1
|
Akiyama M. Expression of Elastin, F-Box and WD-40 Domain-Containing Protein 2, Fibrillin-1, and Alpha-Smooth Muscle Actin in Utilized Blood Vessels for explant culture-A New 3D in Vitro Vascular Model from Bovine Legs. Cell Biochem Biophys 2024:10.1007/s12013-024-01647-5. [PMID: 39731648 DOI: 10.1007/s12013-024-01647-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 12/30/2024]
Abstract
Elastic fibers of the internal and external elastic laminae maintain blood vessel shapes. Impairment of smooth muscle cell function leads to vascular disease development. F-box and WD-40 domain-containing protein 2 (FBXW2) is associated with elastic fibers and osteocalcin expression for bone regeneration in the periosteum. Here, it is hypothesized that FBXW2 has different roles in periosteum and blood vessels. Furthermore, if FBXW2 would be a component of elastic fiber of blood vessels, FBXW2 would be expressed where the well-known components elastin and fibrillin-1 are expressed. For this purpose, explant culture of blood vessels from bovine legs were performed for 5 weeks. It was found that elastin and FBXW2 were expressed within the elastic laminae, whereas fibrillin-1 was expressed around them. After explant culture, elastin and FBXW2 sustained the shape of the elastic fibers in the elastic lamina, whereas the fibrillin-1-rich layer became wide range and encompass toward intima and adventitia layers. Hematoxylin Eosin staining and immunohistochemistry of alpha-smooth muscle actin (α-SMA) revealed weakened media layer after 5 weeks culture. Although fibrillin-1 is a well-known component of elastic fibers and elastin, this study revealed that the location of fibrillin-1 is different from that of elastin, whereas FBXW2 is present in the same region as elastin from day 0 to week 5. In blood vessels, fibrillin-1 fibers around the elastic lamina may be oxytalan fibers. Thus, the proposed 3D in vitro model in this study is useful for identifying the mechanisms of vascular degradation.
Collapse
Affiliation(s)
- Mari Akiyama
- Department of Biomaterials/Osaka Dental University, 8-1, Kuzuhahanazono-cho, Osaka, 573-1121, Japan.
| |
Collapse
|
2
|
Leng S, Li H, Zhang P, Dang Z, Shao B, Xue S, Ning Y, Teng X, Zhang L, Wang H, Li N, Zhang F, Yu W. SGK1-Mediated Vascular Smooth Muscle Cell Phenotypic Transformation Promotes Thoracic Aortic Dissection Progression. Arterioscler Thromb Vasc Biol 2024. [PMID: 39633576 DOI: 10.1161/atvbaha.124.321421] [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: 06/20/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND The occurrence of thoracic aortic dissection (TAD) is closely related to the transformation of vascular smooth muscle cells (VSMCs) from a contractile to a synthetic phenotype. The role of SGK1 (serum- and glucocorticoid-regulated kinase 1) in VSMC phenotypic transformation and TAD occurrence is unclear. METHODS Four-week-old male Sgk1F/F (Sgk1 floxed) and Sgk1F/F;TaglnCre (smooth muscle cell-specific Sgk1 knockout) mice were administered β-aminopropionitrile monofumarate for 4 weeks to model TAD. The SGK1 inhibitor GSK650394 was administered daily via intraperitoneal injection to treat the mouse model of TAD. Immunopurification and mass spectrometry were used to identify proteins that interact with SGK1. Immunoprecipitation, immunofluorescence colocalization, and GST (glutathione S-transferase) pull-down were used to detect molecular interactions between SGK1 and SIRT6 (sirtuin 6). RNA-sequencing analysis was performed to evaluate changes in the SIRT6 transcriptome. Quantitative chromatin immunoprecipitation was used to determine the target genes regulated by SIRT6. Functional experiments were also conducted to investigate the role of SGK1-SIRT6-MMP9 (matrix metalloproteinase 9) in VSMC phenotypic transformation. The effect of SGK1 regulation on target genes was evaluated in human and mouse TAD samples. RESULTS Sgk1F/F;TaglnCre or pharmacological blockade of Sgk1 inhibited the formation and rupture of β-aminopropionitrile monofumarate-induced TADs in mice and reduced the degradation of the ECM (extracellular matrix) in vessels. Mechanistically, SGK1 promoted the ubiquitination and degradation of SIRT6 by phosphorylating SIRT6 at Ser338, thereby reducing the expression of the SIRT6 protein. Furthermore, SIRT6 transcriptionally inhibits the expression of MMP9 through epigenetic modification, forming the SGK1-SIRT6-MMP9 regulatory axis, which participates in the ECM signaling pathway. Additionally, our data showed that the lack of SGK1-mediated inhibition of ECM degradation and VSMC phenotypic transformation is partially dependent on the regulatory effect of SIRT6-MMP9. CONCLUSIONS These findings highlight the key role of SGK1 in the pathogenesis of TAD. A lack of SGK1 inhibits VSMC phenotypic transformation by regulating the SIRT6-MMP9 axis, providing insights into potential epigenetic strategies for TAD treatment.
Collapse
Affiliation(s)
- Shuai Leng
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., W.Y.)
| | - Haijie Li
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Pengfei Zhang
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Zhiqiao Dang
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Baowei Shao
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Shishan Xue
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Yansong Ning
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Xilong Teng
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Leilei Zhang
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Honglu Wang
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Na Li
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Fengquan Zhang
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
| | - Wenqian Yu
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.)
