1
|
Jaziri R, Zaghden O, Ghadhab N, Kammoun R, Chaabani I, Ben Alaya T. Radiological insights into mandibuloacral dysplasia: A case report. Radiol Case Rep 2024; 19:5553-5558. [PMID: 39296747 PMCID: PMC11409206 DOI: 10.1016/j.radcr.2024.08.005] [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: 06/02/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/21/2024] Open
Abstract
Mandibulo-acral dysplasia was first described by Young et al in 1971. It is a rare, autosomal genetic disorder describing characteristic general, craniofacial, and oral manifestations. However, the detailed characteristics of this multisystemic disease have not yet been clarified due to its rarity and the limited number of cases described. Through a clinical case, we have tried to expose some of the most common clinical and radiological signs especially in their bucco-facial location, some modalities of management and the importance of an early diagnosis which turns out to be a real challenge especially for dentists in their daily practice.
Collapse
Affiliation(s)
- Rawia Jaziri
- Department of medical imaging, University Dental Clinic, Monastir, Tunisia
- Research laboratory of Bioactive Natural Substances and Biotechnology, Faculty of Dental Medicine, University of Monastir, Monastir, Tunisia LR24ES14
| | - Olfa Zaghden
- Department of medical imaging, University Dental Clinic, Monastir, Tunisia
- Research laboratory of Bioactive Natural Substances and Biotechnology, Faculty of Dental Medicine, University of Monastir, Monastir, Tunisia LR24ES14
| | - Nawres Ghadhab
- Department of medical imaging, University Dental Clinic, Monastir, Tunisia
- Research laboratory of Bioactive Natural Substances and Biotechnology, Faculty of Dental Medicine, University of Monastir, Monastir, Tunisia LR24ES14
| | - Rym Kammoun
- Department of medical imaging, University Dental Clinic, Monastir, Tunisia
- Laboratory of Histology and Embryology, Faculty of Dental Medicine of Monastir, University of Monastir, Monastir, Tunisia
- ABCDF Laboratory for Biological Clinical and Dento-Facial Approach, University of Monastir, Tunisia
| | - Imen Chaabani
- Department of medical imaging, University Dental Clinic, Monastir, Tunisia
- Research laboratory of Bioactive Natural Substances and Biotechnology, Faculty of Dental Medicine, University of Monastir, Monastir, Tunisia LR24ES14
| | - Touhami Ben Alaya
- Department of medical imaging, University Dental Clinic, Monastir, Tunisia
- Research laboratory of Bioactive Natural Substances and Biotechnology, Faculty of Dental Medicine, University of Monastir, Monastir, Tunisia LR24ES14
| |
Collapse
|
2
|
Hartinger R, Singh K, Leverett J, Djabali K. Enhancing Cellular Homeostasis: Targeted Botanical Compounds Boost Cellular Health Functions in Normal and Premature Aging Fibroblasts. Biomolecules 2024; 14:1310. [PMID: 39456243 PMCID: PMC11506649 DOI: 10.3390/biom14101310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/06/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The human skin, the body's largest organ, undergoes continuous renewal but is significantly impacted by aging, which impairs its function and leads to visible changes. This study aimed to identify botanical compounds that mimic the anti-aging effects of baricitinib, a known JAK1/2 inhibitor. Through in silico screening of a botanical compound library, 14 potential candidates were identified, and 7 were further analyzed for their effects on cellular aging. The compounds were tested on both normal aged fibroblasts and premature aging fibroblasts derived from patients with Hutchinson-Gilford Progeria Syndrome (HGPS). Results showed that these botanical compounds effectively inhibited the JAK/STAT pathway, reduced the levels of phosphorylated STAT1 and STAT3, and ameliorated phenotypic changes associated with cellular aging. Treatments improved cell proliferation, reduced senescence markers, and enhanced autophagy without inducing cytotoxicity. Compounds, such as Resveratrol, Bisdemethoxycurcumin, Pinosylvin, Methyl P-Hydroxycinnamate, cis-Pterostilbene, and (+)-Gallocatechin, demonstrated significant improvements in both control and HGPS fibroblasts. These findings suggest that these botanical compounds have the potential to mitigate age-related cellular alterations, offering promising strategies for anti-aging therapies, particularly for skin health. Further in vivo studies are warranted to validate these results and explore their therapeutic applications.
Collapse
Affiliation(s)
- Ramona Hartinger
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany;
| | - Khushboo Singh
- Amway Corporation, Innovation and Science, 7575 Fulton Street East, Ada, MI 49355, USA
| | - Jesse Leverett
- Amway Corporation, Innovation and Science, 7575 Fulton Street East, Ada, MI 49355, USA
| | - Karima Djabali
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany;
| |
Collapse
|
3
|
Yu R, Xue H, Lin W, Collins F, Mount S, Cao K. Progerin mRNA expression in non-HGPS patients is correlated with widespread shifts in transcript isoforms. NAR Genom Bioinform 2024; 6:lqae115. [PMID: 39211333 PMCID: PMC11358823 DOI: 10.1093/nargab/lqae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disease caused primarily by a C1824T mutation in LMNA. This mutation activates a cryptic splice donor site, producing a lamin variant called progerin. Interestingly, progerin has also been detected in cells and tissues of non-HGPS patients. Here, we investigated progerin expression using publicly available RNA-seq data from non-HGPS patients in the GTEx project. We found that progerin expression is present across all tissue types in non-HGPS patients and correlated with telomere shortening in the skin. Transcriptome-wide correlation analyses suggest that the level of progerin expression is correlated with switches in gene isoform expression patterns. Differential expression analyses show that progerin expression is correlated with significant changes in genes involved in splicing regulation and mitochondrial function. Interestingly, 5' splice sites whose use is correlated with progerin expression have significantly altered frequencies of consensus trinucleotides within the core 5' splice site. Furthermore, introns whose alternative splicing correlates with progerin have reduced GC content. Our study suggests that progerin expression in non-HGPS patients is part of a global shift in splicing patterns.
