1
|
Nguyen TD, Winek MA, Rao MK, Dhyani SP, Lee MY. Nuclear envelope components in vascular mechanotransduction: emerging roles in vascular health and disease. Nucleus 2025; 16:2453752. [PMID: 39827403 DOI: 10.1080/19491034.2025.2453752] [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: 10/08/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
The vascular network, uniquely sensitive to mechanical changes, translates biophysical forces into biochemical signals for vessel function. This process relies on the cell's architectural integrity, enabling uniform responses to physical stimuli. Recently, the nuclear envelope (NE) has emerged as a key regulator of vascular cell function. Studies implicate nucleoskeletal elements (e.g. nuclear lamina) and the linker of nucleoskeleton and cytoskeleton (LINC) complex in force transmission, emphasizing nucleo-cytoskeletal communication in mechanotransduction. The nuclear pore complex (NPC) and its component proteins (i.e. nucleoporins) also play roles in cardiovascular disease (CVD) progression. We herein summarize evidence on the roles of nuclear lamina proteins, LINC complex members, and nucleoporins in endothelial and vascular cell mechanotransduction. Numerous studies attribute NE components in cytoskeletal-related cellular behaviors to insinuate dysregulation of nucleocytoskeletal feedback and nucleocytoplasmic transport as a mechanism of endothelial and vascular dysfunction, and hence implications for aging and vascular pathophysiology.
Collapse
Affiliation(s)
- Tung D Nguyen
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
- The Center for Cardiovascular Research, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Michael A Winek
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Mihir K Rao
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Shaiva P Dhyani
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Monica Y Lee
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
- The Center for Cardiovascular Research, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Rouhi L. Cardiac phenotypes in LMNA mutations. Curr Opin Cardiol 2025; 40:131-138. [PMID: 39998502 PMCID: PMC11968229 DOI: 10.1097/hco.0000000000001209] [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] [Indexed: 02/26/2025]
Abstract
PURPOSE OF REVIEW This review highlights the diverse cardiac manifestations of LMNA mutations, focusing on their underlying molecular mechanisms and clinical implications. As LMNA mutations are implicated in cardiomyopathies, such as dilated cardiomyopathy (DCM), arrhythmogenic cardiomyopathy (ARVC), and conduction system diseases, understanding these phenotypes is critical for advancing diagnosis and management strategies. RECENT FINDINGS Recent studies reveal that LMNA mutations disrupt nuclear envelope stability, activating the DNA damage response (DDR) and compromising chromatin organization and mechanotransduction. Mouse models have elucidated pathways linking LMNA dysfunction to fibrosis, arrhythmias, and myocardial remodeling. Emerging evidence demonstrates that fibroblasts play a crucial role in cardiac phenotypes. Advances in genetic screening have also underscored the importance of early identification and risk stratification, particularly for arrhythmias and sudden cardiac death. SUMMARY The diverse spectrum of LMNA-related cardiac phenotypes, from isolated conduction defects to severe DCM and ARVC, underscores the necessity of personalized care strategies. Bridging insights from molecular studies and clinical research paves the way for targeted therapies to slow disease progression and improve patient outcomes. Future efforts should prioritize translational research on molecular mechanisms with potential in mouse models, alongside a deeper exploration of genotype-phenotype correlations, to refine and implement effective therapeutic interventions.
Collapse
Affiliation(s)
- Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| |
Collapse
|
3
|
Buja LM, McDonald MM, Zhao B, Narula N, Narula J, Barth RF. Insights from autopsy-initiated pathological studies of the pathogenesis and clinical manifestations of atherosclerosis and ischemic heart disease: Part I. Atherosclerosis. Cardiovasc Pathol 2025; 76:107726. [PMID: 39971218 DOI: 10.1016/j.carpath.2025.107726] [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: 09/04/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025] Open
Abstract
CONTEXT Ischemic heart disease (IHD) due to coronary atherosclerosis constitutes the leading cause of morbidity and mortality worldwide. This review was undertaken to document the historical basis for our contemporary understanding of atherosclerosis-based disease and to provide a rationale for continued support for autopsy-based research to make further progress in reducing the morbidity and mortality from atherosclerosis-related disease. OBJECTIVES To analyze the contributions of the autopsy-initiated pathological studies to complement and validate other lines of investigation in determining the pathology and pathogenesis of the leading worldwide cause of morbidity and mortality, namely, atherosclerosis and its major complications of coronary atherosclerosis, ischemic heart disease, coronary thrombosis, acute myocardial infarction, and sudden cardiac death. DATA SOURCES Systematic search on PubMed to gather relevant studies concerning autopsy studies and reviews of the pathology and pathogenesis of atherosclerosis, ischemic heart disease, coronary atherosclerosis, coronary thrombosis, myocardial infarction, and sudden cardiac death CONCLUSIONS: Extensive published reports have confirmed the continuing importance of the autopsy as a powerful tool to understand the pathogenesis, clinical features, and therapeutic options for major diseases. This specifically has been shown by the analysis of atherosclerosis and its major manifestation of ischemic heart disease, as presented in this (Part I) and its companion (Part II) review. Autopsy-initiated pathological studies have documented the prevalence and natural history of atherosclerosis in different human populations in relationship to the prevalence of risk factors and established that the clinically silent phase of the disease begins in the first decades of life. Insights from these studies have been essential in developing and evaluating strategies for continued progress in preventing and controlling the disability and death associated with atherosclerotic heart disease.
Collapse
Affiliation(s)
- L Maximilian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA.
| | | | - Bihong Zhao
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA
| | - Navneet Narula
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA
| | - Jagat Narula
- Division of Cardiology, Department of Internal Medicine, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA
| | - Rolf F Barth
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
4
|
Abe J, Chau K, Mojiri A, Wang G, Oikawa M, Samanthapudi VSK, Osborn AM, Ostos-Mendoza KC, Mariscal-Reyes KN, Mathur T, Jain A, Herrmann J, Yusuf SW, Krishnan S, Deswal A, Lin SH, Kotla S, Cooke JP, Le NT. Impacts of Radiation on Metabolism and Vascular Cell Senescence. Antioxid Redox Signal 2025. [PMID: 40233257 DOI: 10.1089/ars.2024.0741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Significance: This review investigates how radiation therapy (RT) increases the risk of delayed cardiovascular disease (CVD) in cancer survivors. Understanding the mechanisms underlying radiation-induced CVD is essential for developing targeted therapies to mitigate these effects and improve long-term outcomes for patients with cancer. Recent Advances: Recent studies have primarily focused on metabolic alterations induced by irradiation in various cancer cell types. However, there remains a significant knowledge gap regarding the role of chronic metabolic alterations in normal cells, particularly vascular cells, in the progression of CVD after RT. Critical Issues: This review centers on RT-induced metabolic alterations in vascular cells and their contribution to senescence accumulation and chronic inflammation across the vasculature post-RT. We discuss key metabolic pathways, including glycolysis, the tricarboxylic acid cycle, lipid metabolism, glutamine metabolism, and redox metabolism (nicotinamide adenine dinucleotide/Nicotinamide adenine dinucleotide (NADH) and nicotinamide adenine dinucleotide phosphate (NADP+)/NADPH). We further explore the roles of regulatory proteins such as p53, adenosine monophosphate-activated protein kinase, and mammalian target of rapamycin in driving these metabolic dysregulations. The review emphasizes the impact of immune-vascular crosstalk mediated by the senescence-associated secretory phenotype, which perpetuates metabolic dysfunction, enhances chronic inflammation, drives senescence accumulation, and causes vascular damage, ultimately contributing to cardiovascular pathogenesis. Future Directions: Future research should prioritize identifying therapeutic targets within these metabolic pathways or the immune-vascular interactions influenced by RT. Correcting metabolic dysfunction and reducing chronic inflammation through targeted therapies could significantly improve cardiovascular outcomes in cancer survivors. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Junichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Khanh Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anahita Mojiri
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Masayoshi Oikawa
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Venkata S K Samanthapudi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abigail M Osborn
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | - Tammay Mathur
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas, USA
| | - Abhishek Jain
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas, USA
- Department of Medical Physiology, School of Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven H Lin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
5
|
Benedicto I, Hamczyk MR, Dorado B, Andrés V. Vascular cell types in progeria: victims or villains? Trends Mol Med 2025:S1471-4914(25)00056-5. [PMID: 40240194 DOI: 10.1016/j.molmed.2025.03.005] [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: 01/15/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an ultrarare genetic disease caused by progerin, a broadly expressed mutant variant of lamin A protein that accelerates aging and leads to premature death typically in adolescence. Progerin affects many organs and reproduces many characteristics of physiological aging, with the main cause of death in HGPS being atherosclerotic cardiovascular disease (CVD). Due to the rarity of HGPS, advances in understanding the disease and progress toward new therapeutic approaches are crucially dependent on preclinical models. We discuss recent research developments from a variety of HGPS experimental systems, with a special focus on in vivo studies of the role of vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) that are key players in atherosclerosis.
Collapse
Affiliation(s)
- Ignacio Benedicto
- Centro de Investigaciones Biológicas Margarita Salas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.
| | - Magda R Hamczyk
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; Aarhus Institute of Advanced Studies (AIAS), Aarhus University, 8000 Aarhus C, Denmark
| | - Beatriz Dorado
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| |
Collapse
|
6
|
Bonneau L, Rupin-Mas M, Descamps M, Vincent M, Romefort B, Van Bogaert P. Intracranial hypertension in a patient with Hutchinson-Gilford progeria syndrome. Arch Pediatr 2025; 32:210-212. [PMID: 39988546 DOI: 10.1016/j.arcped.2024.12.006] [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: 10/01/2024] [Revised: 12/17/2024] [Accepted: 12/25/2024] [Indexed: 02/25/2025]
Abstract
Hutchinson-Gilford syndrome, also known as progeria, is a rare genetic disorder that causes premature and accelerated ageing from the neonatal period. The disease is caused by de novo mutations in the LMNA gene. Patients present with a range of symptoms, including skin, bone, joint and cardiac disorders, as well as characteristic facial dysmorphia. Intracranial hypertension is not a known symptom of this disease. To the best of our knowledge, no case of a patient with Hutchinson-Gilford syndrome presenting with intracranial hypertension without a traumatic context has been reported in the literature to date. This report presents the case of a child diagnosed with Hutchinson-Gilford syndrome who presented with intracranial hypertension at the age of three years, with no secondary cause identified. Long-term treatment with acetazolamide was required to control the intracranial hypertension. We hypothesise that the intracranial hypertension may be related to vascular abnormalities observed in Hutchinson-Gilford syndrome, which may cause a venous drainage dysfunction. To support the hypothesis of a non-random association between intracranial hypertension and Hutchinson-Gilford syndrome, further reports of similar cases in children with the syndrome are required.
Collapse
Affiliation(s)
- Léa Bonneau
- Department of Pediatric Neurology, Angers University Hospital, 4 rue Larrey, 49100 Angers, France.
| | - Maïlys Rupin-Mas
- Department of Pediatric Neurology, Angers University Hospital, 4 rue Larrey, 49100 Angers, France.
| | - Magali Descamps
- Pediatric ophtalmology, department of Pediatry, Angers University Hospital, 4 rue Larrey, 49100 Angers, France.
| | - Marie Vincent
- Department of Genetics, Nantes University Hospital, 38 bd Jean-Monnet rez-de-chaussée 44093 Nantes, France.
| | - Bénédicte Romefort
- Department of Pediatric Cardiology, Nantes University Hospital, 38 bd Jean-Monnet rez-de-chaussée 44093 Nantes, France.
| | - Patrick Van Bogaert
- Department of Pediatric Neurology, Angers University Hospital, 4 rue Larrey, 49100 Angers, France; Laboratoire Angevin de Recherche en Ingénierie des Systèmes (LARIS), University of Angers, 62 avenue Notre Dame du Lac 49000 Angers, France.
| |
Collapse
|
7
|
Abutaleb NO, Gao XD, Bedapudi A, Choi L, Shores KL, Kennedy C, Duby JE, Cao K, Liu DR, Truskey GA. Adenine base editing rescues pathogenic phenotypes in tissue engineered vascular model of Hutchinson-Gilford progeria syndrome. APL Bioeng 2025; 9:016110. [PMID: 40027545 PMCID: PMC11871533 DOI: 10.1063/5.0244026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/07/2025] [Indexed: 03/05/2025] Open
Abstract
The rare, accelerated aging disease Hutchinson-Gilford Progeria Syndrome (HGPS) is commonly caused by a de novo c.1824 C > T point mutation of the LMNA gene that results in the protein progerin. The primary cause of death is a heart attack or stroke arising from atherosclerosis. A characteristic feature of HGPS arteries is loss of smooth muscle cells. An adenine base editor (ABE7.10max) corrected the point mutation and produced significant improvement in HGPS mouse lifespan, vascular smooth muscle cell density, and adventitial fibrosis. To assess whether base editing correction of human HGPS tissue engineered blood vessels (TEBVs) prevents the HGPS vascular phenotype and to identify the minimum fraction of edited smooth muscle cells needed to effect such changes, we transduced HGPS iPSCs with lentivirus containing ABE7.10max. Endothelial cells (viECs) and smooth muscle cells (viSMCs) obtained by differentiation of edited HGPS iPSCs did not express progerin and had double-stranded DNA breaks and reactive oxygen species at the same levels as healthy viSMCs and viECs. Editing HGPSviECs restored a normal response to shear stress. Normal vasodilation and viSMC density were restored in TEBVs made with edited cells. When TEBVs were prepared with at least 50% edited smooth muscle cells, viSMC proliferation and myosin heavy chain levels significantly improved. Sequencing of TEBV cells after perfusion indicated an enrichment of edited cells after 5 weeks of perfusion when they comprised 50% of the initial number of cells in the TEBVs. Thus, base editing correction of a fraction of HGPS vascular cells improves human TEBV phenotype.