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., W.Y.)
| |
Collapse
|
3
|
Jiang L, Lu L, Xue C, Sun H, Ren K, Zhang L, Zhu H, Zhang B, Wang X, Qiao X, Peng X, Liu J, Duan W. ACE2 deficiency inhibits thoracic aortic dissection by enhancing SIRT3 mediated inhibition of inflammation and VSCMs phenotypic switch. Mol Med 2024; 30:154. [PMID: 39300372 DOI: 10.1186/s10020-024-00926-4] [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: 06/25/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Thoracic aortic dissection (TAD) is an irreversible cardiovascular disorder with high mortality and morbidity. However, the molecular mechanisms remain elusive. Thus, identifying an effective therapeutic target to prevent TAD is especially critical. The purpose of this study is to elucidate the potential mechanism of inflammation and vascular smooth muscle cell (VSMCs) phenotypic switch in β-aminopropionitrile fumarate (BAPN)-induced TAD. METHODS A mouse model of TAD induced by BAPN and IL-1β -stimulated HVSMCs in vivo and in vitro models, respectively. ACE2 Knockdown mice treated with BAPN or without, and the TAD mouse model was treated with or without AAV-ACE2. Transthoracic ultrasound was conducted for assessment the maximum internal diameter of the thoracic aorta arch. RNA sequencing analysis was performed to recapitulate transcriptome profile changes. Western blot were used to detect the expression of MMP2, MMP9, ACE2, SIRT3, OPN, SM22α and other inflammatory markers. The circulating levels of ACE2 was measured by ELISA assay. Histological changes of thoracic aorta tissues were assessed by H&E, EVG and IHC analysis. RESULTS We found that circulating levels of and the protein levels of ACE2 were increased in the TAD mouse model and in patients with TAD. For further evidence, ACE2 deficiency decelerated the formation of TAD. However, overexpression of ACE2 aggravated BAPN-induced aortic injury and VSMCs phenotypic switch via lowered SIRT3 expression and elevated inflammatory cytokine expression. CONCLUSION ACE2 deficiency prevented the development of TAD by inhibiting inflammation and VSMCs phenotypic switch in a SIRT3-dependent manner, suggesting that the ACE2/SIRT3 signaling pathway played a pivotal role in the pathological process of TAD and might be a potential therapeutical target.
Collapse
MESH Headings
- Animals
- Angiotensin-Converting Enzyme 2/metabolism
- Angiotensin-Converting Enzyme 2/genetics
- Mice
- Aortic Dissection/metabolism
- Aortic Dissection/etiology
- Aortic Dissection/genetics
- Aortic Dissection/pathology
- Myocytes, Smooth Muscle/metabolism
- Disease Models, Animal
- Sirtuin 3/metabolism
- Sirtuin 3/genetics
- Sirtuin 3/deficiency
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Inflammation/metabolism
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/etiology
- Aortic Aneurysm, Thoracic/genetics
- Male
- Phenotype
- Humans
- Mice, Knockout
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/drug effects
- Aminopropionitrile/pharmacology
- Mice, Inbred C57BL
- Dissection, Thoracic Aorta
Collapse
Affiliation(s)
- Liqing Jiang
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - Linhe Lu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - Chao Xue
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - He Sun
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - Kai Ren
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - Liyun Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - Hanzhao Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - Xiaoya Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - Xinan Qiao
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - Xiangyan Peng
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China
| | - Jincheng Liu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China.
| | - Weixun Duan
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Military Medical University, 127 Changle West Road, Xi'an, 710032, People's Republic of China.
| |
Collapse
|
4
|
Udugampolage NS, Frolova S, Taurino J, Pini A, Martelli F, Voellenkle C. Coding and Non-Coding Transcriptomic Landscape of Aortic Complications in Marfan Syndrome. Int J Mol Sci 2024; 25:7367. [PMID: 39000474 PMCID: PMC11242319 DOI: 10.3390/ijms25137367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Marfan syndrome (MFS) is a rare congenital disorder of the connective tissue, leading to thoracic aortic aneurysms (TAA) and dissection, among other complications. Currently, the most efficient strategy to prevent life-threatening dissection is preventive surgery. Periodic imaging applying complex techniques is required to monitor TAA progression and to guide the timing of surgical intervention. Thus, there is an acute demand for non-invasive biomarkers for diagnosis and prognosis, as well as for innovative therapeutic targets of MFS. Unraveling the intricate pathomolecular mechanisms underlying the syndrome is vital to address these needs. High-throughput platforms are particularly well-suited for this purpose, as they enable the integration of different datasets, such as transcriptomic and epigenetic profiles. In this narrative review, we summarize relevant studies investigating changes in both the coding and non-coding transcriptome and epigenome in MFS-induced TAA. The collective findings highlight the implicated pathways, such as TGF-β signaling, extracellular matrix structure, inflammation, and mitochondrial dysfunction. Potential candidates as biomarkers, such as miR-200c, as well as therapeutic targets emerged, like Tfam, associated with mitochondrial respiration, or miR-632, stimulating endothelial-to-mesenchymal transition. While these discoveries are promising, rigorous and extensive validation in large patient cohorts is indispensable to confirm their clinical relevance and therapeutic potential.
Collapse
Affiliation(s)
| | - Svetlana Frolova
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
- Department of Biosciences, University of Milan, 20122 Milan, Italy
| | - Jacopo Taurino
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, 20097 Milan, Italy; (N.S.U.); (J.T.); (A.P.)
| | - Alessandro Pini
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, 20097 Milan, Italy; (N.S.U.); (J.T.); (A.P.)
