immunofluorescence tests. Results DM promoted the progression and EndMT occurrence of colon cancer (CC). Regarding the mechanism, DM induced PECAM-1 defection from the cytomembrane, internalization and subsequent accumulation around the cell nucleus in endothelial cells, which promoted β-catenin entry into the nucleus, leading to EndMT occurrence in CC with DM. Additionally, Akt/GSK-3β signaling was enhanced to inhibit the degradation of β-catenin, which regulates the process of EndMT. Conclusions PECAM-1 defects and/or internalization are key events for β-catenin-mediated EndMT, which is significantly boosted by enhanced Akt/GSK-3β signaling in the DM-associated TME. This contributes to the mechanism by which DM promotes the carcinogenesis and progression of CC. Graphical Abstract Video Abstract
- Published
- 2023
- Full Text
- View/download PDF
34. Elucidating the underlying mechanisms of diabetic endotheliopathy using patient specific induced pluripotent stem cells
- Author
-
Eleftheriadou, Magdalini, Margariti, Andriana, and Stitt, Alan
- Subjects
iPSCs ,regenerative medicine ,diabetes ,endothelial cells ,induced pluripotent stem cells ,ROBO4 ,EMT ,EndMT ,angiogenesis ,patient specific cells ,reprogramming ,IPS-ECS ,hind limb ischemia model - Abstract
Diabetic endotheliopathy is the main cause for impaired angiogenesis and reduced neovascularization that lead to microvascular injury and vascular complications. The pathogenesis for vascular complications arising from diabetes is complex. Elucidation of key underlying mechanisms will help the development of novel therapies and the discovery of potential biomarkers. The ability to generate functional endothelial cells (ECs) from induced pluripotent stem cells (iPSCs) is a novel and powerful tool for cell-based therapies. Human iPSC-derived ECs (iPS-ECs) have a broad range of clinical applications including cell-based therapy, disease modelling and drug screening; they can be used in mechanistic studies towards the development of novel therapies and in the discovery of new biomarkers to be applied in regenerative medicine and treatment of diabetic vasculopathy. Here we utilize transcriptomic and proteomic technologies to assess patient-specific iPS-ECs from diabetic (DiPS-ECs) and non-diabetic (NiPS-ECS) donors in order to investigate the mechanisms driving endotheliopathy in diabetes. Our in vitro and in vivo models recapitulate the effects of hyperglycaemia on the vasculature in the clinical setting. RNA-seq data showed differential expression of genes and proteins involved in EC function and angiogenesis were significantly downregulated in DiPS-ECs in comparison to NDiPS-ECs. Moreover, factors involved endothelial-to-mesenchymal transition were increased in DiPS-ECs. DiPS-ECs had significantly reduced impaired tube formation and barrier stability in vitro. DiPS-ECs displayed impaired angiogenic function demonstrated by poor post-transplant engraftment and decreased blood flow recovery (BFR) when injected to the hindlimb of mice following femoral artery ligation. Proteomic and transcriptomic analysis confirmed imbalances in several pathways involved in endothelial function. Pathways involved in metabolism, homeostasis and stress response were found to be underrepresented in DiPS-ECs. RNA-seq revealed differences in the transcriptome of DiPS-ECs. EndMT related gene ontology was enriched in DiPS-ECs. Endothelial specific Roundabout protein 4 (ROBO4) is highly involved in pathways related to angiogenesis, barrier stability and endothelial health. Expression of ROBO4 was found to be impaired in DiPS-ECs and transcriptomic analysis along with in vitro and in vivo studies revealed its importance in vascular specification and angiogenesis. ROBO4 improved ECs function of DiPS-ECs and recovered their lost angiogenic function. Our data suggest that DiPS-ECs carry an imprint of the diabetic milieu which is reflected in their dysfunction. An unregulated EndMT process is proposed to cause a phenotype drift and poor angiogenic functionality observed in DiPS-ECs. ROBO4 seems to act as a fine tuning factor of this process. To the best of our knowledge, we aim to identify and report a novel disease-specific signature in diabetic iPS-ECs. Therefore, our human iPS-ECs model may serve as a valuable tool to study biological pathways and identify new treatments for diabetes-induced endotheliopathy.
- Published
- 2021
35. mTOR Signaling in Pulmonary Vascular Disease: Pathogenic Role and Therapeutic Target.
