1
|
Muela‐Zarzuela I, Suarez‐Rivero JM, Boy‐Ruiz D, López‐Pérez J, Sotelo‐Montoro M, del Mar Navarrete‐Alonso M, Collado IG, Botubol‐Ares JM, Sanz A, Cordero MD. The NLRP3 inhibitor Dapansutrile improves the therapeutic action of lonafarnib on progeroid mice. Aging Cell 2024; 23:e14272. [PMID: 39192596 PMCID: PMC11488313 DOI: 10.1111/acel.14272] [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: 04/10/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 08/29/2024] Open
Abstract
The role of the inflammasomes in aging and progeroid syndromes remain understudied. Recently, MCC950, a NLRP3 inhibitor, was used in Zmpste24-/- mice to ameliorate the phenotypes. However, the safety of MCC950 was questioned due to liver toxicity observed in humans. Nevertheless, inhibition of the inflammasomes would be a beneficial therapy for progeria. Here, we show that OLT1177 (dapansutrile), other NLRP3 inhibitor, improved cellular and animal phenotypes using progeroid fibroblasts and a LmnaG609G/G609G mouse model. In both cases dapansutrile reduced progerin accumulation, NLRP3-inflammasome activation and secretory phenotype of senescence, extended the lifespan of progeroid animals, preserved bodyweight, and reduced kyphosis, inflammation, and senescence. Interestingly, dapansutrile further improved the effect of lonafarnib, the only FDA-approved drug for the progeria. The combination of both drugs reduced the inflammation and senescence, extended survival and ameliorated various progeroid defects both in vitro and in vivo, compared with treatment using lonafarnib alone. These findings and the safety of dapansutrile demonstrated in several clinical trials proposes it as a possible co-adjuvant treatment with lonafarnid in HGPS.
Collapse
Affiliation(s)
- Inés Muela‐Zarzuela
- Department of Molecular Biology and Biochemical EngineeringUniversidad Pablo de OlavideSevilleSpain
| | | | - Daniel Boy‐Ruiz
- Department of Molecular Biology and Biochemical EngineeringUniversidad Pablo de OlavideSevilleSpain
| | - Juan López‐Pérez
- Department of ImmunologyPuerta del Mar HospitalCádizSpain
- Instituto de Investigación e Innovación Biomédica de Cádiz, INiBICAHospital Universitario Puerta del MarCádizSpain
| | - Marta Sotelo‐Montoro
- Department of Molecular Biology and Biochemical EngineeringUniversidad Pablo de OlavideSevilleSpain
| | | | - Isidro G. Collado
- Departamento de Química Orgánica, Facultad de Ciencias, Campus Universitario Río San Pedro s/n, Torre Sur, 4a PlantaUniversity of CádizCádizSpain
| | - José Manuel Botubol‐Ares
- Departamento de Química Orgánica, Facultad de Ciencias, Campus Universitario Río San Pedro s/n, Torre Sur, 4a PlantaUniversity of CádizCádizSpain
| | - Alberto Sanz
- School of Molecular Biosciences, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Mario D. Cordero
- Department of Molecular Biology and Biochemical EngineeringUniversidad Pablo de OlavideSevilleSpain
| |
Collapse
|
2
|
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
|
3
|
Wang M, Zhang J, Qiu J, Ma X, Xu C, Wu Q, Xing S, Chen X, Liu B. Doxycycline decelerates aging in progeria mice. Aging Cell 2024; 23:e14188. [PMID: 38686927 PMCID: PMC11258430 DOI: 10.1111/acel.14188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/26/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
Beyond the antimicrobial activity, doxycycline (DOX) exhibits longevity-promoting effect in nematodes, while its effect on mammals is unclear. Here, we applied a mouse model of Hutchinson-Gilford progeria syndrome (HGPS), Zmpste24 knockout (KO) mice, and analyzed the antiaging effect of DOX. We found that the DOX treatment prolongs lifespan and ameliorates progeroid features of Zmpste24 KO mice, including the decline of body and tissue weight, exercise capacity and cortical bone density, and the shortened colon length. DOX treatment alleviates the abnormal nuclear envelope in multiple tissues, and attenuates cellular senescence and cell death of Zmpste24 KO and HGPS fibroblasts. DOX downregulates the level of proinflammatory IL6 in both serum and tissues. Moreover, the elevated α-tubulin (K40) acetylation mediated by NAT10 in progeria, is rescued by DOX treatment in the aorta tissues in Zmpste24 KO mice and fibroblasts. Collectively, our study uncovers that DOX can decelerate aging in progeria mice via counteracting IL6 expression and NAT10-mediated acetylation of α-tubulin.
