1
|
Joo EY, Park JS, Shin HT, Yoo M, Kim SJ, Lee JE, Choi GS. Mesenchymal Stem Cell Therapy for Hutchinson-Gilford Progeria: Improvements in Arterial Stiffness and Bone Mineral Density in a Single Case. CHILDREN (BASEL, SWITZERLAND) 2025; 12:523. [PMID: 40310235 PMCID: PMC12025413 DOI: 10.3390/children12040523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/24/2025] [Accepted: 04/02/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND/OBJECTIVES Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder that cause premature aging due to LMNA mutations and progerin accumulation. Although lonafarnib, an FDA-approved farnesyltransferase inhibitor, offers modest extension of life, the disease remains progressive. As progeria is associated with stem cell depletion and mesenchymal stem cell (MSC) therapy has shown efficacy in treating atherosclerosis, we aimed to evaluate its efficacy and safety in HGPS. METHODS A 7-year-old male with classic HGPS and preexisting severe cerebrovascular disease received four intravenous infusion of bone marrow-derived MSCs (2.5 × 10⁵ cells/kg) over 8 months. Growth, metabolic, cardiovascular, musculoskeletal, auditory, and inflammatory cytokines were monitored throughout the study. Prophylactic enoxaparin was administered to prevent vascular complications. RESULTS MSC therapy was associated with improved lean body mass (11.5%), bone mineral density (L-spine z-score: 0.55 → 2.03), reduced arterial stiffness (9.98% reductionin pulse wave velocity), joint range of motion, dentition, and decreased sICAM-1 levels. However, Cardiovascular deterioration continued, and the patient passed away 10 months after the fourth dose, likely due to progression of the underlying vascular disease. No severe adverse effects were attributed to MSC therapy. CONCLUSIONS MSC therapy may offer short-term benefits in arterial stiffness, bone health and inflammation in HGPS without notable safety concerns. Further studies are warranted to validate these findings, explore earlier intervention, and determine long-term efficacy and optimal dosing strategies.
Collapse
Affiliation(s)
- Eun-Young Joo
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Ji-Sun Park
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Hyun-Tae Shin
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
- Department of Dermatology, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Myungji Yoo
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Su-Jin Kim
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Ji-Eun Lee
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Gwang-Seong Choi
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Department of Dermatology, Inha University College of Medicine, Incheon 22212, Republic of Korea
| |
Collapse
|
2
|
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
|
3
|
Qi J, Fu X, Zhang L, Tan F, Li N, Sun Q, Hu X, He Z, Xia M, Chai R. Current AAV-mediated gene therapy in sensorineural hearing loss. FUNDAMENTAL RESEARCH 2025; 5:192-202. [PMID: 40166123 PMCID: PMC11955060 DOI: 10.1016/j.fmre.2022.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/04/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
The number of patients with hearing loss is on the rise due to congenital abnormalities, degenerative changes in old age, and acquired injuries such as virus or ototoxic drug-induced diseases. Hearing loss is a refractory and disabling disease that has serious negative effects on quality of life. The pathology of hearing loss in the inner ear is characterized by varying degrees of damage to the cochlear sensory epithelium cells (such as hair cells and supporting cells), stria vascularis (including marginal, intermediate and basal cells) and spiral ganglion neurons. Regeneration or direct repair of damaged cells in the inner ear is an effective way to treat sensorineural deafness. It is currently possible to regenerate hair cells to treat sensorineural hearing loss by FX-322, a small molecule drug in clinical trials. With the development of genetic engineering technology, gene therapy has brought a promising treatment strategy for many previously intractable diseases. Gene therapy has been regarded as a promising method in the treatment and rehabilitation of sensorineural hearing loss, and recombinant adeno-associated virus gene therapy has been widely used in fundamental research into hearing loss treatments. At present, gene therapy for hearing loss is transitioning from feasibility studies to explorations of its safety and its therapeutic potential. The present article reviews the concepts, strategies, and applications of gene therapy mediated by recombinant adeno-associated viruses in the field of hearing loss treatment.
Collapse
Affiliation(s)
- Jieyu Qi
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
- Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- State Key Laboratory of Hearing and Balance Science, Beijing Institute of Technology, Beijing 100081, China
- Advanced Technology Research Institute, Beijing Institute of Technology, Jinan 250300, China
| | - Xiaolong Fu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Liyan Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Fangzhi Tan
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Nianci Li
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Qiuhan Sun
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Xiaojie Hu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Zuhong He
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ming Xia
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
- Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- Southeast University Shenzhen Research Institute, Shenzhen 518063, China
| |
Collapse
|
4
|
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
|
5
|
Kjer-Hansen P, Phan TG, Weatheritt RJ. Protein isoform-centric therapeutics: expanding targets and increasing specificity. Nat Rev Drug Discov 2024; 23:759-779. [PMID: 39232238 DOI: 10.1038/s41573-024-01025-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 09/06/2024]
Abstract
Most protein-coding genes produce multiple protein isoforms; however, these isoforms are commonly neglected in drug discovery. The expression of protein isoforms can be specific to a disease, tissue and/or developmental stage, and this specific expression can be harnessed to achieve greater drug specificity than pan-targeting of all gene products and to enable improved treatments for diseases caused by aberrant protein isoform production. In recent years, several protein isoform-centric therapeutics have been developed. Here, we collate these studies and clinical trials to highlight three distinct but overlapping modes of action for protein isoform-centric drugs: isoform switching, isoform introduction or depletion, and modulation of isoform activity. In addition, we discuss how protein isoforms can be used clinically as targets for cell type-specific drug delivery and immunotherapy, diagnostic biomarkers and sources of cancer neoantigens. Collectively, we emphasize the value of a focus on isoforms as a route to discovering drugs with greater specificity and fewer adverse effects. This approach could enable the targeting of proteins for which pan-inhibition of all isoforms is toxic and poorly tolerated.
Collapse
Affiliation(s)
- Peter Kjer-Hansen
- EMBL Australia, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
- St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Darlinghurst, New South Wales, Australia.
| | - Tri Giang Phan
- St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Darlinghurst, New South Wales, Australia
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Robert J Weatheritt
- EMBL Australia, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
6
|
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
|
7
|
Krüger P, Hartinger R, Djabali K. Navigating Lipodystrophy: Insights from Laminopathies and Beyond. Int J Mol Sci 2024; 25:8020. [PMID: 39125589 PMCID: PMC11311807 DOI: 10.3390/ijms25158020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/06/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
Recent research into laminopathic lipodystrophies-rare genetic disorders caused by mutations in the LMNA gene-has greatly expanded our knowledge of their complex pathology and metabolic implications. These disorders, including Hutchinson-Gilford progeria syndrome (HGPS), Mandibuloacral Dysplasia (MAD), and Familial Partial Lipodystrophy (FPLD), serve as crucial models for studying accelerated aging and metabolic dysfunction, enhancing our understanding of the cellular and molecular mechanisms involved. Research on laminopathies has highlighted how LMNA mutations disrupt adipose tissue function and metabolic regulation, leading to altered fat distribution and metabolic pathway dysfunctions. Such insights improve our understanding of the pathophysiological interactions between genetic anomalies and metabolic processes. This review merges current knowledge on the phenotypic classifications of these diseases and their associated metabolic complications, such as insulin resistance, hypertriglyceridemia, hepatic steatosis, and metabolic syndrome, all of which elevate the risk of cardiovascular disease, stroke, and diabetes. Additionally, a range of published therapeutic strategies, including gene editing, antisense oligonucleotides, and novel pharmacological interventions aimed at addressing defective adipocyte differentiation and lipid metabolism, will be explored. These therapies target the core dysfunctional lamin A protein, aiming to mitigate symptoms and provide a foundation for addressing similar metabolic and genetic disorders.
Collapse
Affiliation(s)
| | | | - 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; (P.K.); (R.H.)
| |
Collapse
|
8
|
Sun Y, Xu L, Li Y, Jia S, Wang G, Cen X, Xu Y, Cao Z, Wang J, Shen N, Hu L, Zhang J, Mao J, Xia H, Liu Z, Fu X. Mitophagy defect mediates the aging-associated hallmarks in Hutchinson-Gilford progeria syndrome. Aging Cell 2024; 23:e14143. [PMID: 38482753 PMCID: PMC11296130 DOI: 10.1111/acel.14143] [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: 10/16/2023] [Revised: 02/03/2024] [Accepted: 03/01/2024] [Indexed: 06/13/2024] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare and fatal disease manifested by premature aging and aging-related phenotypes, making it a disease model for aging. The cellular machinery mediating age-associated phenotypes in HGPS remains largely unknown, resulting in limited therapeutic targets for HGPS. In this study, we showed that mitophagy defects impaired mitochondrial function and contributed to cellular markers associated with aging in mesenchymal stem cells derived from HGPS patients (HGPS-MSCs). Mechanistically, we discovered that mitophagy affected the aging-associated phenotypes of HGPS-MSCs by inhibiting the STING-NF-ĸB pathway and the downstream transcription of senescence-associated secretory phenotypes (SASPs). Furthermore, by utilizing UMI-77, an effective mitophagy inducer, we showed that mitophagy induction alleviated aging-associated phenotypes in HGPS and naturally aged mice. Collectively, our results uncovered that mitophagy defects mediated the aging-associated markers in HGPS, highlighted the function of mitochondrial homeostasis in HGPS progression, and suggested mitophagy as an intervention target for HGPS and aging.