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
| |
Collapse
|
5
|
Van Den Heuvel LJF, Peeters S, Meester JAN, Coucke PJ, Loeys BL. An exploration of alternative therapeutic targets for aortic disease in Marfan syndrome. Drug Discov Today 2024; 29:104023. [PMID: 38750929 DOI: 10.1016/j.drudis.2024.104023] [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: 02/27/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024]
Abstract
Marfan syndrome is a rare connective tissue disorder that causes aortic dissection-related sudden death. Current conventional treatments, beta-blockers, and type 1 angiotensin II receptor blockers are prescribed to slow down aortic aneurysm progression and delay (prophylactic) aortic surgery. However, neither of these treatments ceases aortic growth completely. This review focuses on potential alternative therapeutic leads in the field, ranging from widely used medication with beneficial effects on the aorta to experimental inhibitors with the potential to stop aortic growth in Marfan syndrome. Clinical trials are warranted to uncover their full potential.
Collapse
Affiliation(s)
- Lotte J F Van Den Heuvel
- Center for Medical Genetics Antwerp, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Edegem, Belgium; Center for Medical Genetics Ghent, Ghent University, Ghent, Belgium
| | - Silke Peeters
- Center for Medical Genetics Antwerp, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Edegem, Belgium
| | - Josephina A N Meester
- Center for Medical Genetics Antwerp, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Edegem, Belgium
| | - Paul J Coucke
- Center for Medical Genetics Ghent, Ghent University, Ghent, Belgium
| | - Bart L Loeys
- Center for Medical Genetics Antwerp, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Edegem, Belgium; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
6
|
Antequera-González B, Collell-Hernández R, Martínez-Micaelo N, Marimon-Blanch C, Carbonell-Prat B, Escribano J, Alegret JM. miR-130a expression is related to aortic dilation in bicuspid aortic valve children. Pediatr Res 2024; 95:1741-1748. [PMID: 38273119 DOI: 10.1038/s41390-024-03018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/03/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024]
Abstract
BACKGROUND Bicuspid aortic valve disease (BAV) is present in 0.5-2% of the population and can promote aortic dilation, eventually leading to fatal consequences. Although some biomarkers have been proposed in adults, no studies have tested these candidates in children. We aimed to evaluate four miRNAs previously described to be related to BAV disease and aortic dilation in adults in a paediatric cohort. METHODS Eighty participants ≤17 years old (4-17; mean 12) were included. From the BAV group, 40% had a dilated aorta (z score >2). RT‒qPCR were performed in plasma samples to quantify miR-122, miR-130a, miR-486, and miR-718 using the delta-delta Ct method. Functional and enrichment analyses of miR-130a were also performed. RESULTS miR-130a expression in plasma was found to be significantly lower in BAV patients with a dilated aorta versus nondilated patients (p = 0.008) and healthy TAV controls (p = 0.004). Furthermore, miR-130a expression in plasma was inversely correlated with ascending aorta (r = 0.318, p = 0.004) and aortic root z scores (r = 0.322; p = 0.004). Enrichment analysis showed that miR-130a target genes are related to the TGFβ signalling pathway. CONCLUSIONS miR-130a expression in plasma is decreased in aortic-dilated BAV children compared to nondilated BAV children, helping differentiate low- to high-risk patients. IMPACT miR-130a expression in plasma is related to aortic dilation in bicuspid aortic valve (BAV) children. To our knowledge, this is the first study that analyses miRNA patterns in bicuspid aortic valve children with aortic dilation. miR-130a expression in plasma could be a biomarker in order to help differentiate low-to high-risk BAV children, which is vitally important for advanced care planning.
Collapse
Affiliation(s)
- Borja Antequera-González
- Group of Cardiovascular Research, Pere Virgili Health Research Institute (IISPV), Universitat Rovira i Virgili, 43204, Reus, Spain
| | - Rosa Collell-Hernández
- Pediatric Department, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, 43204, Reus, Spain
| | - Neus Martínez-Micaelo
- Group of Cardiovascular Research, Pere Virgili Health Research Institute (IISPV), Universitat Rovira i Virgili, 43204, Reus, Spain
| | - Cristina Marimon-Blanch
- Pediatric Department, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, 43204, Reus, Spain
| | - Bàrbara Carbonell-Prat
- Group of Cardiovascular Research, Pere Virgili Health Research Institute (IISPV), Universitat Rovira i Virgili, 43204, Reus, Spain
- Cardiology Department, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, 43204, Reus, Spain
| | - Joaquín Escribano
- Pediatric Department, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, 43204, Reus, Spain
| | - Josep M Alegret
- Group of Cardiovascular Research, Pere Virgili Health Research Institute (IISPV), Universitat Rovira i Virgili, 43204, Reus, Spain.
- Cardiology Department, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, 43204, Reus, Spain.