- Author
-
Babicheva, Aleksandra, Makino, Ayako, and Yuan, Jason X-J
- Subjects
Animals ,Humans ,Vascular Diseases ,Signal Transduction ,Clinical Trials as Topic ,TOR Serine-Threonine Kinases ,Molecular Targeted Therapy ,Pulmonary Arterial Hypertension ,EndMT ,RTK/PI3K/AKT/mTOR pathway ,Raptor ,Rictor ,SMC transition ,RTK ,PI3K ,AKT ,mTOR pathway ,Other Chemical Sciences ,Genetics ,Other Biological Sciences ,Chemical Physics - Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal disease without a cure. The exact pathogenic mechanisms of PAH are complex and poorly understood, yet a number of abnormally expressed genes and regulatory pathways contribute to sustained vasoconstriction and vascular remodeling of the distal pulmonary arteries. Mammalian target of rapamycin (mTOR) is one of the major signaling pathways implicated in regulating cell proliferation, migration, differentiation, and protein synthesis. Here we will describe the canonical mTOR pathway, structural and functional differences between mTOR complexes 1 and 2, as well as the crosstalk with other important signaling cascades in the development of PAH. The pathogenic role of mTOR in pulmonary vascular remodeling and sustained vasoconstriction due to its contribution to proliferation, migration, phenotypic transition, and gene regulation in pulmonary artery smooth muscle and endothelial cells will be discussed. Despite the progress in our elucidation of the etiology and pathogenesis of PAH over the two last decades, there is a lack of effective therapeutic agents to treat PAH patients representing a significant unmet clinical need. In this review, we will explore the possibility and therapeutic potential to use inhibitors of mTOR signaling cascade to treat PAH.
- Published
- 2021
36. Regulation of Partial and Reversible Endothelial-to-Mesenchymal Transition in Angiogenesis
- Author
-
Fang, Jennifer S, Hultgren, Nan W, and Hughes, Christopher CW
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Aetiology ,2.1 Biological and endogenous factors ,EndoMT ,EndMT ,endothelial ,mesenchymal ,cell identity ,cell plasticity ,angiogenesis ,partial EndoMT ,Biological sciences ,Biomedical and clinical sciences - Abstract
During development and in several diseases, endothelial cells (EC) can undergo complete endothelial-to-mesenchymal transition (EndoMT or EndMT) to generate endothelial-derived mesenchymal cells. Emerging evidence suggests that ECs can also undergo a partial EndoMT to generate cells with intermediate endothelial- and mesenchymal-character. This partial EndoMT event is transient, reversible, and supports both developmental and pathological angiogenesis. Here, we discuss possible regulatory mechanisms that may control the EndoMT program to dictate whether cells undergo complete or partial mesenchymal transition, and we further consider how these pathways might be targeted therapeutically in cancer.
- Published
- 2021
37. Diosmetin‐7‐O‐β‐D‐glucopyranoside suppresses endothelial–mesenchymal transformation through endoplasmic reticulum stress in cardiac fibrosis.
- Author
-
Wang, Huahua, Zhang, Xiaoyu, Liu, Yangyang, Zhang, Yunyun, Wang, Yingyu, Peng, Yunru, and Ding, Yongfang
- Subjects
- *
HEART fibrosis , *ENDOPLASMIC reticulum , *UNFOLDED protein response , *GLUCOSE-regulated proteins , *FLAVONOID glycosides , *TRANSMISSION electron microscopy , *PATHOLOGICAL physiology - Abstract
Diosmetin‐7‐O‐β‐D‐glucopyranoside (Diosmetin‐7‐O‐glucoside) is a natural flavonoid glycoside known to have a therapeutic application for cardiovascular diseases. Cardiac fibrosis is the main pathological change in the end stage of cardiovascular diseases. Endothelial‐mesenchymal transformation (EndMT) induced by endoplasmic reticulum stress (ER stress) via Src pathways is involved in the process of cardiac fibrosis. However, it is unclear whether and how diosmetin‐7‐O‐glucoside regulates EndMT and ER stress to treat cardiac fibrosis. In this study, molecular docking results showed that diosmetin‐7‐O‐glucoside bound well to ER stress and Src pathway markers. Diosmetin‐7‐O‐glucoside suppressed cardiac fibrosis induced by isoprenaline (ISO) and reduced the levels of EndMT, ER stress in mice heart. Primary cardiac microvascular endothelial cells (CMECs) were induced by transforming growth factor‐β1 (TGF‐β1) to perform EndMT. Diosmetin‐7‐O‐glucoside could effectively regulate EndMT and diminish the accumulation of collagen I and