Collapse
Affiliation(s)
- Reynold Yu
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Huijing Xue
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Wanru Lin
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Francis S Collins
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen M Mount
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| |
Collapse
|
4
|
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
|
5
|
Shores KL, Truskey GA. Mechanotransduction of the vasculature in Hutchinson-Gilford Progeria Syndrome. Front Physiol 2024; 15:1464678. [PMID: 39239311 PMCID: PMC11374724 DOI: 10.3389/fphys.2024.1464678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disorder that causes severe cardiovascular disease, resulting in the death of patients in their teenage years. The disease pathology is caused by the accumulation of progerin, a mutated form of the nuclear lamina protein, lamin A. Progerin binds to the inner nuclear membrane, disrupting nuclear integrity, and causes severe nuclear abnormalities and changes in gene expression. This results in increased cellular inflammation, senescence, and overall dysfunction. The molecular mechanisms by which progerin induces the disease pathology are not fully understood. Progerin's detrimental impact on nuclear mechanics and the role of the nucleus as a mechanosensor suggests dysfunctional mechanotransduction could play a role in HGPS. This is especially relevant in cells exposed to dynamic, continuous mechanical stimuli, like those of the vasculature. The endothelial (ECs) and smooth muscle cells (SMCs) within arteries rely on physical forces produced by blood flow to maintain function and homeostasis. Certain regions within arteries produce disturbed flow, leading to an impaired transduction of mechanical signals, and a reduction in cellular function, which also occurs in HGPS. In this review, we discuss the mechanics of nuclear mechanotransduction, how this is disrupted in HGPS, and what effect this has on cell health and function. We also address healthy responses of ECs and SMCs to physiological mechanical stimuli and how these responses are impaired by progerin accumulation.
Collapse
Affiliation(s)
- Kevin L Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
6
|
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
|
7
|
Ngubo M, Chen Z, McDonald D, Karimpour R, Shrestha A, Yockell‐Lelièvre J, Laurent A, Besong OTO, Tsai EC, Dilworth FJ, Hendzel MJ, Stanford WL. Progeria-based vascular model identifies networks associated with cardiovascular aging and disease. Aging Cell 2024; 23:e14150. [PMID: 38576084 PMCID: PMC11258467 DOI: 10.1111/acel.14150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Hutchinson-Gilford Progeria syndrome (HGPS) is a lethal premature aging disorder caused by a de novo heterozygous mutation that leads to the accumulation of a splicing isoform of Lamin A termed progerin. Progerin expression deregulates the organization of the nuclear lamina and the epigenetic landscape. Progerin has also been observed to accumulate at low levels during normal aging in cardiovascular cells of adults that do not carry genetic mutations linked with HGPS. Therefore, the molecular mechanisms that lead to vascular dysfunction in HGPS may also play a role in vascular aging-associated diseases, such as myocardial infarction and stroke. Here, we show that HGPS patient-derived vascular smooth muscle cells (VSMCs) recapitulate HGPS molecular hallmarks. Transcriptional profiling revealed cardiovascular disease remodeling and reactive oxidative stress response activation in HGPS VSMCs. Proteomic analyses identified abnormal acetylation programs in HGPS VSMC replication fork complexes, resulting in reduced H4K16 acetylation. Analysis of acetylation kinetics revealed both upregulation of K16 deacetylation and downregulation of K16 acetylation. This correlates with abnormal accumulation of error-prone nonhomologous end joining (NHEJ) repair proteins on newly replicated chromatin. The knockdown of the histone acetyltransferase MOF recapitulates preferential engagement of NHEJ repair activity in control VSMCs. Additionally, we find that primary donor-derived coronary artery vascular smooth muscle cells from aged individuals show similar defects to HGPS VSMCs, including loss of H4K16 acetylation. Altogether, we provide insight into the molecular mechanisms underlying vascular complications associated with HGPS patients and normative aging.
Collapse
Affiliation(s)
- Mzwanele Ngubo
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
| | - Zhaoyi Chen
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Darin McDonald
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Rana Karimpour
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Amit Shrestha
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Julien Yockell‐Lelièvre
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Aurélie Laurent
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Université de StrasbourgStrasbourgFrance
| | - Ojong Tabi Ojong Besong
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- School of BioscienceUniversity of SkövdeSkövdeSweden
| | - Eve C. Tsai
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
- Division of Neurosurgery, Department of Surgery, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - F. Jeffrey Dilworth
- Department of Cell and Regenerative BiologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Michael J. Hendzel
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - William L. Stanford
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Department of Biochemistry, Microbiology & ImmunologyUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
8
|
Katsuumi G, Shimizu I, Suda M, Yoshida Y, Furihata T, Joki Y, Hsiao CL, Jiaqi L, Fujiki S, Abe M, Sugimoto M, Soga T, Minamino T. SGLT2 inhibition eliminates senescent cells and alleviates pathological aging. NATURE AGING 2024; 4:926-938. [PMID: 38816549 PMCID: PMC11257941 DOI: 10.1038/s43587-024-00642-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/02/2024] [Indexed: 06/01/2024]
Abstract
It has been reported that accumulation of senescent cells in various tissues contributes to pathological aging and that elimination of senescent cells (senolysis) improves age-associated pathologies. Here, we demonstrate that inhibition of sodium-glucose co-transporter 2 (SGLT2) enhances clearance of senescent cells, thereby ameliorating age-associated phenotypic changes. In a mouse model of dietary obesity, short-term treatment with the SGLT2 inhibitor canagliflozin reduced the senescence load in visceral adipose tissue and improved adipose tissue inflammation and metabolic dysfunction, but normalization of plasma glucose by insulin treatment had no effect on senescent cells. Canagliflozin extended the lifespan of mice with premature aging even when treatment was started in middle age. Metabolomic analyses revealed that short-term treatment with canagliflozin upregulated 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside, enhancing immune-mediated clearance of senescent cells by downregulating expression of programmed cell death-ligand 1. These findings suggest that inhibition of SGLT2 has an indirect senolytic effect by enhancing endogenous immunosurveillance of senescent cells.