Collapse
Affiliation(s)
- Yingying Sun
- The First Affiliated HospitalZhejiang University School of Medicine, and Liangzhu Laboratory of Zhejiang UniversityHangzhouZhejiangChina
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
| | - Le Xu
- The First Affiliated HospitalZhejiang University School of Medicine, and Liangzhu Laboratory of Zhejiang UniversityHangzhouZhejiangChina
| | - Yi Li
- The First Affiliated HospitalZhejiang University School of Medicine, and Liangzhu Laboratory of Zhejiang UniversityHangzhouZhejiangChina
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
| | - Shunze Jia
- The First Affiliated HospitalZhejiang University School of Medicine, and Liangzhu Laboratory of Zhejiang UniversityHangzhouZhejiangChina
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
| | - Gang Wang
- National Clinical Research Center of Kidney Diseases, Jinling HospitalNanjing University School of MedicineNanjingJiangsuChina
| | - Xufeng Cen
- The First Affiliated HospitalZhejiang University School of Medicine, and Liangzhu Laboratory of Zhejiang UniversityHangzhouZhejiangChina
| | - Yuyan Xu
- The First Affiliated HospitalZhejiang University School of Medicine, and Liangzhu Laboratory of Zhejiang UniversityHangzhouZhejiangChina
| | - Zhongkai Cao
- Department of Nephrology, The Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouZhejiangChina
| | - Jingjing Wang
- Department of Nephrology, The Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouZhejiangChina
| | - Ning Shen
- The First Affiliated HospitalZhejiang University School of Medicine, and Liangzhu Laboratory of Zhejiang UniversityHangzhouZhejiangChina
| | - Lidan Hu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouZhejiangChina
| | - Jin Zhang
- The First Affiliated HospitalZhejiang University School of Medicine, and Liangzhu Laboratory of Zhejiang UniversityHangzhouZhejiangChina
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical SciencesZhejiang University School of MedicineHangzhouChina
| | - Jianhua Mao
- Department of Nephrology, The Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouZhejiangChina
| | - Hongguang Xia
- The First Affiliated HospitalZhejiang University School of Medicine, and Liangzhu Laboratory of Zhejiang UniversityHangzhouZhejiangChina
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling HospitalNanjing University School of MedicineNanjingJiangsuChina
| | - Xudong Fu
- The First Affiliated HospitalZhejiang University School of Medicine, and Liangzhu Laboratory of Zhejiang UniversityHangzhouZhejiangChina
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
- Department of Geriatrics, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
9
|
Wu Z, Qu J, Zhang W, Liu GH. Stress, epigenetics, and aging: Unraveling the intricate crosstalk. Mol Cell 2024; 84:34-54. [PMID: 37963471 DOI: 10.1016/j.molcel.2023.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023]
Abstract
Aging, as a complex process involving multiple cellular and molecular pathways, is known to be exacerbated by various stresses. Because responses to these stresses, such as oxidative stress and genotoxic stress, are known to interplay with the epigenome and thereby contribute to the development of age-related diseases, investigations into how such epigenetic mechanisms alter gene expression and maintenance of cellular homeostasis is an active research area. In this review, we highlight recent studies investigating the intricate relationship between stress and aging, including its underlying epigenetic basis; describe different types of stresses that originate from both internal and external stimuli; and discuss potential interventions aimed at alleviating stress and restoring epigenetic patterns to combat aging or age-related diseases. Additionally, we address the challenges currently limiting advancement in this burgeoning field.
Collapse
Affiliation(s)
- Zeming Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; China National Center for Bioinformation, Beijing 100101, China; CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; The Fifth People's Hospital of Chongqing, Chongqing 400062, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
10
|
Hasper J, Welle K, Swovick K, Hryhorenko J, Ghaemmaghami S, Buchwalter A. Long lifetime and tissue-specific accumulation of lamin A/C in Hutchinson-Gilford progeria syndrome. J Cell Biol 2024; 223:e202307049. [PMID: 37966721 PMCID: PMC10651395 DOI: 10.1083/jcb.202307049] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/10/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023] Open
Abstract
LMNA mutations cause laminopathies that afflict the cardiovascular system and include Hutchinson-Gilford progeria syndrome. The origins of tissue specificity in these diseases are unclear as the lamin A/C proteins are broadly expressed. We show that LMNA transcript levels are not predictive of lamin A/C protein levels across tissues and use quantitative proteomics to discover that tissue context and disease mutation each influence lamin A/C protein's lifetime. Lamin A/C's lifetime is an order of magnitude longer in the aorta, heart, and fat, where laminopathy pathology is apparent, than in the liver and intestine, which are spared from the disease. Lamin A/C is especially insoluble in cardiovascular tissues, which may limit degradation and promote protein stability. Progerin is even more long lived than lamin A/C in the cardiovascular system and accumulates there over time. Progerin accumulation is associated with impaired turnover of hundreds of abundant proteins in progeroid tissues. These findings identify impaired lamin A/C protein turnover as a novel feature of laminopathy syndromes.
Collapse
Affiliation(s)
- John Hasper
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Kevin Welle
- Mass Spectrometry Resource Laboratory, University of Rochester, Rochester, NY, USA
| | - Kyle Swovick
- Mass Spectrometry Resource Laboratory, University of Rochester, Rochester, NY, USA
| | - Jennifer Hryhorenko
- Mass Spectrometry Resource Laboratory, University of Rochester, Rochester, NY, USA
| | - Sina Ghaemmaghami
- Mass Spectrometry Resource Laboratory, University of Rochester, Rochester, NY, USA
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Abigail Buchwalter
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Physiology, University of California, San Francisco, CA, USA
| |
Collapse
|
11
|
Macicior J, Fernández D, Ortega-Gutiérrez S. A new fluorescent probe for the visualization of progerin. Bioorg Chem 2024; 142:106967. [PMID: 37979321 DOI: 10.1016/j.bioorg.2023.106967] [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: 09/25/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/20/2023]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) or progeria is a rare genetic disease that causes premature aging, leading to a drastic reduction in the life expectancy of patients. Progeria is mainly caused by the intracellular accumulation of a defective protein called progerin, generated from a mutation in the LMNA gene. Currently, there is only one approved drug for the treatment of progeria, which has limited efficacy. It is believed that progerin levels are the most important biomarker related to the severity of the disease. However, there is a lack of effective tools to directly visualize progerin in the native cellular models, since the commercially available antibodies are not well suited for the direct visualization of progerin in cells from the mouse model of the disease. In this context, an alternative option for the visualization of a protein relies on the use of fluorescent chemical probes, molecules with affinity and specificity towards a protein. In this work we report the synthesis and characterization of a new fluorescent probe (UCM-23079) that allows for the direct visualization of progerin in cells from the most widely used progeroid mouse model. Thus, UCM-23079 is a new tool compound that could help prioritize potential preclinical therapies towards the final goal of finding a definitive cure for progeria.
Collapse
Affiliation(s)
- Jon Macicior
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Plaza de las Ciencias s/n, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Daniel Fernández
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Plaza de las Ciencias s/n, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Silvia Ortega-Gutiérrez
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Plaza de las Ciencias s/n, Universidad Complutense de Madrid, E-28040 Madrid, Spain.
| |
Collapse
|
12
|
Xu D, Guo Y, Qi Z, Hao C, Yu G. An infant with congenital micrognathia and upper airway obstruction was diagnosed as Hutchinson-Gilford progeria syndrome caused by a novel LMNA mutation: Case report and literature review. Heliyon 2023; 9:e20857. [PMID: 37916118 PMCID: PMC10616127 DOI: 10.1016/j.heliyon.2023.e20857] [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: 03/06/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare disease characterized by appearance of premature aging, including the skin, bones, heart, and blood vessels caused by LMNA mutation. In this study, the patient presented with congenital micrognathia and progressively aggravated upper airway obstruction as the initial symptom, which required bilateral mandibular distraction osteogenesis (MDO) surgery intervention. This was not commonly described in the literature, and the primary clinical diagnosis of Pierre Robin sequence (PRS) was made. However, other clinical features included sclerotic skin, dry skin, growth failure, lipoatrophy, joint stiffness, prominent scalp veins, small ear lobes, hair loss, and craniofacial disproportion gradually emerged, the diagnosis of HGPS was preferred when the patient was 5 months old. The genetic testing result with a novel and de novo LMNA mutation (c.1968 + 3_1968+6delGAGT) further confirmed the diagnosis and expanded the clinical and mutational spectrum of HGPS. During the 12-month follow-up period after surgery, the patient no longer suffered dyspnea. Complications of other organs and systems have not happened at the moment. In addition, the pathogenesis, the role of LMNA gene mutation, the progress in clinical treatment, and breakthrough studies about genetic treatment in animals of HGPS are described in the literature review.
Collapse
Affiliation(s)
- Duojiao Xu
- Department of Stomatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yujiao Guo
- Department of Stomatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Zhan Qi
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Rare Disease Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Chanjuan Hao
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Rare Disease Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Guoxia Yu
- Department of Stomatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
13
|
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
|
14
|
Cabral WA, Stephan C, Terajima M, Thaivalappil AA, Blanchard O, Tavarez UL, Narisu N, Yan T, Wincovitch S, Taga Y, Yamauchi M, Kozloff KM, Erdos MR, Collins FS. Bone dysplasia in Hutchinson-Gilford progeria syndrome is associated with dysregulated differentiation and function of bone cell populations. Aging Cell 2023; 22:e13903. [PMID: 37365004 PMCID: PMC10497813 DOI: 10.1111/acel.13903] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disorder affecting tissues of mesenchymal origin. Most individuals with HGPS harbor a de novo c.1824C > T (p.G608G) mutation in the gene encoding lamin A (LMNA), which activates a cryptic splice donor site resulting in production of the toxic "progerin" protein. Clinical manifestations include growth deficiency, lipodystrophy, sclerotic dermis, cardiovascular defects, and bone dysplasia. Here we utilized the LmnaG609G knock-in (KI) mouse model of HGPS to further define mechanisms of bone loss associated with normal and premature aging disorders. Newborn skeletal staining of KI mice revealed altered rib cage shape and spinal curvature, and delayed calvarial mineralization with increased craniofacial and mandibular cartilage content. MicroCT analysis and mechanical testing of adult femurs indicated increased fragility associated with reduced bone mass, recapitulating the progressive bone deterioration that occurs in HGPS patients. We investigated mechanisms of bone loss in KI mice at the cellular level in bone cell populations. Formation of wild-type and KI osteoclasts from marrow-derived precursors was inhibited by KI osteoblast-conditioned media in vitro, suggesting a secreted factor(s) responsible for decreased osteoclasts on KI trabecular surfaces in vivo. Cultured KI osteoblasts exhibited abnormal differentiation characterized by reduced deposition and mineralization of extracellular matrix with increased lipid accumulation compared to wild-type, providing a mechanism for altered bone formation. Furthermore, quantitative analyses of KI transcripts confirmed upregulation of adipogenic genes both in vitro and in vivo. Thus, osteoblast phenotypic plasticity, inflammation and altered cellular cross-talk contribute to abnormal bone formation in HGPS mice.