collagen III. We also showed that the tube formation in CMECs was restored, and the capacity of migration was partially inhibited. Diosmetin‐7‐O‐glucoside also ameliorated ER stress through the three unfolded protein response branches, as evidenced by organelle structure in transmission electron microscopy images and the expression of protein biomarkers like the glucose‐regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP). Further analysis showed that diosmetin‐7‐O‐glucoside could suppress the expression level of Src phosphorylation, then block EndMT with the maintenance of endothelial appearance and endothelial marker expression. These results suggested that the diosmetin‐7‐O‐glucoside can regulate EndMT through ER stress, at least in part via Src‐dependent pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
38. A novel Nanocellulose-Gelatin-AS-IV external stent resists EndMT by activating autophagy to prevent restenosis of grafts
- Author
-
Tianshu Chu, Qingye Li, Chun Dai, Xiang Li, Xiang Kong, Yangming Fan, Hongyan Yin, and Jianjun Ge
- Subjects
Restenosis ,Vein graft ,Extravascular stent ,NC-Gelatin hydrogel ,EndMT ,Materials of engineering and construction. Mechanics of materials ,TA401-492 ,Biology (General) ,QH301-705.5 - Abstract
Vein grafts are widely used for coronary artery bypass grafting and hemodialysis access, but restenosis remains the ''Achilles' heel'' of these treatments. An extravascular stent is one wrapped around the vein graft and provides mechanical strength; it can buffer high arterial pressure and secondary vascular dilation of the vein to prevent restenosis. In this study, we developed a novel Nanocellulose-gelatin hydrogel, loaded with the drug Astragaloside IV (AS-IV) as an extravascular scaffold to investigate its ability to reduce restenosis. We found that the excellent physical and chemical properties of the drug AS-IV loaded Nanocellulose-gelatin hydrogel external stent limit graft vein expansion and make the stent biocompatible. We also found it can prevent restenosis by resisting endothelial-to-mesenchymal transition (EndMT) in vitro. It does so by activating autophagy, and AS-IV can enhance this effect both in vivo and in vitro. This study has added to existing research on the mechanism of extravascular stents in preventing restenosis of grafted veins. Furthermore, we have developed a novel extravascular stent for the prevention and treatment of restenosis. This will help optimize the clinical treatment plan of external stents and improve the prognosis in patients with vein grafts.
- Published
- 2023
- Full Text
- View/download PDF
39. Oscillatory shear stress modulates Notch-mediated endothelial mesenchymal plasticity in cerebral arteriovenous malformations
- Author
-
C. L. Karthika, Vani Venugopal, B. J. Sreelakshmi, S. Krithika, Jaya Mary Thomas, Mathew Abraham, C. C. Kartha, Arumugam Rajavelu, and S. Sumi
- Subjects
Cerebral arteriovenous malformations ,Shear stress ,Endothelial cells ,EndMT ,Notch ,Small-molecule inhibitors ,Cytology ,QH573-671 - Abstract
Abstract Background Cerebral arteriovenous malformations (cAVM) are a significant cause of intracranial hemorrhagic stroke and brain damage. The arteriovenous junctions in AVM nidus are known to have hemodynamic disturbances such as altered shear stress, which could lead to endothelial dysfunction. The molecular mechanisms coupling shear stress and endothelial dysfunction in cAVMs are poorly understood. We speculated that disturbed blood flow in artery–vein junctions activates Notch receptors and promotes endothelial mesenchymal plasticity during cAVM formation. Methods We investigated the expression profile of endothelial mesenchymal transition (EndMT) and cell adhesion markers, as well as activated Notch receptors, in 18 human cAVM samples and 15 control brain tissues, by quantitative real-time PCR (qRT-PCR) and immunohistochemical evaluation. Employing a combination of a microfluidic system, qRT-PCR, immunofluorescence, as well as invasion and inhibitor assays, the effects of various shear stress conditions on Notch-induced EndMT and invasive potential of human cerebral microvascular endothelial cells (hCMEC/d3) were analyzed. Results We found evidence for EndMT and enhanced expression of activated Notch intracellular domain (NICD3 and NICD4) in human AVM nidus samples. The expression of transmembrane adhesion receptor integrin α9/β1 is significantly reduced in