| |
Collapse
|
7
|
Liu X, Deng Y, Liang Z, Qiao D, Zhang W, Wang M, Li F, Liu J, Wu Y, Chen G, Liu Y, Tan W, Xing J, Huang W, Zhao D, Li Y. The alteration of the structure and macroscopic mechanical response of porcine patellar tendon by elastase digestion. Front Bioeng Biotechnol 2024; 12:1374352. [PMID: 38694621 PMCID: PMC11061363 DOI: 10.3389/fbioe.2024.1374352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/01/2024] [Indexed: 05/04/2024] Open
Abstract
Background: The treatment of patellar tendon injury has always been an unsolved problem, and mechanical characterization is very important for its repair and reconstruction. Elastin is a contributor to mechanics, but it is not clear how it affects the elasticity, viscoelastic properties, and structure of patellar tendon. Methods: The patellar tendons from six fresh adult experimental pigs were used in this study and they were made into 77 samples. The patellar tendon was specifically degraded by elastase, and the regional mechanical response and structural changes were investigated by: (1) Based on the previous study of elastase treatment conditions, the biochemical quantification of collagen, glycosaminoglycan and total protein was carried out; (2) The patellar tendon was divided into the proximal, central, and distal regions, and then the axial tensile test and stress relaxation test were performed before and after phosphate-buffered saline (PBS) or elastase treatment; (3) The dynamic constitutive model was established by the obtained mechanical data; (4) The structural relationship between elastin and collagen fibers was analyzed by two-photon microscopy and histology. Results: There was no statistical difference in mechanics between patellar tendon regions. Compared with those before elastase treatment, the low tensile modulus decreased by 75%-80%, the high tensile modulus decreased by 38%-47%, and the transition strain was prolonged after treatment. For viscoelastic behavior, the stress relaxation increased, the initial slope increased by 55%, the saturation slope increased by 44%, and the transition time increased by 25% after enzyme treatment. Elastin degradation made the collagen fibers of patellar tendon become disordered and looser, and the fiber wavelength increased significantly. Conclusion: The results of this study show that elastin plays an important role in the mechanical properties and fiber structure stability of patellar tendon, which supplements the structure-function relationship information of patellar tendon. The established constitutive model is of great significance to the prediction, repair and replacement of patellar tendon injury. In addition, human patellar tendon has a higher elastin content, so the results of this study can provide supporting information on the natural properties of tendon elastin degradation and guide the development of artificial patellar tendon biomaterials.
Collapse
Affiliation(s)
- Xiaoyun Liu
- National Key Discipline of Human Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yuping Deng
- National Key Discipline of Human Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Peking University Shenzhen Graduate School, Shenzhen, China
- Department of Orthopedics and Traumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zeyu Liang
- National Key Discipline of Human Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Dan Qiao
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wentian Zhang
- National Key Discipline of Human Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| | - Mian Wang
- National Key Discipline of Human Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Peking University Shenzhen Graduate School, Shenzhen, China
- Department of Orthopaedics, Pingshan General Hospital of Southern Medical University, Shenzhen, China
| | - Feifei Li
- National Key Discipline of Human Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiannan Liu
- National Key Discipline of Human Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yaobing Wu
- National Key Discipline of Human Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guangxin Chen
- Medical Image College, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Yan Liu
- Department of Anatomy, Gannan Healthcare Vocational College, Ganzhou, China
| | - Wenchang Tan
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jian Xing
- Medical Image College, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Wenhua Huang
- National Key Discipline of Human Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Orthopedics and Traumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dongliang Zhao
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yanbing Li
- National Key Discipline of Human Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Terriaca S, Ferlosio A, Scioli MG, Coppa F, Bertoldo F, Pisano C, Belmonte B, Balistreri CR, Orlandi A. miRNA Regulation of Cell Phenotype and Parietal Remodeling in Atherosclerotic and Non-Atherosclerotic Aortic Aneurysms: Differences and Similarities. Int J Mol Sci 2024; 25:2641. [PMID: 38473887 DOI: 10.3390/ijms25052641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Aortic aneurysms are a serious health concern as their rupture leads to high morbidity and mortality. Abdominal aortic aneurysms (AAAs) and thoracic aortic aneurysms (TAAs) exhibit differences and similarities in their pathophysiological and pathogenetic features. AAA is a multifactorial disease, mainly associated with atherosclerosis, characterized by a relevant inflammatory response and calcification. TAA is rarely associated with atherosclerosis and in some cases is associated with genetic mutations such as Marfan syndrome (MFS) and bicuspid aortic valve (BAV). MFS-related and non-genetic or sporadic TAA share aortic degeneration with endothelial-to-mesenchymal transition (End-Mt) and fibrosis, whereas in BAV TAA, aortic degeneration with calcification prevails. microRNA (miRNAs) contribute to the regulation of aneurysmatic aortic remodeling. miRNAs are a class of non-coding RNAs, which post-transcriptionally regulate gene expression. In this review, we report the involvement of deregulated miRNAs in the different aortic remodeling characterizing AAAs and TAAs. In AAA, miRNA deregulation appears to be involved in parietal inflammatory response, smooth muscle cell (SMC) apoptosis and aortic wall calcification. In sporadic and MFS-related TAA, miRNA deregulation promotes End-Mt, SMC myofibroblastic phenotypic switching and fibrosis with glycosaminoglycan accumulation. In BAV TAA, miRNA deregulation sustains aortic calcification. Those differences may support the development of more personalized therapeutic approaches.