Collapse
Affiliation(s)
- Ming Wang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Jie Zhang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Jiangping Qiu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Xuan Ma
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Chenzhong Xu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Qiuhuan Wu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Shaojun Xing
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| |
Collapse
|
4
|
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
|
5
|
Quintana‐Torres D, Valle‐Cao A, Bousquets‐Muñoz P, Freitas‐Rodríguez S, Rodríguez F, Lucia A, López‐Otín C, López‐Soto A, Folgueras AR. The secretome atlas of two mouse models of progeria. Aging Cell 2023; 22:e13952. [PMID: 37565451 PMCID: PMC10577534 DOI: 10.1111/acel.13952] [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: 06/06/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disease caused by nuclear envelope alterations that lead to accelerated aging and premature death. Several studies have linked health and longevity to cell-extrinsic mechanisms, highlighting the relevance of circulating factors in the aging process as well as in age-related diseases. We performed a global plasma proteomic analysis in two preclinical progeroid models (LmnaG609G/G609G and Zmpste24-/- mice) using aptamer-based proteomic technology. Pathways related to the extracellular matrix, growth factor response and calcium ion binding were among the most enriched in the proteomic signature of progeroid samples compared to controls. Despite the global downregulation trend found in the plasma proteome of progeroid mice, several proteins associated with cardiovascular disease, the main cause of death in HGPS, were upregulated. We also developed a chronological age predictor using plasma proteome data from a cohort of healthy mice (aged 1-30 months), that reported an age acceleration when applied to progeroid mice, indicating that these mice exhibit an "old" plasma proteomic signature. Furthermore, when compared to naturally-aged mice, a great proportion of differentially expressed circulating proteins in progeroid mice were specific to premature aging, highlighting secretome-associated differences between physiological and accelerated aging. This is the first large-scale profiling of the plasma proteome in progeroid mice, which provides an extensive list of candidate circulating plasma proteins as potential biomarkers and/or therapeutic targets for further exploration and hypothesis generation in the context of both physiological and premature aging.
Collapse
Affiliation(s)
- Diego Quintana‐Torres
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| | - Alejandra Valle‐Cao
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| | - Pablo Bousquets‐Muñoz
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
| | - Sandra Freitas‐Rodríguez
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
| | - Francisco Rodríguez
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
| | - Alejandro Lucia
- CIBER of Frailty and Healthy Aging (CIBERFES) and Instituto de Investigación 12 de Octubre (i+12)MadridSpain
- Faculty of Sport SciencesUniversidad EuropeaMadridSpain
| | - Carlos López‐Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
| | - Alejandro López‐Soto
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| | - Alicia R. Folgueras
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| |
Collapse
|
6
|
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
|
7
|
Abutaleb NO, Atchison L, Choi L, Bedapudi A, Shores K, Gete Y, Cao K, Truskey GA. Lonafarnib and everolimus reduce pathology in iPSC-derived tissue engineered blood vessel model of Hutchinson-Gilford Progeria Syndrome. Sci Rep 2023; 13:5032. [PMID: 36977745 PMCID: PMC10050176 DOI: 10.1038/s41598-023-32035-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare, fatal genetic disease that accelerates atherosclerosis. With a limited pool of HGPS patients, clinical trials face unique challenges and require reliable preclinical testing. We previously reported a 3D tissue engineered blood vessel (TEBV) microphysiological system fabricated with iPSC-derived vascular cells from HGPS patients. HGPS TEBVs exhibit features of HGPS atherosclerosis including loss of smooth muscle cells, reduced vasoactivity, excess extracellular matrix (ECM) deposition, inflammatory marker expression, and calcification. We tested the effects of HGPS therapeutics Lonafarnib and Everolimus separately and together, currently in Phase I/II clinical trial, on HGPS TEBVs. Everolimus decreased reactive oxygen species levels, increased proliferation, reduced DNA damage in HGPS vascular cells, and improved vasoconstriction in HGPS TEBVs. Lonafarnib improved shear stress response of HGPS iPSC-derived endothelial cells (viECs) and reduced ECM deposition, inflammation, and calcification in HGPS TEBVs. Combination treatment with Lonafarnib and Everolimus produced additional benefits such as improved endothelial and smooth muscle marker expression and reduced apoptosis, as well as increased TEBV vasoconstriction and vasodilation. These results suggest that a combined trial of both drugs may provide cardiovascular benefits beyond Lonafarnib, if the Everolimus dose can be tolerated.
