1
|
Zhao K, Zeng Z, He Y, Zhao R, Niu J, Sun H, Li S, Dong J, Jing Z, Zhou J. Recent advances in targeted therapy for inflammatory vascular diseases. J Control Release 2024; 372:730-750. [PMID: 38945301 DOI: 10.1016/j.jconrel.2024.06.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
Vascular diseases constitute a significant contributor to worldwide mortality rates, placing a substantial strain on healthcare systems and socio-economic aspects. They are closely associated with inflammatory responses, as sustained inflammation could impact endothelial function, the release of inflammatory mediators, and platelet activation, thus accelerating the progression of vascular diseases. Consequently, directing therapeutic efforts towards mitigating inflammation represents a crucial approach in the management of vascular diseases. Traditional anti-inflammatory medications may have extensive effects on multiple tissues and organs when absorbed through the bloodstream. Conversely, treatments targeting inflammatory vascular diseases, such as monoclonal antibodies, drug-eluting stents, and nano-drugs, can achieve more precise effects, including precise intervention, minimal non-specific effects, and prolonged efficacy. In addition, personalized therapy is an important development trend in targeted therapy for inflammatory vascular diseases. Leveraging advanced simulation algorithms and clinical trial data, treatment strategies are gradually being personalized based on patients' genetic, biomarker, and clinical profiles. It is expected that the application of precision medicine in the field of vascular diseases will have a broader future. In conclusion, targeting therapies offer enhanced safety and efficacy compared to conventional medications; investigating novel targeting therapies and promoting clinical transformation may be a promising direction in improving the prognosis of patients with inflammatory vascular diseases. This article reviews the pathogenesis of inflammatory vascular diseases and presents a comprehensive overview of the potential for targeted therapies in managing this condition.
Collapse
Affiliation(s)
- Kaiwen Zhao
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Zan Zeng
- Department of Vascular Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yuzhen He
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Rong Zhao
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Jinzhu Niu
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Huiying Sun
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Shuangshuang Li
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Jian Dong
- Department of Vascular Surgery, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zaiping Jing
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Jian Zhou
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China; Department of Vascular Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China; Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai, China.
| |
Collapse
|
2
|
Rostami Z, Saharkhiz M, Khorashadizadeh M, Ayadilord M, Naseri M. The immunoregulatory property of mesenchymal stem cells in Crocin treatment by expression modulation of microRNA-155, microRNA-21, microRNA-23b, microRNA-126a, and their target inflammatory genes. Gene 2024; 916:148446. [PMID: 38583816 DOI: 10.1016/j.gene.2024.148446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/14/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Mesenchymal stem cells (MSCs) have high priority in clinical applications for treatment of immune disorders because of their immunomodulatory function. A lot of researches have currently been undertaken to enhance the stemness capacities of the cells and pick an excellent type of MSCs for clinical approaches. This study aims to assess the immunomodulatory related MicroRNAs (miRNAs) expression as well as their target genes in both adipose derived stem cells (Ad-SCs) and dental pulp derived stem cell (DP-SCs) in the presence or lack of Crocin (saffron plant's bioactive compound). For this purpose, first MSCs were extracted from adipose and dental pulp tissues, and then their mesenchymal nature was confirmed using flow cytometry and differentiation tests. Following the cell treatment with an optimal-non-toxic dose of Crocin (Obtained by MTT test), the expression of 4 selected immunomodulatory-related micro-RNAs (Mir-126, -21, -23, and-155) and their target genes (PI3K/ Akt 1 and 2/ NFKB and RELA) were assessed by RT-PCR. Our findings revealed that miRNA-23 and miRNA-126 were up-regulated in both types of cells treated with Crocin, while in the other side, miRNA-21 and miRNA-155 were down-regulated in DP-SCs and were up-regulated in Ad-SCs under treatment. Moreover, the real-time PCR results indicated that Crocin could significantly down regulate the expression of PI3K/ Akt1/ Akt2/ NFKB/ RELA genes in DP-SCs and PI3K/Akt2 genes in Ad-SCs and up regulate the expression of Akt1/ NFKB/ RELA genes in recent cells. Based on the analysis of the obtained data, the immunoregulatory effects of Crocin were higher in DP-SCs than in Ad-SCs. In conclusion, Crocin could control essential signaling pathways related to the inflammation by regulating the expression of related- miRNAs genes that play a key function in the immune regulation pathways in MSCs. Our findings can give an understanding of the mechanisms by which Crocin enhances the immunomodulatory feature of MSCs. According to the research findings, DP-SCs are probably a better immunomodulator in Crocin treatment than Ad-SCs and it may be helpful for MSCs selection in clinical applications for modulation or treatment of autoimmune disorders.
Collapse
Affiliation(s)
- Zeinab Rostami
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran; Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mansoore Saharkhiz
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran; Department of Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Khorashadizadeh
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran; Department of Medical Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Malaksima Ayadilord
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Naseri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran; Department of Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
3
|
Ali I, Zhang H, Zaidi SAA, Zhou G. Understanding the intricacies of cellular senescence in atherosclerosis: Mechanisms and therapeutic implications. Ageing Res Rev 2024; 96:102273. [PMID: 38492810 DOI: 10.1016/j.arr.2024.102273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/16/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Cardiovascular disease is currently the largest cause of mortality and disability globally, surpassing communicable diseases, and atherosclerosis is the main contributor to this epidemic. Aging is intimately linked to atherosclerosis development and progression, however, the mechanism of aging in atherosclerosis is not well known. To emphasize the significant research on the involvement of senescent cells in atherosclerosis, we begin by outlining compelling evidence that indicates various types of senescent cells and SASP factors linked to atherosclerotic phenotypes. We subsequently provide a comprehensive summary of the existing knowledge, shedding light on the intricate mechanisms through which cellular senescence contributes to the pathogenesis of atherosclerosis. Further, we cover that senescence can be identified by both structural changes and several senescence-associated biomarkers. Finally, we discuss that preventing accelerated cellular senescence represents an important therapeutic potential, as permanent changes may occur in advanced atherosclerosis. Together, the review summarizes the relationship between cellular senescence and atherosclerosis, and inspects the molecular knowledge, and potential clinical significance of senescent cells in developing senescent-based therapy, thus providing crucial insights into their biology and potential therapeutic exploration.
Collapse
Affiliation(s)
- Ilyas Ali
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China
| | - Hongliang Zhang
- Shenzhen University General Hospital, Shenzhen University, Shenzhen 518060, PR China
| | - Syed Aqib Ali Zaidi
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China.
| |
Collapse
|
4
|
Han X, Gao C, Lu W, Yan J, Xu H, Guo Z, Qin W, Lu N, Gao J, Zhu W, Fu Y, Jiao K. Macrophage-Derived Extracellular DNA Initiates Heterotopic Ossification. Inflammation 2023; 46:2225-2240. [PMID: 37458919 DOI: 10.1007/s10753-023-01873-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/17/2023] [Accepted: 07/04/2023] [Indexed: 11/25/2023]
Abstract
Heterotopic ossification (HO) severely affects people's lives; however, its pathological mechanism remains poorly understood. Although extracellular DNA (ecDNA) has been shown to play important roles in pathological calcification, its effects in HO development and progression remain unknown. The in vivo rat Achilles tendon injury model and in vitro collagen I calcification model were used to evaluate the effects of ecDNA in the ectopic calcifications and the main cell types involved in those pathological process. Histology, immunofluorescent staining, reverse transcriptase-polymerase chain reaction analysis and micro-computed tomography were used to identify the distribution of macrophage-derived ecDNA and elucidate their roles in HO. The results showed that the amount of ecDNA and ectopic calcification increased significantly and exhibited a strong correlation in the injured tendons of HO model compared with those of the controls, which was accompanied by a significantly increased number of M2 macrophages in the injured tendon. During in vitro co-culture experiments, M2 macrophages calcified the reconstituted type I collagen and ectopic bone collected from the injured tendons of HO rats, while those effects were inhibited by deoxyribonuclease. More importantly, deoxyribonuclease reversed the pathological calcification in the injured rat tendon HO model. The present study showed that ecDNA from M2 macrophages initiates pathological calcification in HO, and the elimination of ecDNA might be developed into a clinical strategy to prevent ectopic mineralization diseases. The use of deoxyribonuclease for the targeted degradation of ecDNA at affected tissue sites provides a potential solution to treat diseases associated with ectopic mineralization.
Collapse
Affiliation(s)
- Xiaoxiao Han
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
- The College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Changhe Gao
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Weicheng Lu
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jianfei Yan
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haoqing Xu
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
- The College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Zhenxing Guo
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenpin Qin
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Naining Lu
- Department of Neurobiology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jialu Gao
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weiwei Zhu
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
- The College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Yutong Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
- The College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Kai Jiao
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
5
|
Rossi GP, Barton M, Dhaun N, Rizzoni D, Seccia TM. Challenges in the evaluation of endothelial cell dysfunction: a statement from the European Society of Hypertension Working Group on Endothelin and Endothelial Factors. J Hypertens 2023; 41:369-379. [PMID: 36728915 DOI: 10.1097/hjh.0000000000003314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Endothelial cell function is mediated by different mechanisms in different vascular beds. Moreover, in humans, endothelial cell dysfunction triggers and accelerates the progression of cardiovascular and chronic kidney diseases. Progression of such diseases can be in part mitigated by the control of cardiovascular risk factors and drugs targeting different systems, including endothelin receptor antagonists (ERAs), renin-angiotensin aldosterone antagonists and agents affecting glucose metabolism, all of which were shown to improve endothelial cell function. In recent years, the microRNAs, which are endogenous regulators of gene expression, have been identified as transmitters of information from endothelial cells to vascular smooth muscle cells, suggesting that they can entail tools to assess the endothelial cell dysfunction in arterial hypertension and target for pharmacologic intervention. This article critically reviews current challenges and limitations of available techniques for the invasive and noninvasive assessment of endothelial cell function, and also discusses therapeutic aspects as well as directions for future research in the areas of endothelial cell biology and pathophysiology in humans.
Collapse
Affiliation(s)
- Gian Paolo Rossi
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Padova, Italy
| | - Matthias Barton
- Molecular Internal Medicine, University of Zürich, and Andreas Grüntzig Foundation, Zürich, Switzerland
| | - Neeraj Dhaun
- University/British Heart Foundation Centre of Research Excellence, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia and Division of Medicine, Istituto Clinico Città di Brescia, Brescia, Italy
| | - Teresa M Seccia
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Padova, Italy
| |
Collapse
|
6
|
Han N, Ma Y, Li Y, Zheng Y, Wu C, Gan T, Li M, Ma L, Zhang J. Imaging and Hemodynamic Characteristics of Vulnerable Carotid Plaques and Artificial Intelligence Applications in Plaque Classification and Segmentation. Brain Sci 2023; 13:brainsci13010143. [PMID: 36672124 PMCID: PMC9856903 DOI: 10.3390/brainsci13010143] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/24/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Stroke is a massive public health problem. The rupture of vulnerable carotid atherosclerotic plaques is the most common cause of acute ischemic stroke (AIS) across the world. Currently, vessel wall high-resolution magnetic resonance imaging (VW-HRMRI) is the most appropriate and cost-effective imaging technique to characterize carotid plaque vulnerability and plays an important role in promoting early diagnosis and guiding aggressive clinical therapy to reduce the risk of plaque rupture and AIS. In recent years, great progress has been made in imaging research on vulnerable carotid plaques. This review summarizes developments in the imaging and hemodynamic characteristics of vulnerable carotid plaques on the basis of VW-HRMRI and four-dimensional (4D) flow MRI, and it discusses the relationship between these characteristics and ischemic stroke. In addition, the applications of artificial intelligence in plaque classification and segmentation are reviewed.