Collapse
Affiliation(s)
- Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Senotherapeutics, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takaaki Furihata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yusuke Joki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Chieh-Lun Hsiao
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Liang Jiaqi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shinya Fujiki
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masataka Sugimoto
- Molecular and Cellular Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, Japan.
| |
Collapse
|
9
|
Gowda SK, Baskaran N, Behera B, Ayyanar P, Vinay K, Thakur V. Reticulate sclerodermoid pigmentation in Hutchinson-Gilford progeria syndrome - A report of two cases. Indian J Dermatol Venereol Leprol 2024; 0:1-3. [PMID: 38841925 DOI: 10.25259/ijdvl_1325_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/12/2024] [Indexed: 06/07/2024]
Affiliation(s)
- Shreya K Gowda
- Department of Dermatology and Venereology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Narayanan Baskaran
- Department of Dermatology, Venereology and Leprology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Biswanath Behera
- Department of Dermatology and Venereology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Pavithra Ayyanar
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Keshavamurthy Vinay
- Department of Dermatology, Venereology and Leprology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Vishal Thakur
- Department of Dermatology and Venereology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| |
Collapse
|
10
|
Bougaran P, Bautch VL. Life at the crossroads: the nuclear LINC complex and vascular mechanotransduction. Front Physiol 2024; 15:1411995. [PMID: 38831796 PMCID: PMC11144885 DOI: 10.3389/fphys.2024.1411995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Vascular endothelial cells line the inner surface of all blood vessels, where they are exposed to polarized mechanical forces throughout their lifespan. Both basal substrate interactions and apical blood flow-induced shear stress regulate blood vessel development, remodeling, and maintenance of vascular homeostasis. Disruption of these interactions leads to dysfunction and vascular pathologies, although how forces are sensed and integrated to affect endothelial cell behaviors is incompletely understood. Recently the endothelial cell nucleus has emerged as a prominent force-transducing organelle that participates in vascular mechanotransduction, via communication to and from cell-cell and cell-matrix junctions. The LINC complex, composed of SUN and nesprin proteins, spans the nuclear membranes and connects the nuclear lamina, the nuclear envelope, and the cytoskeleton. Here we review LINC complex involvement in endothelial cell mechanotransduction, describe unique and overlapping functions of each LINC complex component, and consider emerging evidence that two major SUN proteins, SUN1 and SUN2, orchestrate a complex interplay that extends outward to cell-cell and cell-matrix junctions and inward to interactions within the nucleus and chromatin. We discuss these findings in relation to vascular pathologies such as Hutchinson-Gilford progeria syndrome, a premature aging disorder with cardiovascular impairment. More knowledge of LINC complex regulation and function will help to understand how the nucleus participates in endothelial cell force sensing and how dysfunction leads to cardiovascular disease.
Collapse
Affiliation(s)
- Pauline Bougaran
- Department of Biology, The University of North Carolina, Chapel Hill, NC, United States
| | - Victoria L. Bautch
- Department of Biology, The University of North Carolina, Chapel Hill, NC, United States
- McAllister Heart Institute, The University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
11
|
Jyothi S, Prashanth RR, Nair S, Haribalakrishna A. A Growth-Restricted Neonate with Abnormal Facies and Lax Skin. Neoreviews 2024; 25:e286-e289. [PMID: 38688889 DOI: 10.1542/neo.25-5-e286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Affiliation(s)
- S Jyothi
- Department of Neonatology, Seth G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - R R Prashanth
- Department of Neonatology, Seth G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - Sruthi Nair
- Department of Neonatology, Seth G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - Anitha Haribalakrishna
- Department of Neonatology, Seth G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| |
Collapse
|
12
|
Wang X, Fukumoto T, Noma KI. Therapeutic strategies targeting cellular senescence for cancer and other diseases. J Biochem 2024; 175:525-537. [PMID: 38366629 PMCID: PMC11058315 DOI: 10.1093/jb/mvae015] [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: 11/17/2023] [Revised: 01/12/2024] [Accepted: 01/30/2024] [Indexed: 02/18/2024] Open
Abstract
Cellular senescence occurs in response to endogenous or exogenous stresses and is characterized by stable cell cycle arrest, alterations in nuclear morphology and secretion of proinflammatory factors, referred to as the senescence-associated secretory phenotype (SASP). An increase of senescent cells is associated with the development of several types of cancer and aging-related diseases. Therefore, senolytic agents that selectively remove senescent cells may offer opportunities for developing new therapeutic strategies against such cancers and aging-related diseases. This review outlines senescence inducers and the general characteristics of senescent cells. We also discuss the involvement of senescent cells in certain cancers and diseases. Finally, we describe a series of senolytic agents and their utilization in therapeutic strategies.
Collapse
Affiliation(s)
- Xuebing Wang
- Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo 060-0815, Japan
| | - Takeshi Fukumoto
- Division of Dermatology, Department of Internal Related, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Ken-ichi Noma
- Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo 060-0815, Japan
- Institute of Molecular Biology, University of Oregon, 1370 Franklin Blvd, Eugene, OR 97403, USA
| |
Collapse
|
13
|
Kumar M, Yan P, Kuchel GA, Xu M. Cellular Senescence as a Targetable Risk Factor for Cardiovascular Diseases: Therapeutic Implications: JACC Family Series. JACC Basic Transl Sci 2024; 9:522-534. [PMID: 38680957 PMCID: PMC11055207 DOI: 10.1016/j.jacbts.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 05/01/2024]
Abstract
The prevalence of cardiovascular diseases markedly rises with age. Cellular senescence, a hallmark of aging, is characterized by irreversible cell cycle arrest and the manifestation of a senescence-associated secretory phenotype, which has emerged as a significant contributor to aging, mortality, and a spectrum of chronic ailments. An increasing body of preclinical and clinical research has established connections between senescence, senescence-associated secretory phenotype, and age-related cardiac and vascular pathologies. This review comprehensively outlines studies delving into the detrimental impact of senescence on various cardiovascular diseases, encompassing systemic atherosclerosis (including coronary artery disease, stroke, and peripheral arterial disease), as well as conditions such as hypertension, congestive heart failure, arrhythmias, and valvular heart diseases. In addition, we have preclinical studies demonstrating the beneficial effects of senolytics-a class of drugs designed to eliminate senescent cells selectively across diverse cardiovascular disease scenarios. Finally, we address knowledge gaps on the influence of senescence on cardiovascular systems and discuss the future trajectory of strategies targeting senescence for cardiovascular diseases.
