1
|
Vacante F, Venkateshappa R, Htet M, Yan C, Wu JC. Generation of Marfan syndrome-specific induced pluripotent stem cells harboring FBN1 mutations. Stem Cell Res 2024; 80:103518. [PMID: 39096853 DOI: 10.1016/j.scr.2024.103518] [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: 07/21/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024] Open
Abstract
Marfan syndrome (MFS) is a hereditary condition caused by mutations in the FBN1 gene. Genetic mutations in the FBN1 locus impact the function of the encoded protein, Fibrillin 1, a structural molecule forming microfibrils found in the connective tissue. MFS patients develop severe cardiovascular complications including thoracic aortic aneurysm and aortic dissection, which predispose them to an enhanced risk of premature death. Here, we generated two induced pluripotent stem cell (iPSC) lines harboring mutations in the FBN1 gene (p.C1942C>A and c.1954 T>C), directly derived from MFS patients. We have shown that both iPSC lines displayed expression of pluripotency markers, normal karyotype and ability of trilineage differentiation, representing a valuable tool for the identification of new therapeutic strategies for intervening in this disease.
Collapse
Affiliation(s)
- Francesca Vacante
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ravichandra Venkateshappa
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Min Htet
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
2
|
Jiménez-Berríos GA, Vázquez-Folch SJ, Izquierdo N. Phenotypic Heterogeneity of Patients With Marfan Syndrome in Puerto Rico: A Case Series. Cureus 2024; 16:e68791. [PMID: 39376868 PMCID: PMC11456412 DOI: 10.7759/cureus.68791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2024] [Indexed: 10/09/2024] Open
Abstract
PURPOSE Previous studies have reported on the cardiovascular, ocular, and musculoskeletal findings in patients with Marfan syndrome (MFS). This study aims to report the ocular and genotypic findings in patients with the syndrome in Puerto Rico. PATIENTS AND METHODS A chart review of a cohort of patients with the syndrome from Puerto Rico was done. Patients were examined by at least one of the authors (NJI). Fibrillin-1 (FBN1) full gene sequencing was done to all patients (Laboratory for Molecular Medicine, Center for Genetics and Genomics, Cambridge, MA). This study was approved by the Institutional Review Board of the Universidad Central del Caribe (approval number: 2024-07). Results: Six patients aged 28-79 years were examined. There were seven female and three male patients. The average visual acuity was 0.49 and 0.52 in the right eye (OD) and left eye (OS), respectively. The average refraction (spherical equivalent) was -1.28 sph OD and -1.07 sph OS. The average intraocular pressure was 14 mmHg in both eyes (OU). A patient had a dislocated lens OD; a patient had lens dislocation OU; and a patient had prosthesis OD and aphakia OS. Upon optical coherence tomography (OCT), the retinal nerve fiber layer (RNFL) average was 75.86 µm OD and 81.85 µm OS; the average cup-to-disc (C/D) ratio was 0.41 and 0.35 in the right and left eye, respectively. Upon visual field testing, the average mean deviation (MD) was -6.27 dB OD and -8.55 dB OS. CONCLUSIONS Our findings underscore the significant phenotypic and genotypic heterogeneity of patients with MFS in Puerto Rico. The identification of several mutations in the FBN1 gene in the Puerto Rican population demonstrates the need for an up-to-date approach to diagnose and co-manage patients with the syndrome. This study contributes to a deeper understanding of the genetic heritage of patients with the syndrome and highlights the potential for personalized therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Natalio Izquierdo
- Department of Surgery, School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, PRI
| |
Collapse
|
3
|
Bettini A, Camelliti P, Stuckey DJ, Day RM. Injectable biodegradable microcarriers for iPSC expansion and cardiomyocyte differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404355. [PMID: 38900068 PMCID: PMC11348074 DOI: 10.1002/advs.202404355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/05/2024] [Indexed: 06/21/2024]
Abstract
Cell therapy is a potential novel treatment for cardiac regeneration and numerous studies have attempted to transplant cells to regenerate the myocardium lost during myocardial infarction. To date, only minimal improvements to cardiac function have been reported. This is likely to be the result of low cell retention and survival following transplantation. This study aimed to improve the delivery and engraftment of viable cells by using an injectable microcarrier that provides an implantable, biodegradable substrate for attachment and growth of cardiomyocytes derived from induced pluripotent stem cells (iPSC). We describe the fabrication and characterisation of Thermally Induced Phase Separation (TIPS) microcarriers and their surface modification to enable iPSC-derived cardiomyocyte attachment in xeno-free conditions is described. The selected formulation resulted in iPSC attachment, expansion, and retention of pluripotent phenotype. Differentiation of iPSC into cardiomyocytes on the microcarriers is investigated in comparison with culture on 2D tissue culture plastic surfaces. Microcarrier culture is shown to support culture of a mature cardiomyocyte phenotype, be compatible with injectable delivery, and reduce anoikis. The findings from this study demonstrate that TIPS microcarriers provide a supporting matrix for culturing iPSC and iPSC-derived cardiomyocytes in vitro and are suitable as an injectable cell-substrate for cardiac regeneration.
Collapse
Affiliation(s)
- Annalisa Bettini
- Centre for Advanced Biomedical Imaging, Division of MedicineUniversity College LondonLondonWC1E 6DDUK
- Centre for Precision Healthcare, Division of MedicineUniversity College LondonLondonWC1E 6JFUK
| | - Patrizia Camelliti
- School of Biosciences and MedicineUniversity of SurreyGuildfordSurreyGU2 7XHUK
| | - Daniel J. Stuckey
- Centre for Advanced Biomedical Imaging, Division of MedicineUniversity College LondonLondonWC1E 6DDUK
| | - Richard M. Day
- Centre for Precision Healthcare, Division of MedicineUniversity College LondonLondonWC1E 6JFUK
| |
Collapse
|
4
|
Kwartler CS, Pinelo JEE. Use of iPSC-Derived Smooth Muscle Cells to Model Physiology and Pathology. Arterioscler Thromb Vasc Biol 2024; 44:1523-1536. [PMID: 38695171 PMCID: PMC11209779 DOI: 10.1161/atvbaha.123.319703] [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] [Indexed: 06/28/2024]
Abstract
The implementation of human induced pluripotent stem cell (hiPSC) models has introduced an additional tool for identifying molecular mechanisms of disease that complement animal models. Patient-derived or CRISPR/Cas9-edited induced pluripotent stem cells differentiated into smooth muscle cells (SMCs) have been leveraged to discover novel mechanisms, screen potential therapeutic strategies, and model in vivo development. The field has evolved over almost 15 years of research using hiPSC-SMCs and has made significant strides toward overcoming initial challenges such as the lineage specificity of SMC phenotypes. However, challenges both specific (eg, the lack of specific markers to thoroughly validate hiPSC-SMCs) and general (eg, a lack of transparency and consensus around methodology in the field) remain. In this review, we highlight the recent successes and remaining challenges of the hiPSC-SMC model.
Collapse
Affiliation(s)
- Callie S. Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Jose Emiliano Esparza Pinelo
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
5
|
Biswas PK, Park J. Applications, challenges, and prospects of induced pluripotent stem cells for vascular disease. Mol Cells 2024; 47:100077. [PMID: 38825189 PMCID: PMC11260847 DOI: 10.1016/j.mocell.2024.100077] [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: 04/04/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024] Open
Abstract
Vascular disease, including heart disease, stroke, and peripheral arterial disease, is one of the leading causes of death and disability and represents a significant global health issue. Since the development of human induced pluripotent stem cells (hiPSCs) in 2007, hiPSCs have provided unique and tremendous opportunities for studying human pathophysiology, disease modeling, and drug discovery in the field of regenerative medicine. In this review, we discuss vascular physiology and related diseases, the current methods for generating vascular cells (eg, endothelial cells, smooth muscle cells, and pericytes) from hiPSCs, and describe the opportunities and challenges to the clinical applications of vascular organoids, tissue-engineered blood vessels, and vessels-on-a-chip. We then explore how hiPSCs can be used to study and treat inherited vascular diseases and discuss the current challenges and future prospects. In the future, it will be essential to develop vascularized organoids or tissues that can simultaneously undergo shear stress and cyclic stretching. This development will not only increase their maturity and function but also enable effective and innovative disease modeling and drug discovery.
Collapse
Affiliation(s)
- Polash Kumar Biswas
- Department of Physiology, College of Medicine, Hallym University, Chuncheon-si, Gangwon-do 24252, South Korea
| | - Jinkyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon-si, Gangwon-do 24252, South Korea; Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
6
|
Ganizada BH, Veltrop RJA, Akbulut AC, Koenen RR, Accord R, Lorusso R, Maessen JG, Reesink K, Bidar E, Schurgers LJ. Unveiling cellular and molecular aspects of ascending thoracic aortic aneurysms and dissections. Basic Res Cardiol 2024; 119:371-395. [PMID: 38700707 PMCID: PMC11143007 DOI: 10.1007/s00395-024-01053-1] [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: 02/05/2024] [Revised: 04/03/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024]
Abstract
Ascending thoracic aortic aneurysm (ATAA) remains a significant medical concern, with its asymptomatic nature posing diagnostic and monitoring challenges, thereby increasing the risk of aortic wall dissection and rupture. Current management of aortic repair relies on an aortic diameter threshold. However, this approach underestimates the complexity of aortic wall disease due to important knowledge gaps in understanding its underlying pathologic mechanisms.Since traditional risk factors cannot explain the initiation and progression of ATAA leading to dissection, local vascular factors such as extracellular matrix (ECM) and vascular smooth muscle cells (VSMCs) might harbor targets for early diagnosis and intervention. Derived from diverse embryonic lineages, VSMCs exhibit varied responses to genetic abnormalities that regulate their contractility. The transition of VSMCs into different phenotypes is an adaptive response to stress stimuli such as hemodynamic changes resulting from cardiovascular disease, aging, lifestyle, and genetic predisposition. Upon longer exposure to stress stimuli, VSMC phenotypic switching can instigate pathologic remodeling that contributes to the pathogenesis of ATAA.This review aims to illuminate the current understanding of cellular and molecular characteristics associated with ATAA and dissection, emphasizing the need for a more nuanced comprehension of the impaired ECM-VSMC network.
Collapse
MESH Headings
- Humans
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/physiopathology
- Aortic Dissection/pathology
- Aortic Dissection/genetics
- Aortic Dissection/metabolism
- Animals
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Vascular Remodeling
- Extracellular Matrix/pathology
- Extracellular Matrix/metabolism
- Phenotype
Collapse
Affiliation(s)
- Berta H Ganizada
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Rogier J A Veltrop
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Asim C Akbulut
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Ryan Accord
- Department of Cardiothoracic Surgery, Center for Congenital Heart Disease, University Medical Center Groningen, Groningen, The Netherlands
| | - Roberto Lorusso
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Jos G Maessen
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Koen Reesink
- Department of Biomedical Engineering, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Elham Bidar
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|
7
|
Yang Y, Feng H, Tang Y, Wang Z, Qiu P, Huang X, Chang L, Zhang J, Chen YE, Mizrak D, Yang B. Bioengineered vascular grafts with a pathogenic TGFBR1 variant model aneurysm formation in vivo and reveal underlying collagen defects. Sci Transl Med 2024; 16:eadg6298. [PMID: 38718134 PMCID: PMC11193908 DOI: 10.1126/scitranslmed.adg6298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/17/2024] [Indexed: 05/30/2024]
Abstract
Thoracic aortic aneurysm (TAA) is a life-threatening vascular disease frequently associated with underlying genetic causes. An inadequate understanding of human TAA pathogenesis highlights the need for better disease models. Here, we established a functional human TAA model in an animal host by combining human induced pluripotent stem cells (hiPSCs), bioengineered vascular grafts (BVGs), and gene editing. We generated BVGs from isogenic control hiPSC-derived vascular smooth muscle cells (SMCs) and mutant SMCs gene-edited to carry a Loeys-Dietz syndrome (LDS)-associated pathogenic variant (TGFBR1A230T). We also generated hiPSC-derived BVGs using cells from a patient with LDS (PatientA230T/+) and using genetically corrected cells (Patient+/+). Control and experimental BVGs were then implanted into the common carotid arteries of nude rats. The TGFBR1A230T variant led to impaired mechanical properties of BVGs, resulting in lower burst pressure and suture retention strength. BVGs carrying the variant dilated over time in vivo, resembling human TAA formation. Spatial transcriptomics profiling revealed defective expression of extracellular matrix (ECM) formation genes in PatientA230T/+ BVGs compared with Patient+/+ BVGs. Histological analysis and protein assays validated quantitative and qualitative ECM defects in PatientA230T/+ BVGs and patient tissue, including decreased collagen hydroxylation. SMC organization was also impaired in PatientA230T/+ BVGs as confirmed by vascular contraction testing. Silencing of collagen-modifying enzymes with small interfering RNAs reduced collagen proline hydroxylation in SMC-derived tissue constructs. These studies demonstrated the utility of BVGs to model human TAA formation in an animal host and highlighted the role of reduced collagen modifying enzyme activity in human TAA formation.