Collapse
Affiliation(s)
- Na Han
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
- Second Clinical School, Lanzhou University, Lanzhou 730030, China
| | - Yurong Ma
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Yan Li
- School of Mathematics and Statistics, Lanzhou University, Lanzhou 730030, China
| | - Yu Zheng
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
- Second Clinical School, Lanzhou University, Lanzhou 730030, China
| | - Chuang Wu
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Tiejun Gan
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Min Li
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Laiyang Ma
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
- Second Clinical School, Lanzhou University, Lanzhou 730030, China
| | - Jing Zhang
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
- Correspondence: ; Tel.: +86-139-1999-2479
| |
Collapse
|
7
|
Wen J, Deng J, Xiao T, Liu Y, Meng W. Adipose Rheb deficiency promotes miR-182-5p expression via the cAMP/PPARγ signaling pathway. J Genet Genomics 2023; 50:20-26. [PMID: 35550871 DOI: 10.1016/j.jgg.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 02/06/2023]
Abstract
Dysregulation of microRNAs (miRNAs) in adipocytes plays a critical role in the pathogenesis of obesity. However, the signaling mechanisms regulating miRNAs production in adipose tissue remain largely unclear. Here, we show that adipose tissue-specific knockout of Ras homolog enriched in brain (Rheb), a direct upstream activator of mTOR, increases miR-182-5p level in mouse subcutaneous white adipose tissues. Interestingly, the inhibition of mTOR signaling by rapamycin has no effect on miR-182-5p level in primary subcutaneous white adipocytes, suggesting the presence of a mTOR-independent mechanism regulating Rheb-mediated miR-182-5p expression. Consistent with this view, Rheb-ablation activates the cAMP/PPARγ signaling pathway. In addition, treatment of white adipocytes with pioglitazone, a PPARγ agonist, dramatically upregulates miR-182-5p levels. Our study reveals a unique mechanism by which Rheb regulates miR-182-5p in adipocytes. Given that increasing miR-182-5p in adipose tissue promotes beige fat development, our study also suggests a unique mechanism by which Rheb promotes thermogenesis and energy expenditure.
Collapse
Affiliation(s)
- Jie Wen
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Jiangming Deng
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ting Xiao
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yu Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| | - Wen Meng
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
8
|
A Single-Cell Atlas of the Atherosclerotic Plaque in the Femoral Artery and the Heterogeneity in Macrophage Subtypes between Carotid and Femoral Atherosclerosis. J Cardiovasc Dev Dis 2022; 9:jcdd9120465. [PMID: 36547462 PMCID: PMC9788114 DOI: 10.3390/jcdd9120465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis of femoral arteries can cause the insufficient blood supply to the lower limbs and lead to gangrenous ulcers and other symptoms. Atherosclerosis and inflammatory factors are significantly different from other plaques. Therefore, it is crucial to observe the cellular composition of the femoral atherosclerotic plaque and identify plaque heterogeneity in other arteries. To this end, we performed single-cell sequencing of a human femoral artery plaque. We identified 14 cell types, including endothelial cells, smooth muscle cells, monocytes, three macrophages with four different subtypes of foam cells, three T cells, natural killer cells, and B cells. We then downloaded single-cell sequencing data of carotid atherosclerosis from GEO, which were compared with the one femoral sample. We identified similar cell types, but the femoral artery had significantly more nonspecific immune cells and fewer specific immune cells than the carotid artery. We further compared the differences in the proportion of inflammatory macrophages, and resident macrophages, and the proportion of inflammatory macrophages was greater within the carotid artery. Through comparing one femoral sequencing sample with carotid samples from public datasets, our study reveals the single-cell map of the femoral artery and the heterogeneity of carotid and femoral arteries at the cellular level, laying the foundation for mechanistic and pharmacological studies of the femoral artery.
Collapse
|
9
|
Margiana R, Alsaikhan F, Al-Awsi GRL, Patra I, Sivaraman R, Fadhil AA, Al-Baghdady HFA, Qasim MT, Hameed NM, Mustafa YF, Hosseini-Fard S. Functions and therapeutic interventions of non-coding RNAs associated with TLR signaling pathway in atherosclerosis. Cell Signal 2022; 100:110471. [PMID: 36122884 DOI: 10.1016/j.cellsig.2022.110471] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022]
Abstract
Nowadays, emerging data demonstrate that the toll-like receptor (TLR) signaling pathway plays an important role in the progression of inflammatory atherosclerosis. Indeed, dysregulated TLR signaling pathway could be a cornerstone of inflammation and atherosclerosis, which contributes to the development of cardiovascular diseases. It is interesting to note that this pathway is heavily controlled by several mechanisms, such as epigenetic factors in which the role of non-coding RNAs (ncRNAs), particularly microRNAs and long noncoding RNAs as well as circular RNAs in the pathogenesis of atherosclerosis has been well studied. Recent years have seen a significant surge in the amount of research exploring the interplay between ncRNAs and TLR signaling pathway downstream targets in the development of atherosclerosis; however, there is still considerable room for improvement in this field. The current study was designed to review underlying mechanisms of TLR signaling pathway and ncRNA interactions to shed light on therapeutic implications in patients with atherosclerosis.
Collapse
Affiliation(s)
- Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Dr. Soetomo General Academic Hospital, Surabaya, Jakarta, Indonesia
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | | | - Indrajit Patra
- An Independent Researcher, PhD from NIT Durgapur, Durgapur, West Bengal, India
| | - Ramaswamy Sivaraman
- Dwaraka Doss Goverdhan Doss Vaishnav College, University of Madras, Arumbakkam, Chennai, India
| | | | | | - Maytham T Qasim
- Department of Anesthesia, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq
| | - Noora M Hameed
- Anesthesia techniques, Al-Nisour University College, Baghdad, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | - Seyedreza Hosseini-Fard
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Santovito D, Weber C. Non-canonical features of microRNAs: paradigms emerging from cardiovascular disease. Nat Rev Cardiol 2022; 19:620-638. [PMID: 35304600 DOI: 10.1038/s41569-022-00680-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 02/08/2023]
Abstract
Research showing that microRNAs (miRNAs) are versatile regulators of gene expression has instigated tremendous interest in cardiovascular research. The overwhelming majority of studies are predicated on the dogmatic notion that miRNAs regulate the expression of specific target mRNAs by inhibiting mRNA translation or promoting mRNA decay in the RNA-induced silencing complex (RISC). These efforts mostly identified and dissected contributions of multiple regulatory networks of miRNA-target mRNAs to cardiovascular pathogenesis. However, evidence from studies in the past decade indicates that miRNAs also operate beyond this canonical paradigm, featuring non-conventional regulatory functions and cellular localizations that have a pathophysiological role in cardiovascular disease. In this Review, we highlight the functional relevance of atypical miRNA biogenesis and localization as well as RISC heterogeneity. Moreover, we delineate remarkable non-canonical examples of miRNA functionality, including direct interactions with proteins beyond the Argonaute family and their role in transcriptional regulation in the nucleus and in mitochondria. We scrutinize the relevance of non-conventional biogenesis and non-canonical functions of miRNAs in cardiovascular homeostasis and pathology, and contextualize how uncovering these non-conventional properties can expand the scope of translational research in the cardiovascular field and beyond.
Collapse
Affiliation(s)
- Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany. .,Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, Milan, Italy.
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany. .,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
11
|
Marinescu MC, Lazar AL, Marta MM, Cozma A, Catana CS. Non-Coding RNAs: Prevention, Diagnosis, and Treatment in Myocardial Ischemia-Reperfusion Injury. Int J Mol Sci 2022; 23:ijms23052728. [PMID: 35269870 PMCID: PMC8911068 DOI: 10.3390/ijms23052728] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 12/17/2022] Open
Abstract
Recent knowledge concerning the role of non-coding RNAs (ncRNAs) in myocardial ischemia/reperfusion (I/R) injury provides new insight into their possible roles as specific biomarkers for early diagnosis, prognosis, and treatment. MicroRNAs (miRNAs) have fewer than 200 nucleotides, while long ncRNAs (lncRNAs) have more than 200 nucleotides. The three types of ncRNAs (miRNAs, lncRNAs, and circRNAs) act as signaling molecules strongly involved in cardiovascular disorders (CVD). I/R injury of the heart is the main CVD correlated with acute myocardial infarction (AMI), cardiac surgery, and transplantation. The expression levels of many ncRNAs and miRNAs are highly modified in the plasma of MI patients, and thus they have the potential to diagnose and treat MI. Cardiomyocyte and endothelial cell death is the major trigger for myocardial ischemia–reperfusion syndrome (MIRS). The cardioprotective effect of inflammasome activation in MIRS and the therapeutics targeting the reparative response could prevent progressive post-infarction heart failure. Moreover, the pharmacological and genetic modulation of these ncRNAs has the therapeutic potential to improve clinical outcomes in AMI patients.
Collapse
Affiliation(s)
- Mihnea-Cosmin Marinescu
- County Clinical Emergency Hospital of Brasov Romania, 500326 Brașov, Romania;
- Department of Vascular Surgery, Second Surgical Clinic, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Andrada-Luciana Lazar
- Department of Dermatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Monica Mihaela Marta
- Department of Medical Education, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Angela Cozma
- Department of Internal Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Correspondence:
| | - Cristina-Sorina Catana
- Department of Medical Biochemistry, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
12
|
Shen M, Jiao K, Wang C, Ehrlich H, Wan M, Hao D, Li J, Wan Q, Tonggu L, Yan J, Wang K, Ma Y, Chen J, Tay FR, Niu L. Extracellular DNA: A Missing Link in the Pathogenesis of Ectopic Mineralization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103693. [PMID: 34939364 PMCID: PMC8844461 DOI: 10.1002/advs.202103693] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/10/2021] [Indexed: 05/12/2023]
Abstract
Although deoxyribonucleic acid (DNA) is the genetic coding for the very essence of life, these macromolecules or components thereof are not necessarily lost after a cell dies. There appears to be a link between extracellular DNA and biomineralization. Here the authors demonstrate that extracellular DNA functions as an initiator of collagen intrafibrillar mineralization. This is confirmed with in vitro and in vivo biological mineralization models. Because of their polyanionic property, extracellular DNA molecules are capable of stabilizing supersaturated calcium phosphate solution and mineralizing 2D and 3D collagen matrices completely as early as 24 h. The effectiveness of extracellular DNA in biomineralization of collagen is attributed to the relatively stable formation of amorphous liquid droplets triggered by attraction of DNA to the collagen fibrils via hydrogen bonding. These findings suggest that extracellular DNA is biomimetically significant for fabricating inorganic-organic hybrid materials for tissue engineering. DNA-induced collagen intrafibrillar mineralization provides a clue to the pathogenesis of ectopic mineralization in different body tissues. The use of DNase for targeting extracellular DNA at destined tissue sites provides a potential solution for treatment of diseases associated with ectopic mineralization.
Collapse
Affiliation(s)
- Min‐juan Shen
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Kai Jiao
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Chen‐yu Wang
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Hermann Ehrlich
- Institute of Electronic and Sensor MaterialsTU Bergakademie FreibergFreiberg09599Germany
- Center for Advanced TechnologyAdam Mickiewicz UniversityPoznan61‐614Poland
| | - Mei‐chen Wan
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Dong‐xiao Hao
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- Department of Applied PhysicsXi'an Jiaotong UniversityXi'anShaanxi710049P. R. China
| | - Jing Li
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Qian‐qian Wan
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Lige Tonggu
- School of MedicineUniversity of WashingtonSeattleWA98195USA
| | - Jian‐fei Yan
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Kai‐yan Wang
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Yu‐xuan Ma
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Ji‐hua Chen
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Franklin R. Tay
- The Dental College of GeorgiaAugusta UniversityAugustaGA30912USA
| | - Li‐na Niu
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| |
Collapse
|
13
|
Therapeutic Role of miR-30a in Lipoteichoic Acid-Induced Endometritis via Targeting the MyD88/Nox2/ROS Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:5042048. [PMID: 35003515 PMCID: PMC8741357 DOI: 10.1155/2021/5042048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/01/2021] [Indexed: 01/03/2023]
Abstract
Staphylococcus aureus (S. aureus), a notorious pathogenic bacterium prevalent in the environment, causes a wide range of inflammatory diseases such as endometritis. Endometritis is an inflammatory disease in humans and mammals, which prolongs uterine involution and causes great economic losses. MiR-30a plays an importan trole in the process of inflammation; however, the regulatory role of miR-30a in endometritis is still unknown. Here, we first noticed that there was an increased level of miR-30a in uterine samples of cows with endometritis. And then, bovine endometrial epithelial (BEND) cells stimulated with the virulence factor lipoteichoic acid (LTA) from S. aureus were used as an in vitro endometritis model to explore the potential role of miR-30a in the pathogenesis of endometritis. Our data showed that the induction of the miR-30a expression is dependent on NF-κB activation, and its overexpression significantly decreased the levels of IL-1β and IL-6. Furthermore, we observed that the overexpression of miR-30a inhibited its translation by binding to 3′−UTR of MyD88 mRNA, thus preventing the activation of Nox2 and NF-κB and ROS accumulation. Meanwhile, in vivo studies further revealed that upregulation of miR-30a using chemically synthesized agomirs alleviates the inflammatory conditions in an experimental mouse model of endometritis, as indicated by inhibition of ROS and NF-κB. Taken together, these findings highlight that miR-30a can attenuate LTA-elicited oxidative stress and inflammatory responses through the MyD88/Nox2/ROS/NF-κB pathway and may aid the future development of novel therapies for inflammatory diseases caused by S. aureus, including endometritis.
