1
|
Ma B, Zeng Q, Yang F, Yang H, Li W, Fu R, Cai Z, Zhu G, Shu C, Luo M, Zhou Z. MMP19 in vascular smooth muscle cells protects against thoracic aortic aneurysm and dissection via the MMP19/Aggrecan/Wnt/β-catenin axis. J Mol Cell Cardiol 2025; 202:35-49. [PMID: 39954937 DOI: 10.1016/j.yjmcc.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Thoracic aortic aneurysm and dissection (TAAD) is a life-threatening cardiovascular event characterized by high mortality rates. Previous studies have shown that matrix metalloproteinases 19 (MMP19) was involved in TAAD formation, while the detailed role of MMP19 in TAAD pathogenesis and underlying mechanism remain unclear. METHODS To investigate the role of MMP19 in the progression of TAAD, we generated global Mmp19 knockout mice, as well as VSMCs (vascular smooth muscle cells)-specific Mmp19 knockdown mice, and established a BAPN-induced TAAD model. To elucidate the signaling pathways modulated by Aggrecan, we employed an adeno-associated virus serotype 9 (AAV9) vector encoding Acan short hairpin RNA (shRNA) for VSMC-specific knockdown of Acan. Ultimately, we injected an AAV vector encoding VSMC-specific Mmp19 into BAPN-induced TAAD mice to assess whether MMP19 can mitigate the development of TAAD. RESULTS Our findings revealed elevated mRNA and protein levels of MMP19 in the aortas of both TAAD mice and patients. The systemic ablation of Mmp19, as well as VSMC-specific Mmp19 knockdown, significantly exacerbated BAPN-induced progressive TAAD, and TAAD-related cardiovascular remodeling. Mmp19 deficiency resulted in the accumulation of Acan, but not Vcan, within the aorta, driving the phenotypic switch of VSMCs from contractile to synthetic state through activting Wnt/β-catenin signaling pathway. The selective inhibitor of Wnt/β-catenin signaling, MASB, was effective in reversing the dedifferentiation of VSMCs induced by aggrecan accumulation. Notably, the specific knockdown of Acan in VSMCs restored the contractile phenotype of VSMCs and inhibited Wnt/β-catenin signaling, thereby alleviating BAPN-induced TAAD in Mmp19-/- mice. Additionally, VSMC-specific complementation of MMP19 also alleviated the progressive TAAD phenotype in Mmp19-/- mice. CONCLUSIONS The study underscores that MMP19 deficiency exacerbates TAAD by promoting Acan aggregation and destroying the homeostasis of VSMCs by activating Wnt/β-catenin signaling pathway. These results posit MMP19 as a promising novel therapeutic target for TAAD intervention.
Collapse
MESH Headings
- Humans
- Male
- Female
- Animals
- Mice
- Mice, Knockout
- Matrix Metalloproteinases, Secreted/genetics
- Matrix Metalloproteinases, Secreted/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/surgery
- Mice, Inbred C57BL
- Cells, Cultured
- Wnt Signaling Pathway
- Aggrecans/metabolism
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/surgery
- Myocytes, Smooth Muscle/metabolism
- beta Catenin/metabolism
- Disease Models, Animal
Collapse
Affiliation(s)
- Baihui Ma
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Qingyi Zeng
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China; Department of Medical Genetics/Prenatal Diagnostic Center, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Fangfang Yang
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Hang Yang
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Wenke Li
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Rui Fu
- Emergency Center, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Guoyan Zhu
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Chang Shu
- State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Mingyao Luo
- State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Department of Vascular Surgery, Central-China Branch of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China; Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650102, China.
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
2
|
Zhang J, Ryu JY, Tirado SR, Dickinson LD, Abosch A, Aziz-Sultan MA, Boulos AS, Barrow DL, Batjer HH, Binyamin TR, Blackburn SL, Chang EF, Chen PR, Colby GP, Cosgrove GR, David CA, Day AL, Folkerth RD, Frerichs KU, Howard BM, Jahromi BR, Niemela M, Ojemann SG, Patel NJ, Richardson RM, Shi X, Valle-Giler EP, Wang AC, Welch BG, Williams Z, Zusman EE, Weiss ST, Du R. A Transcriptomic Comparative Study of Cranial Vasculature. Transl Stroke Res 2024; 15:1108-1122. [PMID: 37612482 DOI: 10.1007/s12975-023-01186-w] [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: 07/06/2023] [Revised: 07/06/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023]
Abstract
In genetic studies of cerebrovascular diseases, the optimal vessels to use as controls remain unclear. Our goal is to compare the transcriptomic profiles among 3 different types of control vessels: superficial temporal artery (STA), middle cerebral arteries (MCA), and arteries from the circle of Willis obtained from autopsies (AU). We examined the transcriptomic profiles of STA, MCA, and AU using RNAseq. We also investigated the effects of using these control groups on the results of the comparisons between aneurysms and the control arteries. Our study showed that when comparing pathological cerebral arteries to control groups, all control groups presented similar responses in the activation of immunological processes, the regulation of intracellular signaling pathways, and extracellular matrix productions, despite their intrinsic biological differences. When compared to STA, AU exhibited upregulation of stress and apoptosis genes, whereas MCA showed upregulation of genes associated with tRNA/rRNA processing. Moreover, our results suggest that the matched case-control study design, which involves control STA samples collected from the same subjects of matched aneurysm samples in our study, can improve the identification of non-inherited disease-associated genes. Given the challenges associated with obtaining fresh intracranial arteries from healthy individuals, our study suggests that using MCA, AU, or paired STA samples as controls are feasible strategies for future large-scale studies investigating cerebral vasculopathies. However, the intrinsic differences of each type of control should be taken into consideration when interpreting the results. With the limitations of each control type, it may be most optimal to use multiple tissues as controls.
Collapse
Affiliation(s)
- Jianing Zhang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Jee-Yeon Ryu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Selena-Rae Tirado
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | | | - Aviva Abosch
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - M Ali Aziz-Sultan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Alan S Boulos
- Department of Neurosurgery, Albany Medical Center, Albany, NY, USA
| | - Daniel L Barrow
- Department of Neurosurgery, Emory University, Atlanta, GA, USA
| | - H Hunt Batjer
- Department of Neurosurgery, University of Texas Southwestern, Dallas, TX, USA
| | | | - Spiros L Blackburn
- Department of Neurosurgery, University of Texas Health Science Center, Houston, TX, USA
| | - Edward F Chang
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurosurgery, University of California Los Angeles, Los Angeles, CA, USA
| | - P Roc Chen
- Department of Neurosurgery, University of Texas Health Science Center, Houston, TX, USA
| | - Geoffrey P Colby
- Department of Neurosurgery, University of California Los Angeles, Los Angeles, CA, USA
| | - G Rees Cosgrove
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Carlos A David
- Department of Neurosurgery, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - Arthur L Day
- Department of Neurosurgery, University of Texas Health Science Center, Houston, TX, USA
| | - Rebecca D Folkerth
- Department of Forensic Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Kai U Frerichs
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Brian M Howard
- Department of Neurosurgery, Emory University, Atlanta, GA, USA
| | - Behnam R Jahromi
- Department of Neurosurgery, Helsinki University and Helsinki University Hospital, Helsinki, Finland
| | - Mika Niemela
- Department of Neurosurgery, Helsinki University and Helsinki University Hospital, Helsinki, Finland
| | - Steven G Ojemann
- Department of Neurosurgery, University of Colorado, Denver, CO, USA
| | - Nirav J Patel
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - R Mark Richardson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Xiangen Shi
- Department of Neurosurgery, Affiliated Fuxing Hospital, Capital Medical University, Beijing, China
| | | | - Anthony C Wang
- Department of Neurosurgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Babu G Welch
- Department of Neurosurgery, University of Texas Southwestern, Dallas, TX, USA
| | - Ziv Williams
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | | | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rose Du
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
3
|
Weiss D, Yeung N, Ramachandra AB, Humphrey JD. Transcriptional regulation of postnatal aortic development. Cells Dev 2024; 180:203971. [PMID: 39426523 PMCID: PMC11634634 DOI: 10.1016/j.cdev.2024.203971] [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: 06/24/2024] [Revised: 08/14/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
The aorta exhibits tremendous changes in geometry, composition, and mechanical properties during postnatal development. These changes are necessarily driven by transcriptional changes, both genetically programmed and mechano-responsive, but there has not been a careful comparison of time-course changes in the transcriptional profile and biomechanical phenotype. Here, we show that the greatest period of differential gene expression in the normal postnatal mouse aorta occurs prior to weaning at three weeks of age though with important evolution of many transcripts thereafter. We identify six general temporal patterns, including transcripts that monotonically decrease to lower or increase to higher steady state values as well as those that either peak or dip prior to or near weaning. We show that diverse transcripts within individual groupings correlate well over time, and that sub-sets of these groups correlate well with the developmental progression of different biomechanical metrics that are expected to be involved in mechano-sensing. In particular, expression of genes for elastin and elastin-associated glycoproteins tend to correlate well with the ratio of systolic-to-diastolic stress whereas genes for collagen fibers correlate well with the daily rate of change of systolic stress and genes for mechano-sensing proteins tend to correlate well with the systolic stress itself. We conclude that different groupings of genes having different temporal expression patterns correlate well with different measures of the wall mechanics, hence emphasizing a need for age-dependent, gene-specific computational modeling of postnatal development.
Collapse
Affiliation(s)
- D Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Department of Mechanical & Materials Engineering, University of Denver, Denver, CO, USA
| | - N Yeung
- School of the Biological Sciences, University of Cambridge, Cambridge, UK
| | - A B Ramachandra
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Department of Mechanical Engineering, Iowa State University, Ames, IA, USA
| | - J D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
4
|
Mills AC, Sandhu HK, Ikeno Y, Tanaka A. Heritable thoracic aortic disease: a literature review on genetic aortopathies and current surgical management. Gen Thorac Cardiovasc Surg 2024; 72:293-304. [PMID: 38480670 DOI: 10.1007/s11748-024-02017-x] [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: 12/23/2023] [Accepted: 02/09/2024] [Indexed: 04/16/2024]
Abstract
Heritable thoracic aortic disease puts patients at risk for aortic aneurysms, rupture, and dissections. The diagnosis and management of this heterogenous patient population continues to evolve. Last year, the American Heart Association/American College of Cardiology Joint Committee published diagnosis and management guidelines for aortic disease, which included those with genetic aortopathies. Additionally, evolving research studying the implications of underlying genetic aberrations with new genetic testing continues to become available. In this review, we evaluate the current literature surrounding the diagnosis and management of heritable thoracic aortic disease, as well as novel therapeutic approaches and future directions of research.
Collapse
Affiliation(s)
- Alexander C Mills
- Department of Cardiothoracic and Vascular Surgery, McGovern Medical School at UTHealth Houston, 6400 Fannin St., Ste. #2850, Houston, TX, 77030, USA
| | - Harleen K Sandhu
- Department of Cardiothoracic and Vascular Surgery, McGovern Medical School at UTHealth Houston, 6400 Fannin St., Ste. #2850, Houston, TX, 77030, USA
| | - Yuki Ikeno
- Department of Cardiothoracic and Vascular Surgery, McGovern Medical School at UTHealth Houston, 6400 Fannin St., Ste. #2850, Houston, TX, 77030, USA
| | - Akiko Tanaka
- Department of Cardiothoracic and Vascular Surgery, McGovern Medical School at UTHealth Houston, 6400 Fannin St., Ste. #2850, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Chen Z, Zhao Q, Chen L, Gao S, Meng L, Liu Y, Wang Y, Li T, Xue J. MAGP2 promotes osteogenic differentiation during fracture healing through its crosstalk with the β-catenin pathway. J Cell Physiol 2024; 239:e31183. [PMID: 38348695 DOI: 10.1002/jcp.31183] [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: 10/12/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 04/12/2024]
Abstract
Osteogenic differentiation is important for fracture healing. Microfibrial-associated glycoprotein 2 (MAGP2) is found to function as a proangiogenic regulator in bone formation; however, its role in osteogenic differentiation during bone repair is not clear. Here, a mouse model of critical-sized femur fracture was constructed, and the adenovirus expressing MAGP2 was delivered into the fracture site. Mice with MAGP2 overexpression exhibited increased bone mineral density and bone volume fraction (BV/TV) at Day 14 postfracture. Within 7 days postfracture, overexpression of MAGP2 increased collagen I and II expression at the fracture callus, with increasing chondrogenesis. MAGP2 inhibited collagen II level but elevated collagen I by 14 days following fracture, accompanied by increased endochondral bone formation. In mouse osteoblast precursor MC3T3-E1 cells, MAGP2 treatment elevated the expression of osteoblastic factors (osterix, BGLAP and collagen I) and enhanced ALP activity and mineralization through activating β-catenin signaling after osteogenic induction. Besides, MAGP2 could interact with lipoprotein receptor-related protein 5 (LRP5) and upregulated its expression. Promotion of osteogenic differentiation and β-catenin activation mediated by MAGP2 was partially reversed by LRP5 knockdown. Interestingly, β-catenin/transcription factor 4 (TCF4) increased MAGP2 expression probably by binding to MAGP2 promoter. These findings suggest that MAGP2 may interact with β-catenin/TCF4 to enhance β-catenin/TCF4's function and activate LRP5-activated β-catenin signaling pathway, thus promoting osteogenic differentiation for fracture repair. mRNA sequencing identified the potential targets of MAGP2, providing novel insights into MAGP2 function and the directions for future research.