Collapse
MESH Headings
- Animals
- Humans
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Induced Pluripotent Stem Cells/metabolism
- Collagen/metabolism
- Blood Vessel Prosthesis
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Rats, Nude
- Disease Models, Animal
- Rats
- Bioengineering
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Gene Editing
- Loeys-Dietz Syndrome/genetics
- Loeys-Dietz Syndrome/pathology
- Male
Collapse
Affiliation(s)
- Ying Yang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hao Feng
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
- Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ying Tang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
- Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zhenguo Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ping Qiu
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xihua Huang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lin Chang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yuqing Eugene Chen
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dogukan Mizrak
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bo Yang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
8
|
Curry T, Barrameda ME, Thomas TC, Esfandiarei M. In vivo phenotypic vascular dysfunction extends beyond the aorta in a mouse model for fibrillin-1 (Fbn1) mutation. Sci Rep 2024; 14:5779. [PMID: 38461168 PMCID: PMC10924961 DOI: 10.1038/s41598-024-56438-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: 11/23/2023] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
In individuals with Marfan Syndrome (MFS), fibrillin-1 gene (FBN1) mutations can lead to vascular wall weakening and dysfunction. The experimental mouse model of MFS (Fbn1C1041G/+) has been advantageous in investigating MFS-associated life-threatening aortic aneurysms. It is well established that the MFS mouse model exhibits an accelerated-aging phenotype in elastic organs like the aorta, lung, and skin. However, the impact of Fbn1 mutations on the in vivo function and structure of various artery types with the consideration of sex and age, has not been adequately explored in real-time and a clinically relevant context. In this study, we investigate if Fbn1 mutation contributes to sex-dependent alterations in central and cerebral vascular function similar to phenotypic changes associated with normal aging in healthy control mice. In vivo ultrasound imaging of central and cerebral vasculature was performed in 6-month-old male and female MFS and C57BL/6 mice and sex-matched 12-month-old (middle-aged) healthy control mice. Our findings confirm aortic enlargement (aneurysm) and wall stiffness in MFS mice, but with exacerbation in male diameters. Coronary artery blood flow velocity (BFV) in diastole was not different but left pulmonary artery BFV was decreased in MFS and 12-month-old control mice regardless of sex. At 6 months of age, MFS male mice show decreased posterior cerebral artery BFV as compared to age-matched control males, with no difference observed between female cohorts. Reduced mitral valve early-filling velocities were indicated in MFS mice regardless of sex. Male MFS mice also demonstrated left ventricular hypertrophy. Overall, these results underscore the significance of biological sex in vascular function and structure in MFS mice, while highlighting a trend of pre-mature vascular aging phenotype in MFS mice that is comparable to phenotypes observed in older healthy controls. Furthermore, this research is a vital step in understanding MFS's broader implications and sets the stage for more in-depth future analyses, while providing data-driven preclinical justification for re-evaluating diagnostic approaches and therapeutic efficacy.
Collapse
Affiliation(s)
- T Curry
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA
| | - M E Barrameda
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, 19555 N 59th Ave., Glendale, AZ, 85308, USA
| | - T Currier Thomas
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, 19555 N 59th Ave., Glendale, AZ, 85308, USA.
- Arizona State University, Tempe, AZ, USA.
- Phoenix VA Health Care System, Phoenix, AZ, USA.
| | - M Esfandiarei
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, 19555 N 59th Ave., Glendale, AZ, 85308, USA.
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
9
|
Chi C, Roland TJ, Song K. Differentiation of Pluripotent Stem Cells for Disease Modeling: Learning from Heart Development. Pharmaceuticals (Basel) 2024; 17:337. [PMID: 38543122 PMCID: PMC10975450 DOI: 10.3390/ph17030337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 04/01/2024] Open
Abstract
Heart disease is a pressing public health problem and the leading cause of death worldwide. The heart is the first organ to gain function during embryogenesis in mammals. Heart development involves cell determination, expansion, migration, and crosstalk, which are orchestrated by numerous signaling pathways, such as the Wnt, TGF-β, IGF, and Retinoic acid signaling pathways. Human-induced pluripotent stem cell-based platforms are emerging as promising approaches for modeling heart disease in vitro. Understanding the signaling pathways that are essential for cardiac development has shed light on the molecular mechanisms of congenital heart defects and postnatal heart diseases, significantly advancing stem cell-based platforms to model heart diseases. This review summarizes signaling pathways that are crucial for heart development and discusses how these findings improve the strategies for modeling human heart disease in vitro.
Collapse
Affiliation(s)
- Congwu Chi
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Truman J. Roland
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Kunhua Song
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| |
Collapse
|
10
|
Jiao YC, Wang YX, Liu WZ, Xu JW, Zhao YY, Yan CZ, Liu FC. Advances in the differentiation of pluripotent stem cells into vascular cells. World J Stem Cells 2024; 16:137-150. [PMID: 38455095 PMCID: PMC10915963 DOI: 10.4252/wjsc.v16.i2.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/20/2023] [Accepted: 01/16/2024] [Indexed: 02/26/2024] Open
Abstract
Blood vessels constitute a closed pipe system distributed throughout the body, transporting blood from the heart to other organs and delivering metabolic waste products back to the lungs and kidneys. Changes in blood vessels are related to many disorders like stroke, myocardial infarction, aneurysm, and diabetes, which are important causes of death worldwide. Translational research for new approaches to disease modeling and effective treatment is needed due to the huge socio-economic burden on healthcare systems. Although mice or rats have been widely used, applying data from animal studies to human-specific vascular physiology and pathology is difficult. The rise of induced pluripotent stem cells (iPSCs) provides a reliable in vitro resource for disease modeling, regenerative medicine, and drug discovery because they carry all human genetic information and have the ability to directionally differentiate into any type of human cells. This review summarizes the latest progress from the establishment of iPSCs, the strategies for differentiating iPSCs into vascular cells, and the in vivo transplantation of these vascular derivatives. It also introduces the application of these technologies in disease modeling, drug screening, and regenerative medicine. Additionally, the application of high-tech tools, such as omics analysis and high-throughput sequencing, in this field is reviewed.
Collapse
Affiliation(s)
- Yi-Chang Jiao
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Ying-Xin Wang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Wen-Zhu Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Jing-Wen Xu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Yu-Ying Zhao
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Chuan-Zhu Yan
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao) of Shandong University, Qingdao 266103, Shandong Province, China
- Brain Science Research Institute, Shandong University, Jinan 250012, Shandong Province, China
| | - Fu-Chen Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Brain Science Research Institute, Shandong University, Jinan 250012, Shandong Province, China.
| |
Collapse
|
11
|
Singh AA, Shetty DK, Jacob AG, Bayraktar S, Sinha S. Understanding genomic medicine for thoracic aortic disease through the lens of induced pluripotent stem cells. Front Cardiovasc Med 2024; 11:1349548. [PMID: 38440211 PMCID: PMC10910110 DOI: 10.3389/fcvm.2024.1349548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/31/2024] [Indexed: 03/06/2024] Open
Abstract
Thoracic aortic disease (TAD) is often silent until a life-threatening complication occurs. However, genetic information can inform both identification and treatment at an early stage. Indeed, a diagnosis is important for personalised surveillance and intervention plans, as well as cascade screening of family members. Currently, only 20% of heritable TAD patients have a causative mutation identified and, consequently, further advances in genetic coverage are required to define the remaining molecular landscape. The rapid expansion of next generation sequencing technologies is providing a huge resource of genetic data, but a critical issue remains in functionally validating these findings. Induced pluripotent stem cells (iPSCs) are patient-derived, reprogrammed cell lines which allow mechanistic insights, complex modelling of genetic disease and a platform to study aortic genetic variants. This review will address the need for iPSCs as a frontline diagnostic tool to evaluate variants identified by genomic discovery studies and explore their evolving role in biological insight through to drug discovery.
Collapse
Affiliation(s)
| | | | | | | | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, United Kingdom
| |
Collapse
|
12
|
Caudal A, Snyder MP, Wu JC. Harnessing human genetics and stem cells for precision cardiovascular medicine. CELL GENOMICS 2024; 4:100445. [PMID: 38359791 PMCID: PMC10879032 DOI: 10.1016/j.xgen.2023.100445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/22/2023] [Accepted: 10/25/2023] [Indexed: 02/17/2024]
Abstract
Human induced pluripotent stem cell (iPSC) platforms are valuable for biomedical and pharmaceutical research by providing tissue-specific human cells that retain patients' genetic integrity and display disease phenotypes in a dish. Looking forward, combining iPSC phenotyping platforms with genomic and screening technologies will continue to pave new directions for precision medicine, including genetic prediction, visualization, and treatment of heart disease. This review summarizes the recent use of iPSC technology to unpack the influence of genetic variants in cardiovascular pathology. We focus on various state-of-the-art genomic tools for cardiovascular therapies-including the expansion of genetic toolkits for molecular interrogation, in vitro population studies, and function-based drug screening-and their current applications in patient- and genome-edited iPSC platforms that are heralding new avenues for cardiovascular research.
Collapse
Affiliation(s)
- Arianne Caudal
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Greenstone Biosciences, Palo Alto, CA 94304, USA.
| |
Collapse
|
13
|
Aalders J, Léger L, Van der Meeren L, Sinha S, Skirtach AG, De Backer J, van Hengel J. Three-dimensional co-culturing of stem cell-derived cardiomyocytes and cardiac fibroblasts reveals a role for both cell types in Marfan-related cardiomyopathy. Matrix Biol 2024; 126:14-24. [PMID: 38224822 DOI: 10.1016/j.matbio.2024.01.003] [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: 08/10/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
Pathogenic variants in the FBN1 gene, which encodes the extracellular matrix protein fibrillin-1, cause Marfan syndrome (MFS), which affects multiple organ systems, including the cardiovascular system. Myocardial dysfunction has been observed in a subset of patients with MFS and in several MFS mouse models. However, there is limited understanding of the intrinsic consequences of FBN1 variants on cardiomyocytes (CMs). To elucidate the CM-specific contribution in Marfan's cardiomyopathy, cardiosphere cultures of CMs and cardiac fibroblasts (CFs) are used. CMs and CFs were derived by human induced pluripotent stem cell (iPSC) differentiation from MFS iPSCs with a pathogenic variant in FBN1 (c.3725G>A; p.Cys1242Tyr) and the corresponding CRISPR-corrected iPSC line (Cor). Cardiospheres containing MFS CMs show decreased FBN1, COL1A2 and GJA1 expression. MFS CMs cultured in cardiospheres have fewer binucleated CMs in comparison with Cor CMs. 13% of MFS CMs in cardiospheres are binucleated and 15% and 16% in cardiospheres that contain co-cultures with respectively MFS CFs and Cor CFs, compared to Cor CMs, that revealed up to 23% binucleation when co-cultured with CFs. The sarcomere length of CMs, as a marker of development, is significantly increased in MFS CMs interacting with Cor CF or MFS CF, as compared to monocultured MFS CMs. Nuclear blebbing was significantly more frequent in MFS CFs, which correlated with increased stiffness of the nuclear area compared to Cor CFs. Our cardiosphere model for Marfan-related cardiomyopathy identified a contribution of CFs in Marfan-related cardiomyopathy and suggests that abnormal early development of CMs may play a role in the disease mechanism.