Collapse
Affiliation(s)
- Manish Kumar
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Division of Critical Care Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Pengyi Yan
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - George A. Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Ming Xu
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
14
|
Hanania HL, Hopkins CR, Herd TJ. Skin Hardening and Eczema in a 9-month-old Girl. Pediatr Rev 2024; 45:239-243. [PMID: 38556512 DOI: 10.1542/pir.2022-005701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Affiliation(s)
| | | | - Tiffany J Herd
- Department of Dermatology, Texas Children's Hospital, Houston, TX
| |
Collapse
|
15
|
Messina C. Woodhouse-Sakati syndrome: A review. Rev Neurol (Paris) 2024:S0035-3787(24)00023-7. [PMID: 38320940 DOI: 10.1016/j.neurol.2023.11.008] [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: 03/18/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 02/08/2024]
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a rare and inherited spectrum of movement disorders caused by mutations affecting the function of proteins that participate in the homeostasis of tissue metals such as iron or copper and other metabolic pathways, although the precise function of the proteins encoded are not always known. Woodhouse-Sakati Syndrome (WSS) is one of the rarest NBIAs. Patients with WSS are characterized by endocrinological and neurological manifestations and neuroradiological findings. However, diagnostic criteria have not been published yet. This article reviews updates on the genetic, clinical, biological and imaging findings of WSS and provides a practical guide to recognize this extremely rare disorder.
Collapse
Affiliation(s)
- C Messina
- Department of Medical and Surgical Sciences, and Advanced Technologies, "G.F. Ingrassia", Azienda Ospedaliera Universitaria "G. Rodolico-San Marco", University of Catania, Catania, Italy.
| |
Collapse
|
16
|
Chen Y, Zhen Z, Chen L, Wang H, Wang X, Sun X, Song Z, Wang H, Lin Y, Zhang W, Wu G, Jiang Y, Mao Z. Androgen signaling stabilizes genomes to counteract senescence by promoting XRCC4 transcription. EMBO Rep 2023; 24:e56984. [PMID: 37955230 PMCID: PMC10702805 DOI: 10.15252/embr.202356984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 10/16/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023] Open
Abstract
Aging is accompanied by a decreased DNA repair capacity, which might contribute to age-associated functional decline in multiple tissues. Disruption in hormone signaling, associated with reproductive organ dysfunction, is an early event of age-related tissue degeneration, but whether it impacts DNA repair in nonreproductive organs remains elusive. Using skin fibroblasts derived from healthy donors with a broad age range, we show here that the downregulation of expression of XRCC4, a factor involved in nonhomologous end-joining (NHEJ) repair, which is the dominant pathway to repair somatic double-strand breaks, is mediated through transcriptional mechanisms. We show that the androgen receptor (AR), whose expression is also reduced during aging, directly binds to and enhances the activity of the XRCC4 promoter, facilitating XRCC4 transcription and thus stabilizing the genome. We also demonstrate that dihydrotestosterone (DHT), a powerful AR agonist, restores XRCC4 expression and stabilizes the genome in different models of cellular aging. Moreover, DHT treatment reverses senescence-associated phenotypes, opening a potential avenue to aging interventions in the future.
Collapse
Affiliation(s)
- Yu Chen
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Zhengyi Zhen
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Lingjiang Chen
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Hao Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Xuhui Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Xiaoxiang Sun
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Zhiwei Song
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Haiyan Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yizi Lin
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Wenjun Zhang
- Department of Plastic SurgeryChangzheng HospitalShanghaiChina
| | - Guizhu Wu
- Department of Gynecology, Shanghai First Maternity and Infant HospitalShanghai Tongji University School of MedicineShanghaiChina
| | - Ying Jiang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| |
Collapse
|
17
|
Odinammadu KO, Shilagardi K, Tuminelli K, Judge DP, Gordon LB, Michaelis S. The farnesyl transferase inhibitor (FTI) lonafarnib improves nuclear morphology in ZMPSTE24-deficient fibroblasts from patients with the progeroid disorder MAD-B. Nucleus 2023; 14:2288476. [PMID: 38050983 PMCID: PMC10730222 DOI: 10.1080/19491034.2023.2288476] [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: 10/02/2023] [Accepted: 11/20/2023] [Indexed: 12/07/2023] Open
Abstract
Several related progeroid disorders are caused by defective post-translational processing of prelamin A, the precursor of the nuclear scaffold protein lamin A, encoded by LMNA. Prelamin A undergoes farnesylation and additional modifications at its C-terminus. Subsequently, the farnesylated C-terminal segment is cleaved off by the zinc metalloprotease ZMPSTE24. The premature aging disorder Hutchinson Gilford progeria syndrome (HGPS) and a related progeroid disease, mandibuloacral dysplasia (MAD-B), are caused by mutations in LMNA and ZMPSTE24, respectively, that result in failure to process the lamin A precursor and accumulate permanently farnesylated forms of prelamin A. The farnesyl transferase inhibitor (FTI) lonafarnib is known to correct the aberrant nuclear morphology of HGPS patient cells and improves lifespan in children with HGPS. Importantly, and in contrast to a previous report, we show here that FTI treatment also improves the aberrant nuclear phenotypes in MAD-B patient cells with mutations in ZMPSTE24 (P248L or L425P). As expected, lonafarnib does not correct nuclear defects for cells with lamin A processing-proficient mutations. We also examine prelamin A processing in fibroblasts from two individuals with a prevalent laminopathy mutation LMNA-R644C. Despite the proximity of residue R644 to the prelamin A cleavage site, neither R644C patient cell line shows a prelamin A processing defect, and both have normal nuclear morphology. This work clarifies the prelamin A processing status and role of FTIs in a variety of laminopathy patient cells and supports the FDA-approved indication for the FTI Zokinvy for patients with processing-deficient progeroid laminopathies, but not for patients with processing-proficient laminopathies.