Collapse
Affiliation(s)
- Zhiguang Chen
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qi Zhao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lianghong Chen
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Songlan Gao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lingshuai Meng
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yingjie Liu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yu Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
6
|
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
|
7
|
Jin Y, Li T, Wu S, Liu Z, Li Y. MFAP5 variant-induced multiple giant thoracic aortic aneurysm. Cardiol Young 2024; 34:212-217. [PMID: 38031457 DOI: 10.1017/s1047951123004122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Heritable thoracic aortic aneurysms are complex conditions characterised by the dilation or rupture of the thoracic aorta, often occurring as an autosomal-dominant disorder associated with life-threatening complications. In this case report, we present a de novo variant, MFAP5 c.236_237insA (p.N79Kfs9), which is implicated in the development of inherited thoracic aortic aneurysm. The proband, a 15-year-old male, presented with recurrent cough, dull chest pain, chest distress, vomiting, and reduced activity tolerance, leading to the diagnosis of heritable thoracic aortic aneurysms. Whole-exome sequencing identified a novel heterozygous variant in MFAP5 (NM_003480, c.236_237insA, and p.N79Kfs9). MutationTester and PolyPhen-s predicted this variant to be damaging and disease-causing (probability = 1), while the SFIT score indicated protein damage (0.001). Structural analysis using the AlphaFold Protein structure database revealed that this mutation disrupted the N-linked glycosylation site, resulting in a frameshift, amino acid sequence alteration, and truncation of an essential protein site. To our knowledge, this is the first case report describing a young patient with heritable thoracic aortic aneurysm carrying the novel MFAP5 c.236_237insA (p.N79Kfs*9) variant. This variant represents the third identified mutation site associated with heritable thoracic aortic aneurysm. Given the high mortality and morbidity rates associated with thoracic aortic aneurysms, the prevention of severe and fatal complications is crucial in the clinical management of this condition. Our case highlights the importance of whole-exome sequencing and genetic screening in identifying potential pathogenic or likely pathogenic variants, particularly in early-onset patients with aortic dilation, to inform appropriate management strategies.
Collapse
Affiliation(s)
- Yuxi Jin
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, SC, China
| | - Tiange Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, SC, China
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Shaoying Wu
- Department of Pediatrics, The Second People's Hospital of Liangshan Yi Autonomous Prefecture, Xichang, SC, China
| | - Zhongqiang Liu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, SC, China
| | - Yifei Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, SC, China
| |
Collapse
|
8
|
Gilly A, Park YC, Tsafantakis E, Karaleftheri M, Dedoussis G, Zeggini E. Genome-wide meta-analysis of 92 cardiometabolic protein serum levels. Mol Metab 2023; 78:101810. [PMID: 37778719 PMCID: PMC10582065 DOI: 10.1016/j.molmet.2023.101810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023] Open
Abstract
OBJECTIVES Global cardiometabolic disease prevalence has grown rapidly over the years, making it the leading cause of death worldwide. Proteins are crucial components in biological pathways dysregulated in disease states. Identifying genetic components that influence circulating protein levels may lead to the discovery of biomarkers for early stages of disease or offer opportunities as therapeutic targets. METHODS Here, we carry out a genome-wide association study (GWAS) utilising whole genome sequencing data in 3,005 individuals from the HELIC founder populations cohort, across 92 proteins of cardiometabolic relevance. RESULTS We report 322 protein quantitative trait loci (pQTL) signals across 92 proteins, of which 76 are located in or near the coding gene (cis-pQTL). We link those association signals with changes in protein expression and cardiometabolic disease risk using colocalisation and Mendelian randomisation (MR) analyses. CONCLUSIONS The majority of previously unknown signals we describe point to proteins or protein interactions involved in inflammation and immune response, providing genetic evidence for the contributing role of inflammation in cardiometabolic disease processes.
Collapse
Affiliation(s)
- Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Young-Chan Park
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | | | | | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens, Greece
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine, Munich, Germany.
| |
Collapse
|
9
|
Linssen EC, Demmers J, van Dijk CGM, van Dam R, Nicese MN, Cheng C, de Kort LMO, de Graaf P. Extracellular matrix analysis of fibrosis: A step towards tissue engineering for urethral stricture disease. PLoS One 2023; 18:e0294955. [PMID: 38032942 PMCID: PMC10688748 DOI: 10.1371/journal.pone.0294955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023] Open
Abstract
The urogenital tract is a target for many congenital and acquired diseases, both benign and oncogenic. In males, the urethra that transports urine and semen can be obstructed by a fibrotic disease called urethral stricture disease (USD). In severe USD, the whole organ including the vascular embedding, the corpus spongiosum (CS), is affected. Recurrent or severe USD is treated by reconstructive surgery. Tissue engineering may improve the outcome of urethral reconstruction in patients with complicated USD. Currently in urethral reconstruction only the epithelial layer is replaced, no substitution for the CS is provided, while the CS is important for mechanical support and vascularization. To develop a tissue engineering strategy for the CS, it is necessary to know the protein composition of the CS. As the extracellular matrix (ECM) plays an important role in the formation of fibrosis, we analyzed the distribution and localization of ECM components in human healthy and fibrotic CS tissue using immunohistology. The morphology of components of the elastic network were affected in USD. After decellularization a clear enrichment of proteins belonging to the ECM was found. In the proteomic analysis collagens COL15A1 and COL4A2 as well as inter-alpha-trypsin inhibitor ITIH4 were upregulated in fibrotic samples. The glycoproteins Periostin (POSTN), Microfibrillar-associated protein 5 (MFAP5) and EMILIN2 are downregulated in fibrotic tissue. To our knowledge this is the first proteomic study of ECM proteins of the CS in healthy and in USD. With these results a regenerating approach for tissue engineered CS can be developed, including relevant ECM proteins that reduce fibrosis and promote healthy healing in urethral reconstructive surgery.
Collapse
Affiliation(s)
- Emma C. Linssen
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeroen Demmers
- Department of Proteomics, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Roos van Dam
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maria Novella Nicese
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Cheng
- Department of Nephrology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Petra de Graaf
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
10
|
Zhang T, Li H, Sun S, Zhou W, Zhang T, Yu Y, Wang Q, Wang M. Microfibrillar-associated protein 5 suppresses adipogenesis by inhibiting essential coactivator of PPARγ. Sci Rep 2023; 13:5589. [PMID: 37020143 PMCID: PMC10076305 DOI: 10.1038/s41598-023-32868-y] [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: 01/12/2023] [Accepted: 04/04/2023] [Indexed: 04/07/2023] Open
Abstract
Femoral head necrosis is responsible for severe pain and its incidence is increasing. Abnormal adipogenic differentiation and fat cell hypertrophy of bone marrow mesenchymal stem cells increase intramedullary cavity pressure, leading to osteonecrosis. By analyzing gene expression before and after adipogenic differentiation, we found that Microfibril-Associated Protein 5 (MFAP5) is significantly down-regulated in adipogenesis whilst the mechanism of MFAP5 in regulating the differentiation of bone marrow mesenchymal stem cells is unknown. The purpose of this study was to clarify the role of MAFP5 in adipogenesis and therefore provide a theoretical basis for future therapeutic options of osteonecrosis. By knockdown or overexpression of MFAP5 in C3H10 and 3T3-L1 cells, we found that MFAP5 was significantly down-regulated as a key regulator of adipogenic differentiation, and identified the underlying downstream molecular mechanism. MFAP5 directly bound to and inhibited the expression of Staphylococcal Nuclease And Tudor Domain Containing 1, an essential coactivator of PPARγ, exerting an important regulatory role in adipogenesis.
Collapse
Affiliation(s)
- Tianlong Zhang
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, No128. Ruili Road, Minhang District, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
| | - Haoran Li
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, No128. Ruili Road, Minhang District, Shanghai, 200240, China
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Shiwei Sun
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, No128. Ruili Road, Minhang District, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
| | - Wuling Zhou
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, No128. Ruili Road, Minhang District, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
| | - Tieqi Zhang
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, No128. Ruili Road, Minhang District, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
| | - Yueming Yu
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, No128. Ruili Road, Minhang District, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
| | - Qiang Wang
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, No128. Ruili Road, Minhang District, Shanghai, 200240, China.
- Center of Community-Based Health Research, Fudan University, Shanghai, China.
| | - Minghai Wang
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, No128. Ruili Road, Minhang District, Shanghai, 200240, China.
- Center of Community-Based Health Research, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Korcari A, Nichols AEC, Buckley MR, Loiselle AE. Scleraxis-lineage cells are required for tendon homeostasis and their depletion induces an accelerated extracellular matrix aging phenotype. eLife 2023; 12:e84194. [PMID: 36656751 PMCID: PMC9908079 DOI: 10.7554/elife.84194] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
Aged tendons have disrupted homeostasis, increased injury risk, and impaired healing capacity. Understanding mechanisms of homeostatic disruption is crucial for developing therapeutics to retain tendon health through the lifespan. Here, we developed a novel model of accelerated tendon extracellular matrix (ECM) aging via depletion of Scleraxis-lineage cells in young mice (Scx-DTR). Scx-DTR recapitulates many aspects of tendon aging including comparable declines in cellularity, alterations in ECM structure, organization, and composition. Single-cell RNA sequencing demonstrated a conserved decline in tenocytes associated with ECM biosynthesis in aged and Scx-DTR tendons, identifying the requirement for Scleraxis-lineage cells during homeostasis. However, the remaining cells in aged and Scx-DTR tendons demonstrate functional divergence. Aged tenocytes become pro-inflammatory and lose proteostasis. In contrast, tenocytes from Scx-DTR tendons demonstrate enhanced remodeling capacity. Collectively, this study defines Scx-DTR as a novel model of accelerated tendon ECM aging and identifies novel biological intervention points to maintain tendon function through the lifespan.