Collapse
Affiliation(s)
- Nadia O. Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | | | - Akhil Bedapudi
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Leandro Choi
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Kevin L. Shores
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Crystal Kennedy
- University Program in Genetics and Genomics, Duke University, Durham, North Carolina 27708, USA
| | | | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland 20742, USA
| | | | - George A. Truskey
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| |
Collapse
|
8
|
Zaruba MM, Angermann R, Staggl S, Jeyakumar V, Mair S, Stöckl V, Neyer J, Maurer T, Ungericht M, Gavranovic-Novakovic J, Bauer A, Zehetner C, Messner M. Progerin mRNA Is Associated with Smoking and Signs of Increased Microvascular Damage in Patients with Diabetic Macular Edema. Int J Mol Sci 2025; 26:2099. [PMID: 40076719 PMCID: PMC11900628 DOI: 10.3390/ijms26052099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
The premature aging disease Hutchinson-Gilford Syndrome (HGPS) is caused by defined mutations in the LMNA gene, resulting in the activation of a cryptic splice donor site, which leads to a defective truncated prelamin A protein called progerin. Notably, progerin expression has also been detected in non-mutated healthy individuals, and therefore, its involvement in the physiological aging process has been widely discussed. Since diabetes mellitus is associated with premature aging and increased cardiovascular mortality, we aimed to investigate the role of progerin expression in patients with diabetic retinopathy (DR). mRNA expression of progerin was analyzed in blood samples from 140 patients with DR who received anti-vascular endothelial growth factor (VEGF) therapy. Progerin mRNA levels were significantly lower in female compared to male patients (n = 42 vs. n = 98; 0.67 ± 0.19 vs. 0.89 ± 0.51, p = 0.006) and higher in patients with non-proliferative (NP)DR (n = 87 vs. n = 53; 0.9 ± 0.51 vs. 0.71 ± 0.29, p = 0.013) compared to those with proliferative (P)DR. Additionally, a positive correlation was found between progerin mRNA expression and the number of intravitreal anti-VEGF applications (n = 139, r = 0.21, p = 0.015), central macula thickness (CMT), (n = 137, r = 0.18, p = 0.036) and nicotine consumption (n = 105, r = 0.235, p = 0.002). The nuclear localization and significant upregulation of progerin mRNA and protein levels in dermal fibroblasts from HGPS donors emphasize its role in cellular aging mechanisms. Progerin mRNA levels were higher in patients with NPDR. CMT, number of intravitreal anti-VEGF therapy treatments, and cigarette consumption were positively related to progerin mRNA, suggesting an association with disease progression and premature aging.
Collapse
Affiliation(s)
- Marc-Michael Zaruba
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria (V.J.); (J.G.-N.)
| | - Reinhard Angermann
- Department of Ophthalmology, Medical University Innsbruck, 6020 Innsbruck, Austria
- Department of Ophthalmology, LK Mistelbach/Gänserndorf, 2130 Mistelbach, Austria
| | - Simon Staggl
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria (V.J.); (J.G.-N.)
| | - Vivek Jeyakumar
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria (V.J.); (J.G.-N.)
| | - Sofia Mair
- Department of Ophthalmology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Victoria Stöckl
- Department of Ophthalmology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Julia Neyer
- Department of Ophthalmology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Thomas Maurer
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria (V.J.); (J.G.-N.)
| | - Maria Ungericht
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria (V.J.); (J.G.-N.)
| | - Jasmina Gavranovic-Novakovic
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria (V.J.); (J.G.-N.)
| | - Axel Bauer
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria (V.J.); (J.G.-N.)
| | - Claus Zehetner
- Department of Ophthalmology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Moritz Messner
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria (V.J.); (J.G.-N.)
| |
Collapse
|
9
|
Ho CY, Wu MY, Thammaphet J, Ahmad S, Ho C.S. J, Draganova L, Anderson G, Jonnalagadda US, Hayward R, Shroff R, Wen WTL, Verhulst A, Foo RSY, Shanahan CM. Mineral Stress Drives Loss of Heterochromatin: An Early Harbinger of Vascular Inflammaging and Calcification. Circ Res 2025; 136:379-399. [PMID: 39840455 PMCID: PMC11825498 DOI: 10.1161/circresaha.124.325374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Vascular calcification is a detrimental aging pathology markedly accelerated in patients with chronic kidney disease. PLA (prelamin A) is a biomarker of vascular smooth muscle cell aging that accelerates calcification however the mechanisms remain undefined. METHODS Vascular smooth muscle cells were transduced with PLA using an adenoviral vector and epigenetic modifications were monitored using immunofluorescence and targeted polymerase chain reaction array. Epigenetic findings were verified in vivo using immunohistochemistry in human vessels, in a mouse model of inducible prelamin A expression, and in a rat model of chronic kidney disease-induced calcification. Transcriptomic and chromatin immunoprecipitation followed by sequencing analyses were used to identify gene targets impacted by changes in the epigenetic landscape. Molecular tools and antibody arrays were used to monitor the effects of mineral dysregulation on heterochromatin, inflammation, aging, and calcification. RESULTS Here, we report that depletion of the repressive heterochromatin marks, H3K9me3 (histone H3, lysine 9, trimethylation) and H3K27me3 (histone H3, lysine 27,trimethylation), is an early hallmark of vascular aging induced by both nuclear lamina dysfunction and dysregulated mineral metabolism, which act to modulate the expression of key epigenetic writers and erasers. Global analysis of H3K9me3 and H3K27me3 marks and pathway analysis revealed deregulation of insulin signaling and autophagy pathways as well as cross-talking DNA damage and NF-κB (nuclear factor κB) inflammatory pathways consistent with early activation of the senescence-associated secretory phenotype. Expression of PLA in vivo induced loss of heterochromatin and promoted inflammation and osteogenic differentiation which preceded aging indices, such as DNA damage and senescence. Vessels from children on dialysis and rats with chronic kidney disease showed prelamin A accumulation and accelerated loss of heterochromatin before the onset of calcification. CONCLUSIONS Dysregulated mineral metabolism drives changes in the epigenetic landscape and nuclear lamina dysfunction that together promote early induction of inflammaging pathways priming the vasculature for downstream pathological change.
Collapse
MESH Headings
- Animals
- Heterochromatin/metabolism
- Heterochromatin/pathology
- Heterochromatin/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Humans
- Epigenesis, Genetic
- Rats
- Mice
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cellular Senescence
- Inflammation/metabolism
- Inflammation/pathology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/genetics
- Histones/metabolism
- Mice, Inbred C57BL
- Cells, Cultured
- Disease Models, Animal
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Lamin Type A
Collapse
Affiliation(s)
- Chin Yee Ho
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Meng-Ying Wu
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Jirapath Thammaphet
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Sadia Ahmad
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - James Ho C.S.
- Nanyang Technological University, Singapore (J.H.C.S., U.S.J.)
| | - Lilia Draganova
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Grace Anderson
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | | | - Robert Hayward
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Rukshana Shroff
- Nephrology Unit, Great Ormond Street Hospital and University College London Institute of Child Health, United Kingdom (R.S.)
| | - Wilson Tan Lek Wen
- Cardiovascular Disease Translational Research Programme, National University of Singapore Yong Loo Lin School of Medicine (W.T.L.W., R.F.)
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium (A.V.)
| | - Roger SY. Foo
- Cardiovascular Disease Translational Research Programme, National University of Singapore Yong Loo Lin School of Medicine (W.T.L.W., R.F.)
| | - Catherine M. Shanahan
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| |
Collapse
|
10
|
Vakili S, Izydore EK, Losert L, Cabral WA, Tavarez UL, Shores K, Xue H, Erdos MR, Truskey GA, Collins FS, Cao K. Angiopoietin-2 reverses endothelial cell dysfunction in progeria vasculature. Aging Cell 2025; 24:e14375. [PMID: 39422121 PMCID: PMC11822663 DOI: 10.1111/acel.14375] [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: 05/09/2024] [Revised: 08/31/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disorder in children caused by a point mutation in the lamin A gene, resulting in a toxic form of lamin A called progerin. Accelerated atherosclerosis leading to heart attack and stroke are the major causes of death in these patients. Endothelial cell (EC) dysfunction contributes to the pathogenesis of HGPS related cardiovascular diseases (CVD). Endothelial cell-cell communications are important in the development of the vasculature, and their disruptions contribute to cardiovascular pathology. However, it is unclear how progerin interferes with such communications that lead to vascular dysfunction. An antibody array screening of healthy and HGPS patient EC secretomes identified Angiopoietin-2 (Ang2) as a down-regulated signaling molecule in HGPS ECs. A similar down-regulation of Ang2 mRNA and protein was detected in the aortas from an HGPS mouse model. Addition of Ang2 to HGPS ECs rescues vasculogenesis, normalizes endothelial cell migration and gene expression, and restores nitric oxide bioavailability through eNOS activation. Furthermore, Ang2 addition reverses unfavorable paracrine effects of HGPS ECs on vascular smooth muscle cells. Lastly, by utilizing adenine base editor (ABE)-corrected HGPS ECs and progerin-expressing HUVECs, we demonstrated a negative correlation between progerin and Ang2 expression. Lastly, our results indicated that Ang2 exerts its beneficial effect in ECs through Tie2 receptor binding, activating an Akt-mediated pathway. Together, these results provide molecular insights into EC dysfunction in HGPS and suggest that Ang2 treatment has potential therapeutic effects in HGPS-related CVD.
Collapse
Affiliation(s)
- Sahar Vakili
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| | - Elizabeth K. Izydore
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| | - Leonhard Losert
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| | - Wayne A. Cabral
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Urraca L. Tavarez
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Kevin Shores
- Department of Biomedical EngineeringDuke UniversityDurhamNorth CarolinaUSA
| | - Huijing Xue
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
- Frederick National Laboratory for Cancer ResearchFrederickMarylandUSA
| | - Michael R. Erdos
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - George A. Truskey
- Department of Biomedical EngineeringDuke UniversityDurhamNorth CarolinaUSA
| | - Francis S. Collins
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Kan Cao
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| |
Collapse
|
11
|
Benedicto I, Hamczyk MR, Nevado RM, Barettino A, Carmona RM, Espinós‐Estévez C, Gonzalo P, de la Fuente‐Pérez M, Andrés‐Manzano MJ, González‐Gómez C, Dorado B, Andrés V. Endothelial cell-specific progerin expression does not cause cardiovascular alterations and premature death. Aging Cell 2025; 24:e14389. [PMID: 39479939 PMCID: PMC11822624 DOI: 10.1111/acel.14389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder caused by a mutation in the LMNA gene that provokes the synthesis of progerin, a mutant version of the nuclear protein lamin A that accelerates aging and precipitates death. The most clinically relevant feature of HGPS is the development of cardiac anomalies and severe vascular alterations, including massive loss of vascular smooth muscle cells, increased fibrosis, and generalized atherosclerosis. However, it is unclear if progerin expression in endothelial cells (ECs) causes the cardiovascular manifestations of HGPS. To tackle this question, we generated atherosclerosis-free mice (LmnaLCS/LCSCdh5-CreERT2) and atheroprone mice (Apoe-/-LmnaLCS/LCSCdh5-CreERT2) with EC-specific progerin expression. Like progerin-free controls, LmnaLCS/LCSCdh5-CreERT2 mice did not develop heart fibrosis or cardiac electrical and functional alterations, and had normal vascular structure, body weight, and lifespan. Similarly, atheroprone Apoe-/-LmnaLCS/LCSCdh5-CreERT2 mice showed no alteration in body weight or lifespan versus Apoe-/-LmnaLCS/LCS controls and did not develop vascular alterations or aggravated atherosclerosis. Our results indicate that progerin expression in ECs is not sufficient to cause the cardiovascular phenotype and premature death associated with progeria.