Collapse
|
14
|
Abstract
Regulatory RNAs like microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) control vascular and immune cells' phenotype and thus play a crucial role in atherosclerosis. Moreover, the mutual interactions between miRNAs and lncRNAs link both types of regulatory RNAs in a functional network that affects lesion formation. In this review, we deduce novel concepts of atherosclerosis from the analysis of the current data on regulatory RNAs' role in endothelial cells (ECs) and macrophages. In contrast to arterial ECs, which adopt a stable phenotype by adaptation to high shear stress, macrophages are highly plastic and quickly change their activation status. At predilection sites of atherosclerosis, such as arterial bifurcations, ECs are exposed to disturbed laminar flow, which generates a dysadaptive stress response mediated by miRNAs. Whereas the highly abundant miR-126-5p promotes regenerative proliferation of dysadapted ECs, miR-103-3p stimulates inflammatory activation and impairs endothelial regeneration by aberrant proliferation and micronuclei formation. In macrophages, miRNAs are essential in regulating energy and lipid metabolism, which affects inflammatory activation and foam cell formation.Moreover, lipopolysaccharide-induced miR-155 and miR-146 shape inflammatory macrophage activation through their oppositional effects on NF-kB. Most lncRNAs are not conserved between species, except a small group of very long lncRNAs, such as MALAT1, which blocks numerous miRNAs by providing non-functional binding sites. In summary, regulatory RNAs' roles are highly context-dependent, and therapeutic approaches that target specific functional interactions of miRNAs appear promising against cardiovascular diseases.
Collapse
Affiliation(s)
- Andreas Schober
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany.
| | - Saffiyeh Saboor Maleki
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Maliheh Nazari-Jahantigh
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
15
|
Tang C, Wang L, Sheng Y, Zheng Z, Xie Z, Wu F, You T, Ren L, Xia L, Ruan C, Zhu L. CLEC-2-dependent platelet subendothelial accumulation by flow disturbance contributes to atherogenesis in mice. Theranostics 2021; 11:9791-9804. [PMID: 34815786 PMCID: PMC8581433 DOI: 10.7150/thno.64601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/22/2021] [Indexed: 12/19/2022] Open
Abstract
Rationale: Platelets play an essential role in atherosclerosis, but the underlying mechanisms remain to be addressed. This study is to investigate the role of platelets in d-flow induced vascular inflammation and the underlying mechanism. Methods: We established a disturbed blood flow (d-flow) model by partial carotid ligation (PCL) surgery using atherosclerosis-susceptible mice and wild-type mice to observe the d-flow induced platelet accumulation in the subendothelium or in the plaque by immunostaining or transmission electron microscopy. The mechanism of platelet subendothelial accumulation was further explored by specific gene knockout mice. Results: We observed presence of platelets in atherosclerotic plaques either in the atheroprone area of aortic arch or in carotid artery with d-flow using Ldlr-/- or ApoE-/- mice on high fat diet. Immunostaining showed the subendothelial accumulation of circulating platelets by d-flow in vivo. Transmission electron microscopy demonstrated the accumulation of platelets associated with monocytes in the subendothelial spaces. The subendothelial accumulation of platelet-monocyte/macrophage aggregates reached peak values at 2 days after PCL. In examining the molecules that may mediate the platelet entry, we found that deletion of platelet C-type lectin-like receptor 2 (CLEC-2) reduced the subendothelial accumulation of platelets and monocytes/macrophages by d-flow, and ameliorated plaque formation in Ldlr-/- mice on high fat diet. Supportively, CLEC-2 deficient platelets diminished their promoting effect on the migration of mouse monocyte/macrophage cell line RAW264.7. Moreover, monocyte podoplanin (PDPN), the only ligand of CLEC-2, was upregulated by d-flow, and the myeloid-specific PDPN deletion mitigated the subendothelial accumulation of platelets and monocytes/macrophages. Conclusions: Our results reveal a new CLEC-2-dependent platelet subendothelial accumulation in response to d-flow to regulate vascular inflammation.
Collapse
Affiliation(s)
- Chaojun Tang
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, China
- Suzhou Key Lab for Thrombosis and Vascular Biology, Soochow University, Suzhou, China
| | - Lei Wang
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Yulan Sheng
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Zhong Zheng
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Zhanli Xie
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Fan Wu
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Tao You
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Lijie Ren
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Changgeng Ruan
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, China
| | - Li Zhu
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, China
- Suzhou Key Lab for Thrombosis and Vascular Biology, Soochow University, Suzhou, China
| |
Collapse
|
16
|
Kianmehr A, Qujeq D, Bagheri A, Mahrooz A. Oxidized LDL-regulated microRNAs for evaluating vascular endothelial function: molecular mechanisms and potential biomarker roles in atherosclerosis. Crit Rev Clin Lab Sci 2021; 59:40-53. [PMID: 34523391 DOI: 10.1080/10408363.2021.1974334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
As a simple monolayer, vascular endothelial cells can respond to physicochemical stimuli. In addition to promoting the formation of foam cells, oxidized low-density lipoprotein (ox-LDL) contributes to the atherosclerotic process through different mechanisms, including endothelial cell dysfunction. As conserved noncoding RNAs, microRNAs (miRNAs) naturally lie in different genomic positions and post-transcriptionally regulate the expression of many genes. They participate in integrated networks formed under stress to maintain cellular homeostasis, vascular inflammation, and metabolism. These small RNAs constitute therapeutic targets in different diseases, including atherosclerosis, and their role as biomarkers is crucial given their detectability even years before the emergence of diseases. This review was performed to investigate the role of ox-LDL-regulated miRNAs in atherosclerosis, their molecular mechanisms, and their application as biomarkers of vascular endothelial cell dysfunction.
Collapse
Affiliation(s)
- Anvarsadat Kianmehr
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abouzar Bagheri
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdolkarim Mahrooz
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
17
|
Márquez AB, van der Vorst EPC, Maas SL. Key Chemokine Pathways in Atherosclerosis and Their Therapeutic Potential. J Clin Med 2021; 10:3825. [PMID: 34501271 PMCID: PMC8432216 DOI: 10.3390/jcm10173825] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/20/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022] Open
Abstract
The search to improve therapies to prevent or treat cardiovascular diseases (CVDs) rages on, as CVDs remain a leading cause of death worldwide. Here, the main cause of CVDs, atherosclerosis, and its prevention, take center stage. Chemokines and their receptors have long been known to play an important role in the pathophysiological development of atherosclerosis. Their role extends from the initiation to the progression, and even the potential regression of atherosclerotic lesions. These important regulators in atherosclerosis are therefore an obvious target in the development of therapeutic strategies. A plethora of preclinical studies have assessed various possibilities for targeting chemokine signaling via various approaches, including competitive ligands and microRNAs, which have shown promising results in ameliorating atherosclerosis. Developments in the field also include detailed imaging with tracers that target specific chemokine receptors. Lastly, clinical trials revealed the potential of various therapies but still require further investigation before commencing clinical use. Although there is still a lot to be learned and investigated, it is clear that chemokines and their receptors present attractive yet extremely complex therapeutic targets. Therefore, this review will serve to provide a general overview of the connection between various chemokines and their receptors with atherosclerosis. The different developments, including mouse models and clinical trials that tackle this complex interplay will also be explored.
Collapse
Affiliation(s)
- Andrea Bonnin Márquez
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
18
|
Santovito D, Egea V, Bidzhekov K, Natarelli L, Mourão A, Blanchet X, Wichapong K, Aslani M, Brunßen C, Horckmans M, Hristov M, Geerlof A, Lutgens E, Daemen MJAP, Hackeng T, Ries C, Chavakis T, Morawietz H, Naumann R, von Hundelshausen P, Steffens S, Duchêne J, Megens RTA, Sattler M, Weber C. Noncanonical inhibition of caspase-3 by a nuclear microRNA confers endothelial protection by autophagy in atherosclerosis. Sci Transl Med 2021; 12:12/546/eaaz2294. [PMID: 32493793 DOI: 10.1126/scitranslmed.aaz2294] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 04/02/2020] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) are versatile regulators of gene expression with profound implications for human disease including atherosclerosis, but whether they can exert posttranslational functions to control cell adaptation and whether such noncanonical features harbor pathophysiological relevance is unknown. Here, we show that miR-126-5p sustains endothelial integrity in the context of high shear stress and autophagy. Bound to argonaute-2 (Ago2), miR-126-5p forms a complex with Mex3a, which occurs on the surface of autophagic vesicles and guides its transport into the nucleus. Mutational studies and biophysical measurements demonstrate that Mex3a binds to the central U- and G-rich regions of miR-126-5p with nanomolar affinity via its two K homology domains. In the nucleus, miR-126-5p dissociates from Ago2 and binds to caspase-3 in an aptamer-like fashion with its seed sequence, preventing dimerization of the caspase and inhibiting its activity to limit apoptosis. The antiapoptotic effect of miR-126-5p outside of the RNA-induced silencing complex is important for endothelial integrity under conditions of high shear stress promoting autophagy: ablation of Mex3a or ATG5 in vivo attenuates nuclear import of miR-126-5p, aggravates endothelial apoptosis, and exacerbates atherosclerosis. In human plaques, we found reduced nuclear miR-126-5p and active caspase-3 in areas of disturbed flow. The direct inhibition of caspase-3 by nuclear miR-126-5p reveals a noncanonical mechanism by which miRNAs can modulate protein function.
Collapse
Affiliation(s)
- Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, D-80336 Munich, Germany
| | - Virginia Egea
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany
| | - Kiril Bidzhekov
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany
| | - Lucia Natarelli
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, D-80336 Munich, Germany
| | - André Mourão
- Institute of Structural Biolology, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
| | - Xavier Blanchet
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany
| | - Kanin Wichapong
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229HX Maastricht, Netherlands
| | - Maria Aslani
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, D-80336 Munich, Germany
| | - Coy Brunßen
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine, TU Dresden, D-01307 Dresden, Germany
| | - Michael Horckmans
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany.,Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles (ULB), B-1070 Brussels, Belgium
| | - Michael Hristov
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany
| | - Arie Geerlof
- Institute of Structural Biolology, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
| | - Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, D-80336 Munich, Germany.,Department of Medical Biochemistry and Pathology, Amsterdam University Medical Centers, Amsterdam School of Cardiovascular Sciences (ACS), 1081HZ Amsterdam, Netherlands
| | - Mat J A P Daemen
- Department of Medical Biochemistry and Pathology, Amsterdam University Medical Centers, Amsterdam School of Cardiovascular Sciences (ACS), 1081HZ Amsterdam, Netherlands
| | - Tilman Hackeng
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229HX Maastricht, Netherlands
| | - Christian Ries
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, D-01307 Dresden, Germany
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine, TU Dresden, D-01307 Dresden, Germany
| | - Ronald Naumann
- Max-Planck-Institute of Molecular Cell Biology and Genetics, D-01307 Dresden, Germany
| | - Philipp von Hundelshausen
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, D-80336 Munich, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, D-80336 Munich, Germany
| | - Johan Duchêne
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany
| | - Remco T A Megens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229HX Maastricht, Netherlands
| | - Michael Sattler
- Institute of Structural Biolology, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Center for Integrated Protein Science Munich at Biomolecular NMR Spectroscopy, Department of Chemistry, Technical University of Munich, D-85747 Garching, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, D-80336 Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229HX Maastricht, Netherlands.,Munich Cluster for Systems Neurology (SyNergy), D-81377 Munich, Germany
| |
Collapse
|
19
|
Patients with Coronary Artery Disease Have Lower Levels of Antibody to Heat-Stressed Fibroblast Derived Proteins, versus Normal Subjects. Cardiovasc Ther 2021; 2021:5577218. [PMID: 34239605 PMCID: PMC8225444 DOI: 10.1155/2021/5577218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/03/2021] [Accepted: 06/05/2021] [Indexed: 01/06/2023] Open
Abstract
Cellular stress response plays an important role in the pathophysiology of coronary artery disease (CAD). Inhibition of cellular stress may provide a novel clinical approach regarding the diagnosis and treatment of CAD. Fibroblasts constitute 60-70% of cardiac cells and have a crucial role in cardiovascular function. Hence, the aim of this study was to show a potential therapeutic application of proteins derived from heat-stressed fibroblast in CAD patients. Fibroblasts were isolated from the foreskin and cultured under heat stress conditions. Surprisingly, 1.06% of the cells exhibited a necrotic death pattern. Furthermore, heat-stressed fibroblasts produced higher level of total proteins than control cells. In SDS-PAGE analysis, a 70 kDa protein band was observed in stressed cell culture supernatants which appeared as two acidic spots with close pI in the two-dimensional electrophoresis. To evaluate the immunogenic properties of fibroblast-derived heat shock proteins (HSPs), the serum immunoglobulin-G (IgG) was measured by ELISA in 50 CAD patients and 50 normal subjects who had been diagnosed through angiography. Interestingly, the level of anti-HSP antibody was significantly higher in non-CAD individuals in comparison with the patient's group (p < 0.05). The odds ratio for CAD was 5.06 (95%CI = 2.15‐11.91) in cut-off value of 30 AU/mL of anti-HSP antibody. Moreover, ROC analysis showed that anti-HSP antibodies had a specificity of 74% and a sensitivity of 64%, which is almost equal to 66% sensitivity of exercise stress test (EST) as a CAD diagnostic method. These data revealed that fibroblast-derived HSPs are suitable for the diagnosis and management of CAD through antibody production.