Collapse
Affiliation(s)
- Jeffrey Aalders
- Medical Cell Biology Research Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Laurens Léger
- Medical Cell Biology Research Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Louis Van der Meeren
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Andre G Skirtach
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Julie De Backer
- Centre for Medical Genetics, Ghent University Hospital, Belgium and Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Department of Cardiology, Ghent University Hospital, Ghent, Belgium
| | - Jolanda van Hengel
- Medical Cell Biology Research Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
14
|
Raghavan A, Pirruccello JP, Ellinor PT, Lindsay ME. Using Genomics to Identify Novel Therapeutic Targets for Aortic Disease. Arterioscler Thromb Vasc Biol 2024; 44:334-351. [PMID: 38095107 PMCID: PMC10843699 DOI: 10.1161/atvbaha.123.318771] [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: 06/26/2023] [Accepted: 11/21/2023] [Indexed: 01/04/2024]
Abstract
Aortic disease, including dissection, aneurysm, and rupture, carries significant morbidity and mortality and is a notable cause of sudden cardiac death. Much of our knowledge regarding the genetic basis of aortic disease has relied on the study of individuals with Mendelian aortopathies and, until recently, the genetic determinants of population-level variance in aortic phenotypes remained unclear. However, the application of machine learning methodologies to large imaging datasets has enabled researchers to rapidly define aortic traits and mine dozens of novel genetic associations for phenotypes such as aortic diameter and distensibility. In this review, we highlight the emerging potential of genomics for identifying causal genes and candidate drug targets for aortic disease. We describe how deep learning technologies have accelerated the pace of genetic discovery in this field. We then provide a blueprint for translating genetic associations to biological insights, reviewing techniques for locus and cell type prioritization, high-throughput functional screening, and disease modeling using cellular and animal models of aortic disease.
Collapse
Affiliation(s)
- Avanthi Raghavan
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - James P. Pirruccello
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Patrick T. Ellinor
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Mark E. Lindsay
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Li L, Huang J, Liu Y. The extracellular matrix glycoprotein fibrillin-1 in health and disease. Front Cell Dev Biol 2024; 11:1302285. [PMID: 38269088 PMCID: PMC10806136 DOI: 10.3389/fcell.2023.1302285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/19/2023] [Indexed: 01/26/2024] Open
Abstract
Fibrillin-1 (FBN1) is a large, cysteine-rich, calcium binding extracellular matrix glycoprotein encoded by FBN1 gene. It serves as a structural component of microfibrils and provides force-bearing mechanical support in elastic and nonelastic connective tissue. As such, mutations in the FBN1 gene can cause a wide variety of genetic diseases such as Marfan syndrome, an autosomal dominant disorder characterized by ocular, skeletal and cardiovascular abnormalities. FBN1 also interacts with numerous microfibril-associated proteins, growth factors and cell membrane receptors, thereby mediating a wide range of biological processes such as cell survival, proliferation, migration and differentiation. Dysregulation of FBN1 is involved in the pathogenesis of many human diseases, such as cancers, cardiovascular disorders and kidney diseases. Paradoxically, both depletion and overexpression of FBN1 upregulate the bioavailability and signal transduction of TGF-β via distinct mechanisms in different settings. In this review, we summarize the structure and expression of FBN1 and present our current understanding of the functional role of FBN1 in various human diseases. This knowledge will allow to develop better strategies for therapeutic intervention of FBN1 related diseases.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Junxin Huang
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
16
|
Kalyanaraman H, Casteel DE, Cabriales JA, Tat J, Zhuang S, Chan A, Dretchen KL, Boss GR, Pilz RB. The Antioxidant/Nitric Oxide-Quenching Agent Cobinamide Prevents Aortic Disease in a Mouse Model of Marfan Syndrome. JACC Basic Transl Sci 2024; 9:46-62. [PMID: 38362350 PMCID: PMC10864892 DOI: 10.1016/j.jacbts.2023.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 02/17/2024]
Abstract
Major pathologic changes in the proximal aorta underlie the life-threatening aortic aneurysms and dissections in Marfan Syndrome; current treatments delay aneurysm development without addressing the primary pathology. Because excess oxidative stress and nitric oxide/protein kinase G signaling likely contribute to the aortopathy, we hypothesized that cobinamide, a strong antioxidant that can attenuate nitric oxide signaling, could be uniquely suited to prevent aortic disease. In a well-characterized mouse model of Marfan Syndrome, cobinamide dramatically reduced elastin breaks, prevented excess collagen deposition and smooth muscle cell apoptosis, and blocked DNA, lipid, and protein oxidation and excess nitric oxide/protein kinase G signaling in the ascending aorta. Consistent with preventing pathologic changes, cobinamide diminished aortic root dilation without affecting blood pressure. Cobinamide exhibited excellent safety and pharmacokinetic profiles indicating it could be a practical treatment. We conclude that cobinamide deserves further study as a disease-modifying treatment of Marfan Syndrome.
Collapse
Affiliation(s)
- Hema Kalyanaraman
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Darren E. Casteel
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Justin A. Cabriales
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - John Tat
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Shunhui Zhuang
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Adriano Chan
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | | | - Gerry R. Boss
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Renate B. Pilz
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| |
Collapse
|
17
|
Zhang H, Wu JC. Deciphering Congenital Heart Disease Using Human Induced Pluripotent Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:239-252. [PMID: 38884715 DOI: 10.1007/978-3-031-44087-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Congenital heart disease (CHD) is a leading cause of birth defect-related death. Despite significant advances, the mechanisms underlying the development of CHD are complex and remain elusive due to a lack of efficient, reproducible, and translational model systems. Investigations relied on animal models have inherent limitations due to interspecies differences. Human induced pluripotent stem cells (iPSCs) have emerged as an effective platform for disease modeling. iPSCs allow for the production of a limitless supply of patient-specific somatic cells that enable advancement in cardiovascular precision medicine. Over the past decade, researchers have developed protocols to differentiate iPSCs to multiple cardiovascular lineages, as well as to enhance the maturity and functionality of these cells. With the development of physiologic three-dimensional cardiac organoids, iPSCs represent a powerful platform to mechanistically dissect CHD and serve as a foundation for future translational research.
Collapse
Affiliation(s)
- Hao Zhang
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Stanford, CA, USA.
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
18
|
Tscheuschner L, Tzafriri AR. Cardiovascular Tissue Engineering Models for Atherosclerosis Treatment Development. Bioengineering (Basel) 2023; 10:1373. [PMID: 38135964 PMCID: PMC10740643 DOI: 10.3390/bioengineering10121373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
In the early years of tissue engineering, scientists focused on the generation of healthy-like tissues and organs to replace diseased tissue areas with the aim of filling the gap between organ demands and actual organ donations. Over time, the realization has set in that there is an additional large unmet need for suitable disease models to study their progression and to test and refine different treatment approaches. Increasingly, researchers have turned to tissue engineering to address this need for controllable translational disease models. We review existing and potential uses of tissue-engineered disease models in cardiovascular research and suggest guidelines for generating adequate disease models, aimed both at studying disease progression mechanisms and supporting the development of dedicated drug-delivery therapies. This involves the discussion of different requirements for disease models to test drugs, nanoparticles, and drug-eluting devices. In addition to realistic cellular composition, the different mechanical and structural properties that are needed to simulate pathological reality are addressed.
Collapse
Affiliation(s)
- Linnea Tscheuschner
- Department of Vascular Surgery, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Abraham R. Tzafriri
- Department of Research and Innovation, CBSET Inc., Lexington, MA 02421, USA;
| |
Collapse
|
19
|
Adegunsoye A, Gonzales NM, Gilad Y. Induced Pluripotent Stem Cells in Disease Biology and the Evidence for Their In Vitro Utility. Annu Rev Genet 2023; 57:341-360. [PMID: 37708421 DOI: 10.1146/annurev-genet-022123-090319] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Many human phenotypes are impossible to recapitulate in model organisms or immortalized human cell lines. Induced pluripotent stem cells (iPSCs) offer a way to study disease mechanisms in a variety of differentiated cell types while circumventing ethical and practical issues associated with finite tissue sources and postmortem states. Here, we discuss the broad utility of iPSCs in genetic medicine and describe how they are being used to study musculoskeletal, pulmonary, neurologic, and cardiac phenotypes. We summarize the particular challenges presented by each organ system and describe how iPSC models are being used to address them. Finally, we discuss emerging iPSC-derived organoid models and the potential value that they can bring to studies of human disease.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Genetics, Genomics, and Systems Biology, Section of Pulmonary and Critical Care, and the Department of Medicine, University of Chicago, Chicago, Illinois, USA;
| | - Natalia M Gonzales
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA; ,
| | - Yoav Gilad
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA; ,
- Department of Human Genetics, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
20
|
Redhead C, Taye N, Hubmacher D. En route towards a personalized medicine approach: Innovative therapeutic modalities for connective tissue disorders. Matrix Biol 2023; 122:46-54. [PMID: 37657665 PMCID: PMC10529529 DOI: 10.1016/j.matbio.2023.08.005] [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: 06/28/2023] [Revised: 08/25/2023] [Accepted: 08/27/2023] [Indexed: 09/03/2023]
Abstract
Connective tissue disorders can be caused by pathogenic variants (mutations) in genes encoding extracellular matrix (ECM) proteins. Such disorders typically manifest during development or postnatal growth and result in significant morbidity and mortality. The development of curative treatments for connective tissue disorders is hampered in part by the inability of many mature connective tissues to efficiently regenerate. To be most effective, therapeutic strategies designed to preserve or restore tissue function will likely need to be initiated during phases of significant endogenous connective tissue remodeling and organ sculpting postnatally and directly target the underlying ECM protein mutations. With recent advances in whole exome sequencing, in-vitro and in-vivo disease modeling, and the development of mutation-specific molecular therapeutic modalities, it is now feasible to directly correct disease-causing mutations underlying connective tissue disorders and ameliorate their pathogenic consequences. These technological advances may lead to potentially curative personalized medicine approaches for connective tissue disorders that have previously been considered incurable. In this review, we highlight innovative therapeutic modalities including gene replacement, exon skipping, DNA/mRNA editing, and pharmacological approaches that were used to preserve or restore tissue function in the context of connective tissue disorders. Inherent to a successful application of these approaches is the need to deepen the understanding of mechanisms that regulate ECM formation and homeostasis, and to decipher how individual mutations in ECM proteins compromise ECM and connective tissue development and function.
Collapse
Affiliation(s)
- Charlene Redhead
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nandaraj Taye
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dirk Hubmacher
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
21
|
Ganizada BH, Reesink KD, Parikh S, Ramaekers MJFG, Akbulut AC, Saraber PJMH, Debeij GP, Jaminon AM, Natour E, Lorusso R, Wildberger JE, Mees B, Schurink GW, Jacobs MJ, Cleutjens J, Krapels I, Gombert A, Maessen JG, Accord R, Delhaas T, Schalla S, Schurgers LJ, Bidar E. The Maastricht Acquisition Platform for Studying Mechanisms of Cell-Matrix Crosstalk (MAPEX): An Interdisciplinary and Systems Approach towards Understanding Thoracic Aortic Disease. Biomedicines 2023; 11:2095. [PMID: 37626592 PMCID: PMC10452257 DOI: 10.3390/biomedicines11082095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Current management guidelines for ascending thoracic aortic aneurysms (aTAA) recommend intervention once ascending or sinus diameter reaches 5-5.5 cm or shows a growth rate of >0.5 cm/year estimated from echo/CT/MRI. However, many aTAA dissections (aTAAD) occur in vessels with diameters below the surgical intervention threshold of <55 mm. Moreover, during aTAA repair surgeons observe and experience considerable variations in tissue strength, thickness, and stiffness that appear not fully explained by patient risk factors. To improve the understanding of aTAA pathophysiology, we established a multi-disciplinary research infrastructure: The Maastricht acquisition platform for studying mechanisms of tissue-cell crosstalk (MAPEX). The explicit scientific focus of the platform is on the dynamic interactions between vascular smooth muscle cells and extracellular matrix (i.e., cell-matrix crosstalk), which play an essential role in aortic wall mechanical homeostasis. Accordingly, we consider pathophysiological influences of wall shear stress, wall stress, and smooth muscle cell phenotypic diversity and modulation. Co-registrations of hemodynamics and deep phenotyping at the histological and cell biology level are key innovations of our platform and are critical for understanding aneurysm formation and dissection at a fundamental level. The MAPEX platform enables the interpretation of the data in a well-defined clinical context and therefore has real potential for narrowing existing knowledge gaps. A better understanding of aortic mechanical homeostasis and its derangement may ultimately improve diagnostic and prognostic possibilities to identify and treat symptomatic and asymptomatic patients with existing and developing aneurysms.
Collapse
Affiliation(s)
- Berta H. Ganizada
- Departments of Cardiothoracic Surgery, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands; (B.H.G.)
- Department of Biochemistry, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Koen D. Reesink
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Shaiv Parikh
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Mitch J. F. G. Ramaekers
- Department of Radiology and Nuclear Medicine, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Asim C. Akbulut
- Department of Biochemistry, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
- Stem Cell Research University Maastricht Facility, 6229 ER Maastricht, The Netherlands
| | - Pepijn J. M. H. Saraber
- Department of Biochemistry, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Gijs P. Debeij
- Departments of Cardiothoracic Surgery, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands; (B.H.G.)