Collapse
Affiliation(s)
- Wayne A. Cabral
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Chris Stephan
- Departments of Orthopedic Surgery and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Masahiko Terajima
- Division of Oral and Craniofacial Health Sciences, Adams School of DentistryUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Abhirami A. Thaivalappil
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Owen Blanchard
- Departments of Orthopedic Surgery and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Urraca L. Tavarez
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Narisu Narisu
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Tingfen Yan
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Stephen M. Wincovitch
- Cytogenetics and Microscopy CoreNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Yuki Taga
- Nippi Research Institute of BiomatrixIbarakiJapan
| | - Mitsuo Yamauchi
- Division of Oral and Craniofacial Health Sciences, Adams School of DentistryUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Kenneth M. Kozloff
- Departments of Orthopedic Surgery and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Michael R. Erdos
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Francis S. Collins
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| |
Collapse
|
15
|
Cisneros B, García-Aguirre I, De Ita M, Arrieta-Cruz I, Rosas-Vargas H. Hutchinson-Gilford Progeria Syndrome: Cellular Mechanisms and Therapeutic Perspectives. Arch Med Res 2023; 54:102837. [PMID: 37390702 DOI: 10.1016/j.arcmed.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/26/2023] [Accepted: 06/14/2023] [Indexed: 07/02/2023]
Abstract
In humans, aging is characterized by a gradual decline of physical and psychological functions, with the concomitant onset of chronic-degenerative diseases, which ultimately lead to death. The study of Hutchinson-Gilford progeria syndrome (HGPS), a premature aging disorder that recapitulates several features of natural aging, has provided important insights into deciphering the aging process. The genetic origin of HGPS is a de novo point mutation in the LMNA gene that drives the synthesis of progerin, mutant version of lamin A. Progerin is aberrantly anchored to the nuclear envelope disrupting a plethora of molecular processes; nonetheless, how progerin exerts a cascade of deleterious alterations at the cellular and systemic levels is not fully understood. Over the past decade, the use of different cellular and animal models for HGPS has allowed the identification of the molecular mechanisms underlying HGPS, paving the way towards the development of therapeutic treatments against the disease. In this review, we present an updated overview of the biology of HGPS, including its clinical features, description of key cellular processes affected by progerin (nuclear morphology and function, nucleolar activity, mitochondrial function, protein nucleocytoplasmic trafficking and telomere homeostasis), as well as discussion of the therapeutic strategies under development.
Collapse
Affiliation(s)
- Bulmaro Cisneros
- Genetics and Molecular Biology Department, Research and Advanced Studies Center, National Polytechnical Institute, Mexico City, Mexico
| | - Ian García-Aguirre
- Genetics and Molecular Biology Department, Research and Advanced Studies Center, National Polytechnical Institute, Mexico City, Mexico; Bioengineering Department, School of Engineering and Sciences, Tecnológico de Monterrey, Mexico City, Mexico
| | - Marlon De Ita
- Genetics and Molecular Biology Department, Research and Advanced Studies Center, National Polytechnical Institute, Mexico City, Mexico; Medical Research Unit in Human Genetics, Pediatrics Hospital, 21st Century National Medical Center, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Isabel Arrieta-Cruz
- Basic Research Department, Research Direction, National Institute of Geriatrics, Ministry of Health, Mexico City, Mexico
| | - Haydeé Rosas-Vargas
- Medical Research Unit in Human Genetics, Pediatrics Hospital, 21st Century National Medical Center, Instituto Mexicano del Seguro Social, Mexico City, Mexico.
| |
Collapse
|
16
|
Eriksson M, Haugaa K, Revêchon G. Readily Available Tools to Detect Progerin and Cardiac Disease Progression in Hutchinson-Gilford Progeria Syndrome. Circulation 2023; 147:1745-1747. [PMID: 37276251 DOI: 10.1161/circulationaha.123.064765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Affiliation(s)
- Maria Eriksson
- Departments of Biosciences and Nutrition (M.E., G.R.), Karolinska Institutet, Huddinge, Sweden
| | - Kristina Haugaa
- Medicine Huddinge (K.H.), Karolinska Institutet, Huddinge, Sweden
| | - Gwladys Revêchon
- Departments of Biosciences and Nutrition (M.E., G.R.), Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
17
|
Preußner M, Smith HL, Hughes D, Zhang M, Emmerichs A, Scalzitti S, Peretti D, Swinden D, Neumann A, Haltenhof T, Mallucci GR, Heyd F. ASO targeting RBM3 temperature-controlled poison exon splicing prevents neurodegeneration in vivo. EMBO Mol Med 2023; 15:e17157. [PMID: 36946385 PMCID: PMC10165353 DOI: 10.15252/emmm.202217157] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 03/23/2023] Open
Abstract
Neurodegenerative diseases are increasingly prevalent in the aging population, yet no disease-modifying treatments are currently available. Increasing the expression of the cold-shock protein RBM3 through therapeutic hypothermia is remarkably neuroprotective. However, systemic cooling poses a health risk, strongly limiting its clinical application. Selective upregulation of RBM3 at normothermia thus holds immense therapeutic potential. Here we identify a poison exon within the RBM3 gene that is solely responsible for its cold-induced expression. Genetic removal or antisense oligonucleotide (ASO)-mediated manipulation of this exon yields high RBM3 levels independent of cooling. Notably, a single administration of ASO to exclude the poison exon, using FDA-approved chemistry, results in long-lasting increased RBM3 expression in mouse brains. In prion-diseased mice, this treatment leads to remarkable neuroprotection, with prevention of neuronal loss and spongiosis despite high levels of disease-associated prion protein. Our promising results in mice support the possibility that RBM3-inducing ASOs might also deliver neuroprotection in humans in conditions ranging from acute brain injury to Alzheimer's disease.
Collapse
Affiliation(s)
- Marco Preußner
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
| | - Heather L Smith
- UK Dementia Research Institute and Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Altos LabsCambridge Institute of ScienceCambridgeUK
| | - Daniel Hughes
- UK Dementia Research Institute and Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Min Zhang
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
| | - Ann‐Kathrin Emmerichs
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
| | - Silvia Scalzitti
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
| | - Diego Peretti
- UK Dementia Research Institute and Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Dean Swinden
- UK Dementia Research Institute and Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Altos LabsCambridge Institute of ScienceCambridgeUK
| | - Alexander Neumann
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
- Omiqa BioinformaticsBerlinGermany
| | - Tom Haltenhof
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
- Omiqa BioinformaticsBerlinGermany
| | - Giovanna R Mallucci
- UK Dementia Research Institute and Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Altos LabsCambridge Institute of ScienceCambridgeUK
| | - Florian Heyd
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
| |
Collapse
|
18
|
Liu FX, Zhang DP, Ma YM, Zhang HL, Liu XZ, Zhang ZQ, Sun RQ, Zhang YK, Miao JX, Wu ZX, Liu YL, Feng YC. Effect of Jiawei Tongqiao Huoxue decoction in basilar artery dolichoectasia mice through yes-associated protein/transcriptional Co-activator with PDZ-binding motif pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116599. [PMID: 37149070 DOI: 10.1016/j.jep.2023.116599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Jiawei Tongqiao Huoxue decoction (JTHD), composed of Acorus calamus var. angustatus Besser, Paeonia lactiflora Pall., Conioselinum anthriscoides 'Chuanxiong', Prunus persica (L.) Batsch, Ziziphus jujuba Mill., Carthamus tinctorius L., Pueraria montana var. lobata (Willd.) Maesen & S.M.Almeida ex Sanjappa & Predeep, Zingiber officinale Roscoe, Leiurus quinquestriatus, and Moschus berezovskii Flerov, was developed based on Tongqiao Huoxue decoction in Wang Qingren's "Yilin Gaicuo" in the Qing Dynasty. It has the effect of improving not only the blood flow velocity of vertebral and basilar arteries but also the blood flow parameters and wall shear stress. Especially in recent years, the potential efficacy of traditional Chinese medicine (TCM) for the treatment of basilar artery dolichoectasia (BAD) has attracted great attention as there are still no specific remedies for this disease. However, its molecular mechanism has not been elucidated. To identify the potential mechanisms of JTHD will help to intervene BAD and provide a reference for its clinical application. AIM OF THE STUDY This study aims to establish a mouse model of BAD and explore the mechanism of JTHD regulating yes-associated protein/transcriptional co-activator with PDZ-binding motif (YAP/TAZ) pathway for attenuating BAD mice development. MATERIALS AND METHODS Sixty post-modeling C57/BL6 female mice were randomly divided into sham-operated, model, atorvastatin calcium tablet, low-dose JTHD, and high-dose JTHD groups. After 14 days of modeling, the pharmacological intervention was given for 2 months. Then, JTHD was analyzed by liquid chromatography-tandem mass spectrometry (LC-MS). ELISA was utilized to detect the changes in vascular endothelial growth factor (VEGF) and lipoprotein a (Lp-a) in serum. EVG staining was conducted to observe the pathological changes of blood vessels. TUNEL method was employed to detect the apoptosis rate of vascular smooth muscle cells (VSMCs). Micro-CT and ImagePro Plus software were used to observe and calculate the tortuosity index, lengthening index, percentage increase in vessel diameter, and tortuosity of the basilar artery vessels in mice. Western blot analysis was performed to detect the expression levels of YAP and TAZ proteins in the vascular tissues of mice. RESULTS Many effective compounds such as choline, tryptophan, and leucine with anti-inflammation and vascular remodeling were identified in the Chinese medicine formula by LC-MS analysis. The serum levels of VEGF in the model mice decreased significantly while the levels of Lp-a increased obviously compared with those in the sham-operated group. The intima-media of the basilar artery wall showed severe disruption of the internal elastic layer, atrophy of the muscular layer, and hyaline changes of the connective tissue. Apoptosis of VSMCs added. Dilatation, elongation, and tortuosity of the basilar artery became notable, and tortuosity index, lengthening index, percentage increase in vessel diameter, and bending angle remarkably improved. The expression levels of YAP and TAZ protein in blood vessels elevated conspicuously (P < 0.05, P < 0.01). JTHD group markedly reduced the lengthening, bending angle, percentage increase in vessel diameter, and tortuosity index of basilar artery compared with the model group after 2 months of pharmacological intervention. The group also decreased the secretion of Lp-a and increased the content of VEGF. It inhibited the destruction of the internal elastic layer, muscular atrophy, and hyaline degeneration of connective tissue in basilar artery wall. The apoptosis of VSMCs was decreased, and the expression levels of YAP and TAZ proteins were abated (P < 0.05, P < 0.01). CONCLUSIONS The mechanism of inhibition of basilar artery elongation, dilation, and tortuosity by JTHD, which has various anti-BAD effective compound components, may be related to the reduction in VSMCs apoptosis and downregulation of YAP/TAZ pathway expression.