Collapse
Affiliation(s)
- Kamsi O. Odinammadu
- Department of Cell Biology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Khurts Shilagardi
- Department of Cell Biology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Daniel P. Judge
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Leslie B. Gordon
- The Progeria Research Foundation, Peabody, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Division of Genetics, Hasbro Children’s Hospital and Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Susan Michaelis
- Department of Cell Biology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Ferreira‐Marques M, Carvalho A, Franco AC, Leal A, Botelho M, Carmo‐Silva S, Águas R, Cortes L, Lucas V, Real AC, López‐Otín C, Nissan X, de Almeida LP, Cavadas C, Aveleira CA. Ghrelin delays premature aging in Hutchinson-Gilford progeria syndrome. Aging Cell 2023; 22:e13983. [PMID: 37858983 PMCID: PMC10726901 DOI: 10.1111/acel.13983] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare and fatal genetic condition that arises from a single nucleotide alteration in the LMNA gene, leading to the production of a defective lamin A protein known as progerin. The accumulation of progerin accelerates the onset of a dramatic premature aging phenotype in children with HGPS, characterized by low body weight, lipodystrophy, metabolic dysfunction, skin, and musculoskeletal age-related dysfunctions. In most cases, these children die of age-related cardiovascular dysfunction by their early teenage years. The absence of effective treatments for HGPS underscores the critical need to explore novel safe therapeutic strategies. In this study, we show that treatment with the hormone ghrelin increases autophagy, decreases progerin levels, and alleviates other cellular hallmarks of premature aging in human HGPS fibroblasts. Additionally, using a HGPS mouse model (LmnaG609G/G609G mice), we demonstrate that ghrelin administration effectively rescues molecular and histopathological progeroid features, prevents progressive weight loss in later stages, reverses the lipodystrophic phenotype, and extends lifespan of these short-lived mice. Therefore, our findings uncover the potential of modulating ghrelin signaling offers new treatment targets and translational approaches that may improve outcomes and enhance the quality of life for patients with HGPS and other age-related pathologies.
Collapse
Affiliation(s)
- Marisa Ferreira‐Marques
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - André Carvalho
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Ana Catarina Franco
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Ana Leal
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Mariana Botelho
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Sara Carmo‐Silva
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Rodolfo Águas
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Luísa Cortes
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Vasco Lucas
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Ana Carolina Real
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Carlos López‐Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de OncologíaUniversidad de OviedoOviedoSpain
| | - Xavier Nissan
- CECS, I‐StemCorbeil‐EssonnesFrance
- INSERM U861, I‐StemCorbeil‐EssonnesFrance
- UEVE U861, I‐StemCorbeil‐EssonnesFrance
| | - Luís Pereira de Almeida
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Cláudia Cavadas
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Célia A. Aveleira
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- MIA‐Portugal – Multidisciplinar Institute of AgeingUniversity of CoimbraCoimbraPortugal
| |
Collapse
|
19
|
Lian J, Du L, Li Y, Yin Y, Yu L, Wang S, Ma H. Hutchinson-Gilford progeria syndrome: Cardiovascular manifestations and treatment. Mech Ageing Dev 2023; 216:111879. [PMID: 37832833 DOI: 10.1016/j.mad.2023.111879] [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: 07/04/2023] [Revised: 09/04/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS), also known as hereditary progeria syndrome, is caused by mutations in the LMNA gene and the expression of progerin, which causes accelerated aging and premature death, with most patients dying of heart failure or other cardiovascular complications in their teens. HGPS patients are able to exhibit cardiovascular phenotypes similar to physiological aging, such as extensive atherosclerosis, smooth muscle cell loss, vascular lesions, and electrical and functional abnormalities of the heart. It also excludes the traditional risk causative factors of cardiovascular disease, making HGPS a new model for studying aging-related cardiovascular disease. Here, we analyzed the pathogenesis and pathophysiological characteristics of HGPS and the relationship between HGPS and cardiovascular disease, provided insight into the molecular mechanisms of cardiovascular disease pathogenesis in HGPS patients and treatment strategies for this disease. Moreover, we summarize the disease models used in HGPS studies to improve our understanding of the pathological mechanisms of cardiovascular aging in HGPS patients.
Collapse
Affiliation(s)
- Jing Lian
- Medical School of Yan'an University, Yan'an, China
| | - Linfang Du
- Medical School of Yan'an University, Yan'an, China
| | - Yang Li
- School of Basic Medical Sciences, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Lu Yu
- Department of Pathology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | | | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
20
|
Chen Y, Huang S, Cui Z, Sun X, Tang Y, Zhang H, Chen Z, Jiang R, Zhang W, Li X, Chen J, Liu B, Jiang Y, Wei K, Mao Z. Impaired end joining induces cardiac atrophy in a Hutchinson-Gilford progeria mouse model. Proc Natl Acad Sci U S A 2023; 120:e2309200120. [PMID: 37967221 PMCID: PMC10666128 DOI: 10.1073/pnas.2309200120] [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] [Accepted: 10/14/2023] [Indexed: 11/17/2023] Open
Abstract
Patients with Hutchinson-Gilford progeria syndrome (HGPS) present with a number of premature aging phenotypes, including DNA damage accumulation, and many of them die of cardiovascular complications. Although vascular pathologies have been reported, whether HGPS patients exhibit cardiac dysfunction and its underlying mechanism is unclear, rendering limited options for treating HGPS-related cardiomyopathy. In this study, we reported a cardiac atrophy phenotype in the LmnaG609G/G609G mice (hereafter, HGPS mice). Using a GFP-based reporter system, we demonstrated that the efficiency of nonhomologous end joining (NHEJ) declined by 50% in HGPS cardiomyocytes in vivo, due to the attenuated interaction between γH2AX and Progerin, the causative factor of HGPS. As a result, genomic instability in cardiomyocytes led to an increase of CHK2 protein level, promoting the LKB1-AMPKα interaction and AMPKα phosphorylation, which further led to the activation of FOXO3A-mediated transcription of atrophy-related genes. Moreover, inhibiting AMPK enlarged cardiomyocyte sizes both in vitro and in vivo. Most importantly, our proof-of-concept study indicated that isoproterenol treatment significantly reduced AMPKα and FOXO3A phosphorylation in the heart, attenuated the atrophy phenotype, and extended the mean lifespan of HGPS mice by ~21%, implying that targeting cardiac atrophy may be an approach to HGPS treatment.