Collapse
Affiliation(s)
- Antonion Korcari
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Anne EC Nichols
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
| | - Mark R Buckley
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Alayna E Loiselle
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| |
Collapse
|
12
|
Bax M, Romanov V, Junday K, Giannoulatou E, Martinac B, Kovacic JC, Liu R, Iismaa SE, Graham RM. Arterial dissections: Common features and new perspectives. Front Cardiovasc Med 2022; 9:1055862. [PMID: 36561772 PMCID: PMC9763901 DOI: 10.3389/fcvm.2022.1055862] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
Arterial dissections, which involve an abrupt tear in the wall of a major artery resulting in the intramural accumulation of blood, are a family of catastrophic disorders causing major, potentially fatal sequelae. Involving diverse vascular beds, including the aorta or coronary, cervical, pulmonary, and visceral arteries, each type of dissection is devastating in its own way. Traditionally they have been studied in isolation, rather than collectively, owing largely to the distinct clinical consequences of dissections in different anatomical locations - such as stroke, myocardial infarction, and renal failure. Here, we review the shared and unique features of these arteriopathies to provide a better understanding of this family of disorders. Arterial dissections occur commonly in the young to middle-aged, and often in conjunction with hypertension and/or migraine; the latter suggesting they are part of a generalized vasculopathy. Genetic studies as well as cellular and molecular investigations of arterial dissections reveal striking similarities between dissection types, particularly their pathophysiology, which includes the presence or absence of an intimal tear and vasa vasorum dysfunction as a cause of intramural hemorrhage. Pathway perturbations common to all types of dissections include disruption of TGF-β signaling, the extracellular matrix, the cytoskeleton or metabolism, as evidenced by the finding of mutations in critical genes regulating these processes, including LRP1, collagen genes, fibrillin and TGF-β receptors, or their coupled pathways. Perturbances in these connected signaling pathways contribute to phenotype switching in endothelial and vascular smooth muscle cells of the affected artery, in which their physiological quiescent state is lost and replaced by a proliferative activated phenotype. Of interest, dissections in various anatomical locations are associated with distinct sex and age predilections, suggesting involvement of gene and environment interactions in disease pathogenesis. Importantly, these cellular mechanisms are potentially therapeutically targetable. Consideration of arterial dissections as a collective pathology allows insight from the better characterized dissection types, such as that involving the thoracic aorta, to be leveraged to inform the less common forms of dissections, including the potential to apply known therapeutic interventions already clinically available for the former.
Collapse
Affiliation(s)
- Monique Bax
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Valentin Romanov
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Keerat Junday
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Eleni Giannoulatou
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Jason C. Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- St. Vincent’s Hospital, Darlinghurst, NSW, Australia
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, United States
| | - Renjing Liu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Siiri E. Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Robert M. Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- St. Vincent’s Hospital, Darlinghurst, NSW, Australia
| |
Collapse
|
13
|
Rodrigues Bento J, Meester J, Luyckx I, Peeters S, Verstraeten A, Loeys B. The Genetics and Typical Traits of Thoracic Aortic Aneurysm and Dissection. Annu Rev Genomics Hum Genet 2022; 23:223-253. [PMID: 36044906 DOI: 10.1146/annurev-genom-111521-104455] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Genetic predisposition and risk factors such as hypertension and smoking can instigate the development of thoracic aortic aneurysm (TAA), which can lead to highly lethal aortic wall dissection and/or rupture. Monogenic defects in multiple genes involved in the elastin-contractile unit and the TGFβ signaling pathway have been associated with TAA in recent years, along with several genetic modifiers and risk-conferring polymorphisms. Advances in omics technology have also provided significant insights into the processes behind aortic wall degeneration: inflammation, epigenetics, vascular smooth muscle phenotype change and depletion, reactive oxygen species generation, mitochondrial dysfunction, and angiotensin signaling dysregulation. These recent advances and findings might pave the way for a therapy that is capable of stopping and perhaps even reversing aneurysm progression.
Collapse
Affiliation(s)
- Jotte Rodrigues Bento
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium;
| | - Josephina Meester
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium;
| | - Ilse Luyckx
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; .,Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Silke Peeters
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium;
| | - Aline Verstraeten
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium;
| | - Bart Loeys
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; .,Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
14
|
Comparative Risks of Initial Aortic Events Associated With Genetic Thoracic Aortic Disease. J Am Coll Cardiol 2022; 80:857-869. [PMID: 36007983 DOI: 10.1016/j.jacc.2022.05.054] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/26/2022] [Accepted: 05/20/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pathogenic variants in 11 genes predispose individuals to heritable thoracic aortic disease (HTAD), but limited data are available to stratify the risk for aortic events associated with these genes. OBJECTIVES This study sought to compare the risk of first aortic event, specifically thoracic aortic aneurysm surgery or an aortic dissection, among 7 HTAD genes and variant types within each gene. METHODS A retrospective cohort of probands and relatives with rare variants in 7 genes for HTAD (n = 1,028) was assessed for the risk of first aortic events based on the gene altered, pathogenic variant type, sex, proband status, and location of recruitment. RESULTS Significant differences in aortic event risk were identified among the smooth muscle contraction genes (ACTA2, MYLK, and PRKG1; P = 0.002) and among the genes for Loeys-Dietz syndrome, which encode proteins in the transforming growth factor (TGF)-β pathway (SMAD3, TGFB2, TGFBR1, and TGFBR2;P < 0.0001). Cumulative incidence of type A aortic dissection was higher than elective aneurysm surgery in patients with variants in ACTA2, MYLK, PRKG1, and SMAD3; in contrast, patients with TGFBR2 variants had lower cumulative incidence of type A aortic dissection than elective aneurysm surgery. Cumulative incidence of type B aortic dissection was higher for ACTA2, PRKG1, and TGFBR2 than other genes. After adjusting for proband status, sex, and recruitment location, specific variants in ACTA2 and TGFBR2 were associated with substantially higher risk of aortic event with childhood onset. CONCLUSIONS Gene- and variant-specific data on aortic events in individuals with HTAD support personalized aortic surveillance and clinical management.
Collapse
|
15
|
Li Z, Zhao B, Qin C, Wang Y, Li T, Wang W. Chromatin Dynamics in Digestive System Cancer: Commander and Regulator. Front Oncol 2022; 12:935877. [PMID: 35965507 PMCID: PMC9372441 DOI: 10.3389/fonc.2022.935877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022] Open
Abstract
Digestive system tumors have a poor prognosis due to complex anatomy, insidious onset, challenges in early diagnosis, and chemoresistance. Epidemiological statistics has verified that digestive system tumors rank first in tumor-related death. Although a great number of studies are devoted to the molecular biological mechanism, early diagnostic markers, and application of new targeted drugs in digestive system tumors, the therapeutic effect is still not satisfactory. Epigenomic alterations including histone modification and chromatin remodeling are present in human cancers and are now known to cooperate with genetic changes to drive the cancer phenotype. Chromatin is the carrier of genetic information and consists of DNA, histones, non-histone proteins, and a small amount of RNA. Chromatin and nucleosomes control the stability of the eukaryotic genome and regulate DNA processes such as transcription, replication, and repair. The dynamic structure of chromatin plays a key role in this regulatory function. Structural fluctuations expose internal DNA and thus provide access to the nuclear machinery. The dynamic changes are affected by various complexes and epigenetic modifications. Variation of chromatin dynamics produces early and superior regulation of the expression of related genes and downstream pathways, thereby controlling tumor development. Intervention at the chromatin level can change the process of cancer earlier and is a feasible option for future tumor diagnosis and treatment. In this review, we introduced chromatin dynamics including chromatin remodeling, histone modifications, and chromatin accessibility, and current research on chromatin regulation in digestive system tumors was also summarized.
Collapse
|
16
|
Cheng P, Wirka RC, Kim JB, Kim HJ, Nguyen T, Kundu R, Zhao Q, Sharma D, Pedroza A, Nagao M, Iyer D, Fischbein MP, Quertermous T. Smad3 regulates smooth muscle cell fate and mediates adverse remodeling and calcification of the atherosclerotic plaque. NATURE CARDIOVASCULAR RESEARCH 2022; 1:322-333. [PMID: 36246779 PMCID: PMC9560061 DOI: 10.1038/s44161-022-00042-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 03/01/2022] [Indexed: 04/20/2023]
Abstract
Atherosclerotic plaques consist mostly of smooth muscle cells (SMC), and genes that influence SMC phenotype can modulate coronary artery disease (CAD) risk. Allelic variation at 15q22.33 has been identified by genome-wide association studies to modify the risk of CAD and is associated with the expression of SMAD3 in SMC. However, the mechanism by which this gene modifies CAD risk remains poorly understood. Here we show that SMC-specific deletion of Smad3 in a murine atherosclerosis model resulted in greater plaque burden, more outward remodelling and increased vascular calcification. Single-cell transcriptomic analyses revealed that loss of Smad3 altered SMC transition cell state toward two fates: a SMC phenotype that governs both vascular remodelling and recruitment of inflammatory cells, as well as a chondromyocyte fate. Together, the findings reveal that Smad3 expression in SMC inhibits the emergence of specific SMC phenotypic transition cells that mediate adverse plaque features, including outward remodelling, monocyte recruitment, and vascular calcification.
Collapse
Affiliation(s)
- Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Robert C. Wirka
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Juyong Brian Kim
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Hyun-Jung Kim
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Trieu Nguyen
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Ramendra Kundu
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Quanyi Zhao
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Disha Sharma
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Albert Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Manabu Nagao
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Dharini Iyer
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
17
|
Rombouts KB, van Merrienboer TAR, Ket JCF, Bogunovic N, van der Velden J, Yeung KK. The role of vascular smooth muscle cells in the development of aortic aneurysms and dissections. Eur J Clin Invest 2022; 52:e13697. [PMID: 34698377 PMCID: PMC9285394 DOI: 10.1111/eci.13697] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/12/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Aortic aneurysms (AA) are pathological dilations of the aorta, associated with an overall mortality rate up to 90% in case of rupture. In addition to dilation, the aortic layers can separate by a tear within the layers, defined as aortic dissections (AD). Vascular smooth muscle cells (vSMC) are the predominant cell type within the aortic wall and dysregulation of vSMC functions contributes to AA and AD development and progression. However, since the exact underlying mechanism is poorly understood, finding potential therapeutic targets for AA and AD is challenging and surgery remains the only treatment option. METHODS In this review, we summarize current knowledge about vSMC functions within the aortic wall and give an overview of how vSMC functions are altered in AA and AD pathogenesis, organized per anatomical location (abdominal or thoracic aorta). RESULTS Important functions of vSMC in healthy or diseased conditions are apoptosis, phenotypic switch, extracellular matrix regeneration and degradation, proliferation and contractility. Stressors within the aortic wall, including inflammatory cell infiltration and (epi)genetic changes, modulate vSMC functions and cause disturbance of processes within vSMC, such as changes in TGF-β signalling and regulatory RNA expression. CONCLUSION This review underscores a central role of vSMC dysfunction in abdominal and thoracic AA and AD development and progression. Further research focused on vSMC dysfunction in the aortic wall is necessary to find potential targets for noninvasive AA and AD treatment options.
Collapse
Affiliation(s)
- Karlijn B Rombouts
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Tara A R van Merrienboer
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | | | - Natalija Bogunovic
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands.,Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Update on the molecular landscape of thoracic aortic aneurysmal disease. Curr Opin Cardiol 2022; 37:201-211. [PMID: 35175228 DOI: 10.1097/hco.0000000000000954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF THE REVIEW Thoracic aortic aneurysms and dissections (TAADs) are a major health problem in the Western population. This review summarises recent discoveries in the genetic landscape of TAAD disease, discusses current challenges in clinical practice, and describes the molecular road ahead in TAAD research. Disorders, in which aneurysmal disease is not observed in the thoracic aorta, are not discussed. RECENT FINDINGS Current gene discovery studies have pinpointed about 40 genes associated with TAAD risk, accounting for about 30% of the patients. Importantly, novel genes, and their subsequent functional characterisation, have expanded the knowledge on disease-related pathways providing crucial information on key elements in this disease, and it pinpoints new therapeutic targets. Moreover, current molecular evidence also suggests the existence of less monogenic nature of TAAD disease, in which the presentation of a diseased patient is most likely influenced by a multitude of genetic and environmental factors. SUMMARY CLINICAL PRACTICE/RELEVANCE Ongoing molecular genetic research continues to expand our understanding on the pathomechanisms underlying TAAD disease in order to improve molecular diagnosis, optimise risk stratification, advance therapeutic strategies and facilitate counselling of TAAD patients and their families.