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - MUMC-TAA Student Team
- Departments of Cardiothoracic Surgery, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands; (B.H.G.)
| | - Armand M. Jaminon
- Department of Biochemistry, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Ehsan Natour
- Departments of Cardiothoracic Surgery, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands; (B.H.G.)
| | - Roberto Lorusso
- Departments of Cardiothoracic Surgery, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands; (B.H.G.)
| | - Joachim E. Wildberger
- Department of Radiology and Nuclear Medicine, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Barend Mees
- Department of Vascular Surgery, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Geert Willem Schurink
- Department of Vascular Surgery, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Michael J. Jacobs
- Department of Vascular Surgery, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Jack Cleutjens
- Department of Pathology, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Ingrid Krapels
- Department of Clinical Genetics, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Alexander Gombert
- Department of Vascular Surgery, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Jos G. Maessen
- Departments of Cardiothoracic Surgery, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands; (B.H.G.)
| | - Ryan Accord
- Department of Cardiothoracic Surgery, Center for Congenital Heart Diseases, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Tammo Delhaas
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Simon Schalla
- Department of Radiology and Nuclear Medicine, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
| | - Leon J. Schurgers
- Department of Biochemistry, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands
- Stem Cell Research University Maastricht Facility, 6229 ER Maastricht, The Netherlands
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, 52074 Aachen, Germany
| | - Elham Bidar
- Departments of Cardiothoracic Surgery, CARIM School for Cardiovascular Diseases, Heart and Vascular Center, Maastricht University Medical Center (MUMC+), 6229 ER Maastricht, The Netherlands; (B.H.G.)
| |
Collapse
|
22
|
Rega S, Farina F, Bouhuis S, de Donato S, Chiesa M, Poggio P, Cavallotti L, Bonalumi G, Giambuzzi I, Pompilio G, Perrucci GL. Multi-omics in thoracic aortic aneurysm: the complex road to the simplification. Cell Biosci 2023; 13:131. [PMID: 37475058 DOI: 10.1186/s13578-023-01080-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 07/05/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Thoracic aortic aneurysm (TAA) is a serious condition that affects the aorta, characterized by the dilation of its first segment. The causes of TAA (e.g., age, hypertension, genetic syndromes) are heterogeneous and contribute to the weakening of the aortic wall. This complexity makes treating this life-threatening aortopathy challenging, as there are currently no etiological therapy available, and pharmacological strategies, aimed at avoiding surgical aortic replacement, are merely palliative. Recent studies on novel therapies for TAA have focused on identifying biological targets and etiological mechanisms of the disease by using advanced -omics techniques, including epigenomics, transcriptomics, proteomics, and metabolomics approaches. METHODS This review presents the latest findings from -omics approaches and underscores the importance of integrating multi-omics data to gain more comprehensive understanding of TAA. RESULTS Literature suggests that the alterations in TAA mediators frequently involve members of pro-fibrotic process (i.e., TGF-β signaling pathways) or proteins associated with cell/extracellular structures (e.g., aggrecans). Further analyses often reported the importance in TAA of processes as inflammation (PCR, CD3, leukotriene compounds), oxidative stress (chromatin OXPHOS, fatty acids), mitochondrial respiration and glycolysis/gluconeogenesis (e.g., PPARs and HIF1a). Of note, more recent metabolomics studies added novel molecular markers to the list of TAA-specific detrimental mediators (proteoglycans). CONCLUSION It is increasingly clear that integrating data from different -omics branches, along with clinical data, is essential as well as complicated both to reveal hidden relevant information and to address complex diseases such as TAA. Importantly, recent progresses in metabolomics highlighted novel potential and unprecedented marks in TAA diagnosis and therapy.
Collapse
Affiliation(s)
- Sara Rega
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Unit for the Study of Aortic, Valvular and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Floriana Farina
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Silvia Bouhuis
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Silvia de Donato
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Mattia Chiesa
- Bioinformatics and Artificial Intelligence Facility, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Electronics, Information and Biomedical Engineering, Politecnico Di Milano, Milan, Italy
| | - Paolo Poggio
- Unit for the Study of Aortic, Valvular and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Laura Cavallotti
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giorgia Bonalumi
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Ilaria Giambuzzi
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, Milan, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università Degli Studi Di Milano, Milan, Italy
| | - Gianluca L Perrucci
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy.
| |
Collapse
|
23
|
Nakamura K, Dalal AR, Yokoyama N, Pedroza AJ, Kusadokoro S, Mitchel O, Gilles C, Masoudian B, Leipzig M, Casey KM, Hiesinger W, Uchida T, Fischbein MP. Lineage-Specific Induced Pluripotent Stem Cell-Derived Smooth Muscle Cell Modeling Predicts Integrin Alpha-V Antagonism Reduces Aortic Root Aneurysm Formation in Marfan Syndrome Mice. Arterioscler Thromb Vasc Biol 2023; 43:1134-1153. [PMID: 37078287 PMCID: PMC10330156 DOI: 10.1161/atvbaha.122.318448] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 04/05/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND The role of increased smooth muscle cell (SMC) integrin αv signaling in Marfan syndrome (MFS) aortic aneurysm remains unclear. Herein, we examine the mechanism and potential efficacy of integrin αv blockade as a therapeutic strategy to reduce aneurysm progression in MFS. METHODS Induced pluripotent stem cells (iPSCs) were differentiated into aortic SMCs of the second heart field (SHF) and neural crest (NC) lineages, enabling in vitro modeling of MFS thoracic aortic aneurysms. The pathological role of integrin αv during aneurysm formation was confirmed by blockade of integrin αv with GLPG0187 in Fbn1C1039G/+ MFS mice. RESULTS iPSC-derived MFS SHF SMCs overexpress integrin αv relative to MFS NC and healthy control SHF cells. Furthermore, integrin αv downstream targets (FAK [focal adhesion kinase]/AktThr308/mTORC1 [mechanistic target of rapamycin complex 1]) were activated, especially in MFS SHF. Treatment of MFS SHF SMCs with GLPG0187 reduced p-FAK/p-AktThr308/mTORC1 activity back to control SHF levels. Functionally, MFS SHF SMCs had increased proliferation and migration compared to MFS NC SMCs and control SMCs, which normalized with GLPG0187 treatment. In the Fbn1C1039G/+ MFS mouse model, integrin αv, p-AktThr308, and downstream targets of mTORC1 proteins were elevated in the aortic root/ascending segment compared to littermate wild-type control. Mice treated with GLPG0187 (age 6-14 weeks) had reduced aneurysm growth, elastin fragmentation, and reduction of the FAK/AktThr308/mTORC1 pathway. GLPG0187 treatment reduced the amount and severity of SMC modulation assessed by single-cell RNA sequencing. CONCLUSIONS The integrin αv-FAK-AktThr308 signaling pathway is activated in iPSC SMCs from MFS patients, specifically from the SHF lineage. Mechanistically, this signaling pathway promotes SMC proliferation and migration in vitro. As biological proof of concept, GLPG0187 treatment slowed aneurysm growth and p-AktThr308 signaling in Fbn1C1039G/+ mice. Integrin αv blockade via GLPG0187 may be a promising therapeutic approach to inhibit MFS aneurysmal growth.
Collapse
Affiliation(s)
- Ken Nakamura
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Alex R. Dalal
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Nobu Yokoyama
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Sho Kusadokoro
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Olivia Mitchel
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Casey Gilles
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Bahar Masoudian
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Matthew Leipzig
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Kerriann M. Casey
- Department of Comparative Medicine, Stanford University School of Medicine. Stanford CA, USA
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Tetsuro Uchida
- Second Department of Surgery, Yamagata University Faculty of Medicine. Yamagata, Japan
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| |
Collapse
|
24
|
Granata A. Functional genomics in stroke: current and future applications of iPSCs and gene editing to dissect the function of risk variants. BMC Cardiovasc Disord 2023; 23:223. [PMID: 37120540 PMCID: PMC10148993 DOI: 10.1186/s12872-023-03227-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 04/04/2023] [Indexed: 05/01/2023] Open
Abstract
Stroke is an important disease with unmet clinical need. To uncover novel paths for treatment, it is of critical importance to develop relevant laboratory models that may help to shed light on the pathophysiological mechanisms of stroke. Induced pluripotent stem cells (iPSCs) technology has enormous potential to advance our knowledge into stroke by creating novel human models for research and therapeutic testing. iPSCs models generated from patients with specific stroke types and specific genetic predisposition in combination with other state of art technologies including genome editing, multi-omics, 3D system, libraries screening, offer the opportunity to investigate disease-related pathways and identify potential novel therapeutic targets that can then be tested in these models. Thus, iPSCs offer an unprecedented opportunity to make rapid progress in the field of stroke and vascular dementia research leading to clinical translation. This review paper summarizes some of the key areas in which patient-derived iPSCs technology has been applied to disease modelling and discusses the ongoing challenges and the future directions for the application of this technology in the field of stroke research.
Collapse
Affiliation(s)
- Alessandra Granata
- Department of Clinical Neurosciences, Victor Phillip Dahdaleh Heart & Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0BB, UK.
| |
Collapse
|
25
|
Liu L, Jouve C, Henry J, Berrandou TE, Hulot JS, Georges A, Bouatia-Naji N. Genomic, Transcriptomic, and Proteomic Depiction of Induced Pluripotent Stem Cells-Derived Smooth Muscle Cells As Emerging Cellular Models for Arterial Diseases. Hypertension 2023; 80:740-753. [PMID: 36655574 DOI: 10.1161/hypertensionaha.122.19733] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Vascular smooth muscle cells (SMCs) plasticity is a central mechanism in cardiovascular health and disease. We aimed at providing cellular phenotyping, epigenomic and proteomic depiction of SMCs derived from induced pluripotent stem cells and evaluating their potential as cellular models in the context of complex diseases. METHODS Human induced pluripotent stem cell lines were differentiated using RepSox (R-SMCs) or PDGF-BB (platelet-derived growth factor-BB) and TGF-β (transforming growth factor beta; TP-SMCs), during a 24-day long protocol. RNA-Seq and assay for transposase accessible chromatin-Seq were performed at 6 time points of differentiation, and mass spectrometry was used to quantify proteins. RESULTS Both induced pluripotent stem cell differentiation protocols generated SMCs with positive expression of SMC markers. TP-SMCs exhibited greater proliferation capacity, migration and lower calcium release in response to contractile stimuli, compared with R-SMCs. Genes involved in the contractile function of arteries were highly expressed in R-SMCs compared with TP-SMCs or primary SMCs. R-SMCs and coronary artery transcriptomic profiles were highly similar, characterized by high expression of genes involved in blood pressure regulation and coronary artery disease. We identified FOXF1 and HAND1 as key drivers of RepSox specific program. Extracellular matrix content contained more proteins involved in wound repair in TP-SMCs and higher secretion of basal membrane constituents in R-SMCs. Open chromatin regions of R-SMCs and TP-SMCs were significantly enriched for variants associated with blood pressure and coronary artery disease. CONCLUSIONS Both induced pluripotent stem cell-derived SMCs models present complementary cellular phenotypes of high relevance to SMC plasticity. These cellular models present high potential to study functional regulation at genetic risk loci of main arterial diseases.