Collapse
Affiliation(s)
- Nadia O Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Leigh Atchison
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Leandro Choi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Akhil Bedapudi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kevin Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yantenew Gete
- 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
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
8
|
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: An expanding universe. Cell 2023; 186:243-278. [PMID: 36599349 DOI: 10.1016/j.cell.2022.11.001] [Citation(s) in RCA: 1397] [Impact Index Per Article: 1397.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/19/2022] [Accepted: 11/01/2022] [Indexed: 01/05/2023]
Abstract
Aging is driven by hallmarks fulfilling the following three premises: (1) their age-associated manifestation, (2) the acceleration of aging by experimentally accentuating them, and (3) the opportunity to decelerate, stop, or reverse aging by therapeutic interventions on them. We propose the following twelve hallmarks of aging: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, disabled macroautophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis. These hallmarks are interconnected among each other, as well as to the recently proposed hallmarks of health, which include organizational features of spatial compartmentalization, maintenance of homeostasis, and adequate responses to stress.
Collapse
Affiliation(s)
- Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Linda Partridge
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK; Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain; Altos Labs, Cambridge, UK
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
9
|
Cercillieux A, Ciarlo E, Canto C. Balancing NAD + deficits with nicotinamide riboside: therapeutic possibilities and limitations. Cell Mol Life Sci 2022; 79:463. [PMID: 35918544 PMCID: PMC9345839 DOI: 10.1007/s00018-022-04499-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/20/2022] [Accepted: 07/20/2022] [Indexed: 12/21/2022]
Abstract
Alterations in cellular nicotinamide adenine dinucleotide (NAD+) levels have been observed in multiple lifestyle and age-related medical conditions. This has led to the hypothesis that dietary supplementation with NAD+ precursors, or vitamin B3s, could exert health benefits. Among the different molecules that can act as NAD+ precursors, Nicotinamide Riboside (NR) has gained most attention due to its success in alleviating and treating disease conditions at the pre-clinical level. However, the clinical outcomes for NR supplementation strategies have not yet met the expectations generated in mouse models. In this review we aim to provide a comprehensive view on NAD+ biology, what causes NAD+ deficits and the journey of NR from its discovery to its clinical development. We also discuss what are the current limitations in NR-based therapies and potential ways to overcome them. Overall, this review will not only provide tools to understand NAD+ biology and assess its changes in disease situations, but also to decide which NAD+ precursor could have the best therapeutic potential.
Collapse
Affiliation(s)
- Angelique Cercillieux
- Nestlé Institute of Health Sciences, Nestlé Research Ltd., EPFL Campus, Innovation Park, Building G, 1015, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Eleonora Ciarlo
- Nestlé Institute of Health Sciences, Nestlé Research Ltd., EPFL Campus, Innovation Park, Building G, 1015, Lausanne, Switzerland
| | - Carles Canto
- Nestlé Institute of Health Sciences, Nestlé Research Ltd., EPFL Campus, Innovation Park, Building G, 1015, Lausanne, Switzerland.
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland.
| |
Collapse
|
10
|
Zhang SL, Lin SZ, Zhou YQ, Wang WQ, Li JY, Wang C, Pang QM. Clinical manifestations and gene analysis of Hutchinson-Gilford progeria syndrome: A case report. World J Clin Cases 2022; 10:5018-5024. [PMID: 35801028 PMCID: PMC9198858 DOI: 10.12998/wjcc.v10.i15.5018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/10/2022] [Accepted: 03/26/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This case report describes a child with Hutchinson-Gilford progeria syndrome (HGPS, OMIM: 176670) caused by LMNA (OMIM: 150330) gene mutation, and we have previously analyzed the clinical manifestations and imaging characteristics of this case. After 1-year treatment and follow-up, we focus on analyzing the changes in the clinical manifestations and genetic diagnosis of the patient.