Collapse
Affiliation(s)
- Ignacio Benedicto
- Centro de Investigaciones Biológicas Margarita Salas (CIB), Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Magda R. Hamczyk
- CIBER en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA)Universidad de OviedoOviedoSpain
- Aarhus Institute of Advanced Studies (AIAS)Aarhus UniversityAarhusDenmark
| | - Rosa M. Nevado
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- CIBER en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Ana Barettino
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- CIBER en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Rosa M. Carmona
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | | | - Pilar Gonzalo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- CIBER en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | | | - María J. Andrés‐Manzano
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- CIBER en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Cristina González‐Gómez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- CIBER en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Beatriz Dorado
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- CIBER en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- CIBER en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| |
Collapse
|
12
|
Wang Z, Wu J, Lv Z, Liang P, Li Q, Li Y, Guo Y. LMNA-related cardiomyopathy: From molecular pathology to cardiac gene therapy. J Adv Res 2025:S2090-1232(25)00001-3. [PMID: 39827909 DOI: 10.1016/j.jare.2025.01.001] [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: 11/19/2024] [Revised: 12/29/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND The genetic variants of LMNA cause an array of diseases that often affect the heart. LMNA-related cardiomyopathy exhibits high-penetrance and early-onset phenotypes that lead to late-stage heart failure or lethal arrhythmia. As a subtype of dilated cardiomyopathy and arrhythmogenic cardiomyopathy, LMNA-related cardiac dysfunction is resistant to existing cardiac therapeutic strategies, leaving a major unmet clinical need in cardiomyopathy management. AIM OF REVIEW Here we comprehensively summarize current knowledge about the genetic basis, disease models and pathological mechanisms of LMNA-related cardiomyopathy. Recent translational studies were highlighted to indicate new therapeutic modalities such as gene supplementation, gene silencing and genome editing therapy, which offer potential opportunities to overcome the difficulties in the development of specific drugs for this disease. KEY SCIENTIFIC CONCEPTS OF REVIEW LMNA-related cardiomyopathy involves many diverse disease mechanisms that preclude small-molecule drugs that target only a small fraction of the mechanisms. Agreeing to this notion, the first-in-human clinical trial for this disease recently reported futility. By contrast, gene therapy offers the new hope to directly intervene LMNA variants and demonstrates a tremendous potential for breakthrough therapy for this disease. Concepts in this review are also applicable to studies of other genetic diseases that lack effective therapeutics.
Collapse
Affiliation(s)
- Ze Wang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Jiahao Wu
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyuan Lv
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Ping Liang
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China.
| | - Qirui Li
- Department of Cardiology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| | - Yifei Li
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Yuxuan Guo
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| |
Collapse
|
13
|
Iskandar M, Xiao Barbero M, Jaber M, Chen R, Gomez-Guevara R, Cruz E, Westerheide S. A Review of Telomere Attrition in Cancer and Aging: Current Molecular Insights and Future Therapeutic Approaches. Cancers (Basel) 2025; 17:257. [PMID: 39858038 PMCID: PMC11764024 DOI: 10.3390/cancers17020257] [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: 11/07/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES As cells divide, telomeres shorten through a phenomenon known as telomere attrition, which leads to unavoidable senescence of cells. Unprotected DNA exponentially increases the odds of mutations, which can evolve into premature aging disorders and tumorigenesis. There has been growing academic and clinical interest in exploring this duality and developing optimal therapeutic strategies to combat telomere attrition in aging and cellular immortality in cancer. The purpose of this review is to provide an updated overview of telomere biology and therapeutic tactics to address aging and cancer. METHODS We used the Rayyan platform to review the PubMed database and examined the ClinicalTrial.gov registry to gain insight into clinical trials and their results. RESULTS Cancer cells activate telomerase or utilize alternative lengthening of telomeres to escape telomere shortening, leading to near immortality. Contrarily, normal cells experience telomeric erosion, contributing to premature aging disorders, such as Werner syndrome and Hutchinson-Gilford Progeria, and (2) aging-related diseases, such as neurodegenerative and cardiovascular diseases. CONCLUSIONS The literature presents several promising therapeutic approaches to potentially balance telomere maintenance in aging and shortening in cancer. This review highlights gaps in knowledge and points to the potential of these optimal interventions in preclinical and clinical studies to inform future research in cancer and aging.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sandy Westerheide
- Department of Molecular Biosciences, University of South Florida, 4202 East Fowler Avenue, ISA2015, Tampa, FL 33620, USA; (M.I.); (M.X.B.); (M.J.); (R.C.); (R.G.-G.); (E.C.)
| |
Collapse
|
14
|
Vakili S, Cao K. Angiopoietin-2: A Therapeutic Target for Vascular Protection in Hutchinson-Gilford Progeria Syndrome. Int J Mol Sci 2024; 25:13537. [PMID: 39769300 PMCID: PMC11676795 DOI: 10.3390/ijms252413537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a pediatric condition characterized by clinical features that resemble accelerated aging. The abnormal accumulation of a toxic form of the lamin A protein known as progerin disrupts cellular functions, leading to various complications, including growth retardation, loss of subcutaneous fat, abnormal skin, alopecia, osteoporosis, and progressive joint contractures. Death primarily occurs as the result of complications from progressive atherosclerosis, especially from cardiac disease, such as myocardial infarction or heart failure, or cerebrovascular disease like stroke. Despite the availability of lonafarnib, the only US Food and Drug Administration-approved treatment for HGPS, cardiovascular complications remain the leading cause of morbidity and mortality in affected patients. Defective angiogenesis-the process of forming new blood vessels from existing ones-plays a crucial role in the development of cardiovascular disease. A recent study suggests that Angiopoietin-2 (Ang2), a pro-angiogenic growth factor that regulates angiogenesis and vascular stability, may offer therapeutic potential for the treatment of HGPS. In this review, we describe the clinical features and key cellular processes impacted by progerin and discuss the therapeutic potential of Ang2 in addressing these challenges.
Collapse
Affiliation(s)
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA;
| |
Collapse
|
15
|
Hemmati F, Akinpelu A, Nweze DC, Mistriotis P. 3D confinement alters smooth muscle cell responses to chemical and mechanical cues. APL Bioeng 2024; 8:046103. [PMID: 39464377 PMCID: PMC11512639 DOI: 10.1063/5.0225569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Smooth muscle cell (SMC) phenotypic switching is a hallmark of many vascular diseases. Although prior work has established that chemical and mechanical cues contribute to SMC phenotypic switching, the impact of three-dimensional (3D) confinement on this process remains elusive. Yet, in vivo, arterial SMCs reside within confined environments. In this study, we designed a microfluidic assay to investigate the interplay between 3D confinement and different environmental stimuli in SMC function. Our results show that tightly, but not moderately, confined SMCs acquire a contractile phenotype when exposed to collagen I. Elevated compressive forces induced by hydrostatic pressure abolish this upregulation of the contractile phenotype and compromise SMC survival, particularly in tightly confined spaces. Transforming growth factor beta 1, which promotes the contractile state in moderate confinement, fails to enhance the contractility of tightly confined cells. Fibronectin and engagement of cadherin 2 suppress the contractile phenotype of SMCs regardless of the degree of confinement. In contrast, homophilic engagement of cadherin 11 upregulates SMC-specific genes and enhances contractility in both moderately and tightly confined cells. Overall, our work introduces 3D confinement as a regulator of SMC phenotypic responses to chemical and mechanical signals.
Collapse
Affiliation(s)
- Farnaz Hemmati
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | - Ayuba Akinpelu
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | - Daniel Chinedu Nweze
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | | |
Collapse
|
16
|
Hamczyk MR, Nevado RM, Gonzalo P, Andrés-Manzano MJ, Nogales P, Quesada V, Rosado A, Torroja C, Sánchez-Cabo F, Dopazo A, Bentzon JF, López-Otín C, Andrés V. Endothelial-to-Mesenchymal Transition Contributes to Accelerated Atherosclerosis in Hutchinson-Gilford Progeria Syndrome. Circulation 2024; 150:1612-1630. [PMID: 39206565 DOI: 10.1161/circulationaha.123.065768] [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: 05/29/2023] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Atherosclerosis is the main medical problem in Hutchinson-Gilford progeria syndrome, a rare premature aging disorder caused by the mutant lamin-A protein progerin. Recently, we found that limiting progerin expression to vascular smooth muscle cells (VSMCs) is sufficient to hasten atherosclerosis and death in Apoe-deficient mice. However, the impact of progerin-driven VSMC defects on endothelial cells (ECs) remained unclear. METHODS Apoe- or Ldlr-deficient C57BL/6J mice with ubiquitous, VSMC-, EC- or myeloid-specific progerin expression fed a normal or high-fat diet were used to study endothelial phenotype during Hutchinson-Gilford progeria syndrome-associated atherosclerosis. Endothelial permeability to low-density lipoproteins was assessed by intravenous injection of fluorescently labeled human low-density lipoprotein and confocal microscopy analysis of the aorta. Leukocyte recruitment to the aortic wall was evaluated by en face immunofluorescence. Endothelial-to-mesenchymal transition (EndMT) was assessed by quantitative polymerase chain reaction and RNA sequencing in the aortic intima and by immunofluorescence in aortic root sections. TGFβ (transforming growth factor β) signaling was analyzed by multiplex immunoassay in serum, by Western blot in the aorta, and by immunofluorescence in aortic root sections. The therapeutic benefit of TGFβ1/SMAD3 pathway inhibition was evaluated in mice by intraperitoneal injection of SIS3 (specific inhibitor of SMAD3), and vascular phenotype was assessed by Oil Red O staining, histology, and immunofluorescence in the aorta and the aortic root. RESULTS Both ubiquitous and VSMC-specific progerin expression in Apoe-null mice provoked alterations in aortic ECs, including increased permeability to low-density lipoprotein and leukocyte recruitment. Atherosclerotic lesions in these progeroid mouse models, but not in EC- and myeloid-specific progeria models, contained abundant cells combining endothelial and mesenchymal features, indicating extensive EndMT triggered by dysfunctional VSMCs. Accordingly, the intima of ubiquitous and VSMC-specific progeroid models at the onset of atherosclerosis presented increased expression of EndMT-linked genes, especially those specific to fibroblasts and extracellular matrix. Aorta in both models showed activation of the TGFβ1/SMAD3 pathway, a major trigger of EndMT, and treatment of VSMC-specific progeroid mice with SIS3 alleviated the aortic phenotype. CONCLUSIONS Progerin-induced VSMC alterations promote EC dysfunction and EndMT through TGFβ1/SMAD3, identifying this process as a candidate target for Hutchinson-Gilford progeria syndrome treatment. These findings also provide insight into the complex role of EndMT during atherogenesis.
Collapse
Affiliation(s)
- Magda R Hamczyk
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología, Universidad de Oviedo, Spain (M.R.H., V.Q., C.L.-O.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
| | - Rosa M Nevado
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Pilar Gonzalo
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - María J Andrés-Manzano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Paula Nogales
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Víctor Quesada
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología, Universidad de Oviedo, Spain (M.R.H., V.Q., C.L.-O.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
- Centro de Investigación Biomédica en Red de Cáncer, Spain (V.Q.)
| | - Aránzazu Rosado
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Carlos Torroja
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Fátima Sánchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Ana Dopazo
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Jacob F Bentzon
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
- Department of Clinical Medicine, Aarhus University, Denmark (J.F.B.)
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología, Universidad de Oviedo, Spain (M.R.H., V.Q., C.L.-O.)
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain (C.L.-O.)
- Centre de Recherche des Cordeliers, Université de Paris Cité, Sorbonne Université, INSERM U1138, France (C.L.-O.)
| | - Vicente Andrés
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| |
Collapse
|
17
|
Bi F, Gao C, Guo H. Epigenetic regulation of cardiovascular diseases induced by behavioral and environmental risk factors: Mechanistic, diagnostic, and therapeutic insights. FASEB Bioadv 2024; 6:477-502. [PMID: 39512842 PMCID: PMC11539034 DOI: 10.1096/fba.2024-00080] [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: 05/27/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 11/15/2024] Open
Abstract
Behavioral and environmental risk factors are critical in the development and progression of cardiovascular disease (CVD). Understanding the molecular mechanisms underlying these risk factors will offer valuable insights for targeted preventive and therapeutic strategies. Epigenetic modifications, including DNA methylation, histone modifications, chromatin remodeling, noncoding RNA (ncRNA) expression, and epitranscriptomic modifications, have emerged as key mediators connecting behavioral and environmental risk factors to CVD risk and progression. These epigenetic alterations can profoundly impact on cardiovascular health and susceptibility to CVD by influencing cellular processes, development, and disease risk over an individual's lifetime and potentially across generations. This review examines how behavioral and environmental risk factors affect CVD risk and health outcomes through epigenetic regulation. We review the epigenetic effects of major behavioral risk factors (such as smoking, alcohol consumption, physical inactivity, unhealthy diet, and obesity) and environmental risk factors (including air and noise pollution) in the context of CVD pathogenesis. Additionally, we explore epigenetic biomarkers, considering their role as causal or surrogate indicators, and discuss epigenetic therapeutics targeting the mechanisms through which these risk factors contribute to CVD. We also address future research directions and challenges in leveraging epigenetic insights to reduce the burden of CVD related to behavioral and environmental factors and improve public health outcomes. This review aims to provide a comprehensive understanding of behavioral and environmental epigenetics in CVD and offer valuable strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Feifei Bi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of UtahSalt Lake CityUtahUSA
- Division of Cardiothoracic Surgery, Department of SurgerySchool of Medicine, University of UtahSalt Lake CityUtahUSA
| | - Chen Gao
- Department of Pharmacology and Systems PhysiologyUniversity of CincinnatiCincinnatiOhioUSA
| | - Hongchao Guo
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of UtahSalt Lake CityUtahUSA
- Division of Cardiothoracic Surgery, Department of SurgerySchool of Medicine, University of UtahSalt Lake CityUtahUSA
| |
Collapse
|
18
|
Talarmin-Gas C, Smolyakov G, Parisi C, Scandola C, Andrianasolonirina V, Lecoq C, Houtart V, Lee SH, Adle-Biassette H, Thiébot B, Ganderton T, Manivet P. Validation of metaxin-2 deficient C. elegans as a model for MandibuloAcral Dysplasia associated to mtx-2 (MADaM) syndrome. Commun Biol 2024; 7:1398. [PMID: 39462037 PMCID: PMC11513083 DOI: 10.1038/s42003-024-06967-z] [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: 11/07/2023] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
MandibuloAcral Dysplasia associated to MTX2 gene (MADaM) is a recently described progeroid syndrome (accelerated aging disease) whose clinical manifestations include skin abnormalities, growth retardation, and cardiovascular diseases. We previously proposed that mtx-2-deficient C. elegans could be used as a model for MADaM and to support this, we present here our comprehensive phenotypic characterization of these worms using atomic force microscopy (AFM), transcriptomic, and oxygen consumption rate analyses. AFM analysis showed that young mtx-2-less worms had a significantly rougher, less elastic cuticle which becomes significantly rougher and less elastic as they age, and abnormal mitochondrial morphology. mtx-2 C. elegans displayed slightly delayed development, decreased pharyngeal pumping, significantly reduced mitochondrial respiratory capacities, and transcriptomic analysis identified perturbations in the aging, TOR, and WNT-signaling pathways. The phenotypic characteristics of mtx-2 worms shown here are analogous to many of the human clinical presentations of MADaM and we believe this validates their use as a model which will allow us to uncover the molecular details of the disease and develop new therapeutics and treatments.