Collapse
Affiliation(s)
- Lu Liu
- Université Paris Cité, Inserm, PARCC, Paris, France (L.L., C.J., J.H., T.-E.B., J.-S.H., A.G., N.B.-N.)
| | - Charlène Jouve
- Université Paris Cité, Inserm, PARCC, Paris, France (L.L., C.J., J.H., T.-E.B., J.-S.H., A.G., N.B.-N.)
| | - Joséphine Henry
- Université Paris Cité, Inserm, PARCC, Paris, France (L.L., C.J., J.H., T.-E.B., J.-S.H., A.G., N.B.-N.)
| | - Takiy-Eddine Berrandou
- Université Paris Cité, Inserm, PARCC, Paris, France (L.L., C.J., J.H., T.-E.B., J.-S.H., A.G., N.B.-N.)
| | - Jean-Sébastien Hulot
- Université Paris Cité, Inserm, PARCC, Paris, France (L.L., C.J., J.H., T.-E.B., J.-S.H., A.G., N.B.-N.)
| | - Adrien Georges
- Université Paris Cité, Inserm, PARCC, Paris, France (L.L., C.J., J.H., T.-E.B., J.-S.H., A.G., N.B.-N.)
| | - Nabila Bouatia-Naji
- Université Paris Cité, Inserm, PARCC, Paris, France (L.L., C.J., J.H., T.-E.B., J.-S.H., A.G., N.B.-N.)
| |
Collapse
|
26
|
Liu G, Li J, Ming Y, Xiang B, Zhou X, Chen Y, Chen N, Abudupataer M, Zhu S, Sun X, Sun Y, Lai H, Feng S, Wang C, Zhu K. A hiPSC-derived lineage-specific vascular smooth muscle cell-on-a-chip identifies aortic heterogeneity across segments. LAB ON A CHIP 2023; 23:1835-1851. [PMID: 36810777 DOI: 10.1039/d2lc01158a] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Aortic aneurysm (AA), a potentially lethal condition with the characteristic of aortic dilatation, can only be treated by surgical or endovascular procedures. The underlying mechanisms of AA are unclear and early preventive treatment is still insufficient due to segmental aortic heterogeneity and the limitations of current disease models. Here, we firstly established a comprehensive lineage-specific vascular smooth muscle cell (SMC)-on-a-chip model using human induced pluripotent stem cells to yield cell lineages representing different segments of the aorta and tested the constructed organ-on-a-chip model under various tensile stress conditions. Bulk RNA sequencing, RT-qPCR, immunofluorescence, western blot and FACS analyses were performed to discover the segmental aortic heterogeneity of response for tensile stress and drug testing. The appropriate stretching frequency for all lineages of SMCs was 1.0 Hz, paraxial mesoderm (PM) SMCs were more sensitive to tensile stress than lateral mesoderm (LM) SMCs and neural crest (NC) SMCs. These differences may be related to the different transcriptional profiles of the tension-stressed distinct lineage-specific vascular SMCs, specifically in relation to the PI3K-Akt signaling pathway. Also, the organ-on-a-chip displayed contractile physiology, perfect fluid coordination, and was conducive to drug testing, displaying heterogeneous segmental aortic responses. Compared with LM-SMCs and NC-SMCs, PM-SMCs were more sensitive to ciprofloxacin. The model is evaluated as a novel and suitable supplement to AA animal models for determining differential physiology and drug response in different parts of the aorta. Furthermore, this system could pave the way for disease modeling, drug testing, and the personalized treatment of patients with AA in the future.
Collapse
Affiliation(s)
- Gang Liu
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Jun Li
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yang Ming
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Bitao Xiang
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Xiaonan Zhou
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yabin Chen
- School of Life Science and Technology, Shanghai Tech University, 319 Yueyang Road, Shanghai, 200031, China
| | - Nan Chen
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Mieradilijiang Abudupataer
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Shichao Zhu
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Xiaoning Sun
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yongxin Sun
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Sisi Feng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
27
|
Jia WN, Wang QY, Niu LL, Chen ZX, Jiang YX. Morphometric assessment of the ciliary body in patients with Marfan syndrome and ectopia lentis: A quantitative study using ultrasound biomicroscopy: Ciliary body morphology in Marfan syndrome and ectopia lentis. Am J Ophthalmol 2023; 251:24-31. [PMID: 36948371 DOI: 10.1016/j.ajo.2023.03.003] [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: 11/08/2022] [Revised: 02/28/2023] [Accepted: 03/05/2023] [Indexed: 03/24/2023]
Abstract
PURPOSE To explore the biometric characteristics of the ciliary body in patients with Marfan syndrome (MFS) and ectopia lentis (EL). DESIGN Cross-sectional study. METHODS Seventy-two consecutive MFS patients with EL and 72 non-disease controls were recruited. Ciliary body biometric parameters such as ciliary muscle cross-sectional area at 2000 μm from the scleral spur (CMA2000), ciliary muscle thickness at 1000 μm from the scleral spur (CMT1000), and maximum ciliary body thickness (CBTmax) were measured from multiple directions with ultrasound biomicroscopy (UBM). The relationship between ciliary body parameters and other ocular characteristics was also evaluated. RESULTS Average CMA2000, CMT1000, and CBTmax were 0.692 ± 0.015 mm2, 0.405 ± 0.010 mm, and 0.855±0.023 mm in MFS eyes, respectively, and were significantly smaller than controls (all p < 0.001). The prevalence of ciliary body thinning was 22.2% in the MFS group versus 0 in controls (p < 0.001); eyes with more severe EL had smaller CMA2000 (p = 0.050), thinner CMT1000 (p = 0.022) and shorter CBTmax (p = 0.015). Patients with microspherophakia (MSP) had even smaller CMA2000 (p = 0.033) and CMT1000 (p = 0.044) than those without MSP. The most common subluxation direction was in the superonasal quadrant (25, 39.7%), which probably correlates with the thinnest CMT1000 in the inferotemporal quadrant (p = 0.005). CONCLUSIONS MFS patients with EL had thinner ciliary muscles, shorter ciliary processes, and a higher prevalence of ciliary body thinning, especially those with MSP. Both the extent and direction of subluxation were associated with ciliary body biometry.
Collapse
Affiliation(s)
- Wan-Nan Jia
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Qian-Yi Wang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Ling-Ling Niu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Ze-Xu Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.
| | - Yong-Xiang Jiang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.
| |
Collapse
|
28
|
Davaapil H, McNamara M, Granata A, Macrae RGC, Hirano M, Fitzek M, Aragon-Martin JA, Child A, Smith DM, Sinha S. A phenotypic screen of Marfan syndrome iPSC-derived vascular smooth muscle cells uncovers GSK3β as a new target. Stem Cell Reports 2023; 18:555-569. [PMID: 36669494 PMCID: PMC9968988 DOI: 10.1016/j.stemcr.2022.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 01/20/2023] Open
Abstract
Marfan syndrome (MFS) is a rare connective tissue disorder caused by mutations in FBN1. Patients with MFS notably suffer from aortic aneurysm and dissection. Despite considerable effort, animal models have proven to be poorly predictive for therapeutic intervention in human aortic disease. Patient-derived induced pluripotent stem cells can be differentiated into vascular smooth muscle cells (VSMCs) and recapitulate major features of MFS. We have screened 1,022 small molecules in our in vitro model, exploiting the highly proteolytic nature of MFS VSMCs, and identified 36 effective compounds. Further analysis identified GSK3β as a recurring target in the compound screen. GSK3β inhibition/knockdown did not ameliorate the proliferation defect in MFS-VSMCs but improved MFS-VSMC proteolysis and apoptosis and partially rescued fibrillin-1 deposition. To conclude, we have identified GSK3β as a novel target for MFS, forming the foundation for future work in MFS and other aortic diseases.
Collapse
Affiliation(s)
- Hongorzul Davaapil
- Department of Medicine and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Madeline McNamara
- Department of Medicine and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Alessandra Granata
- Stroke Research Group, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Robyn G C Macrae
- Department of Medicine and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK; Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Mei Hirano
- Department of Medicine and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Martina Fitzek
- Emerging Innovations, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - J A Aragon-Martin
- Department of Surgery and Cancer, Imperial College, Guy Scadding Building, London SW3 6LY, UK; The Marfan Trust, Guy Scadding Building, London SW3 6LY, UK
| | - Anne Child
- Department of Surgery and Cancer, Imperial College, Guy Scadding Building, London SW3 6LY, UK; The Marfan Trust, Guy Scadding Building, London SW3 6LY, UK
| | - David M Smith
- Emerging Innovations, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Sanjay Sinha
- Department of Medicine and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK.
| |
Collapse
|
29
|
Justin AW, Cammarata F, Guy AA, Estevez SR, Burgess S, Davaapil H, Stavropoulou-Tatla A, Ong J, Jacob AG, Saeb-Parsy K, Sinha S, Markaki AE. Densified collagen tubular grafts for human tissue replacement and disease modelling applications. BIOMATERIALS ADVANCES 2023; 145:213245. [PMID: 36549149 DOI: 10.1016/j.bioadv.2022.213245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
There is a significant need across multiple indications for an off-the-shelf bioengineered tubular graft which fulfils the mechanical and biological requirements for implantation and function but does not necessarily require cells for manufacture or deployment. Herein, we present a tissue-like tubular construct using a cell-free, materials-based method of manufacture, utilizing densified collagen hydrogel. Our tubular grafts are seamless, mechanically strong, customizable in terms of lumen diameter and wall thickness, and display a uniform fibril density across the wall thickness and along the tube length. While the method enables acellular grafts to be generated rapidly, inexpensively, and to a wide range of specifications, the cell-compatible densification process also enables a high density of cells to be incorporated uniformly into the walls of the tubes, which we show can be maintained under perfusion culture. Additionally, the method enables tubes consisting of distinct cell domains with cellular configurations at the boundaries which may be useful for modelling aortic disease. Further, we demonstrate additional steps which allow for luminal surface patterning. These results highlight the universality of this approach and its potential for developing the next generation of bioengineered grafts.
Collapse
Affiliation(s)
- Alexander W Justin
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK.
| | - Federico Cammarata
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK
| | - Andrew A Guy
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK
| | - Silas R Estevez
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK
| | - Sebastian Burgess
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK
| | - Hongorzul Davaapil
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | | | - John Ong
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK; East of England Gastroenterology Speciality Training Program, Cambridge, UK
| | - Aishwarya G Jacob
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Department of Biochemistry, University of Cambridge, Downing Site, Tennis Court Road, Cambridge CB2 1QW, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Sanjay Sinha
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Athina E Markaki
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK.
| |
Collapse
|
30
|
De Paola M, Pischiutta F, Comolli D, Mariani A, Kelk J, Lisi I, Cerovic M, Fumagalli S, Forloni G, Zanier ER. Neural cortical organoids from self-assembling human iPSC as a model to investigate neurotoxicity in brain ischemia. J Cereb Blood Flow Metab 2023; 43:680-693. [PMID: 36655331 PMCID: PMC10108182 DOI: 10.1177/0271678x231152023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Brain ischemia is a common acute injury resulting from impaired blood flow to the brain. Translation of effective drug candidates from experimental models to patients has systematically failed. The use of human induced pluripotent stem cells (iPSC) offers new opportunities to gain translational insights into diseases including brain ischemia. We used a human 3D self-assembling iPSC-derived model (human cortical organoids, hCO) to characterize the effects of ischemia caused by oxygen-glucose deprivation (OGD). hCO exposed to 2 h or 8 h of OGD had neuronal death and impaired neuronal network complexity, measured in whole-mounting microtubule-associated protein 2 (MAP-2) immunostaining. Neuronal vulnerability was reflected by a reduction in MAP-2 mRNA levels, and increased release of neurofilament light chain (NfL) in culture media, proportional to OGD severity. Glial fibrillary acidic protein (GFAP) gene or protein levels did not change in hCO, but their release in medium increased after prolonged OGD. In conclusion, this human 3D iPSC-based in vitro model of brain ischemic injury is characterized by marked neuronal injury reflected by the release of the translational biomarker NfL which is relevant for testing neuroprotective strategies.