CASE SUMMARY In April 2020, a 2-year-old boy with HGPS was found to have an abnormal appearance, and growth and development lagged behind those of children of the same age. The child’s weight did not increase normally, the veins of the head were clearly visible, and he had shallow skin color and sparse yellow hair. Peripheral blood DNA samples obtained from the patient and his parents were sequenced using high-throughput whole-exosome sequencing, which was verified by Sanger sequencing. The results showed that there was a synonymous heterozygous mutation of C.1824 C>T (P. G608G) in the LMNA gene.
CONCLUSION Mutation of the LMNA gene provides a molecular basis for diagnosis of HGPS and genetic counseling of the family.
Collapse
Affiliation(s)
- Su-Li Zhang
- Department of Neuroscience, Hainan Women and Children's Medical Center, Haikou 570100, Hainan Province, China
| | - Shuang-Zhu Lin
- Diagnosis and Treatment Center for Children, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Yan-Qiu Zhou
- Diagnosis and Treatment Center for Children, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Wan-Qi Wang
- Changchun University of Chinese Medicine, Changchun 130000, Jilin Province, China
| | - Jia-Yi Li
- Changchun University of Chinese Medicine, Changchun 130000, Jilin Province, China
| | - Cui Wang
- Department of Neuroscience, Hainan Women and Children's Medical Center, Haikou 570100, Hainan Province, China
| | - Qi-Ming Pang
- Department of Neuroscience, Hainan Women and Children's Medical Center, Haikou 570100, Hainan Province, China
| |
Collapse
|
11
|
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]
|
12
|
Jiang Y, Ji JY. Progerin-Induced Impairment in Wound Healing and Proliferation in Vascular Endothelial Cells. FRONTIERS IN AGING 2022; 3:844885. [PMID: 35821855 PMCID: PMC9261432 DOI: 10.3389/fragi.2022.844885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/27/2022] [Indexed: 11/29/2022]
Abstract
Progerin as a mutated isoform of lamin A protein was first known to induce premature atherosclerosis progression in patients with Hutchinson-Gilford progeria syndrome (HGPS), and its role in provoking an inflammatory response in vascular cells and accelerating cell senescence has been investigated recently. However, how progerin triggers endothelial dysfunction that often occurs at the early stage of atherosclerosis in a mechanical environment has not been studied intensively. Here, we generated a stable endothelial cell line that expressed progerin and examined its effects on endothelial wound repair under laminar flow. We found decreased wound healing rate in progerin-expressing ECs under higher shear stress compared with those under low shear. Furthermore, the decreased wound recovery could be due to reduced number of cells at late mitosis, suggesting potential interference by progerin with endothelial proliferation. These findings provided insights into how progerin affects endothelial mechanotransduction and may contribute to the disruption of endothelial integrity in HGPS vasculature, as we continue to examine the mechanistic effect of progerin in shear-induced endothelial functions.
Collapse
|
13
|
Abdelnour SA, Xie L, Hassanin AA, Zuo E, Lu Y. The Potential of CRISPR/Cas9 Gene Editing as a Treatment Strategy for Inherited Diseases. Front Cell Dev Biol 2022; 9:699597. [PMID: 34977000 PMCID: PMC8715006 DOI: 10.3389/fcell.2021.699597] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) is a promising innovative technology for genomic editing that offers scientists the chance to edit DNA structures and change gene function. It has several possible uses consisting of editing inherited deficiencies, treating, and reducing the spread of disorders. Recently, reports have demonstrated the creation of synthetic RNA molecules and supplying them alongside Cas9 into genome of eukaryotes, since distinct specific regions of the genome can be manipulated and targeted. The therapeutic potential of CRISPR/Cas9 technology is great, especially in gene therapy, in which a patient-specific mutation is genetically edited, or in the treating of human disorders that are untreatable with traditional treatments. This review focused on numerous, in vivo, in vitro, and ex vivo uses of the CRISPR/Cas9 technology in human inherited diseases, discovering the capability of this versatile in medicine and examining some of the main limitations for its upcoming use in patients. In addition to introducing a brief impression of the biology of the CRISPR/Cas9 scheme and its mechanisms, we presented the utmost recent progress in the uses of CRISPR/Cas9 technology in editing and treating of human genetic diseases.