Collapse
Affiliation(s)
- Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Shiqi Huang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Zhen Cui
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Xiaoxiang Sun
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Yansong Tang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Hongjie Zhang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Zhixi Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Rui Jiang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Weina Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Xue Li
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Jiayu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Baohua Liu
- National Engineering Research Center for Biotechnology (Shenzhen), Carson International Cancer Center, Medical Research Center, Shenzhen University Health Science Center, Shenzhen518055, China
| | - Ying Jiang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Ke Wei
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Tsingtao Advanced Research Institute, Tongji University, Qingdao266071, China
| |
Collapse
|
21
|
Majumder S, Chattopadhyay A, Wright JM, Guan P, Buja LM, Kwartler CS, Milewicz DM. Pericentrin deficiency in smooth muscle cells augments atherosclerosis through HSF1-driven cholesterol biosynthesis and PERK activation. JCI Insight 2023; 8:e173247. [PMID: 37937642 PMCID: PMC10721278 DOI: 10.1172/jci.insight.173247] [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: 06/21/2023] [Accepted: 09/27/2023] [Indexed: 11/09/2023] Open
Abstract
Microcephalic osteodysplastic primordial dwarfism type II (MOPDII) is caused by biallelic loss-of-function variants in pericentrin (PCNT), and premature coronary artery disease (CAD) is a complication of the syndrome. Histopathology of coronary arteries from patients with MOPDII who died of CAD in their 20s showed extensive atherosclerosis. Hyperlipidemic mice with smooth muscle cell-specific (SMC-specific) Pcnt deficiency (PcntSMC-/-) exhibited significantly greater atherosclerotic plaque burden compared with similarly treated littermate controls despite similar serum lipid levels. Loss of PCNT in SMCs induced activation of heat shock factor 1 (HSF1) and consequently upregulated the expression and activity of HMG-CoA reductase (HMGCR), the rate-limiting enzyme in cholesterol biosynthesis. The increased cholesterol biosynthesis in PcntSMC-/- SMCs augmented PERK signaling and phenotypic modulation compared with control SMCs. Treatment with the HMGCR inhibitor, pravastatin, blocked the augmented SMC modulation and reduced plaque burden in hyperlipidemic PcntSMC-/- mice to that of control mice. These data support the notion that Pcnt deficiency activates cellular stress to increase SMC modulation and plaque burden, and targeting this pathway with statins in patients with MOPDII has the potential to reduce CAD in these individuals. The molecular mechanism uncovered further emphasizes SMC cytosolic stress and HSF1 activation as a pathway driving atherosclerotic plaque formation independently of cholesterol levels.
Collapse
Affiliation(s)
- Suravi Majumder
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, and
| | - Abhijnan Chattopadhyay
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, and
| | - Jamie M. Wright
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, and
| | - Pujun Guan
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, and
| | - L. Maximilian Buja
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Callie S. Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, and
| | - Dianna M. Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, and
| |
Collapse
|
22
|
Buxboim A, Kronenberg-Tenga R, Salajkova S, Avidan N, Shahak H, Thurston A, Medalia O. Scaffold, mechanics and functions of nuclear lamins. FEBS Lett 2023; 597:2791-2805. [PMID: 37813648 DOI: 10.1002/1873-3468.14750] [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: 06/16/2023] [Revised: 09/05/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023]
Abstract
Nuclear lamins are type-V intermediate filaments that are involved in many nuclear processes. In mammals, A- and B-type lamins assemble into separate physical meshwork underneath the inner nuclear membrane, the nuclear lamina, with some residual fraction localized within the nucleoplasm. Lamins are the major part of the nucleoskeleton, providing mechanical strength and flexibility to protect the genome and allow nuclear deformability, while also contributing to gene regulation via interactions with chromatin. While lamins are the evolutionary ancestors of all intermediate filament family proteins, their ultimate filamentous assembly is markedly different from their cytoplasmic counterparts. Interestingly, hundreds of genetic mutations in the lamina proteins have been causally linked with a broad range of human pathologies, termed laminopathies. These include muscular, neurological and metabolic disorders, as well as premature aging diseases. Recent technological advances have contributed to resolving the filamentous structure of lamins and the corresponding lamina organization. In this review, we revisit the multiscale lamin organization and discuss its implications on nuclear mechanics and chromatin organization within lamina-associated domains.
Collapse
Affiliation(s)
- Amnon Buxboim
- The Rachel and Selim Benin School of Computer Science and Engineering and The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| | | | - Sarka Salajkova
- Department of Biochemistry, University of Zurich, Switzerland
| | - Nili Avidan
- The Rachel and Selim Benin School of Computer Science and Engineering and The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| | - Hen Shahak
- The Rachel and Selim Benin School of Computer Science and Engineering and The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| | - Alice Thurston
- Department of Biochemistry, University of Zurich, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Switzerland
| |
Collapse
|
23
|
Brandt A, Petrovsky R, Kriebel M, Großhans J. Use of Farnesyl Transferase Inhibitors in an Ageing Model in Drosophila. J Dev Biol 2023; 11:40. [PMID: 37987370 PMCID: PMC10660854 DOI: 10.3390/jdb11040040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/22/2023] Open
Abstract
The presence of farnesylated proteins at the inner nuclear membrane (INM), such as the Lamins or Kugelkern in Drosophila, leads to specific changes in the nuclear morphology and accelerated ageing on the organismal level reminiscent of the Hutchinson-Gilford progeria syndrome (HGPS). Farnesyl transferase inhibitors (FTIs) can suppress the phenotypes of the nuclear morphology in cultured fibroblasts from HGPS patients and cultured cells overexpressing farnesylated INM proteins. Similarly, FTIs have been reported to suppress the shortened lifespan in model organisms. Here, we report an experimental system combining cell culture and Drosophila flies for testing the activity of substances on the HGPS-like nuclear morphology and lifespan, with FTIs as an experimental example. Consistent with previous reports, we show that FTIs were able to ameliorate the nuclear phenotypes induced by the farnesylated nuclear proteins Progerin, Kugelkern, or truncated Lamin B in cultured cells. The subsequent validation in Drosophila lifespan assays demonstrated the applicability of the experimental system: treating adult Drosophila with the FTI ABT-100 reversed the nuclear phenotypes and extended the lifespan of experimentally induced short-lived flies. Since kugelkern-expressing flies have a significantly shorter average lifespan, half the time is needed for testing substances in the lifespan assay.