Collapse
Affiliation(s)
- Massimiliano De Paola
- Biology of Neurodegenerative Diseases Lab, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Francesca Pischiutta
- Acute Brain Injury and Therapeutic Strategies Lab, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Davide Comolli
- Biology of Neurodegenerative Diseases Lab, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Alessandro Mariani
- Biology of Neurodegenerative Diseases Lab, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Joe Kelk
- Biology of Neurodegenerative Diseases Lab, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Ilaria Lisi
- Acute Brain Injury and Therapeutic Strategies Lab, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Milica Cerovic
- Biology of Neurodegenerative Diseases Lab, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Stefano Fumagalli
- Biology of Neurodegenerative Diseases Lab, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Gianluigi Forloni
- Biology of Neurodegenerative Diseases Lab, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Elisa R Zanier
- Acute Brain Injury and Therapeutic Strategies Lab, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| |
Collapse
|
31
|
Grewal N, Dolmaci O, Jansen E, Klautz R, Driessen A, Lindeman J, Poelmann RE. Are acute type A aortic dissections atherosclerotic? Front Cardiovasc Med 2023; 9:1032755. [PMID: 36698948 PMCID: PMC9868270 DOI: 10.3389/fcvm.2022.1032755] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Background Type A aortic dissections (TAAD) are devastating aortic complications. Patients with Marfan syndrome, a bicuspid aortic valve or a thoracic aortic aneurysm have an increased risk to develop a TAAD. These predisposing conditions are characterized by a histologically thin intimal layer and hardly any atherosclerosis. Little is known about the susceptibility for atherosclerosis in patients with a type A aortic dissection. Objective We aim to systematically describe atherosclerotic lesions in TAAD patients. Materials and methods A total of 51 patients with a TAAD (mean age 62.5 ± 10.8 years, 49% females) and 17 control patients (mean age 63 ± 5.5 years, 53% females) were included in this study. Cardiovascular risk factors were assessed clinically. All sections were stained with Movat pentachrome and hematoxylin eosin. Plaque morphology was classified according to the modified AHA classification scheme proposed by Virmani et al. Results In the TAAD group thirty-seven percent were overweight (BMI > 25). Diabetes and peripheral arterial disease were not present in any of the patients. Fifty-nine percent of the patients had a history of hypertension. The intima in TAAD patients was significantly thinner as compared to the control group (mean thickness 143 ± 126.5 μm versus 193 ± 132 μm, p < 0.023). Seven TAAD patients had a normal intima without any form of adaptive or pathological thickening. Twenty-three TAAD patients demonstrated adaptive intimal thickening. Fourteen had an intimal xanthoma, also known as fatty streaks. A minority of 7 TAAD patients had progressive atherosclerotic lesions, 4 of which demonstrated pathological intimal thickening, 3 patients showed early fibroatheroma, late fibroatheroma and thin cap fibroatheroma. In the control group the majority of the patients exhibited progressive atherosclerotic lesions: three pathologic intimal thickening, two early fibroatheroma, six late fibroatheroma, one healed rupture and two fibrotic calcified plaque. Discussion This study shows that TAAD patients hardly exhibit any form of progressive atherosclerosis. The majority of TAAD patients showcase non-progressive intimal lesions, whereas the control group mostly demonstrated progressive intimal atherosclerotic lesions. Findings are independent of age, sex, or the presence of (a history of) hypertension.
Collapse
Affiliation(s)
- Nimrat Grewal
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, Netherlands,Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, Netherlands,Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Nimrat Grewal,
| | - Onur Dolmaci
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, Netherlands,Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Evert Jansen
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Robert Klautz
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, Netherlands,Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Antoine Driessen
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Jan Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Robert E. Poelmann
- Institute of Biology, Animal Sciences and Health, Leiden University, Leiden, Netherlands,Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
32
|
Liu Q, Liu Z, Gu H, Ge Y, Wu X, Zuo F, Du Q, Lei Y, Wang Z, Lin H. Comparative study of differentiating human pluripotent stem cells into vascular smooth muscle cells in hydrogel-based culture methods. Regen Ther 2022; 22:39-49. [PMID: 36618488 PMCID: PMC9798140 DOI: 10.1016/j.reth.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/31/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs), which provides structural integrity and regulates the diameter of vasculature, are of great potential for modeling vascular-associated diseases and tissue engineering. Here, we presented a detailed comparison of differentiating human pluripotent stem cells (hPSCs) into VSMCs (hPSCs-VSMCs) in four different culture methods, including 2-dimensional (2D) culture, 3-dimensional (3D) PNIPAAm-PEG hydrogel culture, 3-dimensional (3D) alginate hydrogel culture, and transferring 3-dimensional alginate hydrogel culture to 2-dimensional (2D) culture. Both hydrogel-based culture methods could mimic in vivo microenvironment to protect cells from shear force, and avoid cells agglomeration, resulting in the extremely high culture efficiency (e.g., high viability, high purity and high yield) compared with 2D culture. We demonstrated hPSC-VSMCs produced from hydrogel-based culture methods had better contractile phenotypes and the potential of vasculature formation. The transcriptome analysis showed the hPSC-VSMCs derived from hydrogel-based culture methods displayed more upregulated genes in vasculature development, angiogenesis and blood vessel development, extracellular matrix compared with 2D culture. Taken together, hPSC-VSMCs produced from hydrogel-based culture system could be applied in various biomedical fields, and further indicated the suitable development of alginate hydrogel for industrial production by taking all aspects into consideration.
Collapse
Affiliation(s)
- Qing Liu
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Zhen Liu
- Department of Neurosurgery, Beijing Shunyi District Hospital, Beijing, 101300, China
| | - Hongyu Gu
- Department of Thoracic Surgery Ward 3, The First Hospital of Qiqihar, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, China
| | - Yuxia Ge
- Department of Neurology, The Second Hospital of Harbin, Harbin, 150056, China
| | - Xuesheng Wu
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Fuxing Zuo
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qian Du
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Yuguo Lei
- Department of Biomedical Engineering, Huck Life Science Institute, Pennsylvania State University, University Park, PA, 16802, USA,Corresponding author.
| | - Zhanqi Wang
- Department of Vascular Surgery, Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China,Corresponding author.
| | - Haishuang Lin
- Department of Neurology, The Second Hospital of Harbin, Harbin, 150056, China,Corresponding author.
| |
Collapse
|
33
|
Wang G, Xu Y, Wang Q, Chai Y, Sun X, Yang F, Zhang J, Wu M, Liao X, Yu X, Sheng X, Liu Z, Zhang J. Rare and undiagnosed diseases: From disease-causing gene identification to mechanism elucidation. FUNDAMENTAL RESEARCH 2022; 2:918-928. [PMID: 38933382 PMCID: PMC11197726 DOI: 10.1016/j.fmre.2022.09.002] [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: 07/16/2022] [Revised: 09/04/2022] [Accepted: 09/05/2022] [Indexed: 11/27/2022] Open
Abstract
Rare and undiagnosed diseases substantially decrease patient quality of life and have increasingly become a heavy burden on healthcare systems. Because of the challenges in disease-causing gene identification and mechanism elucidation, patients are often confronted with difficulty obtaining a precise diagnosis and treatment. Due to advances in sequencing and multiomics analysis approaches combined with patient-derived iPSC models and gene-editing platforms, substantial progress has been made in the diagnosis and treatment of rare and undiagnosed diseases. The aforementioned techniques also provide an operational basis for future precision medicine studies. In this review, we summarize recent progress in identifying disease-causing genes based on GWAS/WES/WGS-guided multiomics analysis approaches. In addition, we discuss recent advances in the elucidation of pathogenic mechanisms and treatment of diseases with state-of-the-art iPSC and organoid models, which are improved by cell maturation level and gene editing technology. The comprehensive strategies described above will generate a new paradigm of disease classification that will significantly promote the precision and efficiency of diagnosis and treatment for rare and undiagnosed diseases.
Collapse
Affiliation(s)
- Gang Wang
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Yuyan Xu
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qintao Wang
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yi Chai
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiangwei Sun
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fan Yang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Jian Zhang
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mengchen Wu
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xufeng Liao
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaomin Yu
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xin Sheng
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhihong Liu
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Jin Zhang
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine; Center of Gene/Cell Engineering and Genome Medicine of Zhejiang Province, Hangzhou 310058, China
| |
Collapse
|
34
|
Chen ZX, Jia WN, Jiang YX. Genotype-phenotype correlations of marfan syndrome and related fibrillinopathies: Phenomenon and molecular relevance. Front Genet 2022; 13:943083. [PMID: 36176293 PMCID: PMC9514320 DOI: 10.3389/fgene.2022.943083] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Marfan syndrome (MFS, OMIM: 154700) is a heritable multisystemic disease characterized by a wide range of clinical manifestations. The underlying molecular defect is caused by variants in the FBN1. Meanwhile, FBN1 variants are also detected in a spectrum of connective tissue disorders collectively termed as ‘type I fibrillinopathies’. A multitude of FBN1 variants is reported and most of them are unique in each pedigree. Although MFS is being considered a monogenic disorder, it is speculated that the allelic heterogeneity of FBN1 variants contributes to various manifestations, distinct prognoses, and differential responses to the therapies in affected patients. Significant progress in the genotype–phenotype correlations of MFS have emerged in the last 20 years, though, some of the associations were still in debate. This review aims to update the recent advances in the genotype-phenotype correlations of MFS and related fibrillinopathies. The molecular bases and pathological mechanisms are summarized for better support of the observed correlations. Other factors contributing to the phenotype heterogeneity and future research directions were also discussed. Dissecting the genotype-phenotype correlation of FBN1 variants and related disorders will provide valuable information in risk stratification, prognosis, and choice of therapy.
Collapse
Affiliation(s)
- Ze-Xu Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Wan-Nan Jia
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yong-Xiang Jiang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
35
|
Bulut M, Vila Cuenca M, de Graaf M, van den Hil FE, Mummery CL, Orlova VV. Three-Dimensional Vessels-on-a-Chip Based on hiPSC-derived Vascular Endothelial and Smooth Muscle Cells. Curr Protoc 2022; 2:e564. [PMID: 36250774 DOI: 10.1002/cpz1.564] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Blood vessels are composed of endothelial cells (ECs) that form the inner vessel wall and mural cells that cover the ECs to mediate their stabilization. Crosstalk between ECs and VSMCs while the ECs undergo microfluidic flow is vital for the function and integrity of blood vessels. Here, we describe a protocol to generate three-dimensional (3D) engineered vessels-on-chip (VoCs) composed of vascular cells derived from human induced pluripotent stem cells (hiPSCs). We first describe protocols for robust differentiation of vascular smooth muscle cells (hiPSC-VSMCs) from hiPSCs that are effective across multiple hiPSC lines. Second, we describe the fabrication of a simple microfluidic device consisting of a single collagen lumen that can act as a cell scaffold and support fluid flow using the viscous finger patterning (VFP) technique. After the channel is seeded sequentially with hiPSC-derived ECs (hiPSC-ECs) and hiPSC-VSMCs, a stable EC barrier covered by VSMCs lines the collagen lumen. We demonstrate that this 3D VoC model can recapitulate physiological cell-cell interaction and can be perfused under physiological shear stress using a microfluidic pump. The uniform geometry of the vessel lumens allows precise control of flow dynamics. We have thus developed a robust protocol to generate an entirely isogenic hiPSC-derived 3D VoC model, which could be valuable for studying vessel barrier function and physiology in healthy or disease states. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Differentiation of hiPSC-VSMCs Support Protocol 1: Characterization of hiPSC-NCCs and hiPSC-VSMCs Support Protocol 2: Preparation of cryopreserved hiPSC-VSMCs and hiPSC-ECs for VoC culture Basic Protocol 2: Generation of 3D VoC model composed of hiPSC-ECs and hiPSC-VSMCs Support Protocol 3: Structural characterization of 3D VoC model.
Collapse
Affiliation(s)
- Merve Bulut
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marc Vila Cuenca
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mees de Graaf
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
36
|
Luperchio TR, Kozel BA. Extending the spectrum in aortopathy: stenosis to aneurysm. Curr Opin Genet Dev 2022; 76:101962. [DOI: 10.1016/j.gde.2022.101962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 06/21/2022] [Accepted: 06/25/2022] [Indexed: 11/03/2022]
|
37
|
Ito S, Lu HS, Daugherty A, Sawada H. Embryonic Heterogeneity of Smooth Muscle Cells in the Complex Mechanisms of Thoracic Aortic Aneurysms. Genes (Basel) 2022; 13:genes13091618. [PMID: 36140786 PMCID: PMC9498804 DOI: 10.3390/genes13091618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
Smooth muscle cells (SMCs) are the major cell type of the aortic wall and play a pivotal role in the pathophysiology of thoracic aortic aneurysms (TAAs). TAAs occur in a region-specific manner with the proximal region being a common location. In this region, SMCs are derived embryonically from either the cardiac neural crest or the second heart field. These cells of distinct origins reside in specific locations and exhibit different biological behaviors in the complex mechanism of TAAs. The purpose of this review is to enhance understanding of the embryonic heterogeneity of SMCs in the proximal thoracic aorta and their functions in TAAs.
Collapse
Affiliation(s)
- Sohei Ito
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Correspondence: ; Tel.: +1-(859)-218-1705
| |
Collapse
|
38
|
Engineering Smooth Muscle to Understand Extracellular Matrix Remodeling and Vascular Disease. Bioengineering (Basel) 2022; 9:bioengineering9090449. [PMID: 36134994 PMCID: PMC9495899 DOI: 10.3390/bioengineering9090449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
The vascular smooth muscle is vital for regulating blood pressure and maintaining cardiovascular health, and the resident smooth muscle cells (SMCs) in blood vessel walls rely on specific mechanical and biochemical signals to carry out these functions. Any slight change in their surrounding environment causes swift changes in their phenotype and secretory profile, leading to changes in the structure and functionality of vessel walls that cause pathological conditions. To adequately treat vascular diseases, it is essential to understand how SMCs crosstalk with their surrounding extracellular matrix (ECM). Here, we summarize in vivo and traditional in vitro studies of pathological vessel wall remodeling due to the SMC phenotype and, conversely, the SMC behavior in response to key ECM properties. We then analyze how three-dimensional tissue engineering approaches provide opportunities to model SMCs’ response to specific stimuli in the human body. Additionally, we review how applying biomechanical forces and biochemical stimulation, such as pulsatile fluid flow and secreted factors from other cell types, allows us to study disease mechanisms. Overall, we propose that in vitro tissue engineering of human vascular smooth muscle can facilitate a better understanding of relevant cardiovascular diseases using high throughput experiments, thus potentially leading to therapeutics or treatments to be tested in the future.