Collapse
Affiliation(s)
- Chloé Talarmin-Gas
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France.
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France.
| | - Georges Smolyakov
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Cleo Parisi
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Cyril Scandola
- Institut Pasteur, Université Paris Cité, Ultrastructural Bioimaging Unit, 75015, Paris, France
| | - Valérie Andrianasolonirina
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Cloé Lecoq
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Valentine Houtart
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | | | - Homa Adle-Biassette
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
- AP-HP, DMU DREAM, Service d'Anatomocytopathologie, Hôpital Lariboisière, Paris, France
| | - Bénédicte Thiébot
- CY Cergy Paris Université, Université d'Evry, Université Paris-Saclay, CNRS, LAMBE, F-95000, Cergy, France
| | - Timothy Ganderton
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Philippe Manivet
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France.
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France.
- CeleScreen SAS, Paris, France.
| |
Collapse
|
19
|
Qi H, Wu Y, Zhang W, Yu N, Lu X, Liu J. The syntaxin-binding protein STXBP5 regulates progerin expression. Sci Rep 2024; 14:23376. [PMID: 39379476 PMCID: PMC11461833 DOI: 10.1038/s41598-024-74621-z] [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: 03/31/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
Hutchinson-Gilfor progeria syndrome (HGPS) is caused by a mutation in Lamin A resulting in the production of a protein called progerin. The accumulation of progerin induces inflammation, cellular senescence and activation of the P53 pathway. In this study, through public dataset analysis, we identified Syntaxin Binding Protein 5 (STXBP5) as an influencing factor of progerin expression. STXBP5 overexpression accelerated the onset of senescence, while STXBP5 deletion suppressed progerin expression, delayed senility, and decreased the expression of senescence-related factors. STXBP5 and progerin have synergistic effects and a protein-protein interaction. Through bioinformatics analysis, we found that STXBP5 affects ageing-related signalling pathways such as the mitogen-activated protein kinase (MAPK) pathway, the hippo pathway and the interleukin 17 (IL17) signalling pathway in progerin-expressing cells. In addition, STXBP5 overexpression induced changes in transposable elements (TEs), such as the human endogenous retrovirus H internal coding sequence (HERVH-int) changes. Our protein coimmunoprecipitation (Co-IP) results indicated that STXBP5 bound directly to progerin. Therefore, decreasing STXBP5 expression is a potential new therapeutic strategy for treating ageing-related phenotypes in patients with HGPS.
Collapse
Affiliation(s)
- Hongqian Qi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
- College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Yingying Wu
- College of Artificial Intelligence, Nankai University, Tianjin, 300350, China
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Nankai University, Tianjin, 300350, China
| | - Weiyu Zhang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853-2703, USA
| | - Ningbo Yu
- College of Artificial Intelligence, Nankai University, Tianjin, 300350, China
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Nankai University, Tianjin, 300350, China
| | - Xinyi Lu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China.
| | - Jinchao Liu
- College of Artificial Intelligence, Nankai University, Tianjin, 300350, China.
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Nankai University, Tianjin, 300350, China.
| |
Collapse
|
20
|
Cardoso D, Guilbert S, Guigue P, Carabalona A, Harhouri K, Peccate C, Tournois J, Guesmia Z, Ferreira L, Bartoli C, Levy N, Colleaux L, Nissan X, Muchir A. Inhibition of poly(ADP-Ribosyl)ation reduced vascular smooth muscle cells loss and improves aortic disease in a mouse model of human accelerated aging syndrome. Cell Death Dis 2024; 15:723. [PMID: 39353941 PMCID: PMC11448498 DOI: 10.1038/s41419-024-07078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare genetic disorder associated with features of accelerated aging. HGPS is an autosomal dominant disease caused by a de novo mutation of LMNA gene, encoding A-type lamins, resulting in the truncated form of pre-lamin A called progerin. While asymptomatic at birth, patients develop symptoms within the first year of life when they begin to display accelerated aging and suffer from growth retardation, and severe cardiovascular complications including loss of vascular smooth muscle cells (VSMCs). Recent works reported the loss of VSMCs as a major factor triggering atherosclerosis in HGPS. Here, we investigated the mechanisms by which progerin expression leads to massive VSMCs loss. Using aorta tissue and primary cultures of murine VSMCs from a mouse model of HGPS, we showed increased VSMCs death associated with increased poly(ADP-Ribosyl)ation. Poly(ADP-Ribosyl)ation is recognized as a post-translational protein modification that coordinates the repair at DNA damage sites. Poly-ADP-ribose polymerase (PARP) catalyzes protein poly(ADP-Ribosyl)ation by utilizing nicotinamide adenine dinucleotide (NAD+). Our results provided the first demonstration linking progerin accumulation, augmented poly(ADP-Ribosyl)ation and decreased nicotinamide adenine dinucleotide (NAD+) level in VSMCs. Using high-throughput screening on VSMCs differentiated from iPSCs from HGPS patients, we identified a new compound, trifluridine able to increase NAD+ levels through decrease of PARP-1 activity. Lastly, we demonstrate that trifluridine treatment in vivo was able to alleviate aortic VSMCs loss and clinical sign of progeria, suggesting a novel therapeutic approach of cardiovascular disease in progeria.
Collapse
MESH Headings
- Animals
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Disease Models, Animal
- Progeria/pathology
- Progeria/genetics
- Progeria/metabolism
- Mice
- Humans
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Lamin Type A/metabolism
- Lamin Type A/genetics
- Aorta/pathology
- Aorta/drug effects
- Aorta/metabolism
- Poly ADP Ribosylation
- Mice, Inbred C57BL
- Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Déborah Cardoso
- Sorbonne Université, UPMC Paris 06, INSERM UMRS974, Center of Research in Myology, Institut de Myologie, Paris, France
| | - Solenn Guilbert
- Université Paris-Saclay, Université d'Evry, Inserm, IStem UMR861, Corbeil-Essonnes, France
- IStem, CECS, Corbeil-Essonnes, France
| | - Philippe Guigue
- Aix Marseille Université, INSERM, MMG, U1251 Faculté de Médecine Timone, Marseille, France
| | - Aurélie Carabalona
- Aix Marseille Université, INSERM, MMG, U1251 Faculté de Médecine Timone, Marseille, France
| | - Karim Harhouri
- Aix Marseille Université, INSERM, MMG, U1251 Faculté de Médecine Timone, Marseille, France
| | - Cécile Peccate
- Sorbonne Université, UPMC Paris 06, INSERM UMRS974, Center of Research in Myology, Institut de Myologie, Paris, France
| | - Johana Tournois
- Université Paris-Saclay, Université d'Evry, Inserm, IStem UMR861, Corbeil-Essonnes, France
- IStem, CECS, Corbeil-Essonnes, France
| | - Zoheir Guesmia
- Sorbonne Université, UPMC Paris 06, INSERM UMRS974, Center of Research in Myology, Institut de Myologie, Paris, France
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Catherine Bartoli
- Aix Marseille Université, INSERM, MMG, U1251 Faculté de Médecine Timone, Marseille, France
| | - Nicolas Levy
- Aix Marseille Université, INSERM, MMG, U1251 Faculté de Médecine Timone, Marseille, France
| | - Laurence Colleaux
- Aix Marseille Université, INSERM, MMG, U1251 Faculté de Médecine Timone, Marseille, France
| | - Xavier Nissan
- Université Paris-Saclay, Université d'Evry, Inserm, IStem UMR861, Corbeil-Essonnes, France
- IStem, CECS, Corbeil-Essonnes, France
| | - Antoine Muchir
- Sorbonne Université, UPMC Paris 06, INSERM UMRS974, Center of Research in Myology, Institut de Myologie, Paris, France.
| |
Collapse
|
21
|
Barettino A, González-Gómez C, Gonzalo P, Andrés-Manzano MJ, Guerrero CR, Espinosa FM, Carmona RM, Blanco Y, Dorado B, Torroja C, Sánchez-Cabo F, Quintas A, Benguría A, Dopazo A, García R, Benedicto I, Andrés V. Endothelial YAP/TAZ activation promotes atherosclerosis in a mouse model of Hutchinson-Gilford progeria syndrome. J Clin Invest 2024; 134:e173448. [PMID: 39352768 PMCID: PMC11563688 DOI: 10.1172/jci173448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare disease caused by the expression of progerin, an aberrant protein produced by a point mutation in the LMNA gene. HGPS patients show accelerated aging and die prematurely mainly from complications of atherosclerosis such as myocardial infarction, heart failure, or stroke. However, the mechanisms underlying HGPS vascular pathology remain ill-defined. We used single-cell RNA sequencing to characterize the aorta in progerin-expressing LmnaG609G/G609G mice and wild-type controls, with a special focus on endothelial cells (ECs). HGPS ECs showed gene expression changes associated with extracellular matrix alterations, increased leukocyte extravasation, and activation of the yes-associated protein 1/transcriptional activator with PDZ-binding domain (YAP/TAZ) mechanosensing pathway, all validated by different techniques. Atomic force microscopy experiments demonstrated stiffer subendothelial extracellular matrix in progeroid aortae, and ultrasound assessment of live HGPS mice revealed disturbed aortic blood flow, both key inducers of the YAP/TAZ pathway in ECs. YAP/TAZ inhibition with verteporfin reduced leukocyte accumulation in the aortic intimal layer and decreased atherosclerosis burden in progeroid mice. Our findings identify endothelial YAP/TAZ signaling as a key mechanism of HGPS vascular disease and open a new avenue for the development of YAP/TAZ-targeting drugs to ameliorate progerin-induced atherosclerosis.
Collapse
Affiliation(s)
- Ana Barettino
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Cristina González-Gómez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Pilar Gonzalo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - María J. Andrés-Manzano
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | | | - Rosa M. Carmona
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Yaazan Blanco
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Beatriz Dorado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Carlos Torroja
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Fátima Sánchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ana Quintas
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Alberto Benguría
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | - Ignacio Benedicto
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
22
|
Hong Y, Rannou A, Manriquez N, Antich J, Liu W, Fournier M, Omidfar A, Rogers RG. Cardiac and skeletal muscle manifestations in the G608G mouse model of Hutchinson-Gilford progeria syndrome. Aging Cell 2024; 23:e14259. [PMID: 38961628 PMCID: PMC11464102 DOI: 10.1111/acel.14259] [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: 02/02/2024] [Revised: 04/16/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disorder resulting from de novo mutations in the lamin A gene. Children with HGPS typically pass away in their teenage years due to cardiovascular diseases such as atherosclerosis, myocardial infarction, heart failure, and stroke. In this study, we characterized the G608G HGPS mouse model and explored cardiac and skeletal muscle function, along with senescence-associated phenotypes in fibroblasts. Homozygous G608G HGPS mice exhibited cardiac dysfunction, including decreased cardiac output and stroke volume, and impaired left ventricle relaxation. Additionally, skeletal muscle exhibited decreased isometric tetanic torque, muscle atrophy, and increased fibrosis. HGPS fibroblasts showed nuclear abnormalities, decreased proliferation, and increased expression of senescence markers. These findings provide insights into the pathophysiology of the G608G HGPS mouse model and inform potential therapeutic strategies for HGPS.