Collapse
Affiliation(s)
- Sonia Terriaca
- Anatomic Pathology, Policlinico Tor Vergata, 00133 Rome, Italy
| | - Amedeo Ferlosio
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Maria Giovanna Scioli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Francesca Coppa
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Fabio Bertoldo
- Cardiac Surgery Unit, Department of Surgery, Tor Vergata University, 00133 Rome, Italy
| | - Calogera Pisano
- Cardiac Surgery Unit, Department of Surgery, Tor Vergata University, 00133 Rome, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy
- Azienda sanitaria Provinciale di Catania (ASP), 95124 Catania, Italy
| | - Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| |
Collapse
|
9
|
Terriaca S, Monastero R, Orlandi A, Balistreri CR. The key role of miRNA in syndromic and sporadic forms of ascending aortic aneurysms as biomarkers and targets of novel therapeutic strategies. Front Genet 2024; 15:1365711. [PMID: 38450200 PMCID: PMC10915088 DOI: 10.3389/fgene.2024.1365711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 03/08/2024] Open
Abstract
Increasing evidence shows that epigenetics also plays a key role in regulating the pathogenetic mechanism of all types of aortic aneurysms. It is well-known that epigenetic factors modulate gene expression. This mechanism appears to be of interest especially knowing the relevance of genetic susceptibility and genetic factors in the complex pathophysiology of aortic aneurysms, and of sporadic forms; in fact, the latter are the result of a close interaction between genetic and modifiable lifestyle factors (i.e., nutrition, smoking, infections, use of drugs, alcohol, sedentary lifestyle, etc.). Epigenetic factors include DNA methylation, post-translational histone modifications, and non-coding RNA. Here, our attention is focused on the role of miRNA in syndromic and sporadic forms of thoracic aortic aneurysms. They could be both biomarkers and targets of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sonia Terriaca
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Roberto Monastero
- Section of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Augusto Orlandi
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Carmela Rita Balistreri
- Cellular, Molecular, and Clinical Pathological Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi N D), University of Palermo, Palermo, Italy
| |
Collapse
|
10
|
Shi Y, Chen J, Cai L, Zhang X, Chen Z, Yang J, Jiang Y, Lu Y. Uncovering the Hidden World of Aqueous Humor Proteins for Discovery of Biomarkers for Marfan Syndrome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303161. [PMID: 38088571 PMCID: PMC10853735 DOI: 10.1002/advs.202303161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/23/2023] [Indexed: 12/19/2023]
Abstract
Ectopia lentis is a hallmark of Marfan syndrome (MFS), a genetic connective tissue disorder affecting 1/5000 to 1/10 000 individuals worldwide. Early detection in ophthalmology clinics and timely intervention of cardiovascular complications can be lifesaving. In this study, a modified proteomics workflow with liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based data-independent acquisition (DIA) and field asymmetric ion mobility spectrometry (FAIMS) to profile the proteomes of aqueous humor (AH) and lens tissue from MFS children with ectopia lentis is utilized. Over 2300 and 2938 comparable proteins are identified in AH and the lens capsule, respectively. Functional enrichment analyses uncovered dysregulation of complement and coagulation-related pathways, collagen binding, and cell adhesion in MFS. Through weighted correlation network analysis (WGCNA) and machine learning, distinct modules associated with clinical traits are constructed and a unique biomarker panel (Q14376, Q99972, P02760, Q07507; gene names: GALE, MYOC, AMBP, DPT) is defined. These biomarkers are further validated using advanced parallel reaction monitoring (PRM) in an independent patient cohort. The results provide novel insights into the proteome characterization of ectopia lentis and offer a promising approach for developing a valuable biomarker panel to aid in the early diagnosis of Marfan syndrome via AH proteome.
Collapse
Affiliation(s)
- Yumeng Shi
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Jiahui Chen
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Lei Cai
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Xueling Zhang
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Zexu Chen
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Jin Yang
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Yongxiang Jiang
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| | - Yi Lu
- Eye Institute and Department of Ophthalmology, Eye and ENT HospitalFudan UniversityShanghai200031China
- NHC Key Laboratory of MyopiaFudan UniversityShanghai200031China
- Key Laboratory of MyopiaChinese Academy of Medical SciencesShanghai200031China
- Shanghai Key Laboratory of Visual Impairment and RestorationShanghai200031China
| |
Collapse
|
11
|
Wang Y, Panicker IS, Anesi J, Sargisson O, Atchison B, Habenicht AJR. Animal Models, Pathogenesis, and Potential Treatment of Thoracic Aortic Aneurysm. Int J Mol Sci 2024; 25:901. [PMID: 38255976 PMCID: PMC10815651 DOI: 10.3390/ijms25020901] [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: 12/18/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Thoracic aortic aneurysm (TAA) has a prevalence of 0.16-0.34% and an incidence of 7.6 per 100,000 person-years, accounting for 1-2% of all deaths in Western countries. Currently, no effective pharmacological therapies have been identified to slow TAA development and prevent TAA rupture. Large TAAs are treated with open surgical repair and less invasive thoracic endovascular aortic repair, both of which have high perioperative mortality risk. Therefore, there is an urgent medical need to identify the cellular and molecular mechanisms underlying TAA development and rupture to develop new therapies. In this review, we summarize animal TAA models including recent developments in porcine and zebrafish models: porcine models can assess new therapeutic devices or intervention strategies in a large mammal and zebrafish models can employ large-scale small-molecule suppressor screening in microwells. The second part of the review covers current views of TAA pathogenesis, derived from recent studies using these animal models, with a focus on the roles of the transforming growth factor-beta (TGFβ) pathway and the vascular smooth muscle cell (VSMC)-elastin-contractile unit. The last part discusses TAA treatment options as they emerge from recent preclinical studies.
Collapse
Affiliation(s)
- Yutang Wang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia; (I.S.P.)
| | - Indu S. Panicker
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia; (I.S.P.)
| | - Jack Anesi
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia; (I.S.P.)
| | - Owen Sargisson
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia; (I.S.P.)
| | - Benjamin Atchison
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia; (I.S.P.)