Collapse
Affiliation(s)
- Sameh A Abdelnour
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China.,Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Long Xie
- Center for Animal Genomics, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Abdallah A Hassanin
- Genetics Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Erwei Zuo
- Center for Animal Genomics, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| |
Collapse
|
14
|
González‐Dominguez A, Montañez R, Castejón‐Vega B, Nuñez‐Vasco J, Lendines‐Cordero D, Wang C, Mbalaviele G, Navarro‐Pando JM, Alcocer‐Gómez E, Cordero MD. Inhibition of the NLRP3 inflammasome improves lifespan in animal murine model of Hutchinson-Gilford Progeria. EMBO Mol Med 2021; 13:e14012. [PMID: 34448355 PMCID: PMC8495449 DOI: 10.15252/emmm.202114012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 01/09/2023] Open
Abstract
Inflammation is a hallmark of aging and accelerated aging syndromes such as Hutchinson-Gilford progeria syndrome (HGPS). In this study, we present evidence of increased expression of the components of the NLRP3 inflammasome pathway in HGPS skin fibroblasts, an outcome that was associated with morphological changes of the nuclei of the cells. Lymphoblasts from HGPS patients also showed increased basal levels of NLRP3 and caspase 1. Consistent with these results, the expression of caspase 1 and Nlrp3, but not of the other inflammasome receptors was higher in the heart and liver of Zmpste24-/- mice, which phenocopy the human disease. These data were further corroborated in LmnaG609G/G609G mice, another HGPS animal model. We also showed that pharmacological inhibition of the NLRP3 inflammasome by its selective inhibitor, MCC950, improved cellular phenotype, significantly extended the lifespan of progeroid animals, and reduced inflammasome-dependent inflammation. These findings suggest that inhibition of the NLRP3 inflammasome is a potential therapeutic approach for the treatment of HGPS.
Collapse
Affiliation(s)
- Alvaro González‐Dominguez
- Instituto de Investigación e Innovación Biomédica de CádizINiBICAHospital Universitario Puerta del MarCádizSpain
| | - Raúl Montañez
- Instituto de Investigación e Innovación Biomédica de CádizINiBICAHospital Universitario Puerta del MarCádizSpain
| | | | - Jéssica Nuñez‐Vasco
- Instituto de Investigación e Innovación Biomédica de CádizINiBICAHospital Universitario Puerta del MarCádizSpain
| | - Débora Lendines‐Cordero
- Instituto de Investigación e Innovación Biomédica de CádizINiBICAHospital Universitario Puerta del MarCádizSpain
| | - Chun Wang
- Division of Bone and Mineral DiseasesWashington University School of MedicineSt. LouisMOUSA
| | - Gabriel Mbalaviele
- Division of Bone and Mineral DiseasesWashington University School of MedicineSt. LouisMOUSA
| | - José M Navarro‐Pando
- Cátedra de Reproducción y Genética Humana del Instituto para el Estudio de la Biología de la Reproducción Humana (INEBIR)Universidad Europea del Atlántico (UNEATLANTICO)‐Fundación Universitaria Iberoamericana (FUNIBER)SevilleSpain
| | - Elísabet Alcocer‐Gómez
- Departamento de Psicología ExperimentalFacultad de PsicologíaUniversidad de SevillaSevillaSpain
| | - Mario D Cordero
- Instituto de Investigación e Innovación Biomédica de CádizINiBICAHospital Universitario Puerta del MarCádizSpain
| |
Collapse
|
15
|