Collapse
|
19
|
Arnaud P, Mougin Z, Boileau C, Le Goff C. Cooperative Mechanism of ADAMTS/ ADAMTSL and Fibrillin-1 in the Marfan Syndrome and Acromelic Dysplasias. Front Genet 2021; 12:734718. [PMID: 34912367 PMCID: PMC8667168 DOI: 10.3389/fgene.2021.734718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/03/2021] [Indexed: 11/18/2022] Open
Abstract
The term “fibrillinopathies” gathers various diseases with a wide spectrum of clinical features and severity but all share mutations in the fibrillin genes. The first described fibrillinopathy, Marfan syndrome (MFS), is a multisystem disease with a unique combination of skeletal, thoracic aortic aneurysm (TAA) and ocular features. The numerous FBN1 mutations identified in MFS are located all along the gene, leading to the same pathogenic mechanism. The geleophysic/acromicric dysplasias (GD/AD), characterized by short stature, short extremities, and joint limitation are described as “the mirror image” of MFS. Previously, in GD/AD patients, we identified heterozygous FBN1 mutations all affecting TGFβ-binding protein-like domain 5 (TB5). ADAMTS10, ADAMTS17 and, ADAMTSL2 are also involved in the pathogenic mechanism of acromelic dysplasia. More recently, in TAA patients, we identified mutations in THSD4, encoding ADAMTSL6, a protein belonging to the ADAMTSL family suggesting that ADAMTSL proteins are also involved in the Marfanoid spectrum. Together with human genetic data and generated knockout mouse models targeting the involved genes, we provide herein an overview of the role of fibrillin-1 in opposite phenotypes. Finally, we will decipher the potential biological cooperation of ADAMTS-fibrillin-1 involved in these opposite phenotypes.
Collapse
Affiliation(s)
- Pauline Arnaud
- Université de Paris, INSERM U1148, Laboratory for Vascular Translational Science, Hôpital Bichat, Paris, France.,Département de Génétique, AP-HP, Hôpital Bichat, Paris, France
| | - Zakaria Mougin
- Université de Paris, INSERM U1148, Laboratory for Vascular Translational Science, Hôpital Bichat, Paris, France
| | - Catherine Boileau
- Université de Paris, INSERM U1148, Laboratory for Vascular Translational Science, Hôpital Bichat, Paris, France.,Département de Génétique, AP-HP, Hôpital Bichat, Paris, France
| | - Carine Le Goff
- Université de Paris, INSERM U1148, Laboratory for Vascular Translational Science, Hôpital Bichat, Paris, France
| |
Collapse
|
20
|
Abstract
Meester-Loeys syndrome is an X-linked form of syndromic thoracic aortic aneurysm, characterized by the involvement of multiple organ systems. More specifically, the cardiovascular, skeletal, craniofacial, cutaneous and neurological systems are affected. Clear clinical overlap with Marfan syndrome and Loeys-Dietz syndrome is observed. Aortic dissections occur typically at young ages and are most often observed in males. Meester-Loeys syndrome is caused by loss-of-function mutations in BGN, encoding the small leucine-rich proteoglycan biglycan. Although functional consequences of these mutations remain largely elusive, increased TGF-β signaling has been observed. Novel insights will provide opportunities for preventive therapeutic interventions.
Collapse
|
21
|
Connective Tissue Disorders and Cardiovascular Complications: The Indomitable Role of Transforming Growth Factor-β Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:161-184. [PMID: 34807419 DOI: 10.1007/978-3-030-80614-9_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Marfan Syndrome (MFS) and Loeys-Dietz Syndrome (LDS) represent heritable connective tissue disorders that segregate with a similar pattern of cardiovascular defects (thoracic aortic aneurysm, mitral valve prolapse/regurgitation, and aortic dilatation with regurgitation). This pattern of cardiovascular defects appears to be expressed along a spectrum of severity in many heritable connective tissue disorders and raises suspicion of a relationship between the normal development of connective tissues and the cardiovascular system. With overwhelming evidence of the involvement of aberrant Transforming Growth Factor-beta (TGF-β) signaling in MFS and LDS, this signaling pathway may represent the common link in the relationship between connective tissue disorders and their associated cardiovascular complications. To further explore this hypothetical link, this chapter will review the TGF-β signaling pathway, the heritable connective tissue syndromes related to aberrant TGF-β signaling, and will discuss the pathogenic contribution of TGF-β to these syndromes with a primary focus on the cardiovascular system.
Collapse
|
22
|
Yanagisawa H, Yokoyama U. Extracellular matrix-mediated remodeling and mechanotransduction in large vessels during development and disease. Cell Signal 2021; 86:110104. [PMID: 34339854 DOI: 10.1016/j.cellsig.2021.110104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/08/2023]
Abstract
The vascular extracellular matrix (ECM) is synthesized and secreted during embryogenesis and facilitates the growth and remodeling of large vessels. Proper interactions between the ECM and vascular cells are pivotal for building the vasculature required for postnatal dynamic circulation. The ECM serves as a structural component by maintaining the integrity of the vessel wall while also regulating intercellular signaling, which involves cytokines and growth factors. The major ECM component in large vessels is elastic fibers, which include elastin and microfibrils. Elastin is predominantly synthesized by vascular smooth muscle cells (SMCs) and uses microfibrils as a scaffold to lay down and assemble cross-linked elastin. The absence of elastin causes developmental defects that result in the subendothelial proliferation of SMCs and inward remodeling of the vessel wall. Notably, elastic fiber formation is attenuated in the ductus arteriosus and umbilical arteries. These two vessels function during embryogenesis and close after birth via cellular proliferation, migration, and matrix accumulation. In dynamic postnatal mechano-environments, the elastic fibers in large vessels also serve an essential role in proper signal transduction as a component of elastin-contractile units. Disrupted mechanotransduction in SMCs leads to pathological conditions such as aortic aneurysms that exhibit outward remodeling. This review discusses the importance of the ECM-mainly the elastic fiber matrix-in large vessels during developmental remodeling and under pathological conditions. By dissecting the role of the ECM in large vessels, we aim to provide insights into the role of ECM-mediated signal transduction that can provide a basis for seeking new targets for intervention in vascular diseases.
Collapse
Affiliation(s)
- Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, The University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Japan.
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan.
| |
Collapse
|
23
|
Li Y, Fang M, Yang J, Yu C, Kuang J, Sun T, Fan R. Analysis of the contribution of 129 candidate genes to thoracic aortic aneurysm or dissection of a mixed cohort of sporadic and familial cases in South China. Am J Transl Res 2021; 13:4281-4295. [PMID: 34150014 PMCID: PMC8205813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/14/2021] [Indexed: 06/12/2023]
Abstract
Thoracic aortic aneurysm or dissection (TAAD) is a group of life-threatening complex diseases after symptomatic onset with genetic heterogeneity accounting for approximately 20% of cases. Previously, we identified 40 rare variants in 11 TAAD-related core genes among 70 TAAD patients by next-generation sequencing. In this study, we further analyzed the variants in the disease-causing genes in 129 cases of sporadic TAAD and 22 familial cases by whole-exome sequencing. A total of 116 variants in 47 TAAD-related genes were identified, 64.7% (75/116) of which occurred in sporadic TAAD without syndromes, and among these genes, FBN1 was the most common TAAD-related gene. Of the 26.7% (31/116) that were pathogenic or likely pathogenic, almost one third were from sporadic cases without syndromes involving FBN1, SMAD3, SMAD6, MYH11, TGFBR1, MYLK, LOX and LTBP3. Interestingly, the novel VUS (variant of uncertain significance) *879Glu in MCTP2 occurred in two unrelated probands with sporadic acute aortic dissection without a bicuspid aortic valve. Furthermore, more than one variant was detected in 24 patients, and 70.8% (17/24) occurred in sporadic cases. Younger individuals were more likely to carry P/LP (pathogenic or likely pathogenic) variants and harbor more variants. P/LP carriers seem to have a larger aortic diameter, lower D-dimer levels, and a shorter ICU length of stay but longer hospitalization time. In conclusion, we expanded the candidate gene profile of TAAD, especially for sporadic cases without syndromic features. VUSs need further clarification.
Collapse
Affiliation(s)
- Ying Li
- Department of Cardiovascular Surgery, Guangdong Provincial People’s Hospital, School of Medicine, South China University of TechnologyGuangzhou, China
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou, China
| | - Miaoxian Fang
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou, China
| | - Jue Yang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou, China
| | - Changjiang Yu
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou, China
| | - Juntao Kuang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou, China
- The Second School of Clinical Medicine, Southern Medical UniversityGuangzhou, China
| | - Tucheng Sun
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou, China
| | - Ruixin Fan
- Department of Cardiovascular Surgery, Guangdong Provincial People’s Hospital, School of Medicine, South China University of TechnologyGuangzhou, China
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou, China
| |
Collapse
|
24
|
Yap ZJ, Sharif M, Bashir M. Is there an immunogenomic difference between thoracic and abdominal aortic aneurysms? J Card Surg 2021; 36:1520-1530. [PMID: 33604952 DOI: 10.1111/jocs.15440] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND AIM Aortic aneurysms most commonly occur in the infra-renal and proximal thoracic regions. While generally asymptomatic, progressive aneurysmal dilation can become rapidly lethal when dissection or ruptures occurs, highlighting the need for more robust screening. Abdominal aortic aneurysm (AAA) is more prevalent compared to thoracic aortic aneurysm (TAA). The true incidence of TAA is underreported due to the absence of population screening and the silent nature of TAA. To achieve the optimum survival rate in aortic aneurysms, knowledge of natural course, genetic association, and surgical results are needed to be applied with adequate medical treatment and careful selection of patients for operation. The purpose of this paper is to provide a comprehensive review of the literature on natural history, immunology, and genetic differences between thoracic and AAAs. METHOD The literature was collected from OVID, SCOPUS, and PubMed. RESULTS (1) AAA expands faster than TAA. AAA expands at approximately 0.3-0.45 cm annually, depending on various factors (advancing age, diameter of aorta, smoking etc.). TAA expands up to 0.3 cm annually in a non-bicuspid aortic valve patient. (2) An increase in Matrix metallopeptidase 1, 2, 9, 12, 14 led to degrading extracellular matrix of the aortic vessel wall. This significantly contributed to the pathogenesis in AAA, whereas overactive Transforming growth factor-beta played a major role in the pathogenesis of TAA. CONCLUSION In the future, genetic testing may be the gold standard for tackling the geneticheterogeneity of aneurysms, therefore, identifying at-risk individuals developing TAA andAAA earlier.
Collapse
Affiliation(s)
- Zhi Jiun Yap
- Department of Anaesthetic, Dorset County Hospital, Dorset, England
| | - Monira Sharif
- Department of Molecular & Clinical Medicine, Ninewells Hospital and Medical School, Dundee, Scotland
| | - Mohamad Bashir
- Department of Emergency Medicine and Surgery, Royal Blackburn Teaching Hospital, Blackburn, England
| |
Collapse
|
25
|
Creamer TJ, Bramel EE, MacFarlane EG. Insights on the Pathogenesis of Aneurysm through the Study of Hereditary Aortopathies. Genes (Basel) 2021; 12:183. [PMID: 33514025 PMCID: PMC7912671 DOI: 10.3390/genes12020183] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Thoracic aortic aneurysms (TAA) are permanent and localized dilations of the aorta that predispose patients to a life-threatening risk of aortic dissection or rupture. The identification of pathogenic variants that cause hereditary forms of TAA has delineated fundamental molecular processes required to maintain aortic homeostasis. Vascular smooth muscle cells (VSMCs) elaborate and remodel the extracellular matrix (ECM) in response to mechanical and biochemical cues from their environment. Causal variants for hereditary forms of aneurysm compromise the function of gene products involved in the transmission or interpretation of these signals, initiating processes that eventually lead to degeneration and mechanical failure of the vessel. These include mutations that interfere with transduction of stimuli from the matrix to the actin-myosin cytoskeleton through integrins, and those that impair signaling pathways activated by transforming growth factor-β (TGF-β). In this review, we summarize the features of the healthy aortic wall, the major pathways involved in the modulation of VSMC phenotypes, and the basic molecular functions impaired by TAA-associated mutations. We also discuss how the heterogeneity and balance of adaptive and maladaptive responses to the initial genetic insult might contribute to disease.
Collapse
Affiliation(s)
- Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Predoctoral Training in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
26
|
Barallobre-Barreiro J, Loeys B, Mayr M, Rienks M, Verstraeten A, Kovacic JC. Extracellular Matrix in Vascular Disease, Part 2/4: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:2189-2203. [PMID: 32354385 DOI: 10.1016/j.jacc.2020.03.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 01/01/2023]
Abstract
Medium-sized and large arteries consist of 3 layers: the tunica intima, tunica media, and tunica adventitia. The tunica media accounts for the bulk of the vessel wall and is the chief determinant of mechanical compliance. It is primarily composed of circumferentially arranged layers of vascular smooth muscle cells that are separated by concentrically arranged elastic lamellae; a form of extracellular matrix (ECM). The tunica media is separated from the tunica intima and tunica adventitia, the innermost and outermost layers, respectively, by the internal and external elastic laminae. This second part of a 4-part JACC Focus Seminar discusses the contributions of the ECM to vascular homeostasis and pathology. Advances in genetics and proteomics approaches have fostered significant progress in our understanding of vascular ECM. This review highlights the important role of the ECM in vascular disease and the prospect of translating these discoveries into clinical disease biomarkers and potential future therapies.