| | - Andreas J. R. Habenicht
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), 80336 Munich, Germany;
| |
Collapse
|
12
|
Zhang L, Huang QW, Pu YF, Xiao XQ, Song BJ, Zhang XP, Yang YS, Zhang YS, Gong FH. MiR-122 knockdown regulates vascular smooth muscle cells phenotypic switching through enhanced FOXO3 expression. Can J Physiol Pharmacol 2023; 101:652-660. [PMID: 37747048 DOI: 10.1139/cjpp-2022-0549] [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] [Indexed: 09/26/2023]
Abstract
Vascular smooth muscle cells (VSMCs) phenotypic switching is identified as enhanced dedifferentiation, proliferation, and migration ability of VSMCs, in which microRNAs have been identified as important regulators of the process. The present study is aimed to explore the pathophysiological effect of miR-122 on VSMC phenotypic modulation. Here, the result showed that the decreased miR-122 expression was found in VSMCs subjected to platelet-derived growth factor-BB (PDGF-BB) treatment. Next, we investigated the response of miR-122 knockdown in VSMCs with PDGF-BB stimulation. MiR-122 silencing showed increased proliferation and migration capability, whereas attenuated the differentiation markers expression. The above results were reversed by miR-122 overexpression. Finally, we further demonstrated that FOXO3 was an important target for miR-122. Collectively, we demonstrated that miR-122 silencing promoted VSMC phenotypic modulation partially through upregulated FOXO3 expression that indicated miR-122 may be a novel therapeutic target for neointimal formation.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren 554300, China
- Department of Cardiology, The Sixth Affiliated Hospital of Jishou University, Jishou 416000, China
| | - Qian-Wei Huang
- Department of Clinical Laboratory, Tongren Municipal People's Hospital, Tongren 554300, China
| | - Yan-Fen Pu
- Department of Clinical Laboratory, Tongren Municipal People's Hospital, Tongren 554300, China
| | - Xiao-Qiang Xiao
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren 554300, China
| | - Bian-Jing Song
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren 554300, China
| | - Xue-Ping Zhang
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren 554300, China
| | - Yong-Sheng Yang
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren 554300, China
| | - Yu-Song Zhang
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren 554300, China
| | - Fu-Han Gong
- Department of Cardiology, Tongren Municipal People's Hospital, Tongren 554300, China
| |
Collapse
|
13
|
Lau C, Muthu ML, Siddiqui IF, Li L, Reinhardt DP. High-Fat Diet Has a Protective Sex-Dependent Effect on Aortic Aneurysm Severity in a Marfan Syndrome Mouse Model. Can J Cardiol 2023; 39:1553-1567. [PMID: 37482239 DOI: 10.1016/j.cjca.2023.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Marfan syndrome (MFS) is a genetic disorder caused by mutations in fibrillin-1 and is characterized by thoracic aortic aneurysms and other complications. Previous studies revealed sexual dimorphisms in formation of aortic aneurysm in patients with MFS. The current study aimed to investigate the combined role of a high-fat diet (HFD) and biological sex in aortic disease using the mgR/mgR MFS mouse model. METHODS Male and female mgR/mgR mice, as well as wild-type (WT) littermate mice, were fed a control diet (CD [10% fat]) or HFD (60% fat) from 4 to 12 weeks of age. Key aortic disease parameters analyzed included the diameter of the aortic wall; elastic fibre fragmentation; proteoglycan content; mRNA levels of Mmp12, Col1a1, Col3a1, and Fbn1; and fibrillin-1 deposition in the aortic wall. RESULTS HFD-fed female mgR/mgR mice had significantly reduced aortic diameters (35%), elastic fibre fragmentation (56%), pathologically enhanced proteoglycans (45%), and expression of Mmp12 (64%), Col1a1 (41%), and Col3a1 (43%) compared with male mgR/mgR mice on HFD. Fibrillin-1 deposition and Fbn1 mRNA levels were unaffected. The data reveal a protective effect of HFD in female mice. In contrast, CD did not exert any protective effects. CONCLUSIONS This study demonstrates a specific sexual dimorphism in MFS mice, with HFD exerting an explicit protective effect on severity of aortic disease in female mice. These preclinical data may be useful for developing nutritional recommendations for individuals with MFS in the longer term.
Collapse
Affiliation(s)
- Cori Lau
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Muthu L Muthu
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Iram Fatima Siddiqui
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Ling Li
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
14
|
Ding YN, Wang TT, Lv SJ, Tang X, Wei ZY, Yao F, Xu HS, Chen YN, Wang XM, Wang HY, Wang HP, Zhang ZQ, Zhao X, Hao DL, Sun LH, Zhou Z, Wang L, Chen HZ, Liu DP. SIRT6 is an epigenetic repressor of thoracic aortic aneurysms via inhibiting inflammation and senescence. Signal Transduct Target Ther 2023; 8:255. [PMID: 37394473 DOI: 10.1038/s41392-023-01456-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 07/04/2023] Open
Abstract
Thoracic aortic aneurysms (TAAs) develop asymptomatically and are characterized by dilatation of the aorta. This is considered a life-threating vascular disease due to the risk of aortic rupture and without effective treatments. The current understanding of the pathogenesis of TAA is still limited, especially for sporadic TAAs without known genetic mutation. Sirtuin 6 (SIRT6) expression was significantly decreased in the tunica media of sporadic human TAA tissues. Genetic knockout of Sirt6 in mouse vascular smooth muscle cells accelerated TAA formation and rupture, reduced survival, and increased vascular inflammation and senescence after angiotensin II infusion. Transcriptome analysis identified interleukin (IL)-1β as a pivotal target of SIRT6, and increased IL-1β levels correlated with vascular inflammation and senescence in human and mouse TAA samples. Chromatin immunoprecipitation revealed that SIRT6 bound to the Il1b promoter to repress expression partly by reducing the H3K9 and H3K56 acetylation. Genetic knockout of Il1b or pharmacological inhibition of IL-1β signaling with the receptor antagonist anakinra rescued Sirt6 deficiency mediated aggravation of vascular inflammation, senescence, TAA formation and survival in mice. The findings reveal that SIRT6 protects against TAA by epigenetically inhibiting vascular inflammation and senescence, providing insight into potential epigenetic strategies for TAA treatment.