Collapse
|
20
|
Victorino J, Alvarez-Franco A, Manzanares M. Functional genomics and epigenomics of atrial fibrillation. J Mol Cell Cardiol 2021; 157:45-55. [PMID: 33887329 DOI: 10.1016/j.yjmcc.2021.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
Atrial fibrillation is a progressive cardiac arrhythmia that increases the risk of hospitalization and adverse cardiovascular events. Despite years of study, we still do not have a full comprehension of the molecular mechanism responsible for the disease. The recent implementation of large-scale approaches in both patient samples, population studies and animal models has helped us to broaden our knowledge on the molecular drivers responsible for AF and on the mechanisms behind disease progression. Understanding genomic and epigenomic changes that take place during chronification of AF will prove essential to design novel treatments leading to improved patient care.
Collapse
Affiliation(s)
- Jesus Victorino
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Spain
| | - Alba Alvarez-Franco
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Miguel Manzanares
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.
| |
Collapse
|
21
|
Qiu Y, Xu J, Yang L, Zhao G, Ding J, Chen Q, Zhang N, Yang R, Wang J, Li S, Zhang L. MiR-375 silencing attenuates pro-inflammatory macrophage response and foam cell formation by targeting KLF4. Exp Cell Res 2021; 400:112507. [PMID: 33545131 DOI: 10.1016/j.yexcr.2021.112507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/18/2021] [Accepted: 01/26/2021] [Indexed: 12/19/2022]
Abstract
Macrophage mediated inflammation and foam cell formation play crucial roles in the development of atherosclerosis. MiR-375 is a small noncoding RNA that significantly implicated in multiple tumor regulation and has been emerged as a novel biomarker for type 2 diabetes. However, the exact role of miR-375 on macrophage activation remains unknown. In the present study, we observed that miR-375 expression showed an up-regulated expression in atherosclerotic aortas, as well as in bone marrow derived macrophages (BMDMs) and mouse peritoneal macrophages (MPMs) isolated from ApoE deficiency mice and was gradually increased followed the Ox-LDL treated time. Functionally, miR-375 inhibition significantly decreased foam cell formation accompanied by up-regulated genes expression involved in cholesterol efflux but reduced genes expression implicated in cholesterol influx. Moreover, miR-375 silencing increased resolving M2 macrophage but reduced pro-inflammatory M1 macrophage markers expression. Such above effects can be reversed by miR-375 overexpression. Mechanistically, we noticed that miR-375 knockdown promoted KLF4 expression which was required for the ameliorated effect of miR-375 silencing on macrophage activation. Importantly, the consistent results in mRNA expression of M1 and M2 markers were observed in vivo, and miR-375-/-ApoE-/- mice significant decreased atherosclerotic lesions in the whole aorta and aortic sinus. Taken together, these evidences suggested that miR-375 knockdown attenuated macrophage activation partially through activation of KLF4-dependent mechanism.
Collapse
Affiliation(s)
- Yanyan Qiu
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Jinyi Xu
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China.
| | - Lihong Yang
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Guihua Zhao
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Jing Ding
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Qiong Chen
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Na Zhang
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Ruike Yang
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Jijing Wang
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Shuaibing Li
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Luming Zhang
- Department of Cardio-Pulmonary Function, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| |
Collapse
|
22
|
Gong FH, Long L, Yang YS, Shen DH, Zhang YS, Wang XS, Zhang XP, Xiao XQ. Attenuated macrophage activation mediated by microRNA-183 knockdown through targeting NR4A2. Exp Ther Med 2021; 21:300. [PMID: 33717243 DOI: 10.3892/etm.2021.9731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is considered a chronic inflammatory disease, and macrophages function as important mediators in the development of atherogenesis. MicroRNA (miR)-183 is a small non-coding RNA that acts as a novel tumor suppressor and has recently been proposed to affect cardiac hypertrophy. However, the exact role and underlying mechanism of miR-183 in macrophage activation remain unknown. In the present study, miR-183 showed upregulated expression in atheromatous plaques and in bone marrow-derived macrophages (BMDMs) subjected to stimulation with oxidized low-density lipoproteins. Using a miR-183 loss-of-function strategy, it was demonstrated that miR-183 knockdown significantly increased resolving M2 macrophage marker expression but decreased proinflammatory M1 macrophage marker expression, as well as attenuated NF-κB activation. Moreover, decreased foam-cell formation accompanied by upregulation of genes involved in cholesterol efflux and downregulation of genes implicated in cholesterol influx was found in BMDMs transfected with a miR-183 inhibitor. Mechanistically, macrophage activation mediated by miR-183 silencing was partially attributed to direct upregulation of NR4A2 expression in BMDMs. Thus, the present study suggests that neutralizing miR-183 may be a potential therapeutic strategy for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Fu-Han Gong
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren, Guizhou 554300, P.R. China
| | - Li Long
- Department of Clinical Laboratory, Tongren Municipal People's Hospital, Tongren, Guizhou 554300, P.R. China
| | - Yong-Sheng Yang
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren, Guizhou 554300, P.R. China
| | - De-Hong Shen
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren, Guizhou 554300, P.R. China
| | - Yu-Song Zhang
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren, Guizhou 554300, P.R. China
| | - Xue-Sheng Wang
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren, Guizhou 554300, P.R. China
| | - Xue-Ping Zhang
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren, Guizhou 554300, P.R. China
| | - Xiao-Qiang Xiao
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren, Guizhou 554300, P.R. China
| |
Collapse
|
23
|
Golbidi S, Edvinsson L, Laher I. Smoking and Endothelial Dysfunction. Curr Vasc Pharmacol 2020; 18:1-11. [PMID: 30210003 DOI: 10.2174/1573403x14666180913120015] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/12/2018] [Accepted: 09/12/2018] [Indexed: 02/07/2023]
Abstract
Cigarette smoking is one of the most important health concerns worldwide. Even though the rate of smoking is declining in developed countries, it is still experiencing growth in developing regions. Many studies have examined the relationship between smoking, as an established risk factor, and cardiovascular diseases. We provide an updated review of the underlying mechanisms of smokinginduced cardiovascular diseases, with a focus on the relationship between smoking and oxidative stress, particularly from the perspective of endothelial cell dysfunction. We review smoking-induced oxidative stress as a trigger for a generalized vascular inflammation associated with cytokine release, adhesion of inflammatory cells and, ultimately, disruption of endothelial integrity as a protective barrier layer. We also briefly discuss the harms related to the vaping of electronic cigarettes, which many erroneously consider as a safe alternative to smoking. We conclude that even though e-cigarette could be a helpful device during the transition period of cigarette quitting, it is by no means a safe substitute.
Collapse
Affiliation(s)
- Saeid Golbidi
- Department of Family Practice, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lars Edvinsson
- Department of Medicine, Institute of Clinical Sciences, Lund University, Getingevägen, 22185 Lund, Sweden
| | - Ismail Laher
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Hu B, Boakye‐Yiadom KO, Yu W, Yuan Z, Ho W, Xu X, Zhang X. Nanomedicine Approaches for Advanced Diagnosis and Treatment of Atherosclerosis and Related Ischemic Diseases. Adv Healthc Mater 2020; 9:e2000336. [PMID: 32597562 DOI: 10.1002/adhm.202000336] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/30/2020] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases (CVDs) remain one of the major causes of mortality worldwide. In response to this and other worldwide health epidemics, nanomedicine has emerged as a rapidly evolving discipline that involves the development of innovative nanomaterials and nanotechnologies and their applications in therapy and diagnosis. Nanomedicine presents unique advantages over conventional medicines due to the superior properties intrinsic to nanoscopic therapies. Once used mainly for cancer therapies, recently, tremendous progress has been made in nanomedicine that has led to an overall improvement in the treatment and diagnosis of CVDs. This review elucidates the pathophysiology and potential targets of atherosclerosis and associated ischemic diseases. It may be fruitful to pursue future work in the nanomedicine-mediated treatment of CVDs based on these targets. A comprehensive overview is then provided featuring the latest preclinical and clinical outcomes in cardiovascular imaging, biomarker detection, tissue engineering, and nanoscale delivery, with specific emphasis on nanoparticles, nanostructured scaffolds, and nanosensors. Finally, the challenges and opportunities regarding the future development and clinical translation of nanomedicine in related fields are discussed. Overall, this review aims to provide a deep and thorough understanding of the design, application, and future development of nanomedicine for atherosclerosis and related ischemic diseases.
Collapse
Affiliation(s)
- Bin Hu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Kofi Oti Boakye‐Yiadom
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Wei Yu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Zi‐Wei Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - William Ho
- Department of Chemical and Materials EngineeringNew Jersey Institute of Technology Newark NJ 07102 USA
| | - Xiaoyang Xu
- Department of Chemical and Materials EngineeringNew Jersey Institute of Technology Newark NJ 07102 USA
| | - Xue‐Qing Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| |
Collapse
|
25
|
An Overview of Non-coding RNAs and Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:3-45. [PMID: 32285403 DOI: 10.1007/978-981-15-1671-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease management and timely diagnosis remain a major dilemma. Delineating molecular mechanisms of cardiovascular diseases is opening horizon in the field of molecular medicines and in the development of early diagnostic markers. Non-coding RNAs are the highly functional and vibrant nucleic acids and are known to be involved in the regulation of endothelial cells, vascular and smooth muscles cells, cardiac metabolism, ischemia, inflammation and many processes in cardiovascular system. This chapter is comprehensively focusing on the overview of the non-coding RNAs including their discovery, generation, classification and functional regulation. In addition, overview regarding different non-coding RNAs as long non-coding, siRNAs and miRNAs involvement in the cardiovascular diseases is also addressed. Detailed functional analysis of this vast group of highly regulatory molecules will be promising for shaping future drug discoveries.
Collapse
|
26
|
Bao Y, Li S, Ding Y, Du X, Zhang M, Tang W, Zhou S. MiRNA: a potential target for gene diagnosis and treatment of atherosclerotic stroke. Int J Neurosci 2020; 131:283-288. [PMID: 32129115 DOI: 10.1080/00207454.2020.1738428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Stroke is one of the major diseases that endanger the physical health life of middle-aged and old people. It has the characteristics of high incidence, mortality and disability rate. Atherosclerosis is the main intervention target for stroke prevention and treatment. MiRNAs are highly expressed in the cerebral vasculature and play an important regulatory role in the pathogenesis of neuronal damage and ischemic stroke. This article reviews the mechanism of action between miRNAs and atherosclerosis, stroke, ischemia-reperfusion injury, collateral circulation and circRNA molecular networks, providing theoretical support for miRNA in gene diagnosis and drug therapy of atherosclerotic stroke.