Collapse
Affiliation(s)
| | - Bart Loeys
- Center for Medical Genetics, University of Antwerp/Antwerp University Hospital, Antwerp, Belgium; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, United Kingdom; The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Marieke Rienks
- King's British Heart Foundation Centre, King's College London, London, United Kingdom
| | - Aline Verstraeten
- Center for Medical Genetics, University of Antwerp/Antwerp University Hospital, Antwerp, Belgium
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
27
|
Li Y, Gao S, Han Y, Song L, Kong Y, Jiao Y, Huang S, Du J, Li Y. Variants of Focal Adhesion Scaffold Genes Cause Thoracic Aortic Aneurysm. Circ Res 2020; 128:8-23. [PMID: 33092471 DOI: 10.1161/circresaha.120.317361] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Thoracic aortic aneurysm (TAA) leads to substantial mortality worldwide. Familial and syndromic TAAs are highly correlated with genetics. However, the incidence of sporadic isolated TAA (iTAA) is much higher, and the genetic contribution is not yet clear. OBJECTIVE Here, we examined the genetic characteristics of sporadic iTAA. METHODS AND RESULTS We performed a genetic screen of 551 sporadic iTAA cases and 1071 controls via whole-exome sequencing. The prevalence of pathogenic mutations in known causal genes was 5.08% in the iTAA cohort. We selected 100 novel candidate genes using a strict strategy, and the suspected functional variants of these genes were significantly enriched in cases compared with controls and carried by 60.43% of patients. We found more severe phenotypes and a lower proportion of hypertension in cases with pathogenic mutations or suspected functional variants. Among the candidate genes, Testin (TES), which encodes a focal adhesion scaffold protein, was identified as a potential TAA causal gene, accounting for 4 patients with 2 missense variants in the LIM1 domain (c.751T>C encoding p.Y251H; c.838T>C encoding p.Y280H) and highly expressed in the aorta. The 2 variants led to a decrease in TES expression. The thoracic aorta was spontaneously dilated in the TesY249H knock-in and Tes-/- mice. Mechanistically, the p.Y249H variant or knockdown of TES led to the repression of vascular smooth muscle cell contraction genes and disturbed the vascular smooth muscle cell contractile phenotype. Interestingly, suspected functional variants of other focal adhesion scaffold genes, including TLN1 (Talin-1) and ZYX (zyxin), were also significantly enriched in patients with iTAA; moreover, their knockdown resulted in decreased contractility of vascular smooth muscle cells. CONCLUSIONS For the first time, this study revealed the genetic landscape across iTAA and showed that the focal adhesion scaffold genes are critical in the pathogenesis of iTAA.
Collapse
Affiliation(s)
- Yang Li
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Yingchun Han
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Li Song
- BGI Genomics, BGI-Shenzhen, China (Li Song)
| | - Yu Kong
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Yao Jiao
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Shan Huang
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| |
Collapse
|
28
|
Zhao L, Xu L, Hemmerich A, Ferguson NL, Guy CD, McCall SJ, Cardona DM, Westerhoff M, Pai RK, Xiao SY, Liu B, Green CL, Hart J, Zhang X. Reduced MFAP5 expression in stroma of gallbladder adenocarcinoma and its potential diagnostic utility. Virchows Arch 2020; 478:427-434. [PMID: 32895766 DOI: 10.1007/s00428-020-02925-2] [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: 04/18/2020] [Revised: 08/04/2020] [Accepted: 09/02/2020] [Indexed: 11/28/2022]
Abstract
The diagnosis of invasive adenocarcinoma of the gallbladder can sometimes be challenging. The presence of true desmoplastic reaction facilitates the diagnosis of invasion. However, desmoplasia-like changes can be observed in benign gallbladder conditions, and recognition of desmoplasia may be challenging based on morphology. In this study, we tested the expression pattern of microfibril-associated protein 5 (MFAP5), a promising immunohistochemical marker for desmoplasia, in benign gallbladders with desmoplasia-like reaction and gallbladders with invasive adenocarcinoma. We also evaluated the diagnostic utility of MFAP5 in challenging cases with an interobserver agreement study. The results showed that all benign cases retained intact/positive MFAP5 staining pattern in periglandular connective tissue, whereas 79.3% (23 out of 29) of cases of adenocarcinomas demonstrated diffuse and complete loss of MFAP5 staining in the tumor stroma. Interobserver agreement was improved by 2.66 times when images of MFAP5 immunohistochemistry were provided. In conclusion, MFAP5 expression is downregulated in the desmoplastic stroma of gallbladder adenocarcinoma and may provide a useful diagnostic marker in difficult cases.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Liyan Xu
- Department of Pathology, St. Luke's University Health Network, Bethlehem, PA, USA
| | - Amanda Hemmerich
- Department of Pathology, Foundation Medicine, Inc., Morrisville, NC, USA
| | - N Lynn Ferguson
- Department of Pathology, Foundation Medicine, Inc., Morrisville, NC, USA
| | - Cynthia D Guy
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Shannon J McCall
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Diana M Cardona
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Maria Westerhoff
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rish K Pai
- Department of Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Shu-Yuan Xiao
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Beiyu Liu
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Cynthia L Green
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - John Hart
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Xuefeng Zhang
- Department of Pathology, Duke University Medical Center, Durham, NC, USA. .,Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
29
|
Rohde S, Zafar MA, Ziganshin BA, Elefteriades JA. Thoracic aortic aneurysm gene dictionary. Asian Cardiovasc Thorac Ann 2020; 29:682-696. [PMID: 32689806 DOI: 10.1177/0218492320943800] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Thoracic aortic aneurysm is typically clinically silent, with a natural history of progressive enlargement until a potentially lethal complication such as rupture or dissection occurs. Underlying genetic predisposition strongly influences the risk of thoracic aortic aneurysm and dissection. Familial cases are more virulent, have a higher rate of aneurysm growth, and occur earlier in life. To date, over 30 genes have been associated with syndromic and non-syndromic thoracic aortic aneurysm and dissection. The causative genes and their specific variants help to predict the disease phenotype, including age at presentation, risk of dissection at small aortic sizes, and risk of other cardiovascular and systemic manifestations. This genetic "dictionary" is already a clinical reality, allowing us to personalize care based on specific causative mutations for a substantial proportion of these patients. Widespread genetic sequencing of thoracic aortic aneurysm and dissection patients has been and continues to be crucial to the rapid expansion of this dictionary and ultimately, the delivery of truly personalized care to every patient.
Collapse
Affiliation(s)
- Stefanie Rohde
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - Mohammad A Zafar
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - Bulat A Ziganshin
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA.,Department of Cardiovascular and Endovascular Surgery, Kazan State Medical University, Kazan, Russia
| | - John A Elefteriades
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
30
|
Zhu S, Ye L, Bennett S, Xu H, He D, Xu J. Molecular structure and function of microfibrillar-associated proteins in skeletal and metabolic disorders and cancers. J Cell Physiol 2020; 236:41-48. [PMID: 32572962 DOI: 10.1002/jcp.29893] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/16/2022]
Abstract
Microfibrillar-associated proteins (MFAPs) are extracellular matrix glycoproteins, which play a role in microfibril assembly, elastinogenesis, and tissue homeostasis. MFAPs consist of five subfamily members, including MFAP1, MFAP2, MFAP3, MFAP4, and MFAP5. Among these, MFAP2 and MFAP5 are most closely related, and exhibit very limited amino acid sequence homology with MFAP1, MFAP3, and MFAP4. Gene expression profiling analysis reveals that MFAP2, MFAP5, and MFAP4 are specifically expressed in osteoblastic like cells, whereas MFAP1 and MFAP3 are more ubiquitously expressed, indicative of their diverse role in the tropism of tissues. Molecular structural analysis shows that each MFAP family member has distinct features, and functional evidence reveals discrete purposes of individual MFAPs. Animal studies indicate that MFAP2-deficient mice exhibit progressive osteopenia with elevated receptor activator of NF-κB ligand (RANKL) expression, whereas MFAP5-deficient mice are neutropenic, and MFAP4-deficient mice displayed emphysema-like pathology and the impaired formation of neointimal hyperplasia. Emerging data also suggest that MFAPs are involved in cancer progression and fat metabolism. Further understanding of tissue-specific pathophysiology of MFAPs might offer potential novel therapeutic targets for related diseases, such as skeletal and metabolic disorders, and cancers.
Collapse
Affiliation(s)
- Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Lin Ye
- Department of Orthopaedic Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Samuel Bennett
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dengwei He
- Department of Orthopaedic Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| |
Collapse
|
31
|
Verstraeten A, Meester J, Peeters S, Mortier G, Loeys B. Chondrodysplasias and Aneurysmal Thoracic Aortopathy: An Emerging Tale of Molecular Intersection. Trends Mol Med 2020; 26:783-795. [PMID: 32507656 DOI: 10.1016/j.molmed.2020.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/03/2020] [Accepted: 05/13/2020] [Indexed: 12/16/2022]
Abstract
Although at first glance chondrodysplasia and aneurysmal thoracic aortopathy seem oddly dissimilar, recent lines of evidences indicate that they share molecular similarities. Chondrodysplasias are a group of skeletal disorders characterized by genetic defects in hyaline cartilage. Aneurysmal thoracic aortopathy is the pathological enlargement of the thoracic aorta due to wall weakness, along with its ensuing life-threatening complications (i.e., aortic dissection and/or rupture). Extracellular matrix dysregulation, abnormal TGF-β signaling, and, to a more limited extent, endoplasmic reticulum stress emerge as common disease processes. In this review we provide a comprehensive overview of the genetic and pathomechanistic overlap as well as of how these commonalities can guide treatment strategies for both disease entities.
Collapse
Affiliation(s)
- Aline Verstraeten
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium. @uantwerpen.be
| | - Josephina Meester
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Silke Peeters
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Geert Mortier
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Bart Loeys
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium; Department of Human Genetics, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
32
|
van Dijk CG, Louzao-Martinez L, van Mulligen E, Boermans B, Demmers JA, van den Bosch TP, Goumans MJ, Duncker DJ, Verhaar MC, Cheng C. Extracellular Matrix Analysis of Human Renal Arteries in Both Quiescent and Active Vascular State. Int J Mol Sci 2020; 21:E3905. [PMID: 32486169 PMCID: PMC7313045 DOI: 10.3390/ijms21113905] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 01/30/2023] Open
Abstract
In vascular tissue engineering strategies, the addition of vascular-specific extracellular matrix (ECM) components may better mimic the in vivo microenvironment and potentially enhance cell-matrix interactions and subsequent tissue growth. For this purpose, the exact composition of the human vascular ECM first needs to be fully characterized. Most research has focused on characterizing ECM components in mature vascular tissue; however, the developing fetal ECM matches the active environment required in vascular tissue engineering more closely. Consequently, we characterized the ECM protein composition of active (fetal) and quiescent (mature) renal arteries using a proteome analysis of decellularized tissue. The obtained human fetal renal artery ECM proteome dataset contains higher levels of 15 ECM proteins versus the mature renal artery ECM proteome, whereas 16 ECM proteins showed higher levels in the mature tissue compared to fetal. Elastic ECM proteins EMILIN1 and FBN1 are significantly enriched in fetal renal arteries and are mainly produced by cells of mesenchymal origin. We functionally tested the role of EMILIN1 and FBN1 by anchoring the ECM secreted by vascular smooth muscle cells (SMCs) to glass coverslips. This ECM layer was depleted from either EMILIN1 or FBN1 by using siRNA targeting of the SMCs. Cultured endothelial cells (ECs) on this modified ECM layer showed alterations on the transcriptome level of multiple pathways, especially the Rho GTPase controlled pathways. However, no significant alterations in adhesion, migration or proliferation were observed when ECs were cultured on EMILIN1- or FNB1-deficient ECM. To conclude, the proteome analysis identified unique ECM proteins involved in the embryonic development of renal arteries. Alterations in transcriptome levels of ECs cultured on EMILIN1- or FBN1-deficient ECM showed that these candidate proteins could affect the endothelial (regenerative) response.