Collapse
Affiliation(s)
- Yang-Nan Ding
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting-Ting Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuang-Jie Lv
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children, Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi-Yu Wei
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han-Shi Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Nan Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Man Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui-Yu Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - He-Ping Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhu-Qin Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiang Zhao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - De-Long Hao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Hong Sun
- Center for Experimental Animal Research, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Beijing, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, China.
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, China.
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
15
|
Chen M, Cavinato C, Hansen J, Tanaka K, Ren P, Hassab A, Li DS, Youshao E, Tellides G, Iyengar R, Humphrey JD, Schwartz MA. FN (Fibronectin)-Integrin α5 Signaling Promotes Thoracic Aortic Aneurysm in a Mouse Model of Marfan Syndrome. Arterioscler Thromb Vasc Biol 2023; 43:e132-e150. [PMID: 36994727 PMCID: PMC10133209 DOI: 10.1161/atvbaha.123.319120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 03/20/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Marfan syndrome, caused by mutations in the gene for fibrillin-1, leads to thoracic aortic aneurysms (TAAs). Phenotypic modulation of vascular smooth muscle cells (SMCs) and ECM (extracellular matrix) remodeling are characteristic of both nonsyndromic and Marfan aneurysms. The ECM protein FN (fibronectin) is elevated in the tunica media of TAAs and amplifies inflammatory signaling in endothelial and SMCs through its main receptor, integrin α5β1. We investigated the role of integrin α5-specific signals in Marfan mice in which the cytoplasmic domain of integrin α5 was replaced with that of integrin α2 (denoted α5/2 chimera). METHODS We crossed α5/2 chimeric mice with Fbn1mgR/mgR mice (mgR model of Marfan syndrome) to evaluate the survival rate and pathogenesis of TAAs among wild-type, α5/2, mgR, and α5/2 mgR mice. Further biochemical and microscopic analysis of porcine and mouse aortic SMCs investigated molecular mechanisms by which FN affects SMCs and subsequent development of TAAs. RESULTS FN was elevated in the thoracic aortas from Marfan patients, in nonsyndromic aneurysms, and in mgR mice. The α5/2 mutation greatly prolonged survival of Marfan mice, with improved elastic fiber integrity, mechanical properties, SMC density, and SMC contractile gene expression. Furthermore, plating of wild-type SMCs on FN decreased contractile gene expression and activated inflammatory pathways whereas α5/2 SMCs were resistant. These effects correlated with increased NF-kB activation in cultured SMCs and mgR aortas, which was alleviated by the α5/2 mutation or NF-kB inhibition. CONCLUSIONS FN-integrin α5 signaling is a significant driver of TAA in the mgR mouse model. This pathway thus warrants further investigation as a therapeutic target.
Collapse
Affiliation(s)
- Minghao Chen
- Cardiovascular Research Center (M.C., K.T., M.A.S.), Yale School of Medicine, New Haven, CT
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT (C.C., D.S.L., E.Y., J.D.H., M.A.S.)
| | - Jens Hansen
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York (J.H., R.I.)
| | - Keiichiro Tanaka
- Cardiovascular Research Center (M.C., K.T., M.A.S.), Yale School of Medicine, New Haven, CT
| | - Pengwei Ren
- Department of Surgery (P.R., A.H., G.T., M.A.S.), Yale School of Medicine, New Haven, CT
| | - Abdulrahman Hassab
- Department of Surgery (P.R., A.H., G.T., M.A.S.), Yale School of Medicine, New Haven, CT
| | - David S Li
- Department of Biomedical Engineering, Yale University, New Haven, CT (C.C., D.S.L., E.Y., J.D.H., M.A.S.)
| | - Eric Youshao
- Department of Biomedical Engineering, Yale University, New Haven, CT (C.C., D.S.L., E.Y., J.D.H., M.A.S.)
| | - George Tellides
- Department of Surgery (P.R., A.H., G.T., M.A.S.), Yale School of Medicine, New Haven, CT
- Vascular Biology and Therapeutics Program (G.T., J.D.H.), Yale School of Medicine, New Haven, CT
| | - Ravi Iyengar
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York (J.H., R.I.)
| | - Jay D Humphrey
- Vascular Biology and Therapeutics Program (G.T., J.D.H.), Yale School of Medicine, New Haven, CT
- Department of Biomedical Engineering, Yale University, New Haven, CT (C.C., D.S.L., E.Y., J.D.H., M.A.S.)
| | - Martin A Schwartz
- Cardiovascular Research Center (M.C., K.T., M.A.S.), Yale School of Medicine, New Haven, CT
- Department of Surgery (P.R., A.H., G.T., M.A.S.), Yale School of Medicine, New Haven, CT
- Departments of Medicine (Cardiology) and Cell Biology (M.A.S.), Yale School of Medicine, New Haven, CT
- Department of Biomedical Engineering, Yale University, New Haven, CT (C.C., D.S.L., E.Y., J.D.H., M.A.S.)