Collapse
|
39
|
Pedroza AJ, Dalal AR, Shad R, Yokoyama N, Nakamura K, Cheng P, Wirka RC, Mitchel O, Baiocchi M, Hiesinger W, Quertermous T, Fischbein MP. Embryologic Origin Influences Smooth Muscle Cell Phenotypic Modulation Signatures in Murine Marfan Syndrome Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2022; 42:1154-1168. [PMID: 35861960 PMCID: PMC9420801 DOI: 10.1161/atvbaha.122.317381] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 06/15/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Aortic root smooth muscle cells (SMC) develop from both the second heart field (SHF) and neural crest. Disparate responses to disease-causing Fbn1 variants by these lineages are proposed to promote focal aortic root aneurysm formation in Marfan syndrome (MFS), but lineage-stratified SMC analysis in vivo is lacking. METHODS We generated SHF lineage-traced MFS mice and performed integrated multiomic (single-cell RNA and assay for transposase-accessible chromatin sequencing) analysis stratified by embryological origin. SMC subtypes were spatially identified via RNA in situ hybridization. Response to TWIST1 overexpression was determined via lentiviral transduction in human aortic SMCs. RESULTS Lineage stratification enabled nuanced characterization of aortic root cells. We identified heightened SHF-derived SMC heterogeneity including a subset of Tnnt2 (cardiac troponin T)-expressing cells distinguished by altered proteoglycan expression. MFS aneurysm-associated SMC phenotypic modulation was identified in both SHF-traced and nontraced (neural crest-derived) SMCs; however, transcriptomic responses were distinct between lineages. SHF-derived modulated SMCs overexpressed collagen synthetic genes and small leucine-rich proteoglycans while nontraced SMCs activated chondrogenic genes. These modulated SMCs clustered focally in the aneurysmal aortic root at the region of SHF/neural crest lineage overlap. Integrated RNA-assay for transposase-accessible chromatin analysis identified enriched Twist1 and Smad2/3/4 complex binding motifs in SHF-derived modulated SMCs. TWIST1 overexpression promoted collagen and SLRP gene expression in vitro, suggesting TWIST1 may drive SHF-enriched collagen synthesis in MFS aneurysm. CONCLUSIONS SMCs derived from both SHF and neural crest lineages undergo phenotypic modulation in MFS aneurysm but are defined by subtly distinct transcriptional responses. Enhanced TWIST1 transcription factor activity may contribute to enriched collagen synthetic pathways SHF-derived SMCs in MFS.
Collapse
Affiliation(s)
- Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Alex R. Dalal
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Rohan Shad
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Nobu Yokoyama
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Ken Nakamura
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine. Stanford CA, USA
| | - Robert C. Wirka
- Division of Cardiology, UNC School of Medicine, Chapel Hill NC, USA
| | | | - Michael Baiocchi
- Department of Epidemiology and Population Health, Stanford Unviersity School of Medicine. Stanford CA, USA
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine. Stanford CA, USA
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| |
Collapse
|
40
|
Kallenbach K, Remes A, Müller OJ, Arif R, Zaradzki M, Wagner AH. Translational Medicine: Towards Gene Therapy of Marfan Syndrome. J Clin Med 2022; 11:jcm11143934. [PMID: 35887698 PMCID: PMC9319421 DOI: 10.3390/jcm11143934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 12/12/2022] Open
Abstract
Marfan syndrome (MFS) is one of the most common inherited disorders of connective tissue caused by mutations of the fibrillin-1 gene (FBN1). Vascular abnormalities, such as the enlargement of the aorta with the risk of life-threatening rupture are frequently observed. However, current treatment is limited and therapeutic options focus solely on symptomatic therapy. Gene therapy focuses on genetically modifying cells to produce a therapeutic effect and may be a promising treatment option for MFS. Here, we first provide an overview of the historical background and characterization of MFS. Subsequently, we summarise current gene therapy options and possible translational concepts for this inherited disorder that affects connective tissue.
Collapse
Affiliation(s)
- Klaus Kallenbach
- Institute for Cardiac Surgery and Interventional Cardiology (INCCI), Department of Cardiac Surgery, 1210 Luxembourg, Luxembourg;
- VASCERN HTAD European Reference Center, 1210 Luxembourg, Luxembourg
| | - Anca Remes
- Department of Internal Medicine III, University of Kiel and University Hospital Schleswig-Holstein, 24105 Kiel, Germany; (A.R.); (O.J.M.)
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Oliver J. Müller
- Department of Internal Medicine III, University of Kiel and University Hospital Schleswig-Holstein, 24105 Kiel, Germany; (A.R.); (O.J.M.)
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Rawa Arif
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (R.A.); (M.Z.)
| | - Marcin Zaradzki
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (R.A.); (M.Z.)
| | - Andreas H. Wagner
- Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-544062; Fax: +49-6221-544038
| |
Collapse
|
41
|
Patient-derived microphysiological model identifies the therapeutic potential of metformin for thoracic aortic aneurysm. EBioMedicine 2022; 81:104080. [PMID: 35636318 PMCID: PMC9156889 DOI: 10.1016/j.ebiom.2022.104080] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 12/20/2022] Open
Abstract
Background Thoracic aortic aneurysm (TAA) is the permanent dilation of the thoracic aortic wall that predisposes patients to lethal events such as aortic dissection or rupture, for which effective medical therapy remains scarce. Human-relevant microphysiological models serve as a promising tool in drug screening and discovery. Methods We developed a dynamic, rhythmically stretching, three-dimensional microphysiological model. Using patient-derived human aortic smooth muscle cells (HAoSMCs), we tested the biological features of the model and compared them with native aortic tissues. Drug testing was performed on the individualized TAA models, and the potentially effective drug was further tested using β-aminopropionitrile-treated mice and retrospective clinical data. Findings The HAoSMCs on the model recapitulated the expressions of many TAA-related genes in tissue. Phenotypic switching and mitochondrial dysfunction, two disease hallmarks of TAA, were highlighted on the microphysiological model: the TAA-derived HAoSMCs exhibited lower alpha-smooth muscle actin expression, lower mitochondrial membrane potential, lower oxygen consumption rate and higher superoxide accumulation than control cells, while these differences were not evidently reflected in two-dimensional culture flasks. Model-based drug testing demonstrated that metformin partially recovered contractile phenotype and mitochondrial function in TAA patients’ cells. Mouse experiment and clinical investigations also demonstrated better preserved aortic microstructure, higher nicotinamide adenine dinucleotide level and lower aortic diameter with metformin treatment. Interpretation These findings support the application of this human-relevant microphysiological model in studying personalized disease characteristics and facilitating drug discovery for TAA. Metformin may regulate contractile phenotypes and metabolic dysfunctions in diseased HAoSMCs and limit aortic dilation. Funding This work was supported by grants from National Key R&D Program of China (2018YFC1005002), National Natural Science Foundation of China (82070482, 81771971, 81772007, 51927805, and 21734003), the Science and Technology Commission of Shanghai Municipality (20ZR1411700, 18ZR1407000, 17JC1400200, and 20YF1406900), Shanghai Municipal Science and Technology Major Project (2017SHZDZX01), and Shanghai Municipal Education Commission (Innovation Program 2017-01-07-00-07-E00027). Y.S.Z. was not supported by any of these funds; instead, the Brigham Research Institute is acknowledged.
Collapse
|
42
|
Mizrak D, Feng H, Yang B. Dissecting the Heterogeneity of Human Thoracic Aortic Aneurysms Using Single-Cell Transcriptomics. Arterioscler Thromb Vasc Biol 2022; 42:919-930. [PMID: 35708028 PMCID: PMC9339526 DOI: 10.1161/atvbaha.122.317484] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Thoracic aortic aneurysm is a life-threatening condition caused by weakening of the thoracic aorta wall, often developing silently until dissection or rupture occurs. Despite substantial efforts in the past decade, there have been no significant therapeutic advances to prevent or clinically manage diverse forms of thoracic aortic aneurysm and dissection with the only effective treatment being surgical repair. There is an urgent need to understand intra- and inter-aneurysmal heterogeneity underlying thoracic aortic aneurysm and dissection pathogenesis. The human aortic wall consists of many cell types and exhibits significant regional heterogeneity. High-throughput single-cell RNA sequencing has emerged as the principal tool to reveal the complexity in human tissues and clinical specimens. Recent single-cell RNA sequencing studies of different aortic cell populations both in vivo and in vitro began to dissect this complexity and have provided valuable information. In this review, we summarize these findings and discuss the potential applications of single-cell transcriptomics and related high-content technologies in human thoracic aortic aneurysm and dissection research, as well as the challenges associated with it.
Collapse
Affiliation(s)
- Dogukan Mizrak
- Department of Cardiac Surgery, University of Michigan, Ann Arbor (D.M., H.F., B.Y.)
| | - Hao Feng
- Department of Cardiac Surgery, University of Michigan, Ann Arbor (D.M., H.F., B.Y.).,Xiangya School of Medicine, Central South University, Changsha, China (H.F.)
| | - Bo Yang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor (D.M., H.F., B.Y.)
| |
Collapse
|
43
|
Bramel EE, Creamer TJ, Saqib M, Camejo Nunez WA, Bagirzadeh R, Roker LA, Goff LA, MacFarlane EG. Postnatal Smad3 Inactivation in Murine Smooth Muscle Cells Elicits a Temporally and Regionally Distinct Transcriptional Response. Front Cardiovasc Med 2022; 9:826495. [PMID: 35463747 PMCID: PMC9033237 DOI: 10.3389/fcvm.2022.826495] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
Heterozygous, loss of function mutations in positive regulators of the Transforming Growth Factor-β (TGF-β) pathway cause hereditary forms of thoracic aortic aneurysm. It is unclear whether and how the initial signaling deficiency triggers secondary signaling upregulation in the remaining functional branches of the pathway, and if this contributes to maladaptive vascular remodeling. To examine this process in a mouse model in which time-controlled, partial interference with postnatal TGF-β signaling in vascular smooth muscle cells (VSMCs) could be assessed, we used a VSMC-specific tamoxifen-inducible system, and a conditional allele, to inactivate Smad3 at 6 weeks of age, after completion of perinatal aortic development. This intervention induced dilation and histological abnormalities in the aortic root, with minor involvement of the ascending aorta. To analyze early and late events associated with disease progression, we performed a comparative single cell transcriptomic analysis at 10- and 18-weeks post-deletion, when aortic dilation is undetectable and moderate, respectively. At the early time-point, Smad3-inactivation resulted in a broad reduction in the expression of extracellular matrix components and critical components of focal adhesions, including integrins and anchoring proteins, which was reflected histologically by loss of connections between VSMCs and elastic lamellae. At the later time point, however, expression of several transcripts belonging to the same functional categories was normalized or even upregulated; this occurred in association with upregulation of transcripts coding for TGF-β ligands, and persistent downregulation of negative regulators of the pathway. To interrogate how VSMC heterogeneity may influence this transition, we examined transcriptional changes in each of the four VSMC subclusters identified, regardless of genotype, as partly reflecting the proximal-to-distal anatomic location based on in situ RNA hybridization. The response to Smad3-deficiency varied depending on subset, and VSMC subsets over-represented in the aortic root, the site most vulnerable to dilation, most prominently upregulated TGF-β ligands and pro-pathogenic factors such as thrombospondin-1, angiotensin converting enzyme, and pro-inflammatory mediators. These data suggest that Smad3 is required for maintenance of focal adhesions, and that loss of contacts with the extracellular matrix has consequences specific to each VSMC subset, possibly contributing to the regional susceptibility to dilation in the aorta.