Collapse
Affiliation(s)
- Fei Xiang Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Henan Vertigo Disease Diagnosis and Treatment Center, Zhengzhou, China; Institute of Vertigo Disease, Henan University of Chinese Medicine, Zhengzhou, China
| | - Dao Pei Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Henan Vertigo Disease Diagnosis and Treatment Center, Zhengzhou, China; Institute of Vertigo Disease, Henan University of Chinese Medicine, Zhengzhou, China.
| | - Yan Min Ma
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Huai Liang Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Henan Vertigo Disease Diagnosis and Treatment Center, Zhengzhou, China; Institute of Vertigo Disease, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiang Zhe Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhen Qiang Zhang
- College of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Rui Qin Sun
- Research and Experiment Center, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yun Ke Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China.
| | - Jin Xin Miao
- Research and Experiment Center, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhao Xin Wu
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Ya Li Liu
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Yan Chen Feng
- Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
19
|
Hasper J, Welle K, Swovick K, Hryhorenko J, Ghaemmaghami S, Buchwalter A. Long lifetime and selective accumulation of the A-type lamins accounts for the tissue specificity of Hutchinson-Gilford progeria syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.04.527139. [PMID: 37162946 PMCID: PMC10168242 DOI: 10.1101/2023.02.04.527139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Mutations to the LMNA gene cause laminopathies including Hutchinson-Gilford progeria syndrome (HGPS) that severely affect the cardiovascular system. The origins of tissue specificity in these diseases are unclear, as the A-type Lamins are abundant and broadly expressed proteins. We show that A-type Lamin protein and transcript levels are uncorrelated across tissues. As protein-transcript discordance can be caused by variations in protein lifetime, we applied quantitative proteomics to profile protein turnover rates in healthy and progeroid tissues. We discover that tissue context and disease mutation each influence A-type Lamin protein lifetime. Lamin A/C has a weeks-long lifetime in the aorta, heart, and fat, where progeroid pathology is apparent, but a days-long lifetime in the liver and gastrointestinal tract, which are spared from disease. The A-type Lamins are insoluble and densely bundled in cardiovascular tissues, which may present an energetic barrier to degradation and promote long protein lifetime. Progerin is even more long-lived than Lamin A/C in the cardiovascular system and accumulates there over time. Progerin accumulation interferes broadly with protein homeostasis, as hundreds of abundant proteins turn over more slowly in progeroid tissues. These findings indicate that potential gene therapy interventions for HGPS will have significant latency and limited potency in disrupting the long-lived Progerin protein. Finally, we reveal that human disease alleles are significantly over-represented in the long-lived proteome, indicating that long protein lifetime may influence disease pathology and present a significant barrier to gene therapies for numerous human diseases.
Collapse
Affiliation(s)
- John Hasper
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Kevin Welle
- University of Rochester Mass Spectrometry Resource Laboratory, Rochester, NY
| | - Kyle Swovick
- University of Rochester Mass Spectrometry Resource Laboratory, Rochester, NY
| | - Jennifer Hryhorenko
- University of Rochester Mass Spectrometry Resource Laboratory, Rochester, NY
| | - Sina Ghaemmaghami
- University of Rochester Mass Spectrometry Resource Laboratory, Rochester, NY
- Department of Biology, University of Rochester, Rochester, NY
| | - Abigail Buchwalter
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Physiology, University of California, San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub, San Francisco, CA
| |
Collapse
|
20
|
Mu X, Gerhard-Herman MD, Zhang YS. Building Blood Vessel Chips with Enhanced Physiological Relevance. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201778. [PMID: 37693798 PMCID: PMC10489284 DOI: 10.1002/admt.202201778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Indexed: 09/12/2023]
Abstract
Blood vessel chips are bioengineered microdevices, consisting of biomaterials, human cells, and microstructures, which recapitulate essential vascular structure and physiology and allow a well-controlled microenvironment and spatial-temporal readouts. Blood vessel chips afford promising opportunities to understand molecular and cellular mechanisms underlying a range of vascular diseases. The physiological relevance is key to these blood vessel chips that rely on bioinspired strategies and bioengineering approaches to translate vascular physiology into artificial units. Here, we discuss several critical aspects of vascular physiology, including morphology, material composition, mechanical properties, flow dynamics, and mass transport, which provide essential guidelines and a valuable source of bioinspiration for the rational design of blood vessel chips. We also review state-of-art blood vessel chips that exhibit important physiological features of the vessel and reveal crucial insights into the biological processes and disease pathogenesis, including rare diseases, with notable implications for drug screening and clinical trials. We envision that the advances in biomaterials, biofabrication, and stem cells improve the physiological relevance of blood vessel chips, which, along with the close collaborations between clinicians and bioengineers, enable their widespread utility.
Collapse
Affiliation(s)
- Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marie Denise Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
21
|
Murtada SI, Mikush N, Wang M, Ren P, Kawamura Y, Ramachandra AB, Li DS, Braddock DT, Tellides G, Gordon LB, Humphrey JD. Lonafarnib improves cardiovascular function and survival in a mouse model of Hutchinson-Gilford progeria syndrome. eLife 2023; 12:82728. [PMID: 36930696 PMCID: PMC10023154 DOI: 10.7554/elife.82728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Clinical trials have demonstrated that lonafarnib, a farnesyltransferase inhibitor, extends the lifespan in patients afflicted by Hutchinson-Gilford progeria syndrome, a devastating condition that accelerates many characteristics of aging and results in premature death due to cardiovascular sequelae. The US Food and Drug Administration approved Zokinvy (lonafarnib) in November 2020 for treating these patients, yet a detailed examination of drug-associated effects on cardiovascular structure, properties, and function has remained wanting. In this paper, we report encouraging outcomes of daily post-weaning treatment with lonafarnib on the composition and biomechanical phenotype of elastic and muscular arteries as well as associated cardiac function in a well-accepted mouse model of progeria that exhibits severe perimorbid cardiovascular disease. Lonafarnib resulted in 100% survival of the treated progeria mice to the study end-point (time of 50% survival of untreated mice), with associated improvements in arterial structure and function working together to significantly reduce pulse wave velocity and improve left ventricular diastolic function. By contrast, neither treatment with the mTOR inhibitor rapamycin alone nor dual treatment with lonafarnib plus rapamycin improved outcomes over that achieved with lonafarnib monotherapy.
Collapse
Affiliation(s)
- Sae-Il Murtada
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
| | - Nicole Mikush
- Translational Research Imaging Center, Yale UniversityNew HavenUnited States
| | - Mo Wang
- Department of Surgery, Yale UniversityNew HavenUnited States
| | - Pengwei Ren
- Department of Surgery, Yale UniversityNew HavenUnited States
| | - Yuki Kawamura
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
| | | | - David S Li
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
| | | | - George Tellides
- Department of Surgery, Yale UniversityNew HavenUnited States
- Vascular Biology and Therapeutics Program, Yale UniversityNew HavenUnited States
| | - Leslie B Gordon
- Department of Pediatrics, Hasbro Children's Hospital and Warren Albert Medical School, Brown UniversityProvidenceUnited States
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
- Vascular Biology and Therapeutics Program, Yale UniversityNew HavenUnited States
| |
Collapse
|
22
|
Aslesh T, Erkut E, Ren J, Lim KRQ, Woo S, Hatlevig S, Moulton HM, Gosgnach S, Greer J, Maruyama R, Yokota T. DG9-conjugated morpholino rescues phenotype in SMA mice by reaching the CNS via a subcutaneous administration. JCI Insight 2023; 8:160516. [PMID: 36719755 PMCID: PMC10077475 DOI: 10.1172/jci.insight.160516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Antisense oligonucleotide-mediated (AO-mediated) therapy is a promising strategy to treat several neurological diseases, including spinal muscular atrophy (SMA). However, limited delivery to the CNS with AOs administered intravenously or subcutaneously is a major challenge. Here, we demonstrate a single subcutaneous administration of cell-penetrating peptide DG9 conjugated to an AO called phosphorodiamidate morpholino oligomer (PMO) reached the CNS and significantly prolonged the median survival compared with unconjugated PMO and R6G-PMO in a severe SMA mouse model. Treated mice exhibited substantially higher expression of full-length survival of motor neuron 2 in both the CNS and systemic tissues compared with nontreated and unmodified AO-treated mice. The treatment ameliorated the atrophic musculature and improved breathing function accompanied by improved muscle strength and innervation at the neuromuscular junction with no signs of apparent toxicity. We also demonstrated DG9-conjugated PMO localized in nuclei in the spinal cord and brain after subcutaneous injections. Our data identify DG9 peptide conjugation as a powerful way to improve the efficacy of AO-mediated splice modulation. Finally, DG9-PMO is a promising therapeutic option to treat SMA and other neurological diseases, overcoming the necessity for intrathecal injections and treating body-wide tissues without apparent toxicity.
Collapse
Affiliation(s)
| | | | - Jun Ren
- Neuroscience and Mental Health Institute.,Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Susan Hatlevig
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| | - Hong M Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| | - Simon Gosgnach
- Neuroscience and Mental Health Institute.,Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John Greer
- Neuroscience and Mental Health Institute.,Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Toshifumi Yokota
- Neuroscience and Mental Health Institute.,Department of Medical Genetics, and
| |
Collapse
|
23
|
Zhang N, Hu Q, Sui T, Fu L, Zhang X, Wang Y, Zhu X, Huang B, Lu J, Li Z, Zhang Y. Unique progerin C-terminal peptide ameliorates Hutchinson-Gilford progeria syndrome phenotype by rescuing BUBR1. NATURE AGING 2023; 3:185-201. [PMID: 36743663 PMCID: PMC10154249 DOI: 10.1038/s43587-023-00361-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 01/04/2023] [Indexed: 04/30/2023]
Abstract
An accumulating body of evidence indicates an association between mitotic defects and the aging process in Hutchinson-Gilford progeria syndrome (HGPS), which is a premature aging disease caused by progerin accumulation. Here, we found that BUBR1, a core component of the spindle assembly checkpoint, was downregulated during HGPS cellular senescence. The remaining BUBR1 was anchored to the nuclear membrane by binding with the C terminus of progerin, thus further limiting the function of BUBR1. Based on this, we established a unique progerin C-terminal peptide (UPCP) that effectively blocked the binding of progerin and BUBR1 and enhanced the expression of BUBR1 by interfering with the interaction between PTBP1 and progerin. Finally, UPCP significantly inhibited HGPS cellular senescence and ameliorated progeroid phenotypes, extending the lifespan of LmnaG609G/G609G mice. Our findings reveal an essential role for the progerin-PTBP1-BUBR1 axis in HGPS. Therapeutics designed around UPCP may be a beneficial strategy for HGPS treatment.