Rajeev M, Ratan C, Krishnan K, Vijayan M. Hutchinson-Gilford Progeria Syndrome (Hgps) And Application Of Gene Therapy Based Crispr/Cas Technology As A Promising Innovative Treatment Approach. Recent Pat Biotechnol 2021; 15:266-285. [PMID: 34602042 DOI: 10.2174/1872208315666210928114720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/09/2021] [Accepted: 08/23/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Hutchinson-Gilford progeria syndrome (HGPS) also known as progeria of childhood or progeria is a rare, rapid, autosomal dominant genetic disorder characterized by premature aging which occurs shortly after birth. HGPS occurs as a result of de novo point mutation in the gene recognized as LMNA gene that encodes two proteins Lamin A protein and Lamin C protein which are the structural components of the nuclear envelope. Mutations in the gene trigger abnormal splicing and induce internal deletion of 50 amino acids leading to the development of a truncated form of Lamin A protein known as Progerin. Progerin generation can be considered as the crucial step in HGPS since the protein is highly toxic to human cells, permanently farnesylated, and exhibits variation in several biochemical and structural properties within the individual. HGPS also produces complications such as skin alterations, growth failure, atherosclerosis, hair and fat loss, and bone and joint diseases. We have also revised all relevant patents relating to Hutchinson-gilford progeria syndrome and its therapy in the current article. METHOD The goal of the present review article is to provide information about Hutchinson-Gilford progeria syndrome (HGPS) and the use of CRISPR/Cas technology as a promising treatment approach in the treatment of the disease. The review also discusses about different pharmacological and non-pharmacological methods of treatment currently used for HGPS. RESULTS The main limitation associated with progeria is the lack of a definitive cure. The existing treatment modality provides only symptomatic relief. Therefore, it is high time to develop a therapeutic method that hastens premature aging in such patients. CONCLUSION CRISPR/Cas technology is a novel gene-editing tool that allows genome editing at specific loci, and is found to be a promising therapeutic approach for the treatment of genetic disorders such as HGPS where dominant-negative mutations take place.
Collapse
Affiliation(s)
- Mekha Rajeev
- Amrita School of Pharmacy, Amrita Vihwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041. India
| | - Chameli Ratan
- Amrita School of Pharmacy, Amrita Vihwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041. India
| | - Karthik Krishnan
- Amrita School of Pharmacy, Amrita Vihwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041. India
| | - Meenu Vijayan
- Amrita School of Pharmacy, Amrita Vihwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041. India
| |
Collapse
|
16
|
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: 2.3] [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
|
17
|
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: 1.7] [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
|
18
|
Warnecke A, Giesemann A. Embryology, Malformations, and Rare Diseases of the Cochlea. Laryngorhinootologie 2021; 100:S1-S43. [PMID: 34352899 PMCID: PMC8354575 DOI: 10.1055/a-1349-3824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Despite the low overall prevalence of individual rare diseases, cochlear
dysfunction leading to hearing loss represents a symptom in a large
proportion. The aim of this work was to provide a clear overview of rare
cochlear diseases, taking into account the embryonic development of the
cochlea and the systematic presentation of the different disorders. Although
rapid biotechnological and bioinformatic advances may facilitate the
diagnosis of a rare disease, an interdisciplinary exchange is often required
to raise the suspicion of a rare disease. It is important to recognize that
the phenotype of rare inner ear diseases can vary greatly not only in
non-syndromic but also in syndromic hearing disorders. Finally, it becomes
clear that the phenotype of the individual rare diseases cannot be
determined exclusively by classical genetics even in monogenetic
disorders.