cAVM nidal vessels. Cell–cell adhesion proteins such as VE-cadherin and N-cadherin were differentially expressed in AVM nidus compared with control brain tissues. Using well-characterized hCMECs, we show that altered fluid shear stress steers Notch3 nuclear translocation and promotes SNAI1/2 expression and nuclear localization. Oscillatory flow downregulates integrin α9/β1 and VE-cadherin expression, while N-cadherin expression and endothelial cell invasiveness are augmented. Gamma-secretase inhibitor RO4929097, and to a lesser level DAPT, prevent the mesenchymal transition and invasiveness of cerebral microvascular endothelial cells exposed to oscillatory fluid flow. Conclusions Our study provides, for the first time, evidence for the role of oscillatory shear stress in mediating the EndMT process and dysregulated expression of cell adhesion molecules, especially multifunctional integrin α9/β1 in human cAVM nidus. Concomitantly, our findings indicate the potential use of small-molecular inhibitors such as RO4929097 in the less-invasive therapeutic management of cAVMs. Graphical Abstract
- Published
- 2023
- Full Text
- View/download PDF
40. Understanding the role of the long non-coding RNA MIR503HG in endothelial-to-mesenchymal transition during vascular remodelling
- Author
-
Pinho Monteiro, João Pedro, Baker, Andrew, and Caporali, Andrea
- Subjects
vascular remodelling ,EndMT ,endothelial cells ,lncRNA MIR503HG - Abstract
Endothelial-to-mesenchymal transition (EndMT) is a dynamic biological process present during development and involved in a variety of pathological vascular remodelling scenarios. However, despite our growing understanding of the key cellular alterations required, the precise molecular determinants governing this phenotypical transition remain elusive. With long non-coding RNAs (lncRNA) now emerging as powerful regulators of gene expression we sought to understand their role in the process of EndMT. To replicate EndMT in vitro and characterise its molecular signature, human umbilical vein endothelial cells (HUVEC) and human pulmonary artery endothelial cells (HPAEC) were exposed to a continuous co-treatment of transforming growth factor-beta 2 (TGF-β2) and interleukin 1 beta (IL-1β) for a total of 7 days. Using high-throughput RNA-sequencing analysis of these cells, a total of 103 differential expressed lncRNAs were identified. Of these, the downregulation of the lncRNA MIR503HG was found to be a prevalent feature present in multiple human primary EC types undergoing EndMT in vitro. Further analysis revealed that depletion of MIR503HG was sufficient to elicit a robust EndMT phenotype, with a significant increase in the expression of SNAI2, ACTA2 and COL1A1, accompanied by repression of CD31. Conversely, ectopic expression of a single MIR503HG transcript suppressed these hallmark EndMT-associated changes despite TGF-β2 and IL-1β co-treatment. Accompanying RNA-sequencing of these cells showed that the overexpression of MIR503HG alone was able to inhibit over 25% of the EndMT transcriptional profile. Crucially, these changes were found to be independent of the functional regulation of miR-503 and miR-424, found within the MIR503HG locus. Our findings were then confirmed in vivo using a sugen/hypoxia-induced model of pulmonary hypertension (PH) established in endothelial lineage-tracing mice. Here, the expression of the MIR503HG mouse homolog (Gm28730) was significantly downregulated in association with an EndMT profile in the lung. Conversely, targeted up-regulation of MIR503HG in the mouse lung significantly suppressed the appearance of mesenchymal markers in CD31+ cells during PH. Notably, MIR503HG availability was also found to be decreased in lung tissue sections from patients with idiopathic pulmonary arterial hypertension (IPAH) and cultured blood outgrowth ECs isolated from patients with heritable pulmonary arterial hypertension (HPAH). Collectively, our studies identify MIR503HG as essential in maintaining EC phenotypical commitment and preventing EndMT both in vitro and during disease.
- Published
- 2020
- Full Text
- View/download PDF
41. Cardiomyocyte-Restricted Expression of IL11 Causes Cardiac Fibrosis, Inflammation, and Dysfunction.
- Author
-
Sweeney, Mark, O'Fee, Katie, Villanueva-Hayes, Chelsie, Rahman, Ekhlas, Lee, Michael, Vanezis, Konstantinos, Andrew, Ivan, Lim, Wei-Wen, Widjaja, Anissa, Barton, Paul J. R., and Cook, Stuart A.