Collapse
Affiliation(s)
| | - Roman Petrovsky
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| | - Maria Kriebel
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| | - Jörg Großhans
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| |
Collapse
|
24
|
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
|
25
|
Perales S, Sigamani V, Rajasingh S, Czirok A, Rajasingh J. Hutchinson-Gilford progeria patient-derived cardiomyocyte model of carrying LMNA gene variant c.1824 C > T. Cell Tissue Res 2023; 394:189-207. [PMID: 37572165 DOI: 10.1007/s00441-023-03813-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/12/2023] [Indexed: 08/14/2023]
Abstract
Cardiovascular diseases, atherosclerosis, and strokes are the most common causes of death in patients with Hutchinson-Gilford progeria syndrome (HGPS). The LMNA variant c.1824C > T accounts for ~ 90% of HGPS cases. The detailed molecular mechanisms of Lamin A in the heart remain elusive due to the lack of appropriate in vitro models. We hypothesize that HGPS patient's induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iCMCs) will provide a model platform to study the cardio-pathologic mechanisms associated with HGPS. To elucidate the effects of progerin in cardiomyocytes, we first obtained skin fibroblasts (SFs) from a de-identified HGPS patient (hPGP1, proband) and both parents from the Progeria Research Foundation. Through Sanger sequencing and restriction fragment length polymorphism, with the enzyme EciI, targeting Lamin A, we characterized hPGP1-SFs as heterozygous mutants for the LMNA variant c.1824 C > T. Additionally, we performed LMNA exon 11 bisulfite sequencing to analyze the methylation status of the progeria cells. Furthermore, we reprogrammed the three SFs into iPSCs and differentiated them into iCMCs, which gained a beating on day 7. Through particle image velocimetry analysis, we found that hPGP1-iCMCs had an irregular contractile function and decreased cardiac-specific gene and protein expressions by qRT-PCR and Western blot. Our progeria-patient-derived iCMCs were found to be functionally and structurally defective when compared to normal iCMCs. This in vitro model will help in elucidating the role of Lamin A in cardiac diseases and the cardio-pathologic mechanisms associated with progeria. It provides a new platform for researchers to study novel treatment approaches for progeria-associated cardiac diseases.
Collapse
Affiliation(s)
- Selene Perales
- Department of Bioscience Research, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA
| | - Andras Czirok
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA.
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA.
- Department of Medicine-Cardiology, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
26
|
Sharma A, Swarnkar B, Sethuraman G. Hutchinson-Gilford progeria. BMJ Case Rep 2023; 16:e256203. [PMID: 37723091 PMCID: PMC10510932 DOI: 10.1136/bcr-2023-256203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023] Open
Affiliation(s)
- Ananya Sharma
- Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
| | - Bhavya Swarnkar
- Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
| | - Gomathy Sethuraman
- Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
27
|
Chitara N, Rani D, Kanchan T, Krishan K. Odontoma and other congenital dental anomalies: Implications for forensic identification. Congenit Anom (Kyoto) 2023; 63:132-140. [PMID: 37503819 DOI: 10.1111/cga.12533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 07/29/2023]
Abstract
Forensic odontology plays a crucial role in establishing the identity in mass disasters and criminal cases with high accuracy. Dental anomalies and features help in such situations. Congenital and developmental dental anomalies can be easily documented to establish distinctive and individualistic characteristics of an individual. The location, number of teeth involved, and the type of anomaly vary between individuals. Similarly, dental malformations also assist greatly in the identification process. Many types of dental anomalies have been studied in the past for their individualistic characteristics in forensic examinations. One such dental anomaly is odontoma, which is a benign odontogenic malformation. This malformation may also help in the identification of the deceased, when recorded and examined accurately. An odontome is a malformed teeth-like structures consisting of enamel, dentin, and pulpal tissue, formed due to the growth of completely differentiated epithelial and mesenchymal cells. If antemortem (AM) dental records incorporate information regarding odontomes and other dental anomalies, including in radiographs, orthopantomograms or microradiographs, positive identification may be established by comparison of these records with postmortem (PM) records. In the present communication, a rare case of compound composite odontoma in the anterior mandible with multiple denticles has been discussed with a brief overview of congenital and developmental dental anomalies. The authors emphasize the importance of such rare dental anomalies and malformations which may be used for identifying the deceased in mass disasters and forensic identification.
Collapse
Affiliation(s)
- Nandini Chitara
- Department of Anthropology, Panjab University, Chandigarh, India
| | - Deepika Rani
- Department of Anthropology, Panjab University, Chandigarh, India
| | - Tanuj Kanchan
- Department of Forensic Medicine, All India Institute of Medical Sciences, Jodhpur, India
| | - Kewal Krishan
- Department of Anthropology, Panjab University, Chandigarh, India
| |
Collapse
|
28
|
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: 1] [Impact Index Per Article: 1.0] [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
|
29
|
Saadi A, Navarro C, Ozalp O, Lourenco CM, Fayek R, Da Silva N, Chaouch A, Benahmed M, Kubisch C, Munnich A, Lévy N, Roll P, Pacha LA, Chaouch M, Lessel D, De Sandre-Giovannoli A. A recurrent homozygous LMNA missense variant p.Thr528Met causes atypical progeroid syndrome characterized by mandibuloacral dysostosis, severe muscular dystrophy, and skeletal deformities. Am J Med Genet A 2023; 191:2274-2289. [PMID: 37387251 DOI: 10.1002/ajmg.a.63335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 07/01/2023]
Abstract
Atypical progeroid syndromes (APS) are premature aging syndromes caused by pathogenic LMNA missense variants, associated with unaltered expression levels of lamins A and C, without accumulation of wild-type or deleted prelamin A isoforms, as observed in Hutchinson-Gilford progeria syndrome (HGPS) or HGPS-like syndromes. A specific LMNA missense variant, (p.Thr528Met), was previously identified in a compound heterozygous state in patients affected by APS and severe familial partial lipodystrophy, whereas heterozygosity was recently identified in patients affected by Type 2 familial partial lipodystrophy. Here, we report four unrelated boys harboring homozygosity for the p.Thr528Met, variant who presented with strikingly homogeneous APS clinical features, including osteolysis of mandibles, distal clavicles and phalanges, congenital muscular dystrophy with elevated creatine kinase levels, and major skeletal deformities. Immunofluorescence analyses of patient-derived primary fibroblasts showed a high percentage of dysmorphic nuclei with nuclear blebs and typical honeycomb patterns devoid of lamin B1. Interestingly, in some protrusions emerin or LAP2α formed aberrant aggregates, suggesting pathophysiology-associated clues. These four cases further confirm that a specific LMNA variant can lead to the development of strikingly homogeneous clinical phenotypes, in these particular cases a premature aging phenotype with major musculoskeletal involvement linked to the homozygous p.Thr528Met variant.