Collapse
Affiliation(s)
- Yi Bao
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China.,Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Sijing Li
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Yayong Ding
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Xinyu Du
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Miao Zhang
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Wanjuan Tang
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Siqin Zhou
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China
| |
Collapse
|
27
|
Zhang X, Wang Z, Li W, Huang R, Zheng D, Bi G. MicroRNA-217-5p ameliorates endothelial cell apoptosis induced by ox-LDL by targeting CLIC4. Nutr Metab Cardiovasc Dis 2020; 30:523-533. [PMID: 31744714 DOI: 10.1016/j.numecd.2019.09.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND AIMS Endothelial cell apoptosis plays an essential role in the pathogenesis of atherosclerosis. MicroRNAs and chloride intracellular channels (CLICs) have been verified to participate in the endothelial cell apoptosis process, however, the underlying molecular mechanisms are still unclear. The main aim of this study was to investigate the biological effects of microRNA-217-5p (miR-217-5p) and CLIC4 on endothelial cell apoptosis in atherosclerosis. METHODS AND RESULTS An atherosclerotic mouse model (n = 18) was constructed by feeding apolipo protein E knockout ApoE(-/-) mice with high-fat diet for 12 weeks. An atherosclerotic cell model was established by treating human aortic endothelial cells with oxidized low-density lipoprotein (ox-LDL; 50 μg/mL) for 24 h. Quantitative real-time polymerase chain reaction and immunofluorescent staining confirmed the downregulation of miR-217-5p and upregulation of CLIC4 in atherosclerotic endothelial cells. Combined with western blot, flow cytometry assay and Hoechst staining, we demonstrated that miR-217-5p upregulation or CLIC4 knockdown regulated the apoptosis-related genes, ameliorated mitochondrial membrane permeability and therefore inhibited the apoptosis of aortic endothelial cells induced by ox-LDL. We further confirmed that miR-217-5p inhibited apoptosis of endothelial cells through targeting CLIC4 using luciferase report assay and rescue experiments. CONCLUSION We revealed for the first time that miR-217-5p inhibited apoptosis of endothelial cells in atherosclerosis and identified CLIC4 as a novel target of miR-217-5p. Our work provides a potential therapeutic approach for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China
| | - Zhimin Wang
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China
| | - Weishuai Li
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China
| | - Rui Huang
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China
| | - Dongming Zheng
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China
| | - Guorong Bi
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China.
| |
Collapse
|
28
|
Xing X, Li Z, Yang X, Li M, Liu C, Pang Y, Zhang L, Li X, Liu G, Xiao Y. Adipose-derived mesenchymal stem cells-derived exosome-mediated microRNA-342-5p protects endothelial cells against atherosclerosis. Aging (Albany NY) 2020; 12:3880-3898. [PMID: 32096479 PMCID: PMC7066923 DOI: 10.18632/aging.102857] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 02/04/2020] [Indexed: 12/16/2022]
Abstract
Exosomes are reported to mediate several disease-related microRNAs (miRNAs) to affect the progression of diseases, including atherosclerosis. Here, we aimed to screen the atherosclerosis-associated miRNAs and preliminarily investigate the potential regulatory mechanism of atherosclerosis. First, the lesion model for human umbilical vein endothelial cells (HUVECs) was favorably constructed. Later, through RNA-sequencing and bioinformatics analyses, miR-342-5p was identified in lesion model for HUVECs. MiR-342-5p overexpression or knockdown evidently promoted or inhibited the apoptosis of HUVECs impaired by H2O2. Mechanistically, PPP1R12B was found to have great potential as a target of miR-342-5p in HUVECs impaired by H2O2, supported by RNA-sequencing and a series of bioinformatics analyses. Meanwhile, the effect of miR-342-5p on PPP1R12B expression in HUVECs’ lesion model was explored, revealing that miR-342-5p had an inhibitory role in PPP1R12B expression. Additionally, adipose-derived mesenchymal stem cells (ADSCs) in spindle-like shape and their derived exosomes with 30 to 150 nm diameter were characterized. Furthermore, results showed miR-342-5p was evidently decreased in the presence of ADSCs-derived exosomes. These findings indicated ADSCs-derived exosomes restrained the expression of miR-324-5p in lesion model. Collectively, this work demonstrates an atherosclerosis-associated miR-342-5p and reveals a preliminary possible mechanism in which miR-342-5p mediated by ADSCs-derived exosomes protects endothelial cells against atherosclerosis.
Collapse
Affiliation(s)
- Xiaohui Xing
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250000, Shandong Province, P.R. China.,Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Zhongchen Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China.,Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, Shandong Province, P.R. China
| | - Xin Yang
- Department of Otolaryngology, General Hospital of Central Theater Command of PLA, Wuhan 430070, Hubei, China
| | - Mengyou Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Chao Liu
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Yuejiu Pang
- Department of Senile Neurology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, Shandong Province, P.R. China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Xueyuan Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Guangcun Liu
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250000, Shandong Province, P.R. China
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| |
Collapse
|
29
|
Gusev EY, Zotova NV. Cellular Stress and General Pathological Processes. Curr Pharm Des 2020; 25:251-297. [PMID: 31198111 DOI: 10.2174/1381612825666190319114641] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/13/2019] [Indexed: 02/06/2023]
Abstract
From the viewpoint of the general pathology, most of the human diseases are associated with a limited number of pathogenic processes such as inflammation, tumor growth, thrombosis, necrosis, fibrosis, atrophy, pathological hypertrophy, dysplasia and metaplasia. The phenomenon of chronic low-grade inflammation could be attributed to non-classical forms of inflammation, which include many neurodegenerative processes, pathological variants of insulin resistance, atherosclerosis, and other manifestations of the endothelial dysfunction. Individual and universal manifestations of cellular stress could be considered as a basic element of all these pathologies, which has both physiological and pathophysiological significance. The review examines the causes, main phenomena, developmental directions and outcomes of cellular stress using a phylogenetically conservative set of genes and their activation pathways, as well as tissue stress and its role in inflammatory and para-inflammatory processes. The main ways towards the realization of cellular stress and its functional blocks were outlined. The main stages of tissue stress and the classification of its typical manifestations, as well as its participation in the development of the classical and non-classical variants of the inflammatory process, were also described. The mechanisms of cellular and tissue stress are structured into the complex systems, which include networks that enable the exchange of information with multidirectional signaling pathways which together make these systems internally contradictory, and the result of their effects is often unpredictable. However, the possible solutions require new theoretical and methodological approaches, one of which includes the transition to integral criteria, which plausibly reflect the holistic image of these processes.
Collapse
Affiliation(s)
- Eugeny Yu Gusev
- Laboratory of the Immunology of Inflammation, Institute of Immunology and Physiology, Yekaterinburg, Russian Federation
| | - Natalia V Zotova
- Laboratory of the Immunology of Inflammation, Institute of Immunology and Physiology, Yekaterinburg, Russian Federation.,Department of Medical Biochemistry and Biophysics, Ural Federal University named after B.N.Yeltsin, Yekaterinburg, Russian Federation
| |
Collapse
|
30
|
Rashad S, Han X, Saqr K, Tupin S, Ohta M, Niizuma K, Tominaga T. Epigenetic response of endothelial cells to different wall shear stress magnitudes: A report of new mechano-miRNAs. J Cell Physiol 2020; 235:7827-7839. [PMID: 31912899 DOI: 10.1002/jcp.29436] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022]
Abstract
Endothelial cells (ECs) respond to flow stress via a variety of mechanisms, leading to various intracellular responses that can modulate the vessel wall and lead to diseases if the flow is disturbed. Mechano-microRNAs (miRNAs) are a subset of miRNAs in the ECs that are flow responsive. Mechano-miRNAs were shown to be related to atherosclerosis pathophysiology, and a number of them were identified as pathologic. Here, we exposed human carotid ECs to different wall shear stresses (WSS), high and low, and evaluated the response of miRNAs by microarray and quantitative polymerase chain reaction analysis. We discovered five new mechano-miRNAs that were not reported in that context previously to the best of our knowledge. Moreover, functional pathway analysis revealed that under low WSS conditions, several pathways regulating apoptosis are affected. In addition, KLF2 and KLF4, known atheroprotective genes, were downregulated under low WSS and upregulated under high WSS. KLF2 and VCAM1, both angiogenic, were upregulated under high WSS. NOS3, which is vascular protective, was also upregulated with higher WSS. On the contrary, ICAM-1 and E-selectin, both atherogenic and proinflammatory, were upregulated with high WSS. Collectively, the epigenetic landscape with the gene expression analysis reveals that low WSS is associated with a proapoptotic state, while high WSS is associated with a proliferative and proinflammatory state.
Collapse
Affiliation(s)
- Sherif Rashad
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Xiaobo Han
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Khalid Saqr
- Biomedical Flow Dynamics Laboratory, Institute of Fluid Science, Tohoku University, Sendai, Japan
| | - Simon Tupin
- Biomedical Flow Dynamics Laboratory, Institute of Fluid Science, Tohoku University, Sendai, Japan
| | - Makoto Ohta
- Biomedical Flow Dynamics Laboratory, Institute of Fluid Science, Tohoku University, Sendai, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
31
|
Li Q, Xi J, Li B, Li N. MiR‐16, as a potential NF‐κB‐related miRNA, exerts anti‐inflammatory effects on LPS‐induced myocarditis via mediating CD40 expression: A preliminary study. J Biochem Mol Toxicol 2019; 34:e22426. [PMID: 31777165 DOI: 10.1002/jbt.22426] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 09/10/2019] [Accepted: 11/14/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Qiang‐Qiang Li
- Department of Cardiology of Integrated Traditional Chinese and Western MedicineAnqiu People's Hospital Weifang Shandong China
| | - Jing Xi
- Department of CardiologyAnqiu People's Hospital Weifang Shandong China
| | - Bing‐Qiang Li
- Department of CardiologyAnqiu People's Hospital Weifang Shandong China
| | - Ning Li
- Department of CardiologyAnqiu People's Hospital Weifang Shandong China
| |
Collapse
|
32
|
Li N, Liu SF, Dong K, Zhang GC, Huang J, Wang ZH, Wang TJ. Exosome-Transmitted miR-25 Induced by H. pylori Promotes Vascular Endothelial Cell Injury by Targeting KLF2. Front Cell Infect Microbiol 2019; 9:366. [PMID: 31750260 PMCID: PMC6842922 DOI: 10.3389/fcimb.2019.00366] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023] Open
Abstract
Background: Increasing evidence has shown that Helicobacter pylori is associated with coronary heart disease (CHD); however, the underlying mechanism remains unclear. Methods: The expression of miR-25 and mRNAs was measured using qRT-PCR. Protein levels were detected using western blotting and exosomes were assessed with an electron microscope. The target gene of miR-25 was identified using the luciferase report system. Results: H. pylori infection increased the expression of miR-25 in gastric epithelial cells and was associated with increased levels of exosome-transmitted miR-25 in human peripheral blood. Mechanistic investigation showed the Kruppel-like factor 2 (KLF2) was a direct target of exosome-transmitted miR-25 in vascular endothelial cells. In addition, the miR-25/KLF2 axis regulated the NF-κB signaling pathway, resulting in increased expression of interleukin 6 (IL6), monocyte chemoattractant protein-1 (MCP-1), vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1). Conclusion: Our findings suggest that the miR-25/KLF2 axis may be a potential therapeutic target for H. pylori-associated CHD. Furthermore, high levels of exosome-transmitted miR-25 in peripheral blood may pose a potential risk for CHD.