- Subjects
- *
HEART fibrosis , *TRANSFORMING growth factors , *LEFT ventricular dysfunction , *HEART diseases , *TRANSFORMING growth factors-beta , *INFLAMMATION - Abstract
Cardiac fibrosis is a common pathological process in heart disease, representing a therapeutic target. Transforming growth factor β (TGFβ) is the canonical driver of cardiac fibrosis and was recently shown to be dependent on interleukin 11 (IL11) for its profibrotic effects in fibroblasts. In the opposite direction, recombinant human IL11 has been reported as anti-fibrotic and anti-inflammatory in the mouse heart. In this study, we determined the effects of IL11 expression in cardiomyocytes on cardiac pathobiology and function. We used the Cre-loxP system to generate a tamoxifen-inducible mouse with cardiomyocyte-restricted murine Il11 expression. Using protein assays, bulk RNA-sequencing, and in vivo imaging, we analyzed the effects of IL11 on myocardial fibrosis, inflammation, and cardiac function, challenging previous reports suggesting the cardioprotective potential of IL11. TGFβ stimulation of cardiomyocytes caused Il11 upregulation. Compared to wild-type controls, Il11-expressing hearts demonstrated severe cardiac fibrosis and inflammation that was associated with the upregulation of cytokines, chemokines, complement factors, and increased inflammatory cells. IL11 expression also activated a program of endothelial-to-mesenchymal transition and resulted in left ventricular dysfunction. Our data define species-matched IL11 as strongly profibrotic and proinflammatory when secreted from cardiomyocytes and further establish IL11 as a disease factor. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
42. Editorial: Endothelial-to-mesenchymal transition in cardiovascular disease
- Author
-
Mabruka Alfaidi, Paul C. Evans, and J. Geoffrey Pickering
- Subjects
EndMT ,cardiovascular diseases ,pathogenesis ,molecular pathways ,endothelial-to-mesenchymal transition ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Published
- 2023
- Full Text
- View/download PDF
43. PRELP secreted from mural cells protects the function of blood brain barrier through regulation of endothelial cell-cell integrity
- Author
-
Hongorzul Davaapil, Jack Hopkins, Nadia Bonnin, Vasiliki Papadaki, Alex Leung, Hirofumi Kosuge, Takumi Tashima, Makoto Nakakido, Ryohei Sekido, Kouhei Tsumoto, Mandeep S. Sagoo, and Shin-Ichi Ohnuma
- Subjects
PRELP ,vasculature ,neuroinflammation ,integrity ,adhesion ,EndMT ,Biology (General) ,QH301-705.5 - Abstract
Introduction: Proline/arginine-rich end leucine-rich repeat protein (PRELP), is a small secreted proteoglycan expressed by pericytes and vascular smooth muscle cells surrounding the brain vasculature of adult mouse.Methods: We utilised a Prelp knockout (Prelp−/−) mouse model to interrogate vasculature integrity in the brain alongside performing in vitro assays to characterise PRELP application to endothelial cells lines. Our findings were supplemented with RNA expression profiling to elucidate the mechanism of how PRELP maintains neurovasculature function.Results:Prelp−/− mice presented with neuroinflammation and reducedneurovasculature integrity, resulting in IgG and dextran leakage in the cerebellum and cortex. Histological analysis of Prelp−/− mice revealed reducedcell-cell integrity of the blood brain barrier, capillary attachment of pericytes andastrocyte end-feet. RNA-sequencing analysis found that cell-cell adhesion andinflammation are affected in Prelp−/− mice and gene ontology analysis as well as gene set enrichment analysis demonstrated that inflammation related processes and adhesion related processes such as epithelial-mesenchymal transition and apical junctions were significantly affected, suggesting PRELP is a regulator of cell-cell adhesion. Immunofluorescence analysis showed that adhesion junction protein expression levels of cadherin, claudin-5, and ZO-1, was suppressed in Prelp−/− mice neurovasculature. Additionally, in vitro studies revealed that PRELP application to endothelial cells enhances cell-cell integrity, induces mesenchymal-endothelial transition and inhibits TGF-β mediated damage to cell-cell adhesion.Discussion: Our study indicates that PRELP is a novel endogenous secreted regulator of neurovasculature integrity and that PRELP application may be a potential treatment for diseases associated with neurovascular damage.
- Published
- 2023
- Full Text
- View/download PDF
44. Dengue Virus Infection Alters Inter-Endothelial Junctions and Promotes Endothelial–Mesenchymal-Transition-Like Changes in Human Microvascular Endothelial Cells.