Collapse
Affiliation(s)
- Na Zhang
- Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Qianying Hu
- Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Tingting Sui
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Science, Jilin University, Changchun, China
| | - Lu Fu
- Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Xinglin Zhang
- Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Yu Wang
- Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Baiqu Huang
- Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Jun Lu
- Institute of Genetics and Cytology, Northeast Normal University, Changchun, China.
| | - Zhanjun Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Science, Jilin University, Changchun, China.
| | - Yu Zhang
- Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China.
| |
Collapse
|
24
|
Xue H, Cao K. Missing checkpoints in premature aging. NATURE AGING 2023; 3:146-147. [PMID: 37118120 DOI: 10.1038/s43587-022-00351-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- Huijing Xue
- 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
|
25
|
Hamczyk MR, Nevado RM. Vascular smooth muscle cell aging: Insights from Hutchinson-Gilford progeria syndrome. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2023; 35:42-51. [PMID: 35125249 DOI: 10.1016/j.arteri.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/13/2021] [Accepted: 11/03/2021] [Indexed: 02/08/2023]
Abstract
Vascular smooth muscle cells (VSMCs) constitute the principal cellular component of the medial layer of arteries and are responsible for vessel contraction and relaxation in response to blood flow. Alterations in VSMCs can hinder vascular system function, leading to vascular stiffness, calcification and atherosclerosis, which in turn may result in life-threatening complications. Pathological changes in VSMCs typically correlate with chronological age; however, there are certain conditions and diseases, such as Hutchinson-Gilford progeria syndrome (HGPS), that can accelerate this process, resulting in premature vascular aging. HGPS is a rare genetic disorder characterized by severe VSMC loss, accelerated atherosclerosis and death from myocardial infarction or stroke during the adolescence. Because experiments with mouse models have demonstrated that alterations in VSMCs are responsible for early atherosclerosis in HGPS, studies on this disease can provide insights into the mechanisms of vascular aging and assess the relative contribution of VSMCs to this process.
Collapse
Affiliation(s)
- Magda R Hamczyk
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| | - Rosa M Nevado
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| |
Collapse
|
26
|
Coll-Bonfill N, Mahajan U, Shashkova EV, Lin CJ, Mecham RP, Gonzalo S. Progerin induces a phenotypic switch in vascular smooth muscle cells and triggers replication stress and an aging-associated secretory signature. GeroScience 2022; 45:965-982. [PMID: 36482259 PMCID: PMC9886737 DOI: 10.1007/s11357-022-00694-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome is a premature aging disease caused by LMNA gene mutation and the production of a truncated prelamin A protein "progerin" that elicits cellular and organismal toxicity. Progerin accumulates in the vasculature, being especially detrimental for vascular smooth muscle cells (VSMC). Vessel stiffening and aortic atherosclerosis in HGPS patients are accompanied by VSMC depletion in the medial layer, altered extracellular matrix (ECM), and thickening of the adventitial layer. Mechanisms whereby progerin causes massive VSMC loss and vessel alterations remain poorly understood. Mature VSMC retain phenotypic plasticity and can switch to a synthetic/proliferative phenotype. Here, we show that progerin expression in human and mouse VSMC causes a switch towards the synthetic phenotype. This switch elicits some level of replication stress in normal cells, which is exacerbated in the presence of progerin, leading to telomere fragility, genomic instability, and ultimately VSMC death. Calcitriol prevents replication stress, telomere fragility, and genomic instability, reducing VSMC death. In addition, RNA-seq analysis shows induction of a profibrotic and pro-inflammatory aging-associated secretory phenotype upon progerin expression in human primary VSMC. Our data suggest that phenotypic switch-induced replication stress might be an underlying cause of VSMC loss in progeria, which together with loss of contractile features and gain of profibrotic and pro-inflammatory signatures contribute to vascular stiffness in HGPS.
Collapse
Affiliation(s)
- Nuria Coll-Bonfill
- grid.262962.b0000 0004 1936 9342Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1100 S Grand Blvd, St Louis, MO 63104 USA
| | - Urvashi Mahajan
- grid.262962.b0000 0004 1936 9342Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1100 S Grand Blvd, St Louis, MO 63104 USA
| | - Elena V. Shashkova
- grid.262962.b0000 0004 1936 9342Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1100 S Grand Blvd, St Louis, MO 63104 USA
| | - Chien-Jung Lin
- grid.4367.60000 0001 2355 7002Cell Biology and Physiology Department & Department of Medicine, Washington University School of Medicine, St Louis, MO 63108 USA ,grid.262962.b0000 0004 1936 9342Department of Internal Medicine, Cardiovascular Division, Saint Louis University School of Medicine, St Louis, MO 63104 USA
| | - Robert P. Mecham
- grid.4367.60000 0001 2355 7002Cell Biology and Physiology Department & Department of Medicine, Washington University School of Medicine, St Louis, MO 63108 USA
| | - Susana Gonzalo
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1100 S Grand Blvd, St Louis, MO, 63104, USA.
| |
Collapse
|
27
|
Hu Q, Zhang N, Sui T, Li G, Wang Z, Liu M, Zhu X, Huang B, Lu J, Li Z, Zhang Y. Anti-hsa-miR-59 alleviates premature senescence associated with Hutchinson-Gilford progeria syndrome in mice. EMBO J 2022; 42:e110937. [PMID: 36382717 PMCID: PMC9811625 DOI: 10.15252/embj.2022110937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a lethal premature aging disorder without an effective therapeutic regimen. Because of their targetability and influence on gene expression, microRNAs (miRNAs) are attractive therapeutic tools to treat diseases. Here we identified that hsa-miR-59 (miR-59) was markedly upregulated in HGPS patient cells and in multiple tissues of an HGPS mouse model (LmnaG609G/G609G ), which disturbed the interaction between RNAPII and TFIIH, resulting in abnormal expression of cell cycle genes by targeting high-mobility group A family HMGA1 and HMGA2. Functional inhibition of miR-59 alleviated the cellular senescence phenotype of HGPS cells. Treatment with AAV9-mediated anti-miR-59 reduced fibrosis in the quadriceps muscle, heart, and aorta, suppressed epidermal thinning and dermal fat loss, and yielded a 25.5% increase in longevity of LmnaG609G/G609G mice. These results identify a new strategy for the treatment of HGPS and provide insight into the etiology of HGPS disease.
Collapse
Affiliation(s)
- Qianying Hu
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| | - Na Zhang
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| | - Tingting Sui
- The Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal ScienceJilin UniversityChangchunChina
| | - Guanlin Li
- The Institute of Genetics and CytologyNortheast Normal UniversityChangchunChina
| | - Zhiyao Wang
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| | - Mingyue Liu
- The Institute of Genetics and CytologyNortheast Normal UniversityChangchunChina
| | - Xiaojuan Zhu
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| | - Baiqu Huang
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| | - Jun Lu
- The Institute of Genetics and CytologyNortheast Normal UniversityChangchunChina
| | - Zhanjun Li
- The Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal ScienceJilin UniversityChangchunChina
| | - Yu Zhang
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| |
Collapse
|
28
|
Kreda SM. Oligonucleotide-based therapies for cystic fibrosis. Curr Opin Pharmacol 2022; 66:102271. [PMID: 35988291 DOI: 10.1016/j.coph.2022.102271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/10/2022] [Accepted: 07/01/2022] [Indexed: 11/03/2022]
Abstract
In the clinically successful era of CFTR modulators and Theratyping, 10-20% of individuals with cystic fibrosis (CF) may develop disease due to CFTR mutations that remain undruggable. These individuals produce low levels of CFTR mRNA and/or not enough protein to be rescued with modulator drugs. Alternative therapeutic approaches to correct the CFTR defect at the mRNA level using nucleic acid technologies are currently feasible; e.g., oligonucleotides platforms, which are being rapidly developed to correct genetic disorders. Drug-like properties, great specificity, and predictable off-target effects by design make oligonucleotides a valuable approach with fewer clinical and ethical challenges than genomic editing strategies. Together with personalized and precision medicine approaches, oligonucleotides are ideal therapeutics to target CF-causing mutations that affect only a few individuals resilient to modulator therapies.
Collapse
Affiliation(s)
- Silvia M Kreda
- Marsico Lung Institute / Cystic Fibrosis Center, University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill, NC, 27599-7248, USA; Department of Medicine, University of North Carolina at Chapel Hill, NC, 27599-7248, USA; Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, NC, 27599-7248, USA.
| |
Collapse
|
29
|
Della Valle F, Reddy P, Yamamoto M, Liu P, Saera-Vila A, Bensaddek D, Zhang H, Prieto Martinez J, Abassi L, Celii M, Ocampo A, Nuñez Delicado E, Mangiavacchi A, Aiese Cigliano R, Rodriguez Esteban C, Horvath S, Izpisua Belmonte JC, Orlando V. LINE-1 RNA causes heterochromatin erosion and is a target for amelioration of senescent phenotypes in progeroid syndromes. Sci Transl Med 2022; 14:eabl6057. [PMID: 35947677 DOI: 10.1126/scitranslmed.abl6057] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Constitutive heterochromatin is responsible for genome repression of DNA enriched in repetitive sequences, telomeres, and centromeres. During physiological and pathological premature aging, heterochromatin homeostasis is profoundly compromised. Here, we showed that LINE-1 (Long Interspersed Nuclear Element-1; L1) RNA accumulation was an early event in both typical and atypical human progeroid syndromes. L1 RNA negatively regulated the enzymatic activity of the histone-lysine N-methyltransferase SUV39H1 (suppression of variegation 3-9 homolog 1), resulting in heterochromatin loss and onset of senescent phenotypes in vitro. Depletion of L1 RNA in dermal fibroblast cells from patients with different progeroid syndromes using specific antisense oligonucleotides (ASOs) restored heterochromatin histone 3 lysine 9 and histone 3 lysine 27 trimethylation marks, reversed DNA methylation age, and counteracted the expression of senescence-associated secretory phenotype genes such as p16, p21, activating transcription factor 3 (ATF3), matrix metallopeptidase 13 (MMP13), interleukin 1a (IL1a), BTG anti-proliferation factor 2 (BTG2), and growth arrest and DNA damage inducible beta (GADD45b). Moreover, systemic delivery of ASOs rescued the histophysiology of tissues and increased the life span of a Hutchinson-Gilford progeria syndrome mouse model. Transcriptional profiling of human and mouse samples after L1 RNA depletion demonstrated that pathways associated with nuclear chromatin organization, cell proliferation, and transcription regulation were enriched. Similarly, pathways associated with aging, inflammatory response, innate immune response, and DNA damage were down-regulated. Our results highlight the role of L1 RNA in heterochromatin homeostasis in progeroid syndromes and identify a possible therapeutic approach to treat premature aging and related syndromes.