Collapse
Affiliation(s)
- Yeojin Hong
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Alice Rannou
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Nancy Manriquez
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Jack Antich
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Weixin Liu
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Mario Fournier
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Ariel Omidfar
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Russell G. Rogers
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| |
Collapse
|
23
|
Puspitasari YM, Ministrini S, Han J, Karch C, Prisco F, Liberale L, Bengs S, Akhmedov A, Montecucco F, Beer JH, Lüscher TF, Bongiovanni D, Camici GG. Hutchinson-Gilford progeria syndrome mice display accelerated arterial thrombus formation and increased platelet reactivity. Thromb Res 2024; 241:109100. [PMID: 39032390 DOI: 10.1016/j.thromres.2024.109100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION Hutchinson-Gilford Progeria Syndrome (HGPS) is an ultra-rare premature aging genetic disorder caused by a point mutation in the lamin A gene, LMNA. Children with HGPS display short lifespans and typically die due to myocardial infarction or ischemic stroke, both acute cardiovascular events that are tightly linked to arterial thrombosis. Despite this fact, the effect of the classic HGPS LMNA gene mutation on arterial thrombosis remains unknown. METHODS Heterozygous LmnaG609G knock-in (LmnaG609G/+) mice, yielding an equivalent classic mutation observed in HGPS patients (c.1824C>T; pG608G mutation in the human LMNA gene) and corresponding wild-type (WT) control littermates underwent photochemically laser-induced carotid injury to trigger thrombosis. Coagulation and fibrinolytic factors were measured. Furthermore, platelet activation and reactivity were investigated. RESULTS LmnaG609G/+ mice displayed accelerated arterial thrombus formation, as underlined by shortened time to occlusion compared to WT littermates. Levels of factors involved in the coagulation and fibrinolytic system were comparable between groups, while LmnaG609G/+ animals showed higher plasma levels of thrombin-antithrombin complex and lower levels of antithrombin. Bone marrow analysis showed larger megakaryocytes in progeric mice. Lastly, enhanced platelet activation upon adenosine diphosphate, collagen-related peptide, and thrombin stimulation was observed in LmnaG609G/+ animals compared to the WT group, indicating a higher platelet reactivity in progeric animals. CONCLUSIONS LMNA mutation in HGPS mice accelerates arterial thrombus formation, which is mediated, at least in part, by enhanced platelet reactivity, which consequently augments thrombin generation. Given the wide spectrum of antiplatelet agents available clinically, further investigation is warranted to consider the most suitable antiplatelet regimen for children with HGPS to mitigate disease mortality and morbidity.
Collapse
Affiliation(s)
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Jiaying Han
- Department of Internal Medicine I, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Caroline Karch
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Francesco Prisco
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Susan Bengs
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Cardiology, Royal Brompton & Harefield Hospitals, National Heart & Lung Institute, Imperial College, London, United Kingdom
| | - Dario Bongiovanni
- Department of Internal Medicine I, Cardiology, University Hospital Augsburg, University of Augsburg, Augsburg, Germany; Department of Cardiovascular Medicine, Humanitas Clinical and Research Center IRCCS and Humanitas University, Rozzano, Milan, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
24
|
Rolas L, Stein M, Barkaway A, Reglero-Real N, Sciacca E, Yaseen M, Wang H, Vazquez-Martinez L, Golding M, Blacksell IA, Giblin MJ, Jaworska E, Bishop CL, Voisin MB, Gaston-Massuet C, Fossati-Jimack L, Pitzalis C, Cooper D, Nightingale TD, Lopez-Otin C, Lewis MJ, Nourshargh S. Senescent endothelial cells promote pathogenic neutrophil trafficking in inflamed tissues. EMBO Rep 2024; 25:3842-3869. [PMID: 38918502 PMCID: PMC11387759 DOI: 10.1038/s44319-024-00182-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/28/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Cellular senescence is a hallmark of advanced age and a major instigator of numerous inflammatory pathologies. While endothelial cell (EC) senescence is aligned with defective vascular functionality, its impact on fundamental inflammatory responses in vivo at single-cell level remain unclear. To directly investigate the role of EC senescence on dynamics of neutrophil-venular wall interactions, we applied high resolution confocal intravital microscopy to inflamed tissues of an EC-specific progeroid mouse model, characterized by profound indicators of EC senescence. Progerin-expressing ECs supported prolonged neutrophil adhesion and crawling in a cell autonomous manner that additionally mediated neutrophil-dependent microvascular leakage. Transcriptomic and immunofluorescence analysis of inflamed tissues identified elevated levels of EC CXCL1 on progerin-expressing ECs and functional blockade of CXCL1 suppressed the dysregulated neutrophil responses elicited by senescent ECs. Similarly, cultured progerin-expressing human ECs exhibited a senescent phenotype, were pro-inflammatory and prompted increased neutrophil attachment and activation. Collectively, our findings support the concept that senescent ECs drive excessive inflammation and provide new insights into the mode, dynamics, and mechanisms of this response at single-cell level.
Collapse
Affiliation(s)
- Loïc Rolas
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Monja Stein
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anna Barkaway
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Natalia Reglero-Real
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Elisabetta Sciacca
- Centre for Translational Bioinformatics, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mohammed Yaseen
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Haitao Wang
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Laura Vazquez-Martinez
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matthew Golding
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Isobel A Blacksell
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Meredith J Giblin
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Edyta Jaworska
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Cleo L Bishop
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mathieu-Benoit Voisin
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Liliane Fossati-Jimack
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Dianne Cooper
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Thomas D Nightingale
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Carlos Lopez-Otin
- Centre de Recherche des Cordeliers, Inserm U1138, Université Paris Cité, Sorbonne Université, Paris, France
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain
| | - Myles J Lewis
- Centre for Translational Bioinformatics, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sussan Nourshargh
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK.
| |
Collapse
|
25
|
Yu R, Xue H, Lin W, Collins F, Mount S, Cao K. Progerin mRNA expression in non-HGPS patients is correlated with widespread shifts in transcript isoforms. NAR Genom Bioinform 2024; 6:lqae115. [PMID: 39211333 PMCID: PMC11358823 DOI: 10.1093/nargab/lqae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disease caused primarily by a C1824T mutation in LMNA. This mutation activates a cryptic splice donor site, producing a lamin variant called progerin. Interestingly, progerin has also been detected in cells and tissues of non-HGPS patients. Here, we investigated progerin expression using publicly available RNA-seq data from non-HGPS patients in the GTEx project. We found that progerin expression is present across all tissue types in non-HGPS patients and correlated with telomere shortening in the skin. Transcriptome-wide correlation analyses suggest that the level of progerin expression is correlated with switches in gene isoform expression patterns. Differential expression analyses show that progerin expression is correlated with significant changes in genes involved in splicing regulation and mitochondrial function. Interestingly, 5' splice sites whose use is correlated with progerin expression have significantly altered frequencies of consensus trinucleotides within the core 5' splice site. Furthermore, introns whose alternative splicing correlates with progerin have reduced GC content. Our study suggests that progerin expression in non-HGPS patients is part of a global shift in splicing patterns.
Collapse
Affiliation(s)
- Reynold Yu
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Huijing Xue
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Wanru Lin
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Francis S Collins
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen M Mount
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| |
Collapse
|
26
|
Krüger P, Schroll M, Fenzl F, Lederer EM, Hartinger R, Arnold R, Cagla Togan D, Guo R, Liu S, Petry A, Görlach A, Djabali K. Inflammation and Fibrosis in Progeria: Organ-Specific Responses in an HGPS Mouse Model. Int J Mol Sci 2024; 25:9323. [PMID: 39273272 PMCID: PMC11395088 DOI: 10.3390/ijms25179323] [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: 08/10/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is an extremely rare genetic disorder that causes accelerated aging, due to a pathogenic variant in the LMNA gene. This pathogenic results in the production of progerin, a defective protein that disrupts the nuclear lamina's structure. In our study, we conducted a histopathological analysis of various organs in the LmnaG609G/G609G mouse model, which is commonly used to study HGPS. The objective of this study was to show that progerin accumulation drives systemic but organ-specific tissue damage and accelerated aging phenotypes. Our findings show significant fibrosis, inflammation, and dysfunction in multiple organ systems, including the skin, cardiovascular system, muscles, lungs, liver, kidneys, spleen, thymus, and heart. Specifically, we observed severe vascular fibrosis, reduced muscle regeneration, lung tissue remodeling, depletion of fat in the liver, and disruptions in immune structures. These results underscore the systemic nature of the disease and suggest that chronic inflammation and fibrosis play crucial roles in the accelerated aging seen in HGPS. Additionally, our study highlights that each organ responds differently to the toxic effects of progerin, indicating that there are distinct mechanisms of tissue-specific damage.
Collapse
Affiliation(s)
- Peter Krüger
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Moritz Schroll
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Felix Fenzl
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Eva-Maria Lederer
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Ramona Hartinger
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Rouven Arnold
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Deniz Cagla Togan
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Runjia Guo
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Shiyu Liu
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Andreas Petry
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital, Heart Diseases, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80636 Munich, Germany
| | - Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital, Heart Diseases, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80636 Munich, Germany
| | - Karima Djabali
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| |
Collapse
|
27
|
Díaz-López EJ, Sánchez-Iglesias S, Castro AI, Cobelo-Gómez S, Prado-Moraña T, Araújo-Vilar D, Fernandez-Pombo A. Lipodystrophic Laminopathies: From Dunnigan Disease to Progeroid Syndromes. Int J Mol Sci 2024; 25:9324. [PMID: 39273270 PMCID: PMC11395136 DOI: 10.3390/ijms25179324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Lipodystrophic laminopathies are a group of ultra-rare disorders characterised by the presence of pathogenic variants in the same gene (LMNA) and other related genes, along with an impaired adipose tissue pattern and other features that are specific of each of these disorders. The most fascinating traits include their complex genotype-phenotype associations and clinical heterogeneity, ranging from Dunnigan disease, in which the most relevant feature is precisely adipose tissue dysfunction and lipodystrophy, to the other laminopathies affecting adipose tissue, which are also characterised by the presence of signs of premature ageing (Hutchinson Gilford-progeria syndrome, LMNA-atypical progeroid syndrome, mandibuloacral dysplasia types A and B, Nestor-Guillermo progeria syndrome, LMNA-associated cardiocutaneous progeria). This raises several questions when it comes to understanding how variants in the same gene can lead to similar adipose tissue disturbances and, at the same time, to such heterogeneous phenotypes and variable degrees of metabolic abnormalities. The present review aims to gather the molecular basis of adipose tissue impairment in lipodystrophic laminopathies, their main clinical aspects and recent therapeutic strategies. In addition, it also summarises the key aspects for their differential diagnosis.
Collapse
Affiliation(s)
- Everardo Josué Díaz-López
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Sofía Sánchez-Iglesias
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Ana I Castro
- Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), 28029 Madrid, Spain
| | - Silvia Cobelo-Gómez
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Teresa Prado-Moraña
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - David Araújo-Vilar
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Antia Fernandez-Pombo
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| |
Collapse
|
28
|
Shores KL, Truskey GA. Mechanotransduction of the vasculature in Hutchinson-Gilford Progeria Syndrome. Front Physiol 2024; 15:1464678. [PMID: 39239311 PMCID: PMC11374724 DOI: 10.3389/fphys.2024.1464678] [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: 07/14/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disorder that causes severe cardiovascular disease, resulting in the death of patients in their teenage years. The disease pathology is caused by the accumulation of progerin, a mutated form of the nuclear lamina protein, lamin A. Progerin binds to the inner nuclear membrane, disrupting nuclear integrity, and causes severe nuclear abnormalities and changes in gene expression. This results in increased cellular inflammation, senescence, and overall dysfunction. The molecular mechanisms by which progerin induces the disease pathology are not fully understood. Progerin's detrimental impact on nuclear mechanics and the role of the nucleus as a mechanosensor suggests dysfunctional mechanotransduction could play a role in HGPS. This is especially relevant in cells exposed to dynamic, continuous mechanical stimuli, like those of the vasculature. The endothelial (ECs) and smooth muscle cells (SMCs) within arteries rely on physical forces produced by blood flow to maintain function and homeostasis. Certain regions within arteries produce disturbed flow, leading to an impaired transduction of mechanical signals, and a reduction in cellular function, which also occurs in HGPS. In this review, we discuss the mechanics of nuclear mechanotransduction, how this is disrupted in HGPS, and what effect this has on cell health and function. We also address healthy responses of ECs and SMCs to physiological mechanical stimuli and how these responses are impaired by progerin accumulation.
Collapse
Affiliation(s)
- Kevin L Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
29
|
Zaleski KL, Matte GS, Kleinman ME, Prakash A, Stein ML. Anesthetic Management of Cardiopulmonary Bypass in Hutchinson-Gilford Progeria Syndrome: A Case Report. A A Pract 2024; 18:e01842. [PMID: 39177382 DOI: 10.1213/xaa.0000000000001842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is an ultrarare disorder of segmental premature aging that is associated with the development of advanced atherosclerosis and significant cardiovascular and cerebrovascular disease. Treatment with lonafarnib has improved survival in patients with HGPS; however, in extended longitudinal follow-up, there has been an increase in the prevalence of rapidly progressive calcific aortic stenosis. The evolving course of HGPS has prompted reconsideration of conservative management and led to the development of strategies for anatomic treatment. In this case report, we describe the anesthetic management of patients with HGPS undergoing surgical management of aortic stenosis with cardiopulmonary bypass.