Collapse
Affiliation(s)
- Athanasia Warnecke
- Klinik für Hals-, Nasen- und Ohrenheilkunde, Medizinische Hochschule Hannover, Carl-Neuberg-Straße 1, 30625 Hannover.,Deutsche Forschungsgemeinschaft Exzellenzcluster"Hearing4all" - EXC 2177/1 - Project ID 390895286
| | - Anja Giesemann
- Institut für Neuroradiologie, Medizinische Hochschule Hannover, Carl-Neuberg-Straße 1, 30625 Hannover
| |
Collapse
|
19
|
Dreesen O, Kennedy B. Hutchinson-Gilford Progeria paves the way for novel targeted anti-aging therapies. MED 2021; 2:353-354. [DOI: 10.1016/j.medj.2021.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Koblan LW, Erdos MR, Wilson C, Cabral WA, Levy JM, Xiong ZM, Tavarez UL, Davison LM, Gete YG, Mao X, Newby GA, Doherty SP, Narisu N, Sheng Q, Krilow C, Lin CY, Gordon LB, Cao K, Collins FS, Brown JD, Liu DR. In vivo base editing rescues Hutchinson-Gilford progeria syndrome in mice. Nature 2021; 589:608-614. [PMID: 33408413 PMCID: PMC7872200 DOI: 10.1038/s41586-020-03086-7] [Citation(s) in RCA: 258] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS or progeria) is typically caused by a dominant-negative C•G-to-T•A mutation (c.1824 C>T; p.G608G) in LMNA, the gene that encodes nuclear lamin A. This mutation causes RNA mis-splicing that produces progerin, a toxic protein that induces rapid ageing and shortens the lifespan of children with progeria to approximately 14 years1-4. Adenine base editors (ABEs) convert targeted A•T base pairs to G•C base pairs with minimal by-products and without requiring double-strand DNA breaks or donor DNA templates5,6. Here we describe the use of an ABE to directly correct the pathogenic HGPS mutation in cultured fibroblasts derived from children with progeria and in a mouse model of HGPS. Lentiviral delivery of the ABE to fibroblasts from children with HGPS resulted in 87-91% correction of the pathogenic allele, mitigation of RNA mis-splicing, reduced levels of progerin and correction of nuclear abnormalities. Unbiased off-target DNA and RNA editing analysis did not detect off-target editing in treated patient-derived fibroblasts. In transgenic mice that are homozygous for the human LMNA c.1824 C>T allele, a single retro-orbital injection of adeno-associated virus 9 (AAV9) encoding the ABE resulted in substantial, durable correction of the pathogenic mutation (around 20-60% across various organs six months after injection), restoration of normal RNA splicing and reduction of progerin protein levels. In vivo base editing rescued the vascular pathology of the mice, preserving vascular smooth muscle cell counts and preventing adventitial fibrosis. A single injection of ABE-expressing AAV9 at postnatal day 14 improved vitality and greatly extended the median lifespan of the mice from 215 to 510 days. These findings demonstrate the potential of in vivo base editing as a possible treatment for HGPS and other genetic diseases by directly correcting their root cause.
Collapse
Affiliation(s)
- Luke W Koblan
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Michael R Erdos
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Wilson
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Wayne A Cabral
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan M Levy
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Zheng-Mei Xiong
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Urraca L Tavarez
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lindsay M Davison
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yantenew G Gete
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Xiaojing Mao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Sean P Doherty
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Narisu Narisu
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chad Krilow
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles Y Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Therapeutic Innovation Center, Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Kronos, Bio Inc., Cambridge, MA, USA
| | - Leslie B Gordon
- Hasbro Children's Hospital, Alpert Medical School of Brown University, Providence, RI, USA
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Francis S Collins
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Jonathan D Brown
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
21
|
Lai W, Wong W. Progress and trends in the development of therapies for Hutchinson-Gilford progeria syndrome. Aging Cell 2020; 19:e13175. [PMID: 32596971 PMCID: PMC7370734 DOI: 10.1111/acel.13175] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/28/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an autosomal-dominant genetic disease that leads to accelerated aging and often premature death caused by cardiovascular complications. Till now clinical management of HGPS has largely relied on the treatment of manifestations and on the prevention of secondary complications, cure for the disease has not yet been established. Addressing this need cannot only benefit progeria patients but may also provide insights into intervention design for combating physiological aging. By using the systematic review approach, this article revisits the overall progress in the development of strategies for HGPS treatment over the last ten years, from 2010 to 2019. In total, 1,906 articles have been retrieved, of which 56 studies have been included for further analysis. Based on the articles analyzed, the trends in the use of different HGPS models, along with the prevalence, efficiency, and limitations of different reported treatment strategies, have been examined. Emerging strategies for preclinical studies, and possible targets for intervention development, have also been presented as avenues for future research.
Collapse
Affiliation(s)
- Wing‐Fu Lai
- School of Life and Health Sciences The Chinese University of Hong Kong (Shenzhen) Shenzhen China
- Department of Applied Biology and Chemical Technology Hong Kong Polytechnic University Hong Kong Special Administrative Region China
| | - Wing‐Tak Wong
- Department of Applied Biology and Chemical Technology Hong Kong Polytechnic University Hong Kong Special Administrative Region China
| |
Collapse
|