Collapse
Affiliation(s)
- Christian G.M. van Dijk
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Laura Louzao-Martinez
- Center for Proteomics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands; (L.L.-M.); (J.A.A.D.)
| | - Elise van Mulligen
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Bart Boermans
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Jeroen A.A. Demmers
- Center for Proteomics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands; (L.L.-M.); (J.A.A.D.)
| | | | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands;
| | - Dirk J. Duncker
- Experimental Cardiology, Department of Cardiology, Thorax center, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands;
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Caroline Cheng
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
- Experimental Cardiology, Department of Cardiology, Thorax center, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands;
| |
Collapse
|
33
|
Liu B, Granville DJ, Golledge J, Kassiri Z. Pathogenic mechanisms and the potential of drug therapies for aortic aneurysm. Am J Physiol Heart Circ Physiol 2020; 318:H652-H670. [PMID: 32083977 PMCID: PMC7099451 DOI: 10.1152/ajpheart.00621.2019] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/14/2022]
Abstract
Aortic aneurysm is a permanent focal dilation of the aorta. It is usually an asymptomatic disease but can lead to sudden death due to aortic rupture. Aortic aneurysm-related mortalities are estimated at ∼200,000 deaths per year worldwide. Because no pharmacological treatment has been found to be effective so far, surgical repair remains the only treatment for aortic aneurysm. Aortic aneurysm results from changes in the aortic wall structure due to loss of smooth muscle cells and degradation of the extracellular matrix and can form in different regions of the aorta. Research over the past decade has identified novel contributors to aneurysm formation and progression. The present review provides an overview of cellular and noncellular factors as well as enzymes that process extracellular matrix and regulate cellular functions (e.g., matrix metalloproteinases, granzymes, and cathepsins) in the context of aneurysm pathogenesis. An update of clinical trials focusing on therapeutic strategies to slow abdominal aortic aneurysm growth and efforts underway to develop effective pharmacological treatments is also provided.
Collapse
Affiliation(s)
- Bo Liu
- University of Wisconsin, Madison, Department of Surgery, Madison Wisconsin
| | - David J Granville
- International Collaboration on Repair Discoveries Centre and University of British Columbia Centre for Heart Lung Innovation, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan Golledge
- The Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Department of Vascular and Endovascular Surgery, Townsville Hospital and Health Services, Townsville, Queensland, Australia
| | - Zamaneh Kassiri
- University of Alberta, Department of Physiology, Cardiovascular Research Center, Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| |
Collapse
|
34
|
Feifei W, Hui G, Ruiqiang Z, Qunxiang J, Yu'an X. MAGP2, a Component of Extracellular Matrix, Is Upregulated in Colorectal Cancer and Negatively Modulated by miR-200b-3p. Technol Cancer Res Treat 2020; 18:1533033819870777. [PMID: 31426719 PMCID: PMC6702771 DOI: 10.1177/1533033819870777] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Colorectal cancer is one of the leading causes of cancer-related death worldwide, but its mechanism has not been clarified clearly. Microfibrial-associated glycoprotein 2 is mainly located in extracellular matrix, and its role in colorectal cancer is obscure. Methods: Immunohistochemical staining and quantitative real-time polymerase chain reaction were used to compare the expression level of microfibrial-associated glycoprotein 2 in colorectal cancer tissues and adjacent tissues. Western blot was used to detect the expression of microfibrial-associated glycoprotein 2 in colorectal cancer cell lines and normal colonic epithelium cell line. Kaplan-Meier analysis and χ2 test were applied to evaluate the potential of microfibrial-associated glycoprotein 2 to function as cancer biomarker. Lentiviral transduction was used to induce microfibrial-associated glycoprotein 2 overexpression in HCT116 cells and NCM460 cells, followed by detecting cell proliferation, migration, and invasion. Quantitative real-time polymerase chain reaction was used to investigate the changes in downstream genes after microfibrial-associated glycoprotein 2 overexpression. Luciferase assay was conducted to validate whether miR-200b-3p can directly target microfibrial-associated glycoprotein 2. Results: We validated that microfibrial-associated glycoprotein 2 was upregulated in colorectal cancer samples and cells. We also demonstrated its upregulation was associated with several clinicopathologic features such as Dukes stage (P = .048), differentiation status (P = .034), and local lymphatic metastasis (P = .036) of patients with colorectal cancer, and its high expression indicated shorter overall survival of the patients. Microfibrial-associated glycoprotein 2 overexpression remarkably promoted cell proliferation and metastasis via regulating the downstream genes of Notch, including hes family bHLH transcription factor 1 (HES1), Slug, Snail, matrix metalloproteinase 2, matrix metalloproteinase 9, and Kruppel-like factor 4. We also identified miR-200b-3p as a posttranscriptional regulator of microfibrial-associated glycoprotein 2, which partly explain the high expression mechanism of microfibrial-associated glycoprotein 2 in cancer tissues. Conclusion: Microfibrial-associated glycoprotein 2, negatively modulated by miR-200b-3p, is an oncogene of colorectal cancer associated with patients’ prognosis. It may function as a potential biomarker and therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Wei Feifei
- 1 Department of Experimental Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Guo Hui
- 1 Department of Experimental Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Zhao Ruiqiang
- 2 Department of Biochemistry and Molecular Biology, Guangxi Medical University, Nanning, China
| | - Jiang Qunxiang
- 1 Department of Experimental Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Xie Yu'an
- 1 Department of Experimental Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
35
|
Chou EL, Lindsay ME. The genetics of aortopathies: Hereditary thoracic aortic aneurysms and dissections. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2020; 184:136-148. [PMID: 32034893 DOI: 10.1002/ajmg.c.31771] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 01/28/2020] [Indexed: 12/15/2022]
Abstract
Aortopathies encompass a variety of inherited and acquired pathologies that increase risk of life-threatening dissection or rupture. Identifying individuals with hereditary thoracic aortic aneurysm and dissection (HTAAD) for longitudinal monitoring, medical therapy, or elective and preventative repair is paramount to reduce risk of cardiovascular-related mortality and complications from dissection and rupture. Over the past couple of decades, pathogenic variants in numerous genes have been identified in relation to HTAAD. The genetic diagnosis can help stratify patient risk and provide guidance on medical treatment, timing of prophylactic surgical repair, as well as longitudinal surveillance and imaging. Implicated genes and their associated proteins have been found to act on a diverse variety of pathways, cells and structural components linked to transforming growth factor beta (TGF-β) signaling pathways, disruption of the vascular smooth muscle cell contractile apparatus, and primary disruption of extracellular matrix homeostasis. This review describes relevant genetic variants that may help identify and guide the management of hereditary thoracic aortic aneurysms and dissections.
Collapse
Affiliation(s)
- Elizabeth L Chou
- Division of Vascular Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Thoracic Aortic Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mark E Lindsay
- Thoracic Aortic Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Cardiovascular Genetics Program, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Pediatric Cardiology Division, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
36
|
Chiarini A, Dal Prà I, Faggian G, Armato U, Luciani GB. Maladaptive remodeling of pulmonary artery root autografts after Ross procedure: A proteomic study. J Thorac Cardiovasc Surg 2020; 159:621-632.e3. [DOI: 10.1016/j.jtcvs.2019.07.083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/15/2022]
|
37
|
Broekelmann TJ, Bodmer NK, Mecham RP. Identification of the growth factor-binding sequence in the extracellular matrix protein MAGP-1. J Biol Chem 2020; 295:2687-2697. [PMID: 31988245 DOI: 10.1074/jbc.ra119.010540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Microfibril-associated glycoprotein-1 (MAGP-1) is a component of vertebrate extracellular matrix (ECM) microfibrils that, together with the fibrillins, contributes to microfibril function. Many of the phenotypes associated with MAGP-1 gene inactivation are consistent with dysregulation of the transforming growth factor β (TGFβ)/bone morphogenetic protein (BMP) signaling system. We have previously shown that full-length MAGP-1 binds active TGFβ-1 and some BMPs. The work presented here further defines the growth factor-binding domain of MAGP-1. Using recombinant domains and synthetic peptides, along with surface plasmon resonance analysis to measure the kinetics of the MAGP-1-TGFβ-1 interaction, we localized the TGFβ- and BMP-binding site in MAGP-1 to a 19-amino acid-long, highly acidic sequence near the N terminus. This domain was specific for binding active, but not latent, TGFβ-1. Growth factor activity experiments revealed that TGFβ-1 retains signaling activity when complexed with MAGP-1. Furthermore, when bound to fibrillin, MAGP-1 retained the ability to interact with TGFβ-1, and active TGFβ-1 did not bind fibrillin in the absence of MAGP-1. The absence of MAGP was sufficient to raise the amount of total TGFβ stored in the ECM of cultured cells, suggesting that the MAGPs compete with the TGFβ large latent complex for binding to microfibrils. Together, these results indicate that MAGP-1 plays an active role in TGFβ signaling in the ECM.
Collapse
Affiliation(s)
- Thomas J Broekelmann
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Nicholas K Bodmer
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110.
| |
Collapse
|
38
|
Faggion Vinholo T, Brownstein AJ, Ziganshin BA, Zafar MA, Kuivaniemi H, Body SC, Bale AE, Elefteriades JA. Genes Associated with Thoracic Aortic Aneurysm and Dissection: 2019 Update and Clinical Implications. AORTA : OFFICIAL JOURNAL OF THE AORTIC INSTITUTE AT YALE-NEW HAVEN HOSPITAL 2019; 7:99-107. [PMID: 31842235 PMCID: PMC6914358 DOI: 10.1055/s-0039-3400233] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Thoracic aortic aneurysm is a typically silent disease characterized by a lethal natural history. Since the discovery of the familial nature of thoracic aortic aneurysm and dissection (TAAD) almost 2 decades ago, our understanding of the genetics of this disorder has undergone a transformative amplification. To date, at least 37 TAAD-causing genes have been identified and an estimated 30% of the patients with familial nonsyndromic TAAD harbor a pathogenic mutation in one of these genes. In this review, we present our yearly update summarizing the genes associated with TAAD and the ensuing clinical implications for surgical intervention. Molecular genetics will continue to bolster this burgeoning catalog of culprit genes, enabling the provision of personalized aortic care.
Collapse
Affiliation(s)
- Thais Faggion Vinholo
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut
| | - Adam J Brownstein
- Department of Medicine, Johns Hopkins Hospital and Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Bulat A Ziganshin
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut.,Department of Cardiovascular and Endovascular Surgery, Kazan State Medical University, Kazan, Russia
| | - Mohammad A Zafar
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut
| | - Helena Kuivaniemi
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, and Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Simon C Body
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Allen E Bale
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
| | - John A Elefteriades
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
39
|
Abstract
Dissections or ruptures of aortic aneurysms remain a leading cause of death in the developed world, with the majority of deaths being preventable if individuals at risk are identified and properly managed. Genetic variants predispose individuals to these aortic diseases. In the case of thoracic aortic aneurysm and dissections (thoracic aortic disease), genetic data can be used to identify some at-risk individuals and dictate management of the associated vascular disease. For abdominal aortic aneurysms, genetic associations have been identified, which provide insight on the molecular pathogenesis but cannot be used clinically yet to identify individuals at risk for abdominal aortic aneurysms. This compendium will discuss our current understanding of the genetic basis of thoracic aortic disease and abdominal aortic aneurysm disease. Although both diseases share several pathogenic similarities, including proteolytic elastic tissue degeneration and smooth muscle dysfunction, they also have several distinct differences, including population prevalence and modes of inheritance.
Collapse
Affiliation(s)
- Amélie Pinard
- From the Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School; University of Texas Health Science Center at Houston (A.P., D.M.M.)
| | - Gregory T Jones
- Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, New Zealand (G.T.J.)
| | - Dianna M Milewicz
- From the Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School; University of Texas Health Science Center at Houston (A.P., D.M.M.)