Collapse
Affiliation(s)
- Francesco Della Valle
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Sciences and Engineering Division BESE, KAUST Environmental Epigenetics Program, Thuwal, Saudi Arabia
| | - Pradeep Reddy
- Salk Institute for Biological Studies, La Jolla, CA, USA.,Altos Labs, San Diego, CA, USA
| | - Mako Yamamoto
- Salk Institute for Biological Studies, La Jolla, CA, USA.,Altos Labs, San Diego, CA, USA
| | - Peng Liu
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Sciences and Engineering Division BESE, KAUST Environmental Epigenetics Program, Thuwal, Saudi Arabia
| | | | - Dalila Bensaddek
- King Abdullah University of Science and Technology (KAUST), Bioscience Core Lab
| | - Huoming Zhang
- King Abdullah University of Science and Technology (KAUST), Bioscience Core Lab
| | | | - Leila Abassi
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Sciences and Engineering Division BESE, KAUST Environmental Epigenetics Program, Thuwal, Saudi Arabia
| | - Mirko Celii
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Sciences and Engineering Division BESE, KAUST Environmental Epigenetics Program, Thuwal, Saudi Arabia
| | | | | | - Arianna Mangiavacchi
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Sciences and Engineering Division BESE, KAUST Environmental Epigenetics Program, Thuwal, Saudi Arabia
| | | | | | | | | | - Valerio Orlando
- King Abdullah University of Science and Technology (KAUST), Biological Environmental Sciences and Engineering Division BESE, KAUST Environmental Epigenetics Program, Thuwal, Saudi Arabia
| |
Collapse
|
30
|
Zhang Y, Qiao X, Liu L, Han W, Liu Q, Wang Y, Xie T, Tang Y, Wang T, Meng J, Ye A, He S, Chen R, Chen C. Long noncoding RNA MAGI2-AS3 regulates the H2O2 level and cell senescence via HSPA8. Redox Biol 2022; 54:102383. [PMID: 35797800 PMCID: PMC9287730 DOI: 10.1016/j.redox.2022.102383] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Yingmin Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lihui Liu
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wensheng Han
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qinghua Liu
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuanyuan Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ting Xie
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiheng Tang
- University of Chinese Academy of Sciences, Beijing, 100049, China; Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Tiepeng Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiao Meng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Aojun Ye
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shunmin He
- University of Chinese Academy of Sciences, Beijing, 100049, China; Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Runsheng Chen
- University of Chinese Academy of Sciences, Beijing, 100049, China; Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
31
|
Sengupta D, Sengupta K. Lamin A and telomere maintenance in aging: Two to Tango. Mutat Res 2022; 825:111788. [PMID: 35687934 DOI: 10.1016/j.mrfmmm.2022.111788] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 03/28/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
Lamin proteins which constitute the nuclear lamina in almost all higher eukaryotes, are mainly of two types A & B encoded by LMNA and LMNB1/B2 genes respectively. While lamin A remains the principal product of LMNA gene, variants like lamin C, C2 and A∆10 are also formed as alternate splice products. Role of lamin A in aging has been highlighted in recent times due to its association with progeroid or premature aging syndromes which is classified as a type of laminopathy. Progeria caused by accelerated accumulation of lamin A Δ50 or progerin occurs due to a mutation in this LMNA gene leading to defects in post translational modification of lamin A. One of the most common and severe symptoms of progeroid laminopathy is accelerated cellular senescence or aging along with bone resorption, muscle weakness, lipodystrophy and cardiovascular disorders. On the other hand, progerin accumulation and telomere dysfunction merge as common traits in the process of chronological aging. Two major hallmarks of physiological aging in humans include loss of genomic integrity and telomere attrition which can result from defective laminar organization leading to deformed nuclear architecture and culminates into replicative senescence. This also adversely affects epigenetic landscape, mitochondrial dysfunction and several signalling pathways like DNA repair, mTOR, MAPK, TGFβ. In this review, we discuss the telomere-lamina interplay in the context of physiological aging and progeria.
Collapse
Affiliation(s)
- Duhita Sengupta
- Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, West Bengal, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Kaushik Sengupta
- Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, West Bengal, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
32
|
Yang JH, Chang MW, Tsitsipatis D, Yang X, Martindale J, Munk R, Cheng A, Izydore E, Pandey PR, Piao Y, Mazan-Mamczarz K, De S, Abdelmohsen K, Gorospe M. LncRNA OIP5-AS1-directed miR-7 degradation promotes MYMX production during human myogenesis. Nucleic Acids Res 2022; 50:7115-7133. [PMID: 35736212 PMCID: PMC9262585 DOI: 10.1093/nar/gkac524] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/22/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) modulate gene expression programs in physiology and disease. Here, we report a noncoding RNA regulatory network that modulates myoblast fusion into multinucleated myotubes, a process that occurs during muscle development and muscle regeneration after injury. In early stages of human myogenesis, the levels of lncRNA OIP5-AS1 increased, while the levels of miR-7 decreased. Moreover, OIP5-AS1 bound and induced miR-7 decay via target RNA-directed miRNA decay; accordingly, loss of OIP5-AS1 attenuated, while antagonizing miR-7 accelerated, myotube formation. We found that the OIP5-AS1-mediated miR-7 degradation promoted myoblast fusion, as it derepressed the miR-7 target MYMX mRNA, which encodes the fusogenic protein myomixer (MYMX). Remarkably, an oligonucleotide site blocker interfered with the OIP5-AS1-directed miR-7 degradation, allowing miR-7 to accumulate, lowering MYMX production and suppressing myotube formation. These results highlight a mechanism whereby lncRNA OIP5-AS1-mediated miR-7 decay promotes myotube formation by stimulating a myogenic fusion program.
Collapse
Affiliation(s)
- Jen-Hao Yang
- Correspondence may also be addressed to Jen-Hao Yang. Tel: +1 410 454 8392;
| | - Ming-Wen Chang
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Xiaoling Yang
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Jennifer L Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Aiwu Cheng
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Elizabeth Izydore
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Poonam R Pandey
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Yulan Piao
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Krystyna Mazan-Mamczarz
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Supriyo De
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- To whom correspondence should be addressed. Tel: +1 410 454 8412;
| |
Collapse
|
33
|
Ebert T, Tran N, Schurgers L, Stenvinkel P, Shiels PG. Ageing - Oxidative stress, PTMs and disease. Mol Aspects Med 2022; 86:101099. [PMID: 35689974 DOI: 10.1016/j.mam.2022.101099] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022]
Abstract
Post-translational modifications (PTMs) have been proposed as a link between the oxidative stress-inflammation-ageing trinity, thereby affecting several hallmarks of ageing. Phosphorylation, acetylation, and ubiquitination cover >90% of all the reported PTMs. Several of the main PTMs are involved in normal "healthy" ageing and in different age-related diseases, for instance neurodegenerative, metabolic, cardiovascular, and bone diseases, as well as cancer and chronic kidney disease. Ultimately, data from human rare progeroid syndromes, but also from long-living animal species, imply that PTMs are critical regulators of the ageing process. Mechanistically, PTMs target epigenetic and non-epigenetic pathways during ageing. In particular, epigenetic histone modification has critical implications for the ageing process and can modulate lifespan. Therefore, PTM-based therapeutics appear to be attractive pharmaceutical candidates to reduce the burden of ageing-related diseases. Several phosphorylation and acetylation inhibitors have already been FDA-approved for the treatment of other diseases and offer a unique potential to investigate both beneficial effects and possible side-effects. As an example, the most well-studied senolytic compounds dasatinib and quercetin, which have already been tested in Phase 1 pilot studies, also act as kinase inhibitors, targeting cellular senescence and increasing lifespan. Future studies need to carefully determine the best PTM-based candidates for the treatment of the "diseasome of ageing".
Collapse
Affiliation(s)
- Thomas Ebert
- Karolinska Institute, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Stockholm, Sweden; University of Leipzig Medical Center, Medical Department III - Endocrinology, Nephrology, Rheumatology, Leipzig, Germany.
| | - Ngoc Tran
- University of Glasgow, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, Institute of Cancer Sciences, Glasgow, UK
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research School Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Peter Stenvinkel
- Karolinska Institute, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Stockholm, Sweden
| | - Paul G Shiels
- University of Glasgow, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, Institute of Cancer Sciences, Glasgow, UK
| |
Collapse
|
34
|
Benedicto I, Chen X, Bergo MO, Andrés V. Progeria: a perspective on potential drug targets and treatment strategies. Expert Opin Ther Targets 2022; 26:393-399. [DOI: 10.1080/14728222.2022.2078699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Ignacio Benedicto
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Xue Chen
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, China
| | - Martin O. Bergo
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83, Sweden
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Spain
| |
Collapse
|
35
|
Mosevitsky MI. Progerin and Its Role in Accelerated and Natural Aging. Mol Biol 2022. [DOI: 10.1134/s0026893322020091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
36
|
Aldalaqan S, Dalgliesh C, Luzzi S, Siachisumo C, Reynard LN, Ehrmann I, Elliott DJ. Cryptic splicing: common pathological mechanisms involved in male infertility and neuronal diseases. Cell Cycle 2021; 21:219-227. [PMID: 34927545 PMCID: PMC8855859 DOI: 10.1080/15384101.2021.2015672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
High levels of transcription and alternative splicing are recognized hallmarks of gene expression in the testis and largely driven by cells in meiosis. Because of this, the male meiosis stage of the cell cycle is often viewed as having a relatively permissive environment for gene expression. In this review, we highlight recent findings that identify the RNA binding protein RBMXL2 as essential for male meiosis. RBMXL2 functions as a “guardian of the transcriptome” that protects against the use of aberrant (or “cryptic”) splice sites that would disrupt gene expression. This newly discovered protective role during meiosis links with a wider field investigating mechanisms of cryptic splicing control that protect neurons from amyotrophic lateral sclerosis and Alzheimer’s disease. We discuss how the mechanism repressing cryptic splicing patterns during meiosis evolved, and why it may be essential for sperm production and male fertility.