Collapse
Affiliation(s)
- Na Li
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Shi-Feng Liu
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Kai Dong
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Gui-Chun Zhang
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Jing Huang
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Zhi-Heng Wang
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Tong-Jian Wang
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| |
Collapse
|
33
|
Xu F, Liu Y, Zhu X, Li S, Shi X, Li Z, Ai M, Sun J, Hou B, Cai W, Sun H, Ni L, Zhou Y, Qiu L. Protective Effects and Mechanisms of Vaccarin on Vascular Endothelial Dysfunction in Diabetic Angiopathy. Int J Mol Sci 2019; 20:ijms20184587. [PMID: 31533227 PMCID: PMC6769517 DOI: 10.3390/ijms20184587] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/29/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular complications are a major leading cause of mortality in patients suffering from type 2 diabetes mellitus (T2DM). Vascular endothelial dysfunction is a core pathophysiological event in the early stage of T2DM and eventually leads to cardiovascular disease. Vaccarin (VAC), an active flavonoid glycoside extracted from vaccariae semen, exhibits extensive biological activities including vascular endothelial cell protection effects. However, little is known about whether VAC is involved in endothelial dysfunction regulation under high glucose (HG) or hyperglycemia conditions. Here, in an in vivo study, we found that VAC attenuated increased blood glucose, increased glucose and insulin tolerance, relieved the disorder of lipid metabolism and oxidative stress, and improved endothelium-dependent vasorelaxation in STZ/HFD-induced T2DM mice. Furthermore, in cultured human microvascular endothelial cell-1 (HMEC-1) cells, we showed that pretreatment with VAC dose-dependently increased nitric oxide (NO) generation and the phosphorylation of eNOS under HG conditions. Mechanistically, VAC-treated HMEC-1 cells exhibited higher AMPK phosphorylation, which was attenuated by HG stimulation. Moreover, HG-triggered miRNA-34a upregulation was inhibited by VAC pretreatment, which is in accordance with pretreatment with AMPK inhibitor compound C (CC). In addition, both reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC) and VAC abolished HG-evoked dephosphorylation of AMPK and eNOS, increased miRNA-34a expression, and decreased NO production. These results suggest that VAC impedes HG-induced endothelial dysfunction via inhibition of the ROS/AMPK/miRNA-34a/eNOS signaling cascade.
Collapse
Affiliation(s)
- Fei Xu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Yixiao Liu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Xuexue Zhu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Shuangshuang Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Xuelin Shi
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Zhongjie Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Min Ai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Jiangnan Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Bao Hou
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Weiwei Cai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Haijian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Lulu Ni
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Yuetao Zhou
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Liying Qiu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| |
Collapse
|
34
|
Zaiou M. Circular RNAs in hypertension: challenges and clinical promise. Hypertens Res 2019; 42:1653-1663. [PMID: 31239534 DOI: 10.1038/s41440-019-0294-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/03/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022]
Abstract
Hypertension (HT), or high blood pressure (BP), is a chronic disease that is common among populations worldwide. The occurrence of HT is one of the leading causes of cardiovascular morbidity and mortality in adults. Although multiple studies have stressed the multifactorial and multigenic nature of HT, uncertainties about its etiology persist, and current diagnostic biomarkers can explain only a small part of the phenotypic variance of BP. Hence, the search for novel biomarkers that enable early disease prevention and guided therapy is warranted. Regulatory circRNAs have emerged as the newest player in HT-related gene networks and hold promise for improving the accuracy of diagnosis. These RNAs are genome products that are formed through back-splicing of specific regions of pre-mRNAs. Evidence suggests that these RNA species are involved in various metabolic diseases. Recent studies have revealed that aberrant expression of circRNAs is relevant to the occurrence and development of HT. Accordingly, circRNAs are proposed as a new generation of predictive biomarkers and potential therapeutic targets for different forms of HT, including pulmonary hypertension and preeclampsia. This paper presents an overview of the findings from current research focusing on the emerging role of circRNAs in the pathogenesis of hypertension. Furthermore, some of the challenges encountered by circRNA studies are highlighted, and perspectives are provided on the future of research in this area.
Collapse
Affiliation(s)
- Mohamed Zaiou
- University of Lorraine, Department of Biochemistry and Molecular Biology, 7 Avenue de la Foret de Haye, BP 90170, 54505, Vandoeuvre les Nancy Cedex, France.
| |
Collapse
|
35
|
miR-29b Mediates the Chronic Inflammatory Response in Radiotherapy-Induced Vascular Disease. JACC Basic Transl Sci 2019; 4:72-82. [PMID: 30847421 PMCID: PMC6390501 DOI: 10.1016/j.jacbts.2018.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/18/2018] [Accepted: 10/18/2018] [Indexed: 12/17/2022]
Abstract
Radiotherapy is a powerful treatment strategy in patients with oncological diseases. Radiation-induced vasculopathy can dose dependently increase the risk of ischemic cardiovascular diseases (e.g., myocardial infarction, heart failure, stroke). The microRNA miR-29b is repressed in radiation-induced vasculopathy (human irradiated vs. nonirradiated tissue specimen, as well as in murine and cell culture models of irradiation). Pentraxin-3 and dipeptidyl-peptidase 4 are the main downstream effectors of miR-29b in radiation-induced vasculopathy. miR-29b mimics were able to limit pentraxin-3 and dipeptidyl-peptidase 4 levels in the irradiated vasculature (murine model) and to constrain the burden of vascular inflammation.
As a consequence of the success of present-day cancer treatment, radiotherapy-induced vascular disease is emerging. This disease is caused by chronic inflammatory activation and is likely orchestrated in part by microRNAs. In irradiated versus nonirradiated conduit arteries from patients receiving microvascular free tissue transfer reconstructions, irradiation resulted in down-regulation of miR-29b and up-regulation of miR-146b. miR-29b affected inflammation and adverse wound healing through its targets pentraxin-3 and dipeptidyl-peptidase 4. In vitro and in vivo, we showed that miR-29b overexpression therapy, through inhibition of pentraxin-3 and dipeptidyl-peptidase 4, could dampen the vascular inflammatory response.
Collapse
Key Words
- Apoe–/–, apolipoprotein E knockout
- DIG, digoxigenin
- DPP4, Dpp4, dipeptidyl-peptidase 4
- FFT, free flap tissue transfer
- HCtAEC, human carotid artery endothelial cell
- HCtASMC, human carotid artery smooth muscle cell
- NR, nonirradiated
- PTX3, Ptx3, pentraxin-3
- RNA, ribonucleic acid
- SMC, smooth muscle cell
- TGF, tumor growth factor
- arteriosclerosis
- inflammation
- mRNA, messenger ribonucleic acid
- miRNA, microRNA
- microRNA
- radiotherapy
- vRTx, radiation vasculopathy
Collapse
|
36
|
Huang R, Hu Z, Cao Y, Li H, Zhang H, Su W, Xu Y, Liang L, Melgiri ND, Jiang L. MiR-652-3p inhibition enhances endothelial repair and reduces atherosclerosis by promoting Cyclin D2 expression. EBioMedicine 2019; 40:685-694. [PMID: 30674440 PMCID: PMC6413686 DOI: 10.1016/j.ebiom.2019.01.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 01/12/2019] [Accepted: 01/14/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Atherosclerosis is a hyperlipidemia-induced condition affecting the arterial wall that damages healthy endothelial cell (EC) function, leading to enhanced risk of atherothrombotic events. Certain microRNAs regulate EC dysfunction in response to hyperlipidemia and may be suitable therapeutic targets to combat atherosclerosis. METHODS miRNA expression in human ECs was analyzed under various conditions to identify key microRNAs. High-cholesterol diet (HCD)-fed Mir652-/-Apoe-/- (Mir652-/-) mice and matching Mir652+/+Apoe-/- (Mir652+/+) mice were subjected to carotid injury to analyze the effects of miR-652 knockdown on endothelial repair. In silico analysis followed by in vitro and in vivo experiments were applied to identify miR-652's target gene Ccnd2 and investigate the pair's effects on ECs. miR-652-5p and miR-652-3p antagomir therapies were tested in Mir652+/+ mice under normal and HCD diet to assess their effect on endothelial repair. FINDINGS miR-652-3p, which is upregulated in human and murine atherosclerotic plaques, suppresses expression of the endothelial repair gene Ccnd2, thereby enhancing atherosclerotic lesion formation. Post-denudation recovery of ECs was promoted in Mir652-/- mice due to enhanced EC proliferation attributable to de-repression of miR-652-3p's (but not miR-652-5p's) regulation of Ccnd2 expression. Under hyperlipidemic conditions at non-predilection sites, miR-652-3p produces anti-proliferative effects in ECs, such that Mir652-/- mice display reduced atherosclerotic progression. In contrast, neither miR-652-3p nor Ccnd2 displayed significant effects on the endothelium at predilection sites or under disturbed flow conditions. Administration of a miR-652-3p antagomir rescued the proliferation of ECs in vivo, thereby limiting atherosclerotic development. INTERPRETATION miR-652-3p blockade may be a potential therapeutic strategy against atherosclerosis.
Collapse
Affiliation(s)
- Rongzhong Huang
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Zicheng Hu
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yu Cao
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Hongrong Li
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Hong Zhang
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Wenhua Su
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Yu Xu
- Statistical Laboratory, Chuangxu Institute of Lifescience, Chongqing, China
| | - Liwen Liang
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - N D Melgiri
- Impactys Foundation for Biomedical Research, San Diego, CA, USA.
| | - Lihong Jiang
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China.
| |
Collapse
|
37
|
Summerhill V, Karagodin V, Grechko A, Myasoedova V, Orekhov A. Vasculoprotective Role of Olive Oil Compounds via Modulation of Oxidative Stress in Atherosclerosis. Front Cardiovasc Med 2018; 5:188. [PMID: 30622950 PMCID: PMC6308304 DOI: 10.3389/fcvm.2018.00188] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022] Open
Abstract
Existing evidence supports the significant role of oxidative stress in the endothelial injury, and there is a direct link between increased oxidative stress, and the development of endothelial dysfunction. Endothelial dysfunction precedes the development of atherosclerosis and subsequent cardiovascular disease (CVD). The overproduction of reactive oxygen species facilitates the processes, such as oxidative modification of low-density lipoproteins and phospholipids, reduction in the NOS-derived nitric oxide, and the functional disruption of high-density lipids that are profoundly involved in atherogenesis, inflammation, and thrombus formation in vascular cells. Thus, under oxidative stress conditions, endothelial dysfunction was found to be associated with the following endothelial alterations: reduced nitric oxide bioavailability, increased anticoagulant properties, increased platelet aggregation, increased expression of adhesion molecules, chemokines, and cytokines. In this review, we summarized the evidence indicating that endothelial damage triggered by oxidation can be diminished or reversed by the compounds of olive oil, a readily available antioxidant food source. Olive oil bioactive compounds exhibited a potent capability to attenuate oxidative stress and improve endothelial function through their anti-inflammatory, anti-oxidant, and anti-thrombotic properties, therefore reducing the risk and progression of atherosclerosis. Also, their molecular mechanisms of action were explored to establish the potential preventive and/or therapeutic alternatives to the pharmacological remedies available.
Collapse
Affiliation(s)
- Volha Summerhill
- Skolkovo Innovative Center, Institute for Atherosclerosis Research Moscow, Moscow, Russia
| | - Vasilyi Karagodin
- Department of Commodity Research and Expertise, Plekhanov Russian University of Economics, Moscow, Russia
| | - Andrey Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow, Russia
| | - Veronika Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander Orekhov
- Skolkovo Innovative Center, Institute for Atherosclerosis Research Moscow, Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
38
|
Donaldson CJ, Lao KH, Zeng L. The salient role of microRNAs in atherogenesis. J Mol Cell Cardiol 2018; 122:98-113. [DOI: 10.1016/j.yjmcc.2018.08.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 12/17/2022]
|
39
|
Poller W, Dimmeler S, Heymans S, Zeller T, Haas J, Karakas M, Leistner DM, Jakob P, Nakagawa S, Blankenberg S, Engelhardt S, Thum T, Weber C, Meder B, Hajjar R, Landmesser U. Non-coding RNAs in cardiovascular diseases: diagnostic and therapeutic perspectives. Eur Heart J 2018; 39:2704-2716. [PMID: 28430919 PMCID: PMC6454570 DOI: 10.1093/eurheartj/ehx165] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/14/2017] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
Recent research has demonstrated that the non-coding genome plays a key role in genetic programming and gene regulation during development as well as in health and cardiovascular disease. About 99% of the human genome do not encode proteins, but are transcriptionally active representing a broad spectrum of non-coding RNAs (ncRNAs) with important regulatory and structural functions. Non-coding RNAs have been identified as critical novel regulators of cardiovascular risk factors and cell functions and are thus important candidates to improve diagnostics and prognosis assessment. Beyond this, ncRNAs are rapidly emgerging as fundamentally novel therapeutics. On a first level, ncRNAs provide novel therapeutic targets some of which are entering assessment in clinical trials. On a second level, new therapeutic tools were developed from endogenous ncRNAs serving as blueprints. Particularly advanced is the development of RNA interference (RNAi) drugs which use recently discovered pathways of endogenous short interfering RNAs and are becoming versatile tools for efficient silencing of protein expression. Pioneering clinical studies include RNAi drugs targeting liver synthesis of PCSK9 resulting in highly significant lowering of LDL cholesterol or targeting liver transthyretin (TTR) synthesis for treatment of cardiac TTR amyloidosis. Further novel drugs mimicking actions of endogenous ncRNAs may arise from exploitation of molecular interactions not accessible to conventional pharmacology. We provide an update on recent developments and perspectives for diagnostic and therapeutic use of ncRNAs in cardiovascular diseases, including atherosclerosis/coronary disease, post-myocardial infarction remodelling, and heart failure.