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW To review the literature and provide a summary of management of syndromic and nonsyndromic aortopathies. RECENT FINDINGS The number of newly identified genetic causes for aortopathies have continued to increase over the past 10 years. The number of reported individuals with most hereditary aneurysm genes is small but increasing with more publications focusing describing the natural history caused by each gene. SUMMARY Aortopathy can present as an isolated finding or present as part of a larger genetic syndrome. Advances in genetic testing technology has shed light on the increasing importance of molecular diagnostics in the evaluation and management of patients with hereditary aortic disease. Molecular diagnostics and family phenotyping can aide in the diagnosis and management of pediatric patients with aortic disease.
Collapse
Affiliation(s)
| | - Rocio T Moran
- Division of Genetics and Genomics, The MetroHealth System, Cleveland, Ohio, USA
| |
Collapse
|
41
|
Li JH, Zhu XX, Li FX, Huang CS, Huang XT, Wang JQ, Gao ZX, Li SJ, Xu QC, Zhao W, Yin XY. MFAP5 facilitates the aggressiveness of intrahepatic Cholangiocarcinoma by activating the Notch1 signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:476. [PMID: 31775892 PMCID: PMC6882185 DOI: 10.1186/s13046-019-1477-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/11/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is the second most common primary liver cancer. The dismal outcome of ICC patients is due to lack of early diagnosis, the aggressive biological behavior of ICC and the lack of effective therapeutic options. Early diagnosis and prognosis of ICC by non-invasive methods would be helpful in providing valuable information and developing effective treatment strategies. METHODS Expression of microfibrillar-associated protein 5 (MFAP5) in the serum of ICC patients was detected by ELISA. Human ICC specimens were immunostained by MFAP5 antibodies. The growth rate of human ICC cell lines treated with MFAP5 or MFAP5 shRNAs was examined by CCK8 and colony formation assays. Cell cycle analysis was performed with PI staining. The effect of MFAP5 inhibition was assessed by xenograft models in nude mice. RNA-seq and ATAC-seq analyses were used to dissect the molecular mechanism by which MFAP5 promoted ICC aggressiveness. RESULTS We identified MFAP5 as a biomarker for the diagnosis and prognosis of ICC. Upregulated MFAP5 is a common feature in aggressive ICC patients' tissues. Importantly, MFAP5 level in the serum of ICC patients and healthy individuals showed significant differential expression profiles. Furthermore, we showed that MFAP5 promoted ICC cell growth and G1 to S-phase transition. Using RNA-seq expression and ATAC-seq chromatin accessibility profiling of ICC cells with suppressed MFAP5 secretion, we showed that MFAP5 regulated the expression of genes involved in the Notch1 signaling pathway. Furthermore, FLI-06, a Notch signaling inhibitor, completely abolished the MFAP5-dependent transcriptional programs. CONCLUSIONS Raised MFAP5 serum level is useful for differentiating ICC patients from healthy individuals, and could be helpful in ICC diagnosis, prognosis and therapies.
Collapse
Affiliation(s)
- Jian-Hui Li
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiao-Xu Zhu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Fu-Xi Li
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China
| | - Chen-Song Huang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xi-Tai Huang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Jie-Qin Wang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhuo-Xing Gao
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China
| | - Shi-Jin Li
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Qiong-Cong Xu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Wei Zhao
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China.
| | - Xiao-Yu Yin
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
42
|
Hascoet S, Edouard T, Plaisancie J, Arnoult F, Milleron O, Stheneur C, Chevallier B, Zordan C, Odent S, Bal L, Faivre L, Leheup B, Dupuis-Girod S, Ruidavets JB, Acar P, Ferrieres J, Jondeau G, Dulac Y. Incidence of cardiovascular events and risk markers in a prospective study of children diagnosed with Marfan syndrome. Arch Cardiovasc Dis 2019; 113:40-49. [PMID: 31735609 DOI: 10.1016/j.acvd.2019.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 07/21/2019] [Accepted: 09/30/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Little is known about the incidence of cardiovascular events (CVEs) and their associated risk markers in children with Marfan syndrome (MFS). AIMS To assess the incidence of CVEs and determine risk markers in a cohort diagnosed with Marfan syndrome during childhood and followed for several years. METHODS From a French multicentre nationwide database, 462 patients with MFS diagnosed during childhood were included prospectively. Patients' files were screened for a period of 20 years (1993-2013). CVEs (e.g. death, aortic dissection, cardiac valve or aortic root surgery) were assessed during the prospective follow-up. RESULTS Median (interquartile range) age at the end of follow-up was 17.2 (11.1-21.3) years. CVEs were reported for 35 participants (7.6%; 95% confidence interval [CI] 5.3-10.4%). First CVEs were prophylactic aortic root surgery (n=29), aortic dissection (n=4; two aged <18 years) and death (n=2). Kaplan-Meier cumulative incidence of CVEs was 5.3% (95% CI 3.3-8.7%) during childhood (aged≤18 years) and 19.4% (95% CI 13.3-27.9%) at 25years of age. The cumulative rate of CVEs was higher in case of Valsalva sinus Z-score increase of≥0.1 per year (P=0.0003), maximal Valsalva sinus diameter growth speed ≥5mm per year (P=0.03), aortic regurgitation≥2 (P=0.0005) and maximal Valsalva sinus Z-score≥3 before 16 years of age (P<0.0001). In a multivariable Cox proportional analysis, the Valsalva sinus Z-score remained significantly related to outcome. Considering aortic root evolution, aortic regurgitation, age at diagnosis and beta-blocker therapy were related to Valsalva sinus Z-score evolution during follow-up. CONCLUSIONS CVEs in children with MFS are mainly related to prophylactic aortic root surgery. Aortic dissections are rarely observed in children. The Valsalva sinus Z-score is a strong indicator of subsequent CVEs in children with MFS. Attention to follow-up and beta-blocker observance may be warranted in high-risk children.
Collapse
Affiliation(s)
- Sebastien Hascoet
- Department of Paediatric Cardiology and Paediatric Endocrinology, Centre de Référence pour le Syndrome de Marfan et Apparentés, Hôpital des Enfants, CHU Toulouse, 31300 Toulouse, France; Department of Congenital Heart Diseases, Centre Constitutif Réseau M3C (Maladies Rares Cardiopathies Congénitales Complexes), Hôpital Marie Lannelongue, Paris-Sud Université, 92350 Plessis-Robinson, France.
| | - Thomas Edouard
- Department of Paediatric Cardiology and Paediatric Endocrinology, Centre de Référence pour le Syndrome de Marfan et Apparentés, Hôpital des Enfants, CHU Toulouse, 31300 Toulouse, France
| | - Julie Plaisancie
- Department of Paediatric Cardiology and Paediatric Endocrinology, Centre de Référence pour le Syndrome de Marfan et Apparentés, Hôpital des Enfants, CHU Toulouse, 31300 Toulouse, France
| | - Florence Arnoult
- Department of Cardiology, Centre de Référence pour le Syndrome de Marfan et Apparentés, Hôpital Bichat, AP-HP, 75018 Paris, France
| | - Olivier Milleron
- Department of Cardiology, Centre de Référence pour le Syndrome de Marfan et Apparentés, Hôpital Bichat, AP-HP, 75018 Paris, France; Inserm U1148, 75877 Paris, France
| | - Chantal Stheneur
- Department of Cardiology, Centre de Référence pour le Syndrome de Marfan et Apparentés, Hôpital Bichat, AP-HP, 75018 Paris, France; Inserm U1148, 75877 Paris, France; Department of Paediatrics, Hôpital Ambroise Paré, AP-HP, 92100 Boulogne-Billancourt, France
| | | | - Cécile Zordan
- Department of Clinical Genetics, Centre de Compétence pour le Syndrome de Marfan et Apparentés, CHU Bordeaux, 33000 Bordeaux, France
| | - Sylvie Odent
- Department of Clinical Genetics, Centre de Compétence pour le Syndrome de Marfan et Apparentés, Hôpital Sud CHU, 35200 Rennes, France
| | - Laurence Bal
- Department of Clinical Genetics and Aortic Diseases, Centre de Compétence pour le Syndrome de Marfan et Apparentés, Hôpital de la Timone, AP-HM, 13005 Marseille, France
| | - Laurence Faivre
- Department of Clinical Genetics, Centre de Compétence pour le Syndrome de Marfan et Apparentés, Hôpital d'enfants, CHU, Université de Bourgogne Franche Comté, 21070 Dijon, France
| | - Bruno Leheup
- Department of Clinical Genetics, Centre de Compétence pour le Syndrome de Marfan et Apparentés, Hôpital des Enfants Brabois, CHU Nancy, 54500 Vandoeuvre-lès-Nancy, France
| | - Sophie Dupuis-Girod
- Department of Clinical Genetics, Hospices Civils de Lyon, Centre de Compétence pour le Syndrome de Marfan et Apparentés, Hôpital Femme-Mère-Enfants, 69677 Bron, France
| | - Jean-Bernard Ruidavets
- Department of Epidemiology, 31073 Toulouse; Inserm/UPS UMR 1048, I2MC, 31432 Toulouse, France; CHU de Toulouse, 31059 Toulouse, France
| | - Philippe Acar
- Department of Paediatric Cardiology and Paediatric Endocrinology, Centre de Référence pour le Syndrome de Marfan et Apparentés, Hôpital des Enfants, CHU Toulouse, 31300 Toulouse, France
| | - Jean Ferrieres
- Department of Epidemiology, 31073 Toulouse; Inserm/UPS UMR 1048, I2MC, 31432 Toulouse, France; CHU de Toulouse, 31059 Toulouse, France
| | - Guillaume Jondeau
- Department of Cardiology, Centre de Référence pour le Syndrome de Marfan et Apparentés, Hôpital Bichat, AP-HP, 75018 Paris, France; Inserm U1148, 75877 Paris, France
| | - Yves Dulac
- Department of Paediatric Cardiology and Paediatric Endocrinology, Centre de Référence pour le Syndrome de Marfan et Apparentés, Hôpital des Enfants, CHU Toulouse, 31300 Toulouse, France
| |
Collapse
|
43
|
Harky A, Fan KS, Fan KH. The genetics and biomechanics of thoracic aortic diseases. VASCULAR BIOLOGY 2019; 1:R13-R25. [PMID: 32923967 PMCID: PMC7439919 DOI: 10.1530/vb-19-0027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/15/2019] [Indexed: 12/20/2022]
Abstract
Thoracic aortic aneurysms and aortic dissections (TAAD) are highly fatal emergencies within cardiothoracic surgery. With increasing age, thoracic aneurysms become more prevalent and pose an even greater threat when they develop into aortic dissections. Both diseases are multifactorial and are influenced by a multitude of physiological and biomechanical processes. Structural stability of aorta can be disrupted by genes, such as those for extracellular matrix and contractile protein, as well as telomere dysfunction, which leads to senescence of smooth muscle and endothelial cells. Biomechanical changes such as increased luminal pressure imposed by hypertension are also very prevalent and lead to structural instability. Furthermore, ageing is associated with a pro-inflammatory state that exacerbates degeneration of vessel wall, facilitating the development of both aortic aneurysms and aortic dissection. This literature review provides an overview of the aetiology and pathophysiology of both thoracic aneurysms and aortic dissections. With an improved understanding, new therapeutic targets may eventually be identified to facilitate treatment and prevention of these diseases.