Collapse
Affiliation(s)
- Saad Aldalaqan
- Newcastle University Bioscience Institute, Newcastle University, Central Parkway Newcastle, UK
| | - Caroline Dalgliesh
- Newcastle University Bioscience Institute, Newcastle University, Central Parkway Newcastle, UK
| | - Sara Luzzi
- Newcastle University Bioscience Institute, Newcastle University, Central Parkway Newcastle, UK
| | - Chileleko Siachisumo
- Newcastle University Bioscience Institute, Newcastle University, Central Parkway Newcastle, UK
| | - Louise N Reynard
- Newcastle University Bioscience Institute, Newcastle University, Central Parkway Newcastle, UK
| | - Ingrid Ehrmann
- Newcastle University Bioscience Institute, Newcastle University, Central Parkway Newcastle, UK
| | - David J Elliott
- Newcastle University Bioscience Institute, Newcastle University, Central Parkway Newcastle, UK
| |
Collapse
|
37
|
Sánchez-López A, Espinós-Estévez C, González-Gómez C, Gonzalo P, Andrés-Manzano MJ, Fanjul V, Riquelme-Borja R, Hamczyk MR, Macías Á, Del Campo L, Camafeita E, Vázquez J, Barkaway A, Rolas L, Nourshargh S, Dorado B, Benedicto I, Andrés V. Cardiovascular Progerin Suppression and Lamin A Restoration Rescue Hutchinson-Gilford Progeria Syndrome. Circulation 2021; 144:1777-1794. [PMID: 34694158 PMCID: PMC8614561 DOI: 10.1161/circulationaha.121.055313] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Hutchinson-Gilford progeria syndrome (HGPS) is a rare disorder characterized by premature aging and death mainly because of myocardial infarction, stroke, or heart failure. The disease is provoked by progerin, a variant of lamin A expressed in most differentiated cells. Patients look healthy at birth, and symptoms typically emerge in the first or second year of life. Assessing the reversibility of progerin-induced damage and the relative contribution of specific cell types is critical to determining the potential benefits of late treatment and to developing new therapies. METHODS We used CRISPR-Cas9 technology to generate LmnaHGPSrev/HGPSrev (HGPSrev) mice engineered to ubiquitously express progerin while lacking lamin A and allowing progerin suppression and lamin A restoration in a time- and cell type-specific manner on Cre recombinase activation. We characterized the phenotype of HGPSrev mice and crossed them with Cre transgenic lines to assess the effects of suppressing progerin and restoring lamin A ubiquitously at different disease stages as well as specifically in vascular smooth muscle cells and cardiomyocytes. RESULTS Like patients with HGPS, HGPSrev mice appear healthy at birth and progressively develop HGPS symptoms, including failure to thrive, lipodystrophy, vascular smooth muscle cell loss, vascular fibrosis, electrocardiographic anomalies, and precocious death (median lifespan of 15 months versus 26 months in wild-type controls, P<0.0001). Ubiquitous progerin suppression and lamin A restoration significantly extended lifespan when induced in 6-month-old mildly symptomatic mice and even in severely ill animals aged 13 months, although the benefit was much more pronounced on early intervention (84.5% lifespan extension in mildly symptomatic mice, P<0.0001, and 6.7% in severely ill mice, P<0.01). It is remarkable that major vascular alterations were prevented and lifespan normalized in HGPSrev mice when progerin suppression and lamin A restoration were restricted to vascular smooth muscle cells and cardiomyocytes. CONCLUSIONS HGPSrev mice constitute a new experimental model for advancing knowledge of HGPS. Our findings suggest that it is never too late to treat HGPS, although benefit is much more pronounced when progerin is targeted in mice with mild symptoms. Despite the broad expression pattern of progerin and its deleterious effects in many organs, restricting its suppression to vascular smooth muscle cells and cardiomyocytes is sufficient to prevent vascular disease and normalize lifespan.
Collapse
Affiliation(s)
- Amanda Sánchez-López
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.)
| | - Carla Espinós-Estévez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.)
| | - Cristina González-Gómez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.)
| | - Pilar Gonzalo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.)
| | - María J Andrés-Manzano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.)
| | - Víctor Fanjul
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.)
| | - Raquel Riquelme-Borja
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.)
| | - Magda R Hamczyk
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.).,Now with Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología, Universidad de Oviedo, Spain (M.R.H.)
| | - Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.)
| | - Lara Del Campo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.).,Now with Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain (L.d.C.)
| | - Emilio Camafeita
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.)
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.)
| | - Anna Barkaway
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (A.B., L.R., S.N.)
| | - Loïc Rolas
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (A.B., L.R., S.N.)
| | - Sussan Nourshargh
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (A.B., L.R., S.N.)
| | - Beatriz Dorado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.)
| | - Ignacio Benedicto
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (A.S.-L., C.E.-E., C.G.-G., P.G., M.J.A.-M., V.F., R.R.-B., M.R.H., A.M., L.d.C., E.C., J.V., B.D., I.B., V.A.)
| | - Vicente Andrés
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (A.S.-L., C.G.-G., P.G., M.J.A.-M., V.F., M.R.H., A.M., L.d.C., E.C., J.V., B.D., V.A.)
| |
Collapse
|
38
|
Chen J, Zhao H, Huang Y, Li Y, Fan J, Wang R, Han Z, Yang Z, Wu L, Wu D, Luo Y, Ji X. Dysregulation of Principal Circulating miRNAs in Non-human Primates Following Ischemic Stroke. Front Neurosci 2021; 15:738576. [PMID: 34539341 PMCID: PMC8441133 DOI: 10.3389/fnins.2021.738576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/13/2021] [Indexed: 11/13/2022] Open
Abstract
Despite the recent interest in plasma microRNA (miRNA) biomarkers in acute ischemic stroke patients, there is limited knowledge about the miRNAs directly related to stroke itself due to the multiple complications in patients, which has hindered the research progress of biomarkers and therapeutic targets of ischemic stroke. Therefore, in this study, we compared the differentially expressed miRNA profiles in the plasma of three rhesus monkeys pre- and post-cerebral ischemia. After cerebral ischemia, Rfam sequence category revealed increased ribosomic RNA (rRNA) and decreased transfer RNAs (tRNAs) in plasma. Of the 2049 miRNAs detected after cerebral ischemia, 36 were upregulated, and 76 were downregulated (fold change ≥2.0, P < 0.05). For example, mml-miR-191-5p, miR-421, miR-409-5p, and let-7g-5p were found to be significantly overexpressed, whereas mml-miR-128a-5p_R − 2, miR-431_R − 1, and let-7g-3p_1ss22CT were significantly downregulated. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that these differentially expressed miRNAs were implicated in the regulation of ubiquitin-mediated proteolysis and signaling pathways in cancer, glioma, chronic myeloid leukemia, and chemokine signaling. miRNA clustering analysis showed that mml-let-7g-5p and let-7g-3p_1ss22CT, which share three target genes [RB1-inducible coiled-coil 1 (RB1CC1), G-protein subunit γ 5 (GNG5), and chemokine (C-X-C motif) receptor 4 (CXCR4)], belong to one cluster, were altered in opposite directions following ischemia. These data suggest that circulating mml-let-7g may serve as a therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Jian Chen
- Department of Neurosurgery, Institute of Cerebrovascular Diseases Research, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haiping Zhao
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Yuyou Huang
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Yuqian Li
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Junfen Fan
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Rongliang Wang
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Ziping Han
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Zhenhong Yang
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Longfei Wu
- Department of Neurosurgery, Institute of Cerebrovascular Diseases Research, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Di Wu
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Yumin Luo
- Beijing Institute for Brain Disorders, Beijing, China
| | - Xunming Ji
- Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
39
|
Kato H, Maezawa Y. Atherosclerosis and Cardiovascular Diseases in Progeroid Syndromes. J Atheroscler Thromb 2021; 29:439-447. [PMID: 34511576 PMCID: PMC9100459 DOI: 10.5551/jat.rv17061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) and Werner syndrome (WS) are two of the representative genetic progeroid syndromes and have been widely studied in the field of aging research. HGPS is a pediatric disease in which premature aging symptoms appear in early childhood, and death occurs at an average age of 14.5 years, mainly due to cardiovascular disease (CVD). Conversely, WS patients exhibit accelerated aging phenotypes after puberty and die in their 50s due to CVD and malignant tumors. Both diseases are models of human aging, leading to a better understanding of the aging-associated development of CVD. In this review, we discuss the pathogenesis and treatment of atherosclerotic diseases presented by both progeroid syndromes with the latest findings.
Collapse
Affiliation(s)
- Hisaya Kato
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine.,Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine.,Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital
| |
Collapse
|
40
|
井原 健. [Hutchinson-Gilford progeria syndrome]. Nihon Ronen Igakkai Zasshi 2021; 58:409-412. [PMID: 34483167 DOI: 10.3143/geriatrics.58.409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
41
|
Villanueva MT. Antisense approach slows progeria. Nat Rev Drug Discov 2021; 20:343. [PMID: 33772219 DOI: 10.1038/d41573-021-00056-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
42
|
Dang Y, van Heusden C, Nickerson V, Chung F, Wang Y, Quinney NL, Gentzsch M, Randell SH, Moulton HM, Kole R, Ni A, Juliano RL, Kreda SM. Enhanced delivery of peptide-morpholino oligonucleotides with a small molecule to correct splicing defects in the lung. Nucleic Acids Res 2021; 49:6100-6113. [PMID: 34107015 PMCID: PMC8216463 DOI: 10.1093/nar/gkab488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/13/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary diseases offer many targets for oligonucleotide therapeutics. However, effective delivery of oligonucleotides to the lung is challenging. For example, splicing mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) affect a significant cohort of Cystic Fibrosis (CF) patients. These individuals could potentially benefit from treatment with splice switching oligonucleotides (SSOs) that can modulate splicing of CFTR and restore its activity. However, previous studies in cell culture used oligonucleotide transfection methods that cannot be safely translated in vivo. In this report, we demonstrate effective correction of a splicing mutation in the lung of a mouse model using SSOs. Moreover, we also demonstrate effective correction of a CFTR splicing mutation in a pre-clinical CF patient-derived cell model. We utilized a highly effective delivery strategy for oligonucleotides by combining peptide-morpholino (PPMO) SSOs with small molecules termed OECs. PPMOs distribute broadly into the lung and other tissues while OECs potentiate the effects of oligonucleotides by releasing them from endosomal entrapment. The combined PPMO plus OEC approach proved to be effective both in CF patient cells and in vivo in the mouse lung and thus may offer a path to the development of novel therapeutics for splicing mutations in CF and other lung diseases.