Collapse
Affiliation(s)
| | | | - Monica E Kleinman
- From the Departments of Anesthesiology, Critical Care and Pain Medicine
| | - Ashwin Prakash
- Cardiology, Boston Children's Hospital, Boston, Massachusetts
| | - Mary Lyn Stein
- From the Departments of Anesthesiology, Critical Care and Pain Medicine
| |
Collapse
|
30
|
Kiskin FN, Yang Y, Yang H, Zhang JZ. Cracking the code of the cardiovascular enigma: hPSC-derived endothelial cells unveil the secrets of endothelial dysfunction. J Mol Cell Cardiol 2024; 192:65-78. [PMID: 38761989 DOI: 10.1016/j.yjmcc.2024.05.005] [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: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Endothelial dysfunction is a central contributor to the development of most cardiovascular diseases and is characterised by the reduced synthesis or bioavailability of the vasodilator nitric oxide together with other abnormalities such as inflammation, senescence, and oxidative stress. The use of patient-specific and genome-edited human pluripotent stem cell-derived endothelial cells (hPSC-ECs) has shed novel insights into the role of endothelial dysfunction in cardiovascular diseases with strong genetic components such as genetic cardiomyopathies and pulmonary arterial hypertension. However, their utility in studying complex multifactorial diseases such as atherosclerosis, metabolic syndrome and heart failure poses notable challenges. In this review, we provide an overview of the different methods used to generate and characterise hPSC-ECs before comprehensively assessing their effectiveness in cardiovascular disease modelling and high-throughput drug screening. Furthermore, we explore current obstacles that will need to be overcome to unleash the full potential of hPSC-ECs in facilitating patient-specific precision medicine. Addressing these challenges holds great promise in advancing our understanding of intricate cardiovascular diseases and in tailoring personalised therapeutic strategies.
Collapse
Affiliation(s)
- Fedir N Kiskin
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Yuan Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Hao Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Joe Z Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
31
|
Ngubo M, Chen Z, McDonald D, Karimpour R, Shrestha A, Yockell‐Lelièvre J, Laurent A, Besong OTO, Tsai EC, Dilworth FJ, Hendzel MJ, Stanford WL. Progeria-based vascular model identifies networks associated with cardiovascular aging and disease. Aging Cell 2024; 23:e14150. [PMID: 38576084 PMCID: PMC11258467 DOI: 10.1111/acel.14150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Hutchinson-Gilford Progeria syndrome (HGPS) is a lethal premature aging disorder caused by a de novo heterozygous mutation that leads to the accumulation of a splicing isoform of Lamin A termed progerin. Progerin expression deregulates the organization of the nuclear lamina and the epigenetic landscape. Progerin has also been observed to accumulate at low levels during normal aging in cardiovascular cells of adults that do not carry genetic mutations linked with HGPS. Therefore, the molecular mechanisms that lead to vascular dysfunction in HGPS may also play a role in vascular aging-associated diseases, such as myocardial infarction and stroke. Here, we show that HGPS patient-derived vascular smooth muscle cells (VSMCs) recapitulate HGPS molecular hallmarks. Transcriptional profiling revealed cardiovascular disease remodeling and reactive oxidative stress response activation in HGPS VSMCs. Proteomic analyses identified abnormal acetylation programs in HGPS VSMC replication fork complexes, resulting in reduced H4K16 acetylation. Analysis of acetylation kinetics revealed both upregulation of K16 deacetylation and downregulation of K16 acetylation. This correlates with abnormal accumulation of error-prone nonhomologous end joining (NHEJ) repair proteins on newly replicated chromatin. The knockdown of the histone acetyltransferase MOF recapitulates preferential engagement of NHEJ repair activity in control VSMCs. Additionally, we find that primary donor-derived coronary artery vascular smooth muscle cells from aged individuals show similar defects to HGPS VSMCs, including loss of H4K16 acetylation. Altogether, we provide insight into the molecular mechanisms underlying vascular complications associated with HGPS patients and normative aging.
Collapse
Affiliation(s)
- Mzwanele Ngubo
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
| | - Zhaoyi Chen
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Darin McDonald
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Rana Karimpour
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Amit Shrestha
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Julien Yockell‐Lelièvre
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Aurélie Laurent
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Université de StrasbourgStrasbourgFrance
| | - Ojong Tabi Ojong Besong
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- School of BioscienceUniversity of SkövdeSkövdeSweden
| | - Eve C. Tsai
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
- Division of Neurosurgery, Department of Surgery, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - F. Jeffrey Dilworth
- Department of Cell and Regenerative BiologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Michael J. Hendzel
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - William L. Stanford
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Department of Biochemistry, Microbiology & ImmunologyUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
32
|
Hernandez-Sanchez D, Comtois-Bona M, Muñoz M, Ruel M, Suuronen EJ, Alarcon EI. Manufacturing and validation of small-diameter vascular grafts: A mini review. iScience 2024; 27:109845. [PMID: 38799581 PMCID: PMC11126982 DOI: 10.1016/j.isci.2024.109845] [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] [Indexed: 05/29/2024] Open
Abstract
The field of small-diameter vascular grafts remains a challenge for biomaterials scientists. While decades of research have brought us much closer to developing biomimetic materials for regenerating tissues and organs, the physiological challenges involved in manufacturing small conduits that can transport blood while not inducing an immune response or promoting blood clots continue to limit progress in this area. In this short review, we present some of the most recent methods and advancements made by researchers working in the field of small-diameter vascular grafts. We also discuss some of the most critical aspects biomaterials scientists should consider when developing lab-made small-diameter vascular grafts.
Collapse
Affiliation(s)
- Deyanira Hernandez-Sanchez
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
| | - Maxime Comtois-Bona
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
| | - Marcelo Muñoz
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
| | - Marc Ruel
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, 451 Smyth Road, Ottawa ON K1H8M5, Canada
| | - Erik J. Suuronen
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, 451 Smyth Road, Ottawa ON K1H8M5, Canada
| | - Emilio I. Alarcon
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H8M5, Canada
| |
Collapse
|
33
|
Le A, Peng H, Golinsky D, Di Scipio M, Lali R, Paré G. What Causes Premature Coronary Artery Disease? Curr Atheroscler Rep 2024; 26:189-203. [PMID: 38573470 DOI: 10.1007/s11883-024-01200-y] [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] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
PURPOSE OF REVIEW This review provides an overview of genetic and non-genetic causes of premature coronary artery disease (pCAD). RECENT FINDINGS pCAD refers to coronary artery disease (CAD) occurring before the age of 65 years in women and 55 years in men. Both genetic and non-genetic risk factors may contribute to the onset of pCAD. Recent advances in the genetic epidemiology of pCAD have revealed the importance of both monogenic and polygenic contributions to pCAD. Familial hypercholesterolemia (FH) is the most common monogenic disorder associated with atherosclerotic pCAD. However, clinical overreliance on monogenic genes can result in overlooked genetic causes of pCAD, especially polygenic contributions. Non-genetic factors, notably smoking and drug use, are also important contributors to pCAD. Cigarette smoking has been observed in 25.5% of pCAD patients relative to 12.2% of non-pCAD patients. Finally, myocardial infarction (MI) associated with spontaneous coronary artery dissection (SCAD) may result in similar clinical presentations as atherosclerotic pCAD. Recognizing the genetic and non-genetic causes underlying pCAD is important for appropriate prevention and treatment. Despite recent progress, pCAD remains incompletely understood, highlighting the need for both awareness and research.
Collapse
Affiliation(s)
- Ann Le
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Helen Peng
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Danielle Golinsky
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- School of Nursing, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Matteo Di Scipio
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Ricky Lali
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Guillaume Paré
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada.
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada.
- Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada.
| |
Collapse
|
34
|
Castilho RM, Castilho LS, Palomares BH, Squarize CH. Determinants of Chromatin Organization in Aging and Cancer-Emerging Opportunities for Epigenetic Therapies and AI Technology. Genes (Basel) 2024; 15:710. [PMID: 38927646 PMCID: PMC11202709 DOI: 10.3390/genes15060710] [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: 03/31/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 06/28/2024] Open
Abstract
This review article critically examines the pivotal role of chromatin organization in gene regulation, cellular differentiation, disease progression and aging. It explores the dynamic between the euchromatin and heterochromatin, coded by a complex array of histone modifications that orchestrate essential cellular processes. We discuss the pathological impacts of chromatin state misregulation, particularly in cancer and accelerated aging conditions such as progeroid syndromes, and highlight the innovative role of epigenetic therapies and artificial intelligence (AI) in comprehending and harnessing the histone code toward personalized medicine. In the context of aging, this review explores the use of AI and advanced machine learning (ML) algorithms to parse vast biological datasets, leading to the development of predictive models for epigenetic modifications and providing a framework for understanding complex regulatory mechanisms, such as those governing cell identity genes. It supports innovative platforms like CEFCIG for high-accuracy predictions and tools like GridGO for tailored ChIP-Seq analysis, which are vital for deciphering the epigenetic landscape. The review also casts a vision on the prospects of AI and ML in oncology, particularly in the personalization of cancer therapy, including early diagnostics and treatment optimization for diseases like head and neck and colorectal cancers by harnessing computational methods, AI advancements and integrated clinical data for a transformative impact on healthcare outcomes.
Collapse
Affiliation(s)
- Rogerio M. Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA; (L.S.C.); (C.H.S.)
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | - Leonard S. Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA; (L.S.C.); (C.H.S.)
| | - Bruna H. Palomares
- Oral Diagnosis Department, Piracicaba School of Dentistry, State University of Campinas, Piracicaba 13414-903, Sao Paulo, Brazil;
| | - Cristiane H. Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA; (L.S.C.); (C.H.S.)
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109-1078, USA
| |
Collapse
|
35
|
Bougaran P, Bautch VL. Life at the crossroads: the nuclear LINC complex and vascular mechanotransduction. Front Physiol 2024; 15:1411995. [PMID: 38831796 PMCID: PMC11144885 DOI: 10.3389/fphys.2024.1411995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Vascular endothelial cells line the inner surface of all blood vessels, where they are exposed to polarized mechanical forces throughout their lifespan. Both basal substrate interactions and apical blood flow-induced shear stress regulate blood vessel development, remodeling, and maintenance of vascular homeostasis. Disruption of these interactions leads to dysfunction and vascular pathologies, although how forces are sensed and integrated to affect endothelial cell behaviors is incompletely understood. Recently the endothelial cell nucleus has emerged as a prominent force-transducing organelle that participates in vascular mechanotransduction, via communication to and from cell-cell and cell-matrix junctions. The LINC complex, composed of SUN and nesprin proteins, spans the nuclear membranes and connects the nuclear lamina, the nuclear envelope, and the cytoskeleton. Here we review LINC complex involvement in endothelial cell mechanotransduction, describe unique and overlapping functions of each LINC complex component, and consider emerging evidence that two major SUN proteins, SUN1 and SUN2, orchestrate a complex interplay that extends outward to cell-cell and cell-matrix junctions and inward to interactions within the nucleus and chromatin. We discuss these findings in relation to vascular pathologies such as Hutchinson-Gilford progeria syndrome, a premature aging disorder with cardiovascular impairment. More knowledge of LINC complex regulation and function will help to understand how the nucleus participates in endothelial cell force sensing and how dysfunction leads to cardiovascular disease.
Collapse
Affiliation(s)
- Pauline Bougaran
- Department of Biology, The University of North Carolina, Chapel Hill, NC, United States
| | - Victoria L. Bautch
- Department of Biology, The University of North Carolina, Chapel Hill, NC, United States
- McAllister Heart Institute, The University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
36
|
Gordon LB, Basso S, Maestranzi J, Aikawa E, Clift CL, Cammardella AG, Danesi TH, del Nido PJ, Edelman ER, Hamdy A, Hegde SM, Kleinman ME, Maschietto N, Mehra MR, Mukundan S, Musumeci F, Russo M, Rybicki FJ, Shah PB, Suarez WA, Tuminelli K, Zaleski K, Prakash A, Gerhard-Herman M. Intervention for critical aortic stenosis in Hutchinson-Gilford progeria syndrome. Front Cardiovasc Med 2024; 11:1356010. [PMID: 38725831 PMCID: PMC11079313 DOI: 10.3389/fcvm.2024.1356010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is an ultra-rare genetic premature aging disease that is historically fatal in teenage years, secondary to severe accelerated atherosclerosis. The only approved treatment is the farnesyltransferase inhibitor lonafarnib, which improves vascular structure and function, extending average untreated lifespan of 14.5 years by 4.3 years (30%). With this longer lifespan, calcific aortic stenosis (AS) was identified as an emerging critical risk factor for cardiac death in older patients. Intervention to relieve critical AS has the potential for immediate improvement in healthspan and lifespan. However, HGPS patient-device size mismatch, pervasive peripheral arterial disease, skin and bone abnormalities, and lifelong failure to thrive present unique challenges to intervention. An international group of experts in HGPS, pediatric and adult cardiology, cardiac surgery, and pediatric critical care convened to identify strategies for successful treatment. Candidate procedures were evaluated by in-depth examination of 4 cases that typify HGPS clinical pathology. Modified transcatheter aortic valve replacement (TAVR) and left ventricular Apico-Aortic Conduit (AAC) placement were deemed high risk but viable options. Two cases received TAVR and 2 received AAC post-summit. Three were successful and 1 patient died perioperatively due to cardiovascular disease severity, highlighting the importance of intervention timing and comparative risk stratification. These breakthrough interventions for treating critical aortic stenosis in HGPS patients could rewrite the current clinical perspective on disease course by greatly improving late-stage quality of life and increasing lifespan. Expanding worldwide medical and surgical competency for this ultra-rare disease through expert information-sharing could have high impact on treatment success.