- Author
-
Escudero-Flórez, Manuela, Torres-Hoyos, David, Miranda-Brand, Yaneth, Gallego-Gómez, Juan Carlos, and Vicente-Manzanares, Miguel
- Subjects
- *
DENGUE viruses , *ENDOTHELIAL cells , *VIRUS diseases , *CELL junctions , *HYPOVOLEMIC anemia , *TEMPOROPARIETAL junction - Abstract
Dengue virus (DENV) is a pathogenic arbovirus that causes human disease. The most severe stage of the disease (severe dengue) is characterized by vascular leakage, hypovolemic shock, and organ failure. Endothelial dysfunction underlies these phenomena, but the causal mechanisms of endothelial dysfunction are poorly characterized. This study investigated the role of c-ABL kinase in DENV-induced endothelial dysfunction. Silencing c-ABL with artificial miRNA or targeting its catalytic activity with imatinib revealed that c-ABL is required for the early steps of DENV infection. DENV-2 infection and conditioned media from DENV-infected cells increased endothelial expression of c-ABL and CRKII phosphorylation, promoted expression of mesenchymal markers, e.g., vimentin and N-cadherin, and decreased the levels of endothelial-specific proteins, e.g., VE-cadherin and ZO-1. These effects were reverted by silencing or inhibiting c-ABL. As part of the acquisition of a mesenchymal phenotype, DENV infection and treatment with conditioned media from DENV-infected cells increased endothelial cell motility in a c-ABL-dependent manner. In conclusion, DENV infection promotes a c-ABL-dependent endothelial phenotypic change that leads to the loss of intercellular junctions and acquisition of motility. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
45. Partial endothelial-to-mesenchymal transition mediated by HIF-induced CD45 in neointima formation upon carotid artery ligation.
- Author
-
Yamashiro, Yoshito, Ramirez, Karina, Nagayama, Kazuaki, Hattori, Naoko, Liu, Yu-Yu, Matsunaga, Shinji, Tomita, Shuhei, Kubota, Yoshiaki, and Yanagisawa, Hiromi
- Subjects
- *
CAROTID artery , *CD45 antigen , *CADHERINS , *VASCULAR cell adhesion molecule-1 , *VASCULAR remodeling , *COBALT chloride , *PHOSPHATASE inhibitors - Abstract
Aims Endothelial-to-mesenchymal transition (EndMT) is a fundamental process in vascular remodelling. However, the precise regulatory mechanism of vascular remodelling during neointima formation and the source of neointima cells are not entirely understood. Methods and results To investigate the origin of neointima cells and their relevance to vascular wall remodelling, we used an endothelial cell (EC)-specific lineage tracing system [VE-Cadherin (Cdh5)-BAC-CreERT2 mice] and carotid artery ligation model and showed evidence that resident ECs transdifferentiate into neointima cells with the expression of CD45. During the early stages of neointima formation, ECs transiently expressed CD45, a haematopoietic marker, accompanied by a host of EndMT markers, and CD31 and αSMA were prominently expressed in developing neointima. In vitro , CD45-positive EndMT was induced by stabilization of HIF1α with cobalt chloride or with a VHL inhibitor in human primary ECs, which mimicked the hypoxic condition of the ligated artery, and promoted the formation of an integrin α11-shank-associated RH domain-interacting protein (SHARPIN) complex. Notably, a CD45 phosphatase inhibitor disrupted this integrin α11-SHARPIN complex, thereby destabilizing cell–cell junctions. Deletion of Hif1α in ECs suppressed expression of CD45 and EndMT markers and ameliorated neointima formation. Conclusion These results suggest that the HIF-induced CD45 expression is normally required for the retention of an EC fate and cell–cell junctions, CD45-positive EndMT (termed as 'partial EndMT') contributes to neointima formation and vascular wall remodelling. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
46. Combined Shikonin-Loaded MPEG-PCL Micelles Inhibits Effective Transition of Endothelial-to-Mesenchymal Cells
- Author
-
Li G, Shang C, Li Q, Chen L, Yue Z, Ren L, Yang J, Zhang J, and Wang W
- Subjects
shikonin ,shikonin-loaded mpeg-pcl micelles ,inflammation ,endmt ,Medicine (General) ,R5-920 - Abstract
Guanglin Li,1,2 Chenxu Shang,3 Qingqing Li,4 Lifang Chen,1,2 Zejun Yue,1,2 Lingxuan Ren,3 Jianjun Yang,3 Jiye Zhang,4 Weirong Wang1,2 1Department of Medical Laboratory Animal Science, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, People’s Republic of China; 2Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, People’s Republic of China; 3Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, People’s Republic of China; 4School of Pharmacy, Xi’an Jiaotong University Health Science Center, Xi’an, People’s Republic of ChinaCorrespondence: Weirong Wang, Xi’an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi’an, 710061, People’s Republic of China, Tel/Fax +86 29 82655362, Email szb2013072@xjtu.edu.cnIntroduction: Shikonin is well known for its anti-inflammatory activity in cardiovascular diseases. However, the application of shikonin is limited by its low water solubility and