Collapse
Affiliation(s)
- Yan Dang
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Catharina van Heusden
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Veronica Nickerson
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Felicity Chung
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Yang Wang
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Nancy L Quinney
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Martina Gentzsch
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Scott H Randell
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Hong M Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Ryszard Kole
- Department of Pharmacology, The University of North Carolina at Chapel Hill, 4010 Genetic Medicine Bldg, Chapel Hill, NC 27599, USA
| | - Aiguo Ni
- Initos Pharmaceuticals, LLC, Chapel Hill, NC 27514, USA
| | | | - Silvia M Kreda
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| |
Collapse
|
43
|
Macicior J, Marcos-Ramiro B, Ortega-Gutiérrez S. Small-Molecule Therapeutic Perspectives for the Treatment of Progeria. Int J Mol Sci 2021; 22:7190. [PMID: 34281245 PMCID: PMC8267806 DOI: 10.3390/ijms22137190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS), or progeria, is an extremely rare disorder that belongs to the class of laminopathies, diseases characterized by alterations in the genes that encode for the lamin proteins or for their associated interacting proteins. In particular, progeria is caused by a point mutation in the gene that codifies for the lamin A gene. This mutation ultimately leads to the biosynthesis of a mutated version of lamin A called progerin, which accumulates abnormally in the nuclear lamina. This accumulation elicits several alterations at the nuclear, cellular, and tissue levels that are phenotypically reflected in a systemic disorder with important alterations, mainly in the cardiovascular system, bones, skin, and overall growth, which results in premature death at an average age of 14.5 years. In 2020, lonafarnib became the first (and only) FDA approved drug for treating progeria. In this context, the present review focuses on the different therapeutic strategies currently under development, with special attention to the new small molecules described in recent years, which may represent the upcoming first-in-class drugs with new mechanisms of action endowed with effectiveness not only to treat but also to cure progeria.
Collapse
Affiliation(s)
| | | | - Silvia Ortega-Gutiérrez
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain; (J.M.); (B.M.-R.)
| |
Collapse
|
44
|
Musunuru K. Adenine base editing to treat progeria syndrome and extend the lifespan. THE JOURNAL OF CARDIOVASCULAR AGING 2021; 1. [PMID: 34308436 PMCID: PMC8302045 DOI: 10.20517/jca.2021.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an exceedingly rare and hitherto incurable and fatal disease marked by accelerated aging simultaneously affecting a number of organs. Most cases of HGPS are caused by a single copy of a specific single-nucleotide mutation, c.C1824T, in the LMNA (lamin A) gene. Different mutations in LMNA are responsible for a variety of disorders affecting a variety of organs, including dilated cardiomyopathy, familial partial lipodystrophy, Emery-Dreifuss muscular dystrophy, limb girdle muscular dystrophy, Charcot-Marie-Tooth disease, and restrictive dermopathy. The unique pathophysiology of HGPS arises from the distinctive nature of the c.C1824T mutation; despite being a synonymous mutation that does not directly change an amino acid in the lamin A protein, it nonetheless exerts a profound effect on the protein by creating a cryptic splice site that causes incorrect splicing of the LMNA mRNA transcript, resulting in production of a truncated form of lamin A termed progerin, which is constitutively farnesylated. The farnesylated protein inappropriately accumulates in cells and causes dysregulation of the nuclear lamina - a structure in which the normal lamin A protein is a key component - that results in cellular dysfunction, senescence, and death. Vascular smooth muscle cells (VSMCs) represent one of the cell types particularly affected by progerin, and cardiovascular complications are the typical cause of death of HGPS patients in their youth.
Collapse
Affiliation(s)
- Kiran Musunuru
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
45
|
Dieterle MP, Husari A, Steinberg T, Wang X, Ramminger I, Tomakidi P. From the Matrix to the Nucleus and Back: Mechanobiology in the Light of Health, Pathologies, and Regeneration of Oral Periodontal Tissues. Biomolecules 2021; 11:824. [PMID: 34073044 PMCID: PMC8228498 DOI: 10.3390/biom11060824] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
Among oral tissues, the periodontium is permanently subjected to mechanical forces resulting from chewing, mastication, or orthodontic appliances. Molecularly, these movements induce a series of subsequent signaling processes, which are embedded in the biological concept of cellular mechanotransduction (MT). Cell and tissue structures, ranging from the extracellular matrix (ECM) to the plasma membrane, the cytosol and the nucleus, are involved in MT. Dysregulation of the diverse, fine-tuned interaction of molecular players responsible for transmitting biophysical environmental information into the cell's inner milieu can lead to and promote serious diseases, such as periodontitis or oral squamous cell carcinoma (OSCC). Therefore, periodontal integrity and regeneration is highly dependent on the proper integration and regulation of mechanobiological signals in the context of cell behavior. Recent experimental findings have increased the understanding of classical cellular mechanosensing mechanisms by both integrating exogenic factors such as bacterial gingipain proteases and newly discovered cell-inherent functions of mechanoresponsive co-transcriptional regulators such as the Yes-associated protein 1 (YAP1) or the nuclear cytoskeleton. Regarding periodontal MT research, this review offers insights into the current trends and open aspects. Concerning oral regenerative medicine or weakening of periodontal tissue diseases, perspectives on future applications of mechanobiological principles are discussed.
Collapse
Affiliation(s)
- Martin Philipp Dieterle
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (M.P.D.); (X.W.); (I.R.); (P.T.)
| | - Ayman Husari
- Center for Dental Medicine, Department of Orthodontics, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany;
- Faculty of Engineering, University of Freiburg, Georges-Köhler-Allee 101, 79110 Freiburg, Germany
| | - Thorsten Steinberg
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (M.P.D.); (X.W.); (I.R.); (P.T.)
| | - Xiaoling Wang
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (M.P.D.); (X.W.); (I.R.); (P.T.)
| | - Imke Ramminger
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (M.P.D.); (X.W.); (I.R.); (P.T.)
| | - Pascal Tomakidi
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (M.P.D.); (X.W.); (I.R.); (P.T.)
| |
Collapse
|
46
|
Molecular and Cellular Mechanisms Driving Cardiovascular Disease in Hutchinson-Gilford Progeria Syndrome: Lessons Learned from Animal Models. Cells 2021; 10:cells10051157. [PMID: 34064612 PMCID: PMC8151355 DOI: 10.3390/cells10051157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/29/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disease that recapitulates many symptoms of physiological aging and precipitates death. Patients develop severe vascular alterations, mainly massive vascular smooth muscle cell loss, vessel stiffening, calcification, fibrosis, and generalized atherosclerosis, as well as electrical, structural, and functional anomalies in the heart. As a result, most HGPS patients die of myocardial infarction, heart failure, or stroke typically during the first or second decade of life. No cure exists for HGPS, and therefore it is of the utmost importance to define the mechanisms that control disease progression in order to develop new treatments to improve the life quality of patients and extend their lifespan. Since the discovery of the HGPS-causing mutation, several animal models have been generated to study multiple aspects of the syndrome and to analyze the contribution of different cell types to the acquisition of the HGPS-associated cardiovascular phenotype. This review discusses current knowledge about cardiovascular features in HGPS patients and animal models and the molecular and cellular mechanisms through which progerin causes cardiovascular disease.
Collapse
|
47
|
Revêchon G, Whisenant D, Eriksson M. Splice-inhibition therapy targets progeria. Nat Med 2021; 27:377-379. [PMID: 33707774 DOI: 10.1038/s41591-021-01267-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Gwladys Revêchon
- Karolinska Institutet, Department of Biosciences and Nutrition, Center for Innovative Medicine, Huddinge, Sweden
| | - Daniel Whisenant
- Karolinska Institutet, Department of Biosciences and Nutrition, Center for Innovative Medicine, Huddinge, Sweden
| | - Maria Eriksson
- Karolinska Institutet, Department of Biosciences and Nutrition, Center for Innovative Medicine, Huddinge, Sweden.
| |
Collapse
|
48
|
Gordon LB, Tuminelli K, Andrés V, Campisi J, Kieran MW, Doucette L, Gordon AS. The progeria research foundation 10 th international scientific workshop; researching possibilities, ExTENding lives - webinar version scientific summary. Aging (Albany NY) 2021; 13:9143-9151. [PMID: 33735109 PMCID: PMC8034973 DOI: 10.18632/aging.202835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/19/2021] [Indexed: 02/06/2023]
Abstract
Progeria is an ultra-rare (prevalence 1 in 20 million), fatal, pediatric autosomal dominant premature aging disease caused by a mutation in the LMNA gene. This mutation results in accumulation of a high level of an aberrant form of the nuclear membrane protein, Lamin A. This aberrant protein, termed progerin, accumulates in many tissues and is responsible for the diverse array of disease phenotypes. Children die predominantly from premature atherosclerotic cardiovascular disease. The Progeria Research Foundation’s 10th International Scientific Workshop took place via webinar on November 2 and 3, 2020. Participants from 30 countries joined in this new, virtual meeting format. Patient family presentations led the program, followed by updates on Progeria’s first-ever application for FDA drug approval as well as initial results from the only current Progeria clinical trial. This was followed by presentations of unpublished preclinical data on drugs in development targeting the disease-causing DNA mutation, the aberrant mRNA, progerin protein, and its downstream effector proteins. Tying bench to bedside, clinicians presented new discoveries on the natural history of disease to inform future clinical trial development and new Progeria aortic valve replacement procedures. The program engaged the Progeria research community as a single unit with a common goal – to treat and cure children with Progeria worldwide.
Collapse
Affiliation(s)
- Leslie B Gordon
- Department of Pediatrics, Division of Genetics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI 02903, USA.,Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.,The Progeria Research Foundation, Peabody, MA 01961, USA
| | | | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid 28029, Spain
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA.,Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | - Lynn Doucette
- The Progeria Research Foundation, Peabody, MA 01961, USA
| | | |
Collapse
|
49
|
Puttaraju M, Jackson M, Klein S, Shilo A, Bennett CF, Gordon L, Rigo F, Misteli T. Systematic screening identifies therapeutic antisense oligonucleotides for Hutchinson-Gilford progeria syndrome. Nat Med 2021; 27:526-535. [PMID: 33707772 PMCID: PMC10167920 DOI: 10.1038/s41591-021-01262-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/25/2021] [Indexed: 11/09/2022]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare, invariably fatal childhood premature aging disorder caused by a pre-messenger RNA (mRNA) splicing defect in the LMNA gene. We used combined in vitro screening and in vivo validation to systematically explore the effects of target sequence, backbone chemistry and mechanism of action to identify optimized antisense oligonucleotides (ASOs) for therapeutic use in HGPS. In a library of 198 ASOs, the most potent ASOs targeted the LMNA exon 12 junction and acted via non-RNase H-mediated mechanisms. Treatment with an optimized lead candidate resulted in extension of lifespan in a mouse model of HGPS. Progerin mRNA levels were robustly reduced in vivo, but the extent of progerin protein reduction differed between tissues, suggesting a long half-life and tissue-specific turnover of progerin in vivo. These results identify a novel therapeutic agent for HGPS and provide insight into the HGPS disease mechanism.
Collapse
Affiliation(s)
- Madaiah Puttaraju
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Asaf Shilo
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Leslie Gordon
- Division of Genetics, Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | - Tom Misteli
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|