Collapse
Affiliation(s)
- Wolfgang Poller
- Department of Cardiology, CBF, CC11, Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Charite Centrum 11 (Cardiovascular Medicine), Hindenburgdamm 20, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Johann Wolfgang Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main, Germany
- DZHK, Site Rhein-Main, Frankfurt, Germany
| | - Stephane Heymans
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Tanja Zeller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Martinistrasse 52, Hamburg, Germany
- DZHK, Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Jan Haas
- Institute for Cardiomyopathies Heidelberg (ICH), Universitätsklinikum Heidelberg, Im Neuenheimer Feld 669, Heidelberg, Germany
- DZHK, Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Mahir Karakas
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Martinistrasse 52, Hamburg, Germany
- DZHK, Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - David-Manuel Leistner
- Department of Cardiology, CBF, CC11, Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Charite Centrum 11 (Cardiovascular Medicine), Hindenburgdamm 20, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany
| | - Philipp Jakob
- Department of Cardiology, CBF, CC11, Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Charite Centrum 11 (Cardiovascular Medicine), Hindenburgdamm 20, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany
| | - Shinichi Nakagawa
- RNA Biology Laboratory, RIKEN Advanced Research Institute, Wako, Saitama, Japan
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo Nishi 6-chome, Kita-ku, Sapporo, Japan
| | - Stefan Blankenberg
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Martinistrasse 52, Hamburg, Germany
- DZHK, Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Stefan Engelhardt
- Institute for Pharmacology and Toxikology, Technische Universität München, Biedersteiner Strasse 29, München, Germany
- DZHK, Site Munich, Munich, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Christian Weber
- DZHK, Site Munich, Munich, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Pettenkoferstrasse 8a/9, Munich, Germany
| | - Benjamin Meder
- Institute for Cardiomyopathies Heidelberg (ICH), Universitätsklinikum Heidelberg, Im Neuenheimer Feld 669, Heidelberg, Germany
- DZHK, Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Roger Hajjar
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ulf Landmesser
- Department of Cardiology, CBF, CC11, Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Charite Centrum 11 (Cardiovascular Medicine), Hindenburgdamm 20, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany
- Berlin Institute of Health, Kapelle-Ufer 2, Berlin, Germany
| |
Collapse
|
40
|
Morbiducci U, Kok AM, Kwak BR, Stone PH, Steinman DA, Wentzel JJ. Atherosclerosis at arterial bifurcations: evidence for the role of haemodynamics and geometry. Thromb Haemost 2018; 115:484-92. [DOI: 10.1160/th15-07-0597] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/13/2015] [Indexed: 11/05/2022]
Abstract
SummaryAtherosclerotic plaques are found at distinct locations in the arterial system, despite the exposure to systemic risk factors of the entire vascular tree. From the study of arterial bifurcation regions, emerges ample evidence that haemodynamics are involved in the local onset and progression of the atherosclerotic disease. This observed co-localisation of disturbed flow regions and lesion prevalence at geometrically predisposed districts such as arterial bifurcations has led to the formulation of a ‘haemodynamic hypothesis’, that in this review is grounded to the most current research concerning localising factors of vascular disease. In particular, this review focuses on carotid and coronary bifurcations because of their primary relevance to stroke and heart attack. We highlight reported relationships between atherosclerotic plaque location, progression and composition, and fluid forces at vessel’s wall, in particular shear stress and its ‘easier-tomeasure’ surrogates, i.e. vascular geometric attributes (because geometry shapes the flow) and intravascular flow features (because they mediate disturbed shear stress), in order to give more insight in plaque initiation and destabilisation. Analogous to Virchow’s triad for thrombosis, atherosclerosis must be thought of as subject to a triad of, and especially interactions among, haemodynamic forces, systemic risk factors, and the biological response of the wall.
Collapse
|
41
|
Kuosmanen SM, Kansanen E, Kaikkonen MU, Sihvola V, Pulkkinen K, Jyrkkänen HK, Tuoresmäki P, Hartikainen J, Hippeläinen M, Kokki H, Tavi P, Heikkinen S, Levonen AL. NRF2 regulates endothelial glycolysis and proliferation with miR-93 and mediates the effects of oxidized phospholipids on endothelial activation. Nucleic Acids Res 2018; 46:1124-1138. [PMID: 29161413 PMCID: PMC5815049 DOI: 10.1093/nar/gkx1155] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/12/2017] [Accepted: 11/01/2017] [Indexed: 01/02/2023] Open
Abstract
Phospholipids, such as 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (PAPC), are the major components of cell membranes. Their exposure to reactive oxygen species creates oxidized phospholipids, which predispose to the development of chronic inflammatory diseases and metabolic disorders through endothelial activation and dysfunction. Although the effects of oxidized PAPC (oxPAPC) on endothelial cells have been previously studied, the underlying molecular mechanisms evoking biological responses remain largely unknown. Here, we investigated the molecular mechanisms of oxPAPC function with a special emphasis on NRF2-regulated microRNAs (miRNAs) in human umbilical vein endothelial cells (HUVECs) utilizing miRNA profiling, global run-on sequencing (GRO-seq), genome-wide NRF2 binding model, and RNA sequencing (RNA-seq) with miRNA overexpression and silencing. We report that the central regulators of endothelial activity, KLF2 for quiescence, PFKFB3 for glycolysis, and VEGFA, FOXO1 and MYC for growth and proliferation, are regulated by transcription factor NRF2 and the NRF2-regulated miR-106b∼25 cluster member, miR-93, in HUVECs. Mechanistically, oxPAPC was found to induce glycolysis and proliferation NRF2-dependently, and oxPAPC-dependent induction of the miR-106b∼25 cluster was mediated by NRF2. Additionally, several regulatory loops were established between NRF2, miR-93 and the essential regulators of healthy endothelium, collectively implying that NRF2 controls the switch between the quiescent and the proliferative endothelial states together with miR-93.
Collapse
Affiliation(s)
- Suvi M Kuosmanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Emilia Kansanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Minna U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Virve Sihvola
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Kati Pulkkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Henna-Kaisa Jyrkkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Pauli Tuoresmäki
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Juha Hartikainen
- School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland
- Heart Centre, Kuopio University Hospital, 70211 Kuopio, Finland
| | | | - Hannu Kokki
- School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland
- Anaesthesia and Operative Services, 70211 Kuopio University Hospital, Kuopio, Finland
| | - Pasi Tavi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
42
|
Wang X, Searle AK, Hohmann JD, Liu AL, Abraham MK, Palasubramaniam J, Lim B, Yao Y, Wallert M, Yu E, Chen YC, Peter K. Dual-Targeted Theranostic Delivery of miRs Arrests Abdominal Aortic Aneurysm Development. Mol Ther 2018. [PMID: 29525742 PMCID: PMC6080135 DOI: 10.1016/j.ymthe.2018.02.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is an often deadly disease without medical, non-invasive treatment options. The upregulation of vascular cell adhesion molecule-1 (VCAM-1) on aortic endothelium provides an early target epitope for a novel biotechnological theranostic approach. MicroRNA-126 was used as a therapeutic agent, based on its capability to downregulate VCAM-1 expression in endothelial cells and thereby reduces leukocyte adhesion and exerts anti-inflammatory effects. Ultrasound microbubbles were chosen as carriers, allowing both molecular imaging as well as targeted therapy of AAA. Microbubbles were coupled with a VCAM-1-targeted single-chain antibody (scFvmVCAM-1) and a microRNA-126 mimic (M126) constituting theranostic microbubbles (TargMB-M126). TargMB-M126 downregulates VCAM-1 expression in vitro and in an in vivo acute inflammatory murine model. Most importantly, using TargMB-M126 and ultrasound-guided burst delivery of M126, the development of AAA in an angiotensin-II-induced mouse model can be prevented. Overall, we describe a unique biotechnological theranostic approach with the potential for early diagnosis and long-sought-after medical therapy of AAA.
Collapse
Affiliation(s)
- Xiaowei Wang
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia; Department of Medicine, Monash University, Melbourne, VIC, Australia.
| | - Amy Kate Searle
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia; Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Jan David Hohmann
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Ao Leo Liu
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia; Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Meike-Kristin Abraham
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Jathushan Palasubramaniam
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Bock Lim
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Yu Yao
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Maria Wallert
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Eefang Yu
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia; Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Yung-Chih Chen
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia; Department of Medicine, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
43
|
Regulatory crosstalk between KLF5, miR-29a and Fbw7/CDC4 cooperatively promotes atherosclerotic development. Biochim Biophys Acta Mol Basis Dis 2017; 1864:374-386. [PMID: 29074464 DOI: 10.1016/j.bbadis.2017.10.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/01/2017] [Accepted: 10/16/2017] [Indexed: 12/13/2022]
Abstract
Atherogenesis is a chronic inflammatory process that involves complex interactions between endothelial dysfunction, lipid deposition and vascular smooth-muscle cell (VSMC) proliferation. However, the molecular mechanism is still unclear. We found that a pro-atherosclerotic factor (oxLDL) induced the expression of Krüppel-like factor 5 (KLF5), which in turn increased miR-29a expression levels. The increased miR-29a was retained within HASMCs and down-regulated Fbw7/CDC4 expression by targeting the 3´UTR of Fbw7/CDC4, subsequently increasing KLF5 stability by reducing the Fbw7/CDC4-dependent ubiquitination of KLF5, forming a positive feedback loop to enhance VSMC proliferation and promote atherogenesis. These results indicate a potentially important role for the oxLDL-activated feedback mechanism in VSMC proliferation and atherogenesis. Suppression of miR-29a may be an effective way to attenuate atherosclerosis. In conclusion, our data are the first to reveal that the regulatory crosstalk between KLF5, miR-29a, and Fbw7/CDC4 cooperatively promotes atherosclerotic development.
Collapse
|
44
|
Nakajima H, Mochizuki N. Flow pattern-dependent endothelial cell responses through transcriptional regulation. Cell Cycle 2017; 16:1893-1901. [PMID: 28820314 DOI: 10.1080/15384101.2017.1364324] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Blood flow provides endothelial cells (ECs) lining the inside of blood vessels with mechanical stimuli as well as humoral stimuli. Fluid shear stress, the frictional force between flowing blood and ECs, is recognized as an essential mechanical cue for vascular growth, remodeling, and homeostasis. ECs differentially respond to distinct flow patterns. High laminar shear flow leads to inhibition of cell cycle progression and stabilizes vessels, whereas low shear flow or disturbed flow leads to increased turnover of ECs and inflammatory responses of ECs prone to atherogenic. These differences of EC responses dependent on flow pattern are mainly ascribed to distinct patterns of gene expression. In this review, we highlight flow pattern-dependent transcriptional regulation in ECs by focusing on KLF2 and NFκB, major transcription factors responding to laminar flow and disturbed flow, respectively. Moreover, we introduce roles of a new flow-responsive transcriptional co-regulator, YAP, in blood vessel maintenance and discuss how these transcriptional regulators are spatiotemporally regulated by flow and then regulate EC functions in normal and pathological conditions.