poor bioavailability. Methoxy poly (ethylene glycol)-b-poly (ϵ-caprolactone) (MPEG-PCL) is considered a promising delivery system for hydrophobic drugs. Therefore, in this study, we prepared shikonin-loaded MPEG-PCL micelles and investigated their effect on endothelial-to-mesenchymal transition (EndMT) induced by inflammatory cytokines.Methods: Shikonin was encapsulated in MPEG-PCL micelles using an anti-solvent method and the physiochemical characteristics of the micelles (particle size, zeta potential, morphology, critical micelle concentration (CMC), drug loading and encapsulation efficiency) were investigated. Cellular uptake of micelles in human umbilical vein endothelial cells (HUVECs) was evaluated using fluorescence microscopy. In vitro EndMT inhibition was explored in HUVECs by quantitative real-time PCR analysis.Results: The average particle size of shikonin-loaded MPEG-PCL micelles was 54.57± 0.13 nm and 60 nm determined by dynamic light scattering and transmission electron microscopy, respectively. The zeta potential was − 6.23± 0.02 mV. The CMC of the micelles was 6.31× 10− 7mol/L. The drug loading and encapsulation efficiency were 0.88± 0.08% and 43.08± 3.77%, respectively. The MPEG-PCL micelles significantly improved the cellular uptake of cargo with low water solubility. Real-time PCR analysis showed that co-treatment with TNF-α and IL-1β successfully induced EndMT in HUVECs, whereas this process was significantly inhibited by shikonin and shikonin-loaded MPEG-PCL micelles, with greater inhibition mediated by the shikonin-loaded MPEG-PCL micelles.Conclusion: Shikonin-loaded MPEG-PCL micelles significantly improved the EndMT-inhibiting effect of the free shikonin. MPEG-PCL is suitable for use more generally as a lipophilic drug carrier.Keywords: shikonin, shikonin-loaded MPEG-PCL micelles, inflammation, EndMT
- Published
- 2022
47. TGFβ signaling pathways in human health and disease
- Author
-
Pei-Yu Chen, Lingfeng Qin, and Michael Simons
- Subjects
TGFβ ,EndMT ,aneurysm ,cell fate ,smooth muscle cell ,endothelail cell ,Biology (General) ,QH301-705.5 - Abstract
Transforming growth factor beta (TGFβ) is named for the function it was originally discovered to perform-transformation of normal cells into aggressively growing malignant cells. It became apparent after more than 30 years of research, however, that TGFβ is a multifaceted molecule with a myriad of different activities. TGFβs are widely expressed with almost every cell in the human body producing one or another TGFβ family member and expressing its receptors. Importantly, specific effects of this growth factor family differ in different cell types and under different physiologic and pathologic conditions. One of the more important and critical TGFβ activities is the regulation of cell fate, especially in the vasculature, that will be the focus of this review.
- Published
- 2023
- Full Text
- View/download PDF
48. The Provenance, Providence, and Position of Endothelial Cells in Injured Spinal Cord Vascular Pathology.
- Author
-
Chopra, Manjeet, Bhagwani, Ankita, and Kumar, Hemant
- Subjects
- *
ENDOTHELIAL cells , *PERICYTES , *SPINAL cord , *CENTRAL nervous system injuries , *PATHOLOGY , *DIVINE providence - Abstract
Endothelial cells (ECs) and pericytes are present in all blood vessels. Their position confers an important role in controlling oxygen and nutrient transportation to the different organs. ECs can adopt different morphologies based on their need and functions. Both ECs and pericytes express different surface markers that help in their identification, but heterogeneity and overlapping between markers among different cells pose a challenge for their precise identification. Spatiotemporal association of ECs and pericytes have great importance in sprout formation and vessel stabilization. Any traumatic injury in CNS may lead to vascular damage along with neuronal damage. Hence, ECs–pericyte interaction by physical contact and paracrine molecules is crucial in recovering the epicenter region by promoting angiogenesis. ECs can transform into other types of cells through endothelial–mesenchymal transition (EndMT), promoting wound healing in the epicenter region. Various signaling pathways mediate the interaction of ECs with pericytes that have an extensive role in angiogenesis. In this review, we discussed ECs and pericytes surface markers, the spatiotemporal association and interaction of ECs–pericytes, and signaling associated with the pathology of traumatic SCI. Linking the brain or spinal cord-specific pathologies and human vascular pathology will pave the way toward identifying new therapeutic targets and developing innovative preventive strategies. Endothelial-pericyte interaction strategic for formation of functional neo-vessels that are crucial for neurological recovery [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
49. Epigallocatechin-3-Gallate Attenuates Myocardial Dysfunction via Inhibition of Endothelial-to-Mesenchymal Transition.