Collapse
Affiliation(s)
- Abdelkrim Saadi
- Service de neurologie, Etablissement Hospitalier Specialisé de Ben Aknoun, Université Benyoucef Benkhedda, Algiers, Algeria
- Laboratoire de Neurosciences, Service de neurologie, Centre Hospitalo Universitaire Mustapha Bacha, Université Benyoucef Benkhedda Alger, Algiers, Algeria
| | - Claire Navarro
- INSERM, MMG, Aix Marseille University, Marseille, France
- Neoflow Therapeutics, 61 boulevard des Dames, 13002, Marseille, France
| | - Ozge Ozalp
- Genetic Diagnosis Center, Adana City Training and Research Hospital University of Health Sciences, Adana, Turkey
| | - Charles Marques Lourenco
- Neurogenetics Unit-Inborn Errors of Metabolism Clinics, National Reference Center for Rare Diseases, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, Brazil
- Department of Specialized Education, Personalized Medicine Area, DLE/Grupo Pardini, Rio de Janeiro, Brazil
| | - Racha Fayek
- INSERM, MMG, Aix Marseille University, Marseille, France
| | | | - Athmane Chaouch
- Service de neurophysiologie, Etablissement Hospitalier Specialisé, Algiers, Algeria
| | - Meryem Benahmed
- Service d'anatomo-pathologie, Centre Pierre Marie Curie, Algiers, Algeria
| | - Christian Kubisch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Arnold Munnich
- Department of Clinical Genetics, Institut de Recherche Necker Enfants Malades, Paris, France
| | - Nicolas Lévy
- INSERM, MMG, Aix Marseille University, Marseille, France
- Department of Medical Genetics, La Timone Hospital, APHM, Marseille, France
| | - Patrice Roll
- INSERM, MMG, Aix Marseille University, Marseille, France
- Cell Biology Laboratory, La Timone Hospital, APHM, Marseille, France
| | - Lamia Ali Pacha
- Laboratoire de Neurosciences, Service de neurologie, Centre Hospitalo Universitaire Mustapha Bacha, Université Benyoucef Benkhedda Alger, Algiers, Algeria
| | - Malika Chaouch
- Service de neurologie, Etablissement Hospitalier Specialisé de Ben Aknoun, Université Benyoucef Benkhedda, Algiers, Algeria
| | - Davor Lessel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Human Genetics, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Annachiara De Sandre-Giovannoli
- INSERM, MMG, Aix Marseille University, Marseille, France
- Department of Medical Genetics, La Timone Hospital, APHM, Marseille, France
- Biological Resource Center (CRB-TAC), La Timone Hospital, APHM, Marseille, France
| |
Collapse
|
30
|
Pande S, Ghosh DK. Nuclear proteostasis imbalance in laminopathy-associated premature aging diseases. FASEB J 2023; 37:e23116. [PMID: 37498235 DOI: 10.1096/fj.202300878r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/15/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
Laminopathies are a group of rare genetic disorders with heterogeneous clinical phenotypes such as premature aging, cardiomyopathy, lipodystrophy, muscular dystrophy, microcephaly, epilepsy, and so on. The cellular phenomena associated with laminopathy invariably show disruption of nucleoskeleton of lamina due to deregulated expression, localization, function, and interaction of mutant lamin proteins. Impaired spatial and temporal tethering of lamin proteins to the lamina or nucleoplasmic aggregation of lamins are the primary molecular events that can trigger nuclear proteotoxicity by modulating differential protein-protein interactions, sequestering quality control proteins, and initiating a cascade of abnormal post-translational modifications. Clearly, laminopathic cells exhibit moderate to high nuclear proteotoxicity, raising the question of whether an imbalance in nuclear proteostasis is involved in laminopathic diseases, particularly in diseases of early aging such as HGPS and laminopathy-associated premature aging. Here, we review nuclear proteostasis and its deregulation in the context of lamin proteins and laminopathies.
Collapse
Affiliation(s)
- Shruti Pande
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Debasish Kumar Ghosh
- Enteric Disease Division, Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
31
|
Wilkie SE, Marcu DE, Carter RN, Morton NM, Gonzalo S, Selman C. Hepatic hydrogen sulfide levels are reduced in mouse model of Hutchinson-Gilford progeria syndrome. Aging (Albany NY) 2023; 15:5266-5278. [PMID: 37354210 PMCID: PMC10333079 DOI: 10.18632/aging.204835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/09/2023] [Indexed: 06/26/2023]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare human disease characterised by accelerated biological ageing. Current treatments are limited, and most patients die before 15 years of age. Hydrogen sulfide (H2S) is an important gaseous signalling molecule that it central to multiple cellular homeostasis mechanisms. Dysregulation of tissue H2S levels is thought to contribute to an ageing phenotype in many tissues across animal models. Whether H2S is altered in HGPS is unknown. We investigated hepatic H2S production capacity and transcript, protein and enzymatic activity of proteins that regulate hepatic H2S production and disposal in a mouse model of HGPS (G609G mice, mutated Lmna gene equivalent to a causative mutation in HGPS patients). G609G mice were maintained on either regular chow (RC) or high fat diet (HFD), as HFD has been previously shown to significantly extend lifespan of G609G mice, and compared to wild type (WT) mice maintained on RC. RC fed G609G mice had significantly reduced hepatic H2S production capacity relative