Collapse
Affiliation(s)
- Hiroyuki Nakajima
- a Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute , Suita , Osaka , Japan
| | - Naoki Mochizuki
- a Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute , Suita , Osaka , Japan.,b AMED-CREST. National Cerebral and Cardiovascular Center , Suita , Osaka , Japan
| |
Collapse
|
45
|
Cheng XW, Wan YF, Zhou Q, Wang Y, Zhu HQ. MicroRNA‑126 inhibits endothelial permeability and apoptosis in apolipoprotein E‑knockout mice fed a high‑fat diet. Mol Med Rep 2017; 16:3061-3068. [PMID: 28713948 PMCID: PMC5548065 DOI: 10.3892/mmr.2017.6952] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 03/21/2017] [Indexed: 02/01/2023] Open
Abstract
Endothelial dysfunction and apoptosis have key roles in the initiation and progression of atherosclerosis (AS). AS has been demonstrated to be associated with a high-fat diet, which may increase endothelial permeability and apoptosis; however, the exact mechanisms underlying the development of AS remain poorly understood. MicroRNAs (miRNAs) are vital for the regulation of cardiovascular disease, and dysregulated miRNAs have been implicated in AS. The present study investigated whether miRNA (miR)-126 regulates high-fat diet-induced endothelial permeability and apoptosis by targeting transforming growth factor β (TGFβ), a secreted protein that controls cellular proliferation and apoptosis. In the present study, apolipoprotein E (apoE)−/− mice were fed a high-fat diet in order to establish a model of AS. Mice were subcutaneously injected with a miR-126 mimic, a miR-126 antagomir or control miRNA. Reverse transcription-quantitative polymerase chain reaction was used to assess miR-126 expression, and a fluorometric assay was used to evaluate caspase-3 activity. The effects of miR-126 on the endothelial permeability of the aortic intima were also explored. Western blotting and immunohistochemical analysis were used to investigate the effects of miR-126 on B-cell lymphoma-2 (Bcl-2) and transforming growth factor (TGF) β protein expression levels. Furthermore, a luciferase assay was performed to verify whether TGFβ may be a direct target gene of miR-126. In apolipoprotein E-knockout mice, a high-fat diet reduced miR-126 expression and induced apoptosis as determined by the upregulation of caspase-3 activity. A miR-126 antagomir increased endothelial permeability and apoptosis in mice fed a high-fat diet. By contrast, an miR-126 mimic attenuated endothelial permeability and apoptosis. The reduction in miR-126 was associated with a reduction in protein expression levels of Bcl-2 and an increase of TGFβ in mice fed a high-fat diet. In addition, the present study demonstrated that miR-126 reduced TGFβ expression following binding to the 3′-untranslated region of TGFβ mRNA. The current study demonstrated a role for miR-126 in AS and identified TGFβ as a direct target of miR-126. Furthermore, the present study demonstrated that miR-126 contributed to endothelial permeability and apoptosis, and suggested that the downregulation of TGFβ may be involved in the molecular mechanisms underlying the actions of miR-126. miR-126 may therefore have potential as a novel therapeutic target for the treatment of AS.
Collapse
Affiliation(s)
- Xiao-Wen Cheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yu-Feng Wan
- Department of Otolaryngology, The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, P.R. China
| | - Qing Zhou
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuan Wang
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Hua-Qing Zhu
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
46
|
Khalyfa A, Kheirandish-Gozal L, Khalyfa AA, Philby MF, Alonso-Álvarez ML, Mohammadi M, Bhattacharjee R, Terán-Santos J, Huang L, Andrade J, Gozal D. Circulating Plasma Extracellular Microvesicle MicroRNA Cargo and Endothelial Dysfunction in Children with Obstructive Sleep Apnea. Am J Respir Crit Care Med 2017; 194:1116-1126. [PMID: 27163713 DOI: 10.1164/rccm.201602-0323oc] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RATIONALE Obese children are at increased risk for developing obstructive sleep apnea (OSA), and both of these conditions are associated with an increased risk for endothelial dysfunction (ED) in children, an early risk factor for atherosclerosis and cardiovascular disease. Although weight loss and treatment of OSA by adenotonsillectomy improve endothelial function, not every obese child or child with OSA develops ED. Exosomes are circulating extracellular vesicles containing functional mRNA and microRNA (miRNA) that can be delivered to other cells, such as endothelial cells. OBJECTIVES To investigate whether circulating exosomal miRNAs of children with OSA differentiate based on endothelial functional status. METHODS Obese children (body mass index z score >1.65) and nonobese children were recruited and underwent polysomnographic testing (PSG), and fasting endothelial function measurements and blood draws in the morning after PSG. Plasma exosomes were isolated from all subjects. Isolated exosomes were then incubated with confluent endothelial cell monolayer cultures. Electric cell-substrate impedance sensing systems were used to determine the ability of exosomes to disrupt the intercellular barrier formed by confluent endothelial cells. In addition, immunofluorescent assessments of zonula occludens-1 tight junction protein cellular distribution were conducted to examine endothelial barrier dysfunction. miRNA and mRNA arrays were also applied to exosomes and endothelial cells, and miRNA inhibitors and mimics were transfected for mechanistic assays. MEASUREMENTS AND MAIN RESULTS Plasma exosomes isolated from either obese children or nonobese children with OSA were primarily derived from endothelial cell sources and recapitulated ED, or its absence, in naive human endothelial cells and also in vivo when injected into mice. Microarrays identified a restricted signature of exosomal miRNAs that readily distinguished ED from normal endothelial function. Among the miRNAs, expression of exosomal miRNA-630 was reduced in children with ED and normalized after therapy along with restoration of endothelial function. Conversely, transfection of exosomes from subjects without ED with an miRNA-630 inhibitor induces the ED functional phenotype. Gene target discovery experiments further revealed that miRNA-630 regulates 416 gene targets in endothelial cells that include the Nrf2, AMP kinase, and tight junction pathways. CONCLUSIONS These observations elucidate a novel role of exosomal miRNA-360 as a putative key mediator of vascular function and cardiovascular disease risk in children with underlying OSA and/or obesity, and identify therapeutic targets.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- 1 Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, and
| | - Leila Kheirandish-Gozal
- 1 Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, and
| | - Ahamed A Khalyfa
- 1 Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, and
| | - Mona F Philby
- 1 Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, and
| | - María Luz Alonso-Álvarez
- 2 Sleep Unit, CIBER of Respiratory Diseases, Instituto Carlos III, CIBERES, Hospital Universitario de Burgos, Burgos, Spain
| | - Meelad Mohammadi
- 1 Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, and
| | - Rakesh Bhattacharjee
- 1 Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, and
| | - Joaquin Terán-Santos
- 2 Sleep Unit, CIBER of Respiratory Diseases, Instituto Carlos III, CIBERES, Hospital Universitario de Burgos, Burgos, Spain
| | - Lei Huang
- 3 Center for Research Informatics, The University of Chicago, Chicago, Illinois; and
| | - Jorge Andrade
- 3 Center for Research Informatics, The University of Chicago, Chicago, Illinois; and
| | - David Gozal
- 1 Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, and
| |
Collapse
|
47
|
Mao YZ, Jiang L. Effects of Notch signalling pathway on the relationship between vascular endothelial dysfunction and endothelial stromal transformation in atherosclerosis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017. [PMID: 28622044 DOI: 10.1080/21691401.2017.1337030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
At present, with the improvement of living standards and population aging, the incidence of cardiovascular and cerebrovascular disease is on the rise and has been a serious threat to human health. Statistics show that the current death caused by cardiovascular and cerebrovascular disease has become the first cause of death has been increasing year by year. Therefore, studies on coronary heart disease and atherosclerosis (AS) have become a hot topic in clinical and basic research. In this study, the question of the effect of Notch signalling pathway on the relationship between endothelial dysfunction and endothelial stromal transformation in AS was studied in depth. Based on our results, we drew conclusions as follows. First, the Notch signalling pathway was activated in the atherosclerotic model; secondly, the Notch signalling pathway was demonstrated to enhance AS by promoting vascular endothelial dysfunction; thirdly, it was demonstrated that the Notch signalling pathway was mediated by promoting endothelial and to enhance AS; finally, we confirmed the endothelial function through the Notch signalling pathway to affect the transformation of endothelial stroma to achieve synergistic AS effect. The results of this study have a good guiding significance for the important role of Notch signalling in AS and indicate the ability to influence endothelial function and endothelial stromal transformation by intervening Notch signalling pathway and can affect the relationship between them, and thus eventually achieve the treatment of AS.
Collapse
Affiliation(s)
- Yong-Zhong Mao
- a Department of Pediatric Surgery Union Hospital , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Ling Jiang
- b Department of Geriatrics , Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| |
Collapse
|
48
|
Sukegawa M, Wang X, Nishioka C, Pan B, Xu K, Ohkawara H, Hamasaki Y, Mita M, Nakamura K, Okamoto M, Shimura H, Ohta M, Ikezoe T. The BCR/ABL tyrosine kinase inhibitor, nilotinib, stimulates expression of IL-1β in vascular endothelium in association with downregulation of miR-3p. Leuk Res 2017; 58:83-90. [PMID: 28501737 DOI: 10.1016/j.leukres.2017.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 10/19/2022]
Abstract
BCR/ABL tyrosine kinase inhibitors (TKIs) have significantly improved the prognosis for in dividuals with chronic myeloid leukemia (CML). However, many patients treated with TKIs suffer from TKI-related complications. In particular, vascular events such as peripheral artery occlusive disease have become aserious clinical problem for patients who receive the TKI, nilotinib. At present, the molecular mechanisms by which TKIs cause vascular endothelial cell insults remain unknown.This study explored the effects of the TKIs, imatinib, nilotinib and dasatinib, on vascular endothelial cells in vitro, and found that only nilotinib induced expression of interleukin-1β (IL-1β) by vascular endothelial cells. Nilotinib-induced IL-1β expression stimulated the adhesion of monocytes to vascular endothelial cells in association with an increase in levels of adhesion molecules. MicroRNA database searching identified miR-3121-3p binding sites in the 3'-UTR of the IL-1β gene. Exposure of endothelial cells to nilotinib caused downregulation of miR-3121-3p in these cells. Importantly, forced-expression of miR-3121-3p counteracted nilotinib-induced expression of IL-1β. Importantly, serum levels if IL-1β were significantly elevated in CML patients receiving nilotinib (n=14) compared to those receiving other TKIs (n=16) (3.76±1.22pg/ml vs 0.27±0.77pg/ml, p<0.05). Taken together, our data suggest that nilotinib decreases levels of miR-3121-3p resulting in an increase in expression of IL-1β and adhesion molecules in vascular endothelial cells. The miR-3121-3p/IL-1β axis could be a potential target to prevent vascular events in CML patients with high risk of vascular events.
Collapse
Affiliation(s)
- Masumi Sukegawa
- Department of Hematology, Fukushima Medical University, Hikarigaoka-1, Fukushima 960-1295, Japan
| | - Xiangmin Wang
- Department of Hematology, Fukushima Medical University, Hikarigaoka-1, Fukushima 960-1295, Japan; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99,West Huaihai Road, 221002 Xuzhou, China
| | - Chie Nishioka
- Department of Hematology and Respiratory Medicine, Kochi University, Oko-cho, Nankoku, Kochi 783-8505, Japan
| | - Bin Pan
- Department of Hematology, Fukushima Medical University, Hikarigaoka-1, Fukushima 960-1295, Japan; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99,West Huaihai Road, 221002 Xuzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99,West Huaihai Road, 221002 Xuzhou, China
| | - Hiroshi Ohkawara
- Department of Hematology, Fukushima Medical University, Hikarigaoka-1, Fukushima 960-1295, Japan
| | - Yoichi Hamasaki
- Department of Hematology, Iwaki Kyoritsu General Hospital, Iwaki, Fukushima 973-8402 Japan
| | - Masayuki Mita
- Department of Hematology, Shirakawa Kosei General Hospital, Shirakawa, 961-0005 Fukushima, Japan
| | - Kenichi Nakamura
- Department of Hematology, Shirakawa Kosei General Hospital, Shirakawa, 961-0005 Fukushima, Japan
| | - Masatoshi Okamoto
- Department of Hematology, YUASA Foundation Jusendo General Hospital, Koriyama, Fukushima 963-8585, Japan
| | - Hiromi Shimura
- Department of Laboratory Medicine, Fukushima Medical University, Hikarigaoka-1, Fukushima 960-1295, Japan
| | - Masatsugu Ohta
- Department of Hematology, Fukushima Medical University Aizu Medical Center, Aizuwakamatsu, 969-3492 Fukushima, Japan
| | - Takayuki Ikezoe
- Department of Hematology, Fukushima Medical University, Hikarigaoka-1, Fukushima 960-1295, Japan.
| |
Collapse
|
49
|
|
50
|
Sajjanar B, Deb R, Raina SK, Pawar S, Brahmane MP, Nirmale AV, Kurade NP, Manjunathareddy GB, Bal SK, Singh NP. Untranslated regions (UTRs) orchestrate translation reprogramming in cellular stress responses. J Therm Biol 2017; 65:69-75. [DOI: 10.1016/j.jtherbio.2017.02.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 11/29/2022]
|