- Author
-
Kim, Sejin, Lee, Hyunjae, Moon, Hanbyeol, Kim, Ran, Kim, Minsuk, Jeong, Seongtae, Kim, Hojin, Kim, Sang Hyeon, Hwang, Soo Seok, Lee, Min Young, Kim, Jongmin, Song, Byeong-Wook, and Chang, Woochul
- Subjects
EPIGALLOCATECHIN gallate ,TRANSFORMING growth factors ,CELL physiology ,MYOCARDIAL infarction ,UMBILICAL veins ,CARDIOVASCULAR diseases - Abstract
Cardiac tissue damage following ischemia leads to cardiomyocyte apoptosis and myocardial fibrosis. Epigallocatechin-3-gallate (EGCG), an active polyphenol flavonoid or catechin, exerts bioactivity in tissues with various diseases and protects ischemic myocardium; however, its association with the endothelial-to-mesenchymal transition (EndMT) is unknown. Human umbilical vein endothelial cells (HUVECs) pretreated with transforming growth factor β2 (TGF-β2) and interleukin 1β (IL-1β) were treated with EGCG to verify cellular function. In addition, EGCG is involved in RhoA GTPase transmission, resulting in reduced cell mobility, oxidative stress, and inflammation-related factors. A mouse myocardial infarction (MI) model was used to confirm the association between EGCG and EndMT in vivo. In the EGCG-treated group, ischemic tissue was regenerated by regulating proteins involved in the EndMT process, and cardioprotection was induced by positively regulating apoptosis and fibrosis of cardiomyocytes. Furthermore, EGCG can reactivate myocardial function due to EndMT inhibition. In summary, our findings confirm that EGCG is an impact activator controlling the cardiac EndMT process derived from ischemic conditions and suggest that supplementation with EGCG may be beneficial in the prevention of cardiovascular disease. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
50. Oscillatory shear stress modulates Notch-mediated endothelial mesenchymal plasticity in cerebral arteriovenous malformations.
- Author
-
Karthika, C. L., Venugopal, Vani, Sreelakshmi, B. J., Krithika, S., Thomas, Jaya Mary, Abraham, Mathew, Kartha, C. C., Rajavelu, Arumugam, and Sumi, S.
- Abstract
Background: Cerebral arteriovenous malformations (cAVM) are a significant cause of intracranial hemorrhagic stroke and brain damage. The arteriovenous junctions in AVM nidus are known to have hemodynamic disturbances such as altered shear stress, which could lead to endothelial dysfunction. The molecular mechanisms coupling shear stress and endothelial dysfunction in cAVMs are poorly understood. We speculated that disturbed blood flow in artery–vein junctions activates Notch receptors and promotes endothelial mesenchymal plasticity during cAVM formation. Methods: We investigated the expression profile of endothelial mesenchymal transition (EndMT) and cell adhesion markers, as well as activated Notch receptors, in 18 human cAVM samples and 15 control brain tissues, by quantitative real-time PCR (qRT-PCR) and immunohistochemical evaluation. Employing a combination of a microfluidic system, qRT-PCR, immunofluorescence, as well as invasion and inhibitor assays, the effects of various shear stress conditions on Notch-induced EndMT and invasive potential of human cerebral microvascular endothelial cells (hCMEC/d3) were analyzed. Results: We found evidence for EndMT and enhanced expression of activated Notch intracellular domain (NICD3 and NICD4) in human AVM nidus samples. The expression of transmembrane adhesion receptor integrin α9/β1 is significantly reduced in cAVM nidal vessels. Cell–cell adhesion proteins such as VE-cadherin and N-cadherin were differentially expressed in AVM nidus compared with control brain tissues. Using well-characterized hCMECs, we show that altered fluid shear stress steers Notch3 nuclear translocation and promotes SNAI1/2 expression and nuclear localization. Oscillatory flow downregulates integrin α9/β1 and VE-cadherin expression, while N-cadherin expression and endothelial cell invasiveness are augmented. Gamma-secretase inhibitor RO4929097, and to a lesser level DAPT, prevent the mesenchymal transition and invasiveness of cerebral microvascular endothelial cells exposed to oscillatory fluid flow. Conclusions: Our study provides, for the first time, evidence for the role of oscillatory shear stress in mediating the EndMT process and dysregulated expression of cell adhesion molecules, especially multifunctional integrin α9/β1 in human cAVM nidus. Concomitantly, our findings indicate the potential use of small-molecular inhibitors such as RO4929097 in the less-invasive therapeutic management of cAVMs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.