Collapse
Affiliation(s)
- Leslie B. Gordon
- Division of Genetics, Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI, United States
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
- The Progeria Research Foundation, Peabody, MA, United States
| | - Sammy Basso
- The Progeria Research Foundation, Peabody, MA, United States
- Associazione Italiana Progeria Sammy Basso, Tezze sul Brenta, Vicenza
- CNR - National Research Council of Italy, Institute of Molecular Genetics Luigi Luca Cavalli-Sforza,Unit 9 of Bologna, Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Elena Aikawa
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, United States
| | - Cassandra L. Clift
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, United States
| | | | - Tommaso Hinna Danesi
- Department of Surgery, Division of Cardiac Surgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Pedro J. del Nido
- Department of Cardiac Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Elazer R. Edelman
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Abeer Hamdy
- Department of Pediatrics, Division of Pediatric Cardiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Sheila M. Hegde
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Monica E. Kleinman
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Nicola Maschietto
- Department of Cardiology, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Mandeep R. Mehra
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Srinivasan Mukundan
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Francesco Musumeci
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, Roma, Italy
| | - Marco Russo
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, Roma, Italy
| | - Frank J. Rybicki
- Department of Radiology, University of Arizona - Phoenix, Phoenix, AZ, United States
| | - Pinak Bipin Shah
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, United States
| | - William A. Suarez
- Division of Pediatric Cardiology, Department of Pediatrics, University of Toledo Medical Center, Toledo, OH, United States
| | | | - Katherine Zaleski
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Ashwin Prakash
- Department of Cardiology, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Marie Gerhard-Herman
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
37
|
Jagaraj CJ, Shadfar S, Kashani SA, Saravanabavan S, Farzana F, Atkin JD. Molecular hallmarks of ageing in amyotrophic lateral sclerosis. Cell Mol Life Sci 2024; 81:111. [PMID: 38430277 PMCID: PMC10908642 DOI: 10.1007/s00018-024-05164-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/21/2024] [Accepted: 02/06/2024] [Indexed: 03/03/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, severely debilitating and rapidly progressing disorder affecting motor neurons in the brain, brainstem, and spinal cord. Unfortunately, there are few effective treatments, thus there remains a critical need to find novel interventions that can mitigate against its effects. Whilst the aetiology of ALS remains unclear, ageing is the major risk factor. Ageing is a slowly progressive process marked by functional decline of an organism over its lifespan. However, it remains unclear how ageing promotes the risk of ALS. At the molecular and cellular level there are specific hallmarks characteristic of normal ageing. These hallmarks are highly inter-related and overlap significantly with each other. Moreover, whilst ageing is a normal process, there are striking similarities at the molecular level between these factors and neurodegeneration in ALS. Nine ageing hallmarks were originally proposed: genomic instability, loss of telomeres, senescence, epigenetic modifications, dysregulated nutrient sensing, loss of proteostasis, mitochondrial dysfunction, stem cell exhaustion, and altered inter-cellular communication. However, these were recently (2023) expanded to include dysregulation of autophagy, inflammation and dysbiosis. Hence, given the latest updates to these hallmarks, and their close association to disease processes in ALS, a new examination of their relationship to pathophysiology is warranted. In this review, we describe possible mechanisms by which normal ageing impacts on neurodegenerative mechanisms implicated in ALS, and new therapeutic interventions that may arise from this.
Collapse
Affiliation(s)
- Cyril Jones Jagaraj
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sina Shadfar
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sara Assar Kashani
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sayanthooran Saravanabavan
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Fabiha Farzana
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Julie D Atkin
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia.
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
38
|
Escudeiro-Lopes S, Filimonenko VV, Jarolimová L, Hozák P. Lamin A/C and PI(4,5)P2-A Novel Complex in the Cell Nucleus. Cells 2024; 13:399. [PMID: 38474363 PMCID: PMC10931150 DOI: 10.3390/cells13050399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/06/2024] [Accepted: 02/10/2024] [Indexed: 03/14/2024] Open
Abstract
Lamins, the nuclear intermediate filaments, are important regulators of nuclear structural integrity as well as nuclear functional processes such as DNA transcription, replication and repair, and epigenetic regulations. A portion of phosphorylated lamin A/C localizes to the nuclear interior in interphase, forming a lamin A/C pool with specific properties and distinct functions. Nucleoplasmic lamin A/C molecular functions are mainly dependent on its binding partners; therefore, revealing new interactions could give us new clues on the lamin A/C mechanism of action. In the present study, we show that lamin A/C interacts with nuclear phosphoinositides (PIPs), and with nuclear myosin I (NM1). Both NM1 and nuclear PIPs have been previously reported as important regulators of gene expression and DNA damage/repair. Furthermore, phosphorylated lamin A/C forms a complex with NM1 in a phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2)-dependent manner in the nuclear interior. Taken together, our study reveals a previously unidentified interaction between phosphorylated lamin A/C, NM1, and PI(4,5)P2 and suggests new possible ways of nucleoplasmic lamin A/C regulation, function, and importance for the formation of functional nuclear microdomains.
Collapse
Affiliation(s)
- Sara Escudeiro-Lopes
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; (S.E.-L.); (V.V.F.)
| | - Vlada V. Filimonenko
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; (S.E.-L.); (V.V.F.)
- Electron Microscopy Core Facility, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Lenka Jarolimová
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; (S.E.-L.); (V.V.F.)
| | - Pavel Hozák
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; (S.E.-L.); (V.V.F.)
| |
Collapse
|
39
|
Macías Á, Nevado RM, González-Gómez C, Gonzalo P, Andrés-Manzano MJ, Dorado B, Benedicto I, Andrés V. Coronary and carotid artery dysfunction and K V7 overexpression in a mouse model of Hutchinson-Gilford progeria syndrome. GeroScience 2024; 46:867-884. [PMID: 37233881 PMCID: PMC10828489 DOI: 10.1007/s11357-023-00808-3] [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: 03/30/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare genetic disease caused by expression of progerin, a lamin A variant that is also expressed at low levels in non-HGPS individuals. Although HGPS patients die predominantly from myocardial infarction and stroke, the mechanisms that provoke pathological alterations in the coronary and cerebral arteries in HGPS remain ill defined. Here, we assessed vascular function in the coronary arteries (CorAs) and carotid arteries (CarAs) of progerin-expressing LmnaG609G/G609G mice (G609G), both in resting conditions and after hypoxic stimulus. Wire myography, pharmacological screening, and gene expression studies demonstrated vascular atony and stenosis, as well as other functional alterations in progeroid CorAs and CarAs and aorta. These defects were associated with loss of vascular smooth muscle cells and overexpression of the KV7 family of voltage-dependent potassium channels. Compared with wild-type controls, G609G mice showed reduced median survival upon chronic isoproterenol exposure, a baseline state of chronic cardiac hypoxia characterized by overexpression of hypoxia-inducible factor 1α and 3α genes, and increased cardiac vascularization. Our results shed light on the mechanisms underlying progerin-induced coronary and carotid artery disease and identify KV7 channels as a candidate target for the treatment of HGPS.
Collapse
Affiliation(s)
- Álvaro Macías
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Rosa M Nevado
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina González-Gómez
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Gonzalo
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - María Jesús Andrés-Manzano
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz Dorado
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Benedicto
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), 28040, Madrid, Spain
| | - Vicente Andrés
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
40
|
Arun A, Nath AR, Thankachan B, Unnikrishnan MK. Hutchinson-Gilford progeria syndrome: unraveling the genetic basis, symptoms, and advancements in therapeutic approaches. THERAPEUTIC ADVANCES IN RARE DISEASE 2024; 5:26330040241305144. [PMID: 39691184 PMCID: PMC11650505 DOI: 10.1177/26330040241305144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024]
Abstract
Hutchinson-Gilford Progeria syndrome (HGPS) serves as a prominent model for Progeroid syndromes, a group of rare genetic disorders characterized by accelerated aging. This review explores the genetic basis, clinical presentation, and complications of HGPS. HGPS is caused by mutations in the LMNA gene, resulting in the production of a defective structural protein, prelamin A. This protein contains a "CAAX" motif, where C represents cysteine, and its abnormal processing is central to the disease's pathology. HGPS leads to multiple organ systems being affected, including cardiovascular, skeletal, neurological, and dermatological systems, causing severe disability and increased mortality. Cardiovascular issues are particularly significant in HGPS and are crucial for developing therapeutic strategies. Recent advances in treatment modalities offer promise for managing HGPS. Farnesyltransferase inhibitors and genetic interventions, such as CRISPR-Cas9, have shown potential in mitigating progerin-associated symptoms, with encouraging results observed in preclinical and clinical studies. Additionally, emerging therapies such as rapamycin, sulforaphane, and MG132 hold promise in targeting underlying disease mechanisms. Comprehensive management approaches, including growth hormone therapy, retinoids, and dental care, are emphasized to enhance overall patient well-being. Despite progress, further research is essential to unravel the complex pathophysiology of Progeroid syndromes and develop effective treatments. Continued focus on therapies that address progerin accumulation and its downstream effects is vital for improving patient care and outcomes for individuals affected by HGPS and related disorders. This review highlights ongoing efforts to understand and combat Progeroid syndromes, aiming to alleviate the burdens imposed by these debilitating conditions.
Collapse
Affiliation(s)
- Akhil Arun
- Department of Pharmacy Practice Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, AIMS Ponekkara P.O., Kochi, KL 682041, India
| | - Athira Rejith Nath
- Department of Pharmacy Practice Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, KL, India
| | - Bonny Thankachan
- Department of Pharmacy Practice Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, KL, India
| | - M. K. Unnikrishnan
- Department of Pharmacy Practice Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, KL, India
| |
Collapse
|
41
|
Liu H, Liu X, Luo S, Ma R, Ge W, Meng S, Gao Y. Lamin A/C mediates microglial activation by modulating cell proliferation and immune response. J Neurosci Res 2024; 102:e25263. [PMID: 38284866 DOI: 10.1002/jnr.25263] [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: 08/06/2023] [Accepted: 10/01/2023] [Indexed: 01/30/2024]
Abstract
Lamin A/C is involved in macrophage activation and premature aging, also known as progeria. As the resident macrophage in brain, overactivation of microglia causes brain inflammation, promoting aging and brain disease. In this study, we investigated the role of Lamin A/C in microglial activation and its impact on progeria using Lmna-/- mice, primary microglia, Lmna knockout (Lmna-KO) and Lmna-knockdown (Lmna-KD) BV2 cell lines. We found that the microglial activation signatures, including cell proliferation, morphology changes, and proinflammatory cytokine secretion (IL-1β, IL-6, and TNF-α), were significantly suppressed in all Lamin A/C-deficient models when stimulated with LPS. TMT-based quantitative proteomic and bioinformatic analysis were further applied to explore the mechanism of Lamin A/C-regulated microglia activation from the proteome level. The results revealed that immune response and phagocytosis were impaired in Lmna-/- microglia. Stat1 was identified as the hub protein in the mechanism by which Lamin A/C regulates microglial activation. Additionally, DNA replication, chromatin organization, and mRNA processing were also altered by Lamin A/C, with Ki67 fulfilling the main hub function. Lamin A/C is a mechanosensitive protein and, the immune- and proliferation-related biological processes are also regulated by mechanotransduction. We speculate that Lamin A/C-mediated mechanotransduction is required for microglial activation. Our study proposes a novel mechanism for microglial activation mediated by Lamin A/C.
Collapse
Affiliation(s)
- Haotian Liu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xinnan Liu
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Shiqi Luo
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Rayna Ma
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Wei Ge
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Shu Meng
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yanpan Gao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| |
Collapse
|
42
|
Jouabadi SM, Ataabadi EA, Golshiri K, Bos D, Stricker BHC, Danser AHJ, Mattace-Raso F, Roks AJM. Clinical Impact and Mechanisms of Nonatherosclerotic Vascular Aging: The New Kid to Be Blocked. Can J Cardiol 2023; 39:1839-1858. [PMID: 37495207 DOI: 10.1016/j.cjca.2023.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/07/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023] Open
Abstract
Ischemic cardiovascular disease and stroke remain the leading cause of global morbidity and mortality. During aging, protective mechanisms in the body gradually deteriorate, resulting in functional, structural, and morphologic changes that affect the vascular system. Because atherosclerotic plaques are not always present along with these alterations, we refer to this kind of vascular aging as nonatherosclerotic vascular aging (NAVA). To maintain proper vascular function during NAVA, it is important to preserve intracellular signalling, prevent inflammation, and block the development of senescent cells. Pharmacologic interventions targeting these components are potential therapeutic approaches for NAVA, with a particular emphasis on inflammation and senescence. This review provides an overview of the pathophysiology of vascular aging and explores potential pharmacotherapies that can improve the function of aged vasculature, focusing on NAVA.