Collapse
Affiliation(s)
- Amer Harky
- Department of Cardiothoracic Surgery, Liverpool Heart and Chest, Liverpool, UK
| | - Ka Siu Fan
- St. George's Medical School, University of London, London, UK
| | - Ka Hay Fan
- Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
44
|
Abstract
Mutations in extracellular matrix and smooth muscle cell contractile proteins predispose to thoracic aortic aneurysms in Marfan syndrome (MFS) and related disorders. These genetic alterations lead to a compromised extracellular matrix-smooth muscle cell contractile unit. The abnormal aortic tissue responds with defective mechanosensing under hemodynamic stress. Aberrant mechanosensing is associated with transforming growth factor-beta (TGF-β) hyperactivity, enhanced angiotensin-II (Ang-II) signaling, and perturbation of other cellular signaling pathways. The downstream consequences include enhanced proteolytic activity, expression of inflammatory cytokines and chemokines, infiltration of inflammatory cells in the aortic wall, vascular smooth muscle cell apoptosis, and medial degeneration. Mouse models highlight aortic inflammation as a contributing factor in the development of aortic aneurysms. Anti-inflammatory drugs and antioxidants can reduce aortic oxidative stress that prevents aggravation of aortic disease in MFS mice. Targeting TGF-β and Ang-II downstream signaling pathways such as ERK1/2, mTOR, PI3/Akt, P38/MAPK, and Rho kinase signaling attenuates disease pathogenesis. Aortic extracellular matrix degradation and medial degeneration were reduced upon inhibition of inflammatory cytokines and matrix metalloproteinases, but the latter lack specificity. Treating inflammation associated with aortic aneurysms in MFS and related disorders could prove to be beneficial in limiting disease pathogenesis.
Collapse
|
45
|
Saito H, Hayashi H, Ueda T, Mine T, Kumita SI. Changes in Aortic Wall Thickness at a Site of Entry Tear on Computed Tomography before Development of Acute Aortic Dissection. Ann Vasc Dis 2019; 12:379-384. [PMID: 31636750 PMCID: PMC6766757 DOI: 10.3400/avd.oa.19-00051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective: To determine if there are changes in the aortic wall before acute aortic dissection (AD) that can be observed on contrast-enhanced computed tomography (CECT). Materials and Methods: Twenty-two patients with AD who underwent CECT before developing AD were retrospectively identified and enrolled as the AD group. Twenty-five consecutive patients who underwent CECT and did not develop AD were enrolled as the control group. In the AD group, the site of entry tear was detected on CECT images; the aortic wall thickness at this site, defined as the dissection-related wall thickness (D-T), was then measured on CECT images acquired before AD. Moreover, the mean thickness of the ascending, thoracic descending, and abdominal aortic walls before AD was defined as the non-dissection-related wall thickness (non-D-T). In the control group, the aortic wall thickness was measured similarly and defined as the control wall thickness (C-T). The D-T, non-D-T, and C-T values were compared using one-way analysis of variance with the Games-Howell pairwise comparison test. Results: The D-T (2.17±0.75 mm) was significantly greater than the non-D-T (1.58±0.22 mm; P<.01) and C-T (1.53±0.15 mm; P<.01). Conclusion: The aortic wall may have become thicker prior to the onset of AD.
Collapse
Affiliation(s)
- Hidemasa Saito
- Department of Radiology, Nippon Medical School, Tokyo, Japan
| | | | - Tatsuo Ueda
- Department of Radiology, Nippon Medical School, Tokyo, Japan
| | - Takahiko Mine
- Department of Radiology, Nippon Medical School, Tokyo, Japan
| | | |
Collapse
|
46
|
Regulation of PDE5 expression in human aorta and thoracic aortic aneurysms. Sci Rep 2019; 9:12206. [PMID: 31434939 PMCID: PMC6704119 DOI: 10.1038/s41598-019-48432-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/25/2019] [Indexed: 01/05/2023] Open
Abstract
Aneurysms and dissections affecting thoracic aorta are associated with smooth muscle cell (SMC) dysfunction. NO/cGMP signaling pathway in smooth muscle cells has been shown to be affected in sporadic thoracic aortic aneurysms. We analyzed the mRNA levels of PDE5, a cGMP-hydrolyzing enzyme highly expressed in aortic SMCs, that regulates arterious vascular tone by lowering cGMP levels. We found that aortic tissue obtained from Marfan, tricuspid and bicuspid thoracic aneurysms expressed lower levels of PDE5 mRNA compared to control aortas. In particular, we found that affected aortas showed lower levels of all the PDE5A isoforms, compared to control aortas. Transfection of vascular SMCs (VSMCs) with NOTCH3 activated domain (NICD3) induced the expression of PDE5A1 and A3 protein isoforms, but not that of the corresponding mRNAs. VSMC stimulation with GSNO, a nitric oxide analogue or with 8-br-cGMP, but not with 8-br-cAMP, up-regulated PDE5 and NOTCH-3 protein levels, indicating a negative feedback loop to protect the arterial wall from excessive relaxation. Finally, we found that PDE5 is expressed early during human aorta development, suggesting that if loss of function mutations of PDE5 occur, they might potentially affect aortic wall development.
Collapse
|
47
|
Loss of microfibril-associated protein 5 (MFAP5) expression in colon cancer stroma. Virchows Arch 2019; 476:383-390. [DOI: 10.1007/s00428-019-02649-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/31/2019] [Accepted: 08/13/2019] [Indexed: 11/26/2022]
|
48
|
Sarret C, Ashkavand Z, Paules E, Dorboz I, Pediaditakis P, Sumner S, Eymard-Pierre E, Francannet C, Krupenko NI, Boespflug-Tanguy O, Krupenko SA. Deleterious mutations in ALDH1L2 suggest a novel cause for neuro-ichthyotic syndrome. NPJ Genom Med 2019; 4:17. [PMID: 31341639 PMCID: PMC6650503 DOI: 10.1038/s41525-019-0092-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/03/2019] [Indexed: 01/06/2023] Open
Abstract
Neuro-ichthyotic syndromes are a group of rare genetic diseases mainly associated with perturbations in lipid metabolism, intracellular vesicle trafficking, or glycoprotein synthesis. Here, we report a patient with a neuro-ichthyotic syndrome associated with deleterious mutations in the ALDH1L2 (aldehyde dehydrogenase 1 family member L2) gene encoding for mitochondrial 10-formyltetrahydrofolate dehydrogenase. Using fibroblast culture established from the ALDH1L2-deficient patient, we demonstrated that the enzyme loss impaired mitochondrial function affecting both mitochondrial morphology and the pool of metabolites relevant to β-oxidation of fatty acids. Cells lacking the enzyme had distorted mitochondria, accumulated acylcarnitine derivatives and Krebs cycle intermediates, and had lower ATP and increased ADP/AMP indicative of a low energy index. Re-expression of functional ALDH1L2 enzyme in deficient cells restored the mitochondrial morphology and the metabolic profile of fibroblasts from healthy individuals. Our study underscores the role of ALDH1L2 in the maintenance of mitochondrial integrity and energy balance of the cell, and suggests the loss of the enzyme as the cause of neuro-cutaneous disease.
Collapse
Affiliation(s)
- Catherine Sarret
- IGCNC, Institut Pascal, UMR CNRS-UCA-SIGMA, Aubière, France
- Department of Clinical Genetics and Medical Cytogenetics, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Zahra Ashkavand
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC USA
| | - Evan Paules
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC USA
- Department of Nutrition, University of North Carolina, Chapel Hill, NC USA
| | - Imen Dorboz
- INSERM UMR1141, DHU PROTECT, PARIS-DIDEROT, University Sorbonne Paris-Cite, Paris, France
| | - Peter Pediaditakis
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC USA
| | - Susan Sumner
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC USA
- Department of Nutrition, University of North Carolina, Chapel Hill, NC USA
| | - Eléonore Eymard-Pierre
- Department of Clinical Genetics and Medical Cytogenetics, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Christine Francannet
- Department of Clinical Genetics and Medical Cytogenetics, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Natalia I. Krupenko
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC USA
- Department of Nutrition, University of North Carolina, Chapel Hill, NC USA
| | - Odile Boespflug-Tanguy
- INSERM UMR1141, DHU PROTECT, PARIS-DIDEROT, University Sorbonne Paris-Cite, Paris, France
- Department of Child Neurology and Metabolic Disorders, LEUKOFRANCE, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sergey A. Krupenko
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC USA
- Department of Nutrition, University of North Carolina, Chapel Hill, NC USA
| |
Collapse
|
49
|
Michel JB, Jondeau G, Milewicz DM. From genetics to response to injury: vascular smooth muscle cells in aneurysms and dissections of the ascending aorta. Cardiovasc Res 2019; 114:578-589. [PMID: 29360940 DOI: 10.1093/cvr/cvy006] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/16/2018] [Indexed: 12/20/2022] Open
Abstract
Vascular smooth muscle cells (vSMCs) play a crucial role in both the pathogenesis of Aneurysms and Dissections of the ascending thoracic aorta (TAAD) in humans and in the associated adaptive compensatory responses, since thrombosis and inflammatory processes are absent in the majority of cases. Aneurysms and dissections share numerous characteristics, including aetiologies and histopathological alterations: vSMC disappearance, medial areas of mucoid degeneration, and extracellular matrix (ECM) breakdown. Three aetiologies predominate in TAAD in humans: (i) genetic causes in heritable familial forms, (ii) an association with bicuspid aortic valves, and (iii) a sporadic degenerative form linked to the aortic aging process. Genetic forms include mutations in vSMC genes encoding for molecules of the ECM or the TGF-β pathways, or participating in vSMC tone. On the other hand, aneurysms and dissections, whatever their aetiologies, are characterized by an increase in wall permeability leading to transmural advection of plasma proteins which could interact with vSMCs and ECM components. In this context, blood-borne plasminogen appears to play an important role, because its outward convection through the wall is increased in TAAD, and it could be converted to active plasmin at the vSMC membrane. Active plasmin can induce vSMC disappearance, proteolysis of adhesive proteins, activation of MMPs and release of TGF-β from its ECM storage sites. Conversely, vSMCs could respond to aneurysmal biomechanical and proteolytic injury by an epigenetic phenotypic switch, including constitutional overexpression and nuclear translocation of Smad2 and an increase in antiprotease and ECM protein synthesis. In contrast, such an epigenetic phenomenon is not observed in dissections. In this context, dysfunction of proteins involved in vSMC tone are interesting to study, particularly in interaction with plasma protein transport through the wall and TGF-β activation, to establish the relationship between these dysfunctions and ECM proteolysis.
Collapse
Affiliation(s)
- Jean-Baptiste Michel
- UMR 1148, Laboratory for Translational Vascular Science, Inserm and Paris 7- Denis Diderot University, Xavier Bichat Hospital, 75018 Paris, France
| | - Guillaume Jondeau
- UMR 1148, Laboratory for Translational Vascular Science, Inserm and Paris 7- Denis Diderot University, Xavier Bichat Hospital, 75018 Paris, France.,Cardiology Department, National Reference Center for Marfan Syndrome and Related Diseases, APHP Hopital Bichat, 75018 Paris
| | - Dianna M Milewicz
- Division of Medical Genetics, Department of Internal Medicine, University of Texas Medical School at Houston, Houston, TX 77030, USA
| |
Collapse
|
50
|
Chen Z, Yan X, Li K, Ling Y, Kang H. Stromal fibroblast-derived MFAP5 promotes the invasion and migration of breast cancer cells via Notch1/slug signaling. Clin Transl Oncol 2019; 22:522-531. [PMID: 31190277 DOI: 10.1007/s12094-019-02156-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The tumor microenvironment (TME) regulates tumor progression, and cancer-associated fibroblasts (CAFs) are the primary stromal components of the TME, with the potential to drive tumor metastasis via the secretion of paracrine factors, but the specific mechanisms driving this process have not been defined. METHODS Proteins secreted from CAFs and normal fibroblasts (NFs) were analyzed via proteomic analysis (fold change > 2, p < 0.05) to identify tumor-promoting proteins secreted by CAFs. RESULTS Proteomic analysis revealed that microfibrillar-associated protein 5 (MFAP5) is preferentially expressed and secreted by CAFs relative to NFs, which was confirmed by Western blotting and RT-qPCR. Transwell and wound healing assays confirmed that MFAP5 is secreted by CAFs, and drives the invasion and migration of MCF7 breast cancer cells. We further found that in MCF7 cells MFAP5 promoted epithelial-mesenchymal transition, activating Notch1 signaling and consequently upregulating NICD1 and slug. When Notch1 was knocked down in MCF7 cells, the ability of MFAP5 to promote invasion and migration decreased. CONCLUSION CAFs promote cancer cells invasion and migration via MFAP5 secretion and activation of the Notch1/slug signaling. These data highlight this pathway as a therapeutic target to disrupt tumor progression through the interference of CAF-tumor crosstalk.
Collapse
Affiliation(s)
- Z Chen
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - X Yan
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - K Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Y Ling
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - H Kang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|