| |
Collapse
|
16
|
Zheng YK, Zhou ZS, Wang GZ, Tu JY, Cheng HB, Ma SZ, Ke C, Wang Y, Jian QP, Shu YH, Wu XW. MiR-122-5p regulates the mevalonate pathway by targeting p53 in non-small cell lung cancer. Cell Death Dis 2023; 14:234. [PMID: 37005437 PMCID: PMC10067850 DOI: 10.1038/s41419-023-05761-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
The 5-year survival rate of non-small cell lung cancer (NSCLC) patients is very low. MicroRNAs (miRNAs) are involved in the occurrence of NSCLC. miR-122-5p interacts with wild-type p53 (wtp53), and wtp53 affects tumor growth by inhibiting the mevalonate (MVA) pathway. Therefore, this study aimed to evaluate the role of these factors in NSCLC. The role of miR-122-5p and p53 was established in samples from NSCLC patients, and human NSCLC cells A549 using the miR-122-5p inhibitor, miR-122-5p mimic, and si-p53. Our results showed that inhibiting miR-122-5p expression led to the activation of p53. This inhibited the progression of the MVA pathway in the NSCLC cells A549, hindered cell proliferation and migration, and promoted apoptosis. miR-122-5p was negatively correlated with p53 expression in p53 wild-type NSCLC patients. The expression of key genes in the MVA pathway in tumors of p53 wild-type NSCLC patients was not always higher than the corresponding normal tissues. The malignancy of NSCLC was positively correlated with the high expression of the key genes in the MVA pathway. Therefore, miR-122-5p regulated NSCLC by targeting p53, providing potential molecular targets for developing targeted drugs.
Collapse
Affiliation(s)
- Yu-Kun Zheng
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Zhong-Shi Zhou
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
- Hubei Engineering Technology Research Center of Chinese Material Medical Processing Technology, Wuhan, 430065, China
| | - Guang-Zhong Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
- Hubei Engineering Technology Research Center of Chinese Material Medical Processing Technology, Wuhan, 430065, China
| | - Ji-Yuan Tu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
- Hubei Engineering Technology Research Center of Chinese Material Medical Processing Technology, Wuhan, 430065, China
| | - Huan-Bo Cheng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Shang-Zhi Ma
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Chang Ke
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yan Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Qi-Pan Jian
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yu-Hang Shu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiao-Wei Wu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
17
|
Lopez JL, Ramirez JL, Phu TA, Duong P, Bouchareychas L, Kuhrau CR, Lin PY, Eckalbar WL, Barczak AJ, Rudolph JD, Maliskova L, Conte MS, Vartanian SM, Raffai RL, Oskowitz AZ. Patients with abdominal aortic aneurysms have reduced levels of microRNA 122-5p in circulating exosomes. PLoS One 2023; 18:e0281371. [PMID: 36787323 PMCID: PMC9928131 DOI: 10.1371/journal.pone.0281371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 01/20/2023] [Indexed: 02/15/2023] Open
Abstract
OBJECTIVE There are currently no specific biomarkers to identify patients with abdominal aortic aneurysms (AAAs). Circulating exosomes contain microRNAs (miRNA) that are potential biomarkers for the presence of disease. This study aimed to characterize the exosomal miRNA expression profile of patients with AAAs in order to identify novel biomarkers of disease. METHODS Patients undergoing duplex ultrasound (US) or computed tomography (CT) for screening or surveillance of an AAA were screened to participate in the study. Cases with AAA were defined as having a max aortic diameter >3 cm. Circulating plasma exosomes were isolated using Cushioned-Density Gradient Ultracentrifugation and total RNA was extracted. Next Generation Sequencing was performed on the Illumina HiSeq4000 SE50. Differential miRNA expression analysis was performed using DESeq2 software with a Benjamini-Hochberg correction. MicroRNA expression profiles were validated by Quantitative Real-Time PCR. RESULTS A total of 109 patients were screened to participate in the study. Eleven patients with AAA and 15 non-aneurysmal controls met study criteria and were enrolled. Ultrasound measured aortic diameter was significantly larger in the AAA group (mean maximum diameter 4.3 vs 2.0 cm, P = 6.45x10-6). More AAA patients had coronary artery disease (5/11 vs 1/15, P = 0.05) as compared to controls, but the groups did not differ significantly in the rates of peripheral arterial disease and chronic obstructive pulmonary disease. A total of 40 miRNAs were differentially expressed (P<0.05). Of these, 18 miRNAs were downregulated and 22 were upregulated in the AAA group compared to controls. After false discovery rate (FDR) adjustment, only miR-122-5p was expressed at significantly different levels in the AAA group compared to controls (fold change = 5.03 controls vs AAA; raw P = 1.8x10-5; FDR P = 0.02). CONCLUSION Plasma exosomes from AAA patients have significantly reduced levels of miRNA-122-5p compared to controls. This is a novel exosome-associated miRNA that warrants further investigation to determine its use as a diagnostic biomarker and potential implications in AAA pathogenesis.
Collapse
Affiliation(s)
- Jose L. Lopez
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Joel L. Ramirez
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
| | - Tuan Anh Phu
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Veterans Affairs, Surgical Service (112G), San Francisco, California, United States of America
| | - Phat Duong
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Veterans Affairs, Surgical Service (112G), San Francisco, California, United States of America
| | - Laura Bouchareychas
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Veterans Affairs, Surgical Service (112G), San Francisco, California, United States of America
| | - Christina R. Kuhrau
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Pei-Yu Lin
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Walter L. Eckalbar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCSF CoLabs, University of California San Francisco, San Francisco, California, United States of America
| | - Andrea J. Barczak
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCSF CoLabs, University of California San Francisco, San Francisco, California, United States of America
| | - Joshua D. Rudolph
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCSF CoLabs, University of California San Francisco, San Francisco, California, United States of America
| | - Lenka Maliskova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCSF CoLabs, University of California San Francisco, San Francisco, California, United States of America
| | - Michael S. Conte
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Shant M. Vartanian
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Robert L. Raffai
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Veterans Affairs, Surgical Service (112G), San Francisco, California, United States of America
| | - Adam Z. Oskowitz
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|