Collapse
Affiliation(s)
- Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Predoctoral Training in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Muzna Saqib
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wendy A. Camejo Nunez
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Predoctoral Training in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Rustam Bagirzadeh
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - LaToya Ann Roker
- School of Medicine Microscope Facility, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Loyal A. Goff
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
44
|
Wilks BT, Evans EB, Howes A, Hopkins CM, Nakhla MN, Williams G, Morgan JR. Quantifying Cell-Derived Changes in Collagen Synthesis, Alignment, and Mechanics in a 3D Connective Tissue Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103939. [PMID: 35102708 PMCID: PMC8981917 DOI: 10.1002/advs.202103939] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/06/2021] [Indexed: 06/14/2023]
Abstract
Dysregulation of extracellular matrix (ECM) synthesis, organization, and mechanics are hallmark features of diseases like fibrosis and cancer. However, most in vitro models fail to recapitulate the three-dimensional (3D) multi-scale hierarchical architecture of collagen-rich tissues and as a result, are unable to mirror native or disease phenotypes. Herein, using primary human fibroblasts seeded into custom fabricated 3D non-adhesive agarose molds, a novel strategy is proposed to direct the morphogenesis of engineered 3D ring-shaped tissue constructs with tensile and histological properties that recapitulate key features of fibrous connective tissue. To characterize the shift from monodispersed cells to a highly-aligned, collagen-rich matrix, a multi-modal approach integrating histology, multiphoton second-harmonic generation, and electron microscopy is employed. Structural changes in collagen synthesis and alignment are then mapped to functional differences in tissue mechanics and total collagen content. Due to the absence of an exogenously added scaffolding material, this model enables the direct quantification of cell-derived changes in 3D matrix synthesis, alignment, and mechanics in response to the addition or removal of relevant biomolecular perturbations. To illustrate this, the effects of nutrient composition, fetal bovine serum, rho-kinase inhibitor, and pro- and anti-fibrotic compounds on ECM synthesis, 3D collagen architecture, and mechanophenotype are quantified.
Collapse
Affiliation(s)
- Benjamin T. Wilks
- Center for Biomedical EngineeringBrown UniversityProvidenceRI02129USA
- Center for Alternatives to Animals in TestingBrown UniversityProvidenceRI02129USA
- Present address:
Center for Engineering in Medicine & SurgeryHarvard Medical School & Massachusetts General HospitalBostonMA02114USA
| | | | - Andrew Howes
- Department of Molecular BiologyCell Biology & BiochemistryBrown UniversityProvidenceRI02129USA
| | - Caitlin M. Hopkins
- Center for Alternatives to Animals in TestingBrown UniversityProvidenceRI02129USA
- Department of Pathology & Laboratory MedicineBrown UniversityProvidenceRI02129USA
| | - Morcos N. Nakhla
- Department of Pathology & Laboratory MedicineBrown UniversityProvidenceRI02129USA
| | - Geoffrey Williams
- Department of Molecular BiologyCell Biology & BiochemistryBrown UniversityProvidenceRI02129USA
| | - Jeffrey R. Morgan
- Center for Biomedical EngineeringBrown UniversityProvidenceRI02129USA
- Center for Alternatives to Animals in TestingBrown UniversityProvidenceRI02129USA
- Department of Pathology & Laboratory MedicineBrown UniversityProvidenceRI02129USA
| |
Collapse
|
45
|
Yu Y, Shen H, Zhu J, Cao X, Li Q, Shao L, Shen Z. Generation of Marfan patient specific iPSCs (ICSSUi001-A) carrying a novel heterozygous mutation in FBN1 gene. Stem Cell Res 2022; 60:102720. [DOI: 10.1016/j.scr.2022.102720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 02/21/2022] [Indexed: 11/29/2022] Open
|
46
|
Development of vascular disease models to explore disease causation and pathomechanisms of rare vascular diseases. Semin Immunopathol 2022; 44:259-268. [PMID: 35233690 PMCID: PMC8887661 DOI: 10.1007/s00281-022-00925-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/10/2022] [Indexed: 12/15/2022]
Abstract
As the field of medicine is striving forward heralded by a new era of next-generation sequencing (NGS) and integrated technologies such as bioprinting and biological material development, the utility of rare monogenetic vascular disease modeling in this landscape is starting to emerge. With their genetic simplicity and broader applicability, these patient-specific models are at the forefront of modern personalized medicine. As a collective, rare diseases are a significant burden on global healthcare systems, and rare vascular diseases make up a significant proportion of this. High costs are due to a lengthy diagnostic process, affecting all ages from infants to adults, as well as the severity and chronic nature of the disease. Their complex nature requires sophisticated disease models and integrated approaches involving multidisciplinary teams. Here, we review these emerging vascular disease models, how they contribute to our understanding of the pathomechanisms in rare vascular diseases and provide useful platforms for therapeutic discovery.
Collapse
|
47
|
Granata A, Kasioulis I, Serrano F, Cooper JD, Traylor M, Sinha S, Markus HS. The Histone Deacetylase 9 Stroke-Risk Variant Promotes Apoptosis and Inflammation in a Human iPSC-Derived Smooth Muscle Cells Model. Front Cardiovasc Med 2022; 9:849664. [PMID: 35433850 PMCID: PMC9005977 DOI: 10.3389/fcvm.2022.849664] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/04/2022] [Indexed: 12/02/2022] Open
Abstract
A common variant in the Histone Deacetylase 9 (HDAC9) gene is the strongest genetic risk for large-vessel stroke, and HDAC9 offers a novel target for therapeutic modulation. However, the mechanisms linking the HDAC9 variant with increased stroke risk is still unclear due to the lack of relevant models to study the underlying molecular mechanisms. We generated vascular smooth muscle cells using human induced pluripotent stem cells with the HDAC9 stroke risk variant to assess HDAC9-mediated phenotypic changes in a relevant cells model and test the efficacy of HDAC inhibitors for potential therapeutic strategies. Our human induced pluripotent stem cells derived vascular smooth muscle cells show enhanced HDAC9 expression and allow us to assess HDAC9-mediated effects on promoting smooth muscle cell dysfunction, including proliferation, migration, apoptosis and response to inflammation. These phenotypes could be reverted by treatment with HDAC inhibitors, including sodium valproate and small molecules inhibitors. By demonstrating the relevance of the model and the efficacy of HDAC inhibitors, our model provides a robust phenotypic screening platform, which could be applied to other stroke-associated genetic variants.
Collapse
Affiliation(s)
- Alessandra Granata
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Ioannis Kasioulis
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Felipe Serrano
- Anne McLaren Laboratory, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - James D Cooper
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Sanjay Sinha
- Department of Medicine, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
48
|
Lu H, Du W, Ren L, Hamblin MH, Becker RC, Chen YE, Fan Y. Vascular Smooth Muscle Cells in Aortic Aneurysm: From Genetics to Mechanisms. J Am Heart Assoc 2021; 10:e023601. [PMID: 34796717 PMCID: PMC9075263 DOI: 10.1161/jaha.121.023601] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Aortic aneurysm, including thoracic aortic aneurysm and abdominal aortic aneurysm, is the second most prevalent aortic disease following atherosclerosis, representing the ninth-leading cause of death globally. Open surgery and endovascular procedures are the major treatments for aortic aneurysm. Typically, thoracic aortic aneurysm has a more robust genetic background than abdominal aortic aneurysm. Abdominal aortic aneurysm shares many features with thoracic aortic aneurysm, including loss of vascular smooth muscle cells (VSMCs), extracellular matrix degradation and inflammation. Although there are limitations to perfectly recapitulating all features of human aortic aneurysm, experimental models provide valuable tools to understand the molecular mechanisms and test novel therapies before human clinical trials. Among the cell types involved in aortic aneurysm development, VSMC dysfunction correlates with loss of aortic wall structural integrity. Here, we discuss the role of VSMCs in aortic aneurysm development. The loss of VSMCs, VSMC phenotypic switching, secretion of inflammatory cytokines, increased matrix metalloproteinase activity, elevated reactive oxygen species, defective autophagy, and increased senescence contribute to aortic aneurysm development. Further studies on aortic aneurysm pathogenesis and elucidation of the underlying signaling pathways are necessary to identify more novel targets for treating this prevalent and clinical impactful disease.
Collapse
Affiliation(s)
- Haocheng Lu
- Department of Internal MedicineCardiovascular CenterUniversity of Michigan Medical CenterAnn ArborMI
| | - Wa Du
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Lu Ren
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Milton H. Hamblin
- Department of PharmacologyTulane University School of MedicineNew OrleansLA
| | - Richard C. Becker
- Division of Cardiovascular Health and DiseaseDepartment of Internal MedicineUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Y. Eugene Chen
- Department of Internal MedicineCardiovascular CenterUniversity of Michigan Medical CenterAnn ArborMI
| | - Yanbo Fan
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOH
- Division of Cardiovascular Health and DiseaseDepartment of Internal MedicineUniversity of Cincinnati College of MedicineCincinnatiOH
| |
Collapse
|
49
|
Zhu G, Luo M, Chen Q, Zhang Y, Zhao K, Zhang Y, Shu C, Yang H, Zhou Z. Novel LTBP3 mutations associated with thoracic aortic aneurysms and dissections. Orphanet J Rare Dis 2021; 16:513. [PMID: 34906192 PMCID: PMC8670144 DOI: 10.1186/s13023-021-02143-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Thoracic aortic aneurysm and dissection (TAAD) is a hidden-onset but life-threatening disorder with high clinical variability and genetic heterogeneity. In recent years, an increasing number of genes have been identified to be related to TAAD. However, some genes remain uncertain because of limited case reports and/or functional studies. LTBP3 was such an ambiguous gene that was previously known for dental and skeletal dysplasia and then noted to be associated with TAAD. More research on individuals or families harboring variants in this gene would be helpful to obtain full knowledge of the disease and clarify its association with TAAD. METHODS A total of 266 TAAD probands with no causative mutations in known genes had been performed wholeexome sequencing (WES) to identify potentially pathogenic variants. In this study, rare LTBP3 variants were the focus of analysis. RESULTS Two compound heterozygous mutations, c.625dup (p.Leu209fs) and c.1965del (p.Arg656fs), in LTBP3 were identified in a TAAD patient along with short stature and dental problems, which was the first TAAD case with biallelic LTBP3 null mutations in an Asian population. Additionally, several rare heterozygous LTBP3 variants were also detected in other sporadic TAAD patients. CONCLUSION The identification of LTBP3 mutations in TAAD patients in our study provided more clinical evidence to support its association with TAAD, which broadens the gene spectrum of LTBP3. LTBP3 should be considered to be incorporated into the routine genetic analysis of heritable aortopathy, which might help to fully understand its phenotypic spectrum and improve the diagnostic rate of TAAD.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Qianlong Chen
- 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
| | - Yinhui Zhang
- 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
| | - Kun Zhao
- 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
| | - Yujing Zhang
- 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
| | - 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.
| | - 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
|
50
|
Gene Editing in Pluripotent Stem Cells and Their Derived Organoids. Stem Cells Int 2021; 2021:8130828. [PMID: 34887928 PMCID: PMC8651378 DOI: 10.1155/2021/8130828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/22/2021] [Indexed: 12/26/2022] Open
Abstract
With the rapid rise in gene-editing technology, pluripotent stem cells (PSCs) and their derived organoids have increasingly broader and practical applications in regenerative medicine. Gene-editing technologies, from large-scale nucleic acid endonucleases to CRISPR, have ignited a global research and development boom with significant implications in regenerative medicine. The development of regenerative medicine technologies, regardless of whether it is PSCs or gene editing, is consistently met with controversy. Are the tools for rewriting the code of life a boon to humanity or a Pandora's box? These technologies raise concerns regarding ethical issues, unexpected mutations, viral infection, etc. These concerns remain even as new treatments emerge. However, the potential negatives cannot obscure the virtues of PSC gene editing, which have, and will continue to, benefit mankind at an unprecedented rate. Here, we briefly introduce current gene-editing technology and its application in PSCs and their derived organoids, while addressing ethical concerns and safety risks and discussing the latest progress in PSC gene editing. Gene editing in PSCs creates visualized in vitro models, providing opportunities for examining mechanisms of known and unknown mutations and offering new possibilities for the treatment of cancer, genetic diseases, and other serious or refractory disorders. From model construction to treatment exploration, the important role of PSCs combined with gene editing in basic and clinical medicine studies is illustrated. The applications, characteristics, and existing challenges are summarized in combination with our lab experiences in this field in an effort to help gene-editing technology better serve humans in a regulated manner. Current preclinical and clinical trials have demonstrated initial safety and efficacy of PSC gene editing; however, for better application in clinical settings, additional investigation is warranted.
Collapse
|