Collapse
Affiliation(s)
- Soroush Mohammadi Jouabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ehsan Ataei Ataabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Keivan Golshiri
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Bruno H C Stricker
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Francesco Mattace-Raso
- Division of Geriatric Medicine, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anton J M Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
43
|
Worman HJ, Michaelis S. Prelamin A and ZMPSTE24 in premature and physiological aging. Nucleus 2023; 14:2270345. [PMID: 37885131 PMCID: PMC10730219 DOI: 10.1080/19491034.2023.2270345] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
As human longevity increases, understanding the molecular mechanisms that drive aging becomes ever more critical to promote health and prevent age-related disorders. Premature aging disorders or progeroid syndromes can provide critical insights into aspects of physiological aging. A major cause of progeroid syndromes which result from mutations in the genes LMNA and ZMPSTE24 is disruption of the final posttranslational processing step in the production of the nuclear scaffold protein lamin A. LMNA encodes the lamin A precursor, prelamin A and ZMPSTE24 encodes the prelamin A processing enzyme, the zinc metalloprotease ZMPSTE24. Progeroid syndromes resulting from mutations in these genes include the clinically related disorders Hutchinson-Gilford progeria syndrome (HGPS), mandibuloacral dysplasia-type B, and restrictive dermopathy. These diseases have features that overlap with one another and with some aspects of physiological aging, including bone defects resembling osteoporosis and atherosclerosis (the latter primarily in HGPS). The progeroid syndromes have ignited keen interest in the relationship between defective prelamin A processing and its accumulation in normal physiological aging. In this review, we examine the hypothesis that diminished processing of prelamin A by ZMPSTE24 is a driver of physiological aging. We review features a new mouse (LmnaL648R/L648R) that produces solely unprocessed prelamin A and provides an ideal model for examining the effects of its accumulation during aging. We also discuss existing data on the accumulation of prelamin A or its variants in human physiological aging, which call out for further validation and more rigorous experimental approaches to determine if prelamin A contributes to normal aging.
Collapse
Affiliation(s)
- Howard J. Worman
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Susan Michaelis
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
44
|
Abdellatif M, Rainer PP, Sedej S, Kroemer G. Hallmarks of cardiovascular ageing. Nat Rev Cardiol 2023; 20:754-777. [PMID: 37193857 DOI: 10.1038/s41569-023-00881-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/18/2023]
Abstract
Normal circulatory function is a key determinant of disease-free life expectancy (healthspan). Indeed, pathologies affecting the cardiovascular system, which are growing in prevalence, are the leading cause of global morbidity, disability and mortality, whereas the maintenance of cardiovascular health is necessary to promote both organismal healthspan and lifespan. Therefore, cardiovascular ageing might precede or even underlie body-wide, age-related health deterioration. In this Review, we posit that eight molecular hallmarks are common denominators in cardiovascular ageing, namely disabled macroautophagy, loss of proteostasis, genomic instability (in particular, clonal haematopoiesis of indeterminate potential), epigenetic alterations, mitochondrial dysfunction, cell senescence, dysregulated neurohormonal signalling and inflammation. We also propose a hierarchical order that distinguishes primary (upstream) from antagonistic and integrative (downstream) hallmarks of cardiovascular ageing. Finally, we discuss how targeting each of the eight hallmarks might be therapeutically exploited to attenuate residual cardiovascular risk in older individuals.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, Austria.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- BioTechMed Graz, Graz, Austria.
| | - Peter P Rainer
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
45
|
Brandt A, Petrovsky R, Kriebel M, Großhans J. Use of Farnesyl Transferase Inhibitors in an Ageing Model in Drosophila. J Dev Biol 2023; 11:40. [PMID: 37987370 PMCID: PMC10660854 DOI: 10.3390/jdb11040040] [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: 08/10/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/22/2023] Open
Abstract
The presence of farnesylated proteins at the inner nuclear membrane (INM), such as the Lamins or Kugelkern in Drosophila, leads to specific changes in the nuclear morphology and accelerated ageing on the organismal level reminiscent of the Hutchinson-Gilford progeria syndrome (HGPS). Farnesyl transferase inhibitors (FTIs) can suppress the phenotypes of the nuclear morphology in cultured fibroblasts from HGPS patients and cultured cells overexpressing farnesylated INM proteins. Similarly, FTIs have been reported to suppress the shortened lifespan in model organisms. Here, we report an experimental system combining cell culture and Drosophila flies for testing the activity of substances on the HGPS-like nuclear morphology and lifespan, with FTIs as an experimental example. Consistent with previous reports, we show that FTIs were able to ameliorate the nuclear phenotypes induced by the farnesylated nuclear proteins Progerin, Kugelkern, or truncated Lamin B in cultured cells. The subsequent validation in Drosophila lifespan assays demonstrated the applicability of the experimental system: treating adult Drosophila with the FTI ABT-100 reversed the nuclear phenotypes and extended the lifespan of experimentally induced short-lived flies. Since kugelkern-expressing flies have a significantly shorter average lifespan, half the time is needed for testing substances in the lifespan assay.
Collapse
Affiliation(s)
| | - Roman Petrovsky
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| | - Maria Kriebel
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| | - Jörg Großhans
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| |
Collapse
|
46
|
van der Linden J, Trap L, Scherer CV, Roks AJM, Danser AHJ, van der Pluijm I, Cheng C. Model Systems to Study the Mechanism of Vascular Aging. Int J Mol Sci 2023; 24:15379. [PMID: 37895059 PMCID: PMC10607365 DOI: 10.3390/ijms242015379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death globally. Within cardiovascular aging, arterial aging holds significant importance, as it involves structural and functional alterations in arteries that contribute substantially to the overall decline in cardiovascular health during the aging process. As arteries age, their ability to respond to stress and injury diminishes, while their luminal diameter increases. Moreover, they experience intimal and medial thickening, endothelial dysfunction, loss of vascular smooth muscle cells, cellular senescence, extracellular matrix remodeling, and deposition of collagen and calcium. This aging process also leads to overall arterial stiffening and cellular remodeling. The process of genomic instability plays a vital role in accelerating vascular aging. Progeria syndromes, rare genetic disorders causing premature aging, exemplify the impact of genomic instability. Throughout life, our DNA faces constant challenges from environmental radiation, chemicals, and endogenous metabolic products, leading to DNA damage and genome instability as we age. The accumulation of unrepaired damages over time manifests as an aging phenotype. To study vascular aging, various models are available, ranging from in vivo mouse studies to cell culture options, and there are also microfluidic in vitro model systems known as vessels-on-a-chip. Together, these models offer valuable insights into the aging process of blood vessels.
Collapse
Affiliation(s)
- Janette van der Linden
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Molecular Genetics, Cancer Genomics Center Netherlands, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Lianne Trap
- Department of Pulmonary Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Caroline V. Scherer
- Department of Molecular Genetics, Cancer Genomics Center Netherlands, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Anton J. M. Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - A. H. Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Cancer Genomics Center Netherlands, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
47
|
Walther BK, Sears AP, Mojiri A, Avazmohammadi R, Gu J, Chumakova OV, Pandian NKR, Dominic A, Martiel JL, Yazdani SK, Cooke JP, Ohayon J, Pettigrew RI. Disrupted Stiffness Ratio Alters Nuclear Mechanosensing. MATTER 2023; 6:3608-3630. [PMID: 37937235 PMCID: PMC10627551 DOI: 10.1016/j.matt.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
The ability of endothelial cells to sense and respond to dynamic changes in blood flow is critical for vascular homeostasis and cardiovascular health. The mechanical and geometric properties of the nuclear and cytoplasmic compartments affect mechanotransduction. We hypothesized that alterations to these parameters have resulting mechanosensory consequences. Using atomic force microscopy and mathematical modeling, we assessed how the nuclear and cytoplasmic compartment stiffnesses modulate shear stress transfer to the nucleus within aging endothelial cells. Our computational studies revealed that the critical parameter controlling shear transfer is not the individual mechanics of these compartments, but the stiffness ratio between them. Replicatively aged cells had a reduced stiffness ratio, attenuating shear transfer, while the ratio was not altered in a genetic model of accelerated aging. We provide a theoretical framework suggesting that dysregulation of the shear stress response can be uniquely imparted by relative mechanical changes in subcellular compartments.
Collapse
Affiliation(s)
- Brandon K. Walther
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- Texas A&M University, Department of Biomedical Engineering, College Station, TX 77843, USA
| | - Adam P. Sears
- Texas A&M University, Department of Biomedical Engineering, College Station, TX 77843, USA
- Houston Methodist Hospital, Houston, TX 77030, USA
| | - Anahita Mojiri
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Reza Avazmohammadi
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- Texas A&M University, Department of Biomedical Engineering, College Station, TX 77843, USA
- Texas A&M University, Department of Mechanical Engineering, College Station, TX 77843, USA
| | - Jianhua Gu
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Olga V. Chumakova
- University of Texas Health Science Center, Department of Integrative Biology and Pharmacology, Houston, TX 77030, USA
| | | | - Abishai Dominic
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | | | - Saami K. Yazdani
- Wake Forest University, Department of Engineering, Winston-Salem, NC 27101, USA
| | - John P. Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- Texas A&M University, Department of Biomedical Engineering, College Station, TX 77843, USA
| | - Jacques Ohayon
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- University Grenoble Alpes, CNRS, TIMC UMR 5525, 38000 Grenoble, France
- Savoie Mont-Blanc University, Polytech Annecy-Chambéry, 73376 Le Bourget du Lac, France
| | - Roderic I. Pettigrew
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- Texas A&M University, Department of Biomedical Engineering, College Station, TX 77843, USA
- Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
48
|
Mannion AJ, Holmgren L. Nuclear mechanosensing of the aortic endothelium in health and disease. Dis Model Mech 2023; 16:dmm050361. [PMID: 37909406 PMCID: PMC10629673 DOI: 10.1242/dmm.050361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
The endothelium, the monolayer of endothelial cells that line blood vessels, is exposed to a number of mechanical forces, including frictional shear flow, pulsatile stretching and changes in stiffness influenced by extracellular matrix composition. These forces are sensed by mechanosensors that facilitate their transduction to drive appropriate adaptation of the endothelium to maintain vascular homeostasis. In the aorta, the unique architecture of the vessel gives rise to changes in the fluid dynamics, which, in turn, shape cellular morphology, nuclear architecture, chromatin dynamics and gene regulation. In this Review, we discuss recent work focusing on how differential mechanical forces exerted on endothelial cells are sensed and transduced to influence their form and function in giving rise to spatial variation to the endothelium of the aorta. We will also discuss recent developments in understanding how nuclear mechanosensing is implicated in diseases of the aorta.
Collapse
Affiliation(s)
- Aarren J. Mannion
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 171 64, Sweden
| | - Lars Holmgren
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 171 64, Sweden
| |
Collapse
|
49
|
Kim BH, Chung YH, Woo TG, Kang SM, Park S, Park BJ. Progerin, an Aberrant Spliced Form of Lamin A, Is a Potential Therapeutic Target for HGPS. Cells 2023; 12:2299. [PMID: 37759521 PMCID: PMC10527460 DOI: 10.3390/cells12182299] [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: 08/08/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare genetic disorder caused by the mutant protein progerin, which is expressed by the abnormal splicing of the LMNA gene. HGPS affects systemic levels, with the exception of cognition or brain development, in children, showing that cellular aging can occur in the short term. Studying progeria could be useful in unraveling the causes of human aging (as well as fatal age-related disorders). Elucidating the clear cause of HGPS or the development of a therapeutic medicine could improve the quality of life and extend the survival of patients. This review aimed to (i) briefly describe how progerin was discovered as the causative agent of HGPS, (ii) elucidate the puzzling observation of the absence of primary neurological disease in HGPS, (iii) present several studies showing the deleterious effects of progerin and the beneficial effects of its inhibition, and (iv) summarize research to develop a therapy for HGPS and introduce clinical trials for its treatment.
Collapse
Affiliation(s)
- Bae-Hoon Kim
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
| | - Yeon-Ho Chung
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
| | - Tae-Gyun Woo
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
| | - So-Mi Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea; (S.-M.K.); (S.P.)
| | - Soyoung Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea; (S.-M.K.); (S.P.)
| | - Bum-Joon Park
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea; (S.-M.K.); (S.P.)
| |
Collapse
|
50
|
Murtada SI, Kawamura Y, Cavinato C, Wang M, Ramachandra AB, Spronck B, Li DS, Tellides G, Humphrey JD. Biomechanical and transcriptional evidence that smooth muscle cell death drives an osteochondrogenic phenotype and severe proximal vascular disease in progeria. Biomech Model Mechanobiol 2023; 22:1333-1347. [PMID: 37149823 PMCID: PMC10544720 DOI: 10.1007/s10237-023-01722-5] [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: 09/09/2022] [Accepted: 04/11/2023] [Indexed: 05/08/2023]
Abstract
Hutchinson-Gilford Progeria Syndrome results in rapid aging and severe cardiovascular sequelae that accelerate near end-of-life. We found a progressive disease process in proximal elastic arteries that was less evident in distal muscular arteries. Changes in aortic structure and function were then associated with changes in transcriptomics assessed via both bulk and single cell RNA sequencing, which suggested a novel sequence of progressive aortic disease: adverse extracellular matrix remodeling followed by mechanical stress-induced smooth muscle cell death, leading a subset of remnant smooth muscle cells to an osteochondrogenic phenotype that results in an accumulation of proteoglycans that thickens the aortic wall and increases pulse wave velocity, with late calcification exacerbating these effects. Increased central artery pulse wave velocity is known to drive left ventricular diastolic dysfunction, the primary diagnosis in progeria children. It appears that mechanical stresses above ~ 80 kPa initiate this progressive aortic disease process, explaining why elastic lamellar structures that are organized early in development under low wall stresses appear to be nearly normal whereas other medial constituents worsen progressively in adulthood. Mitigating early mechanical stress-driven smooth muscle cell loss/phenotypic modulation promises to have important cardiovascular implications in progeria patients.
Collapse
Affiliation(s)
- Sae-Il Murtada
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Yuki Kawamura
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Molly Wang
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | | | - Bart Spronck
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Maastricht University, Maastricht, Netherlands
| | - David S Li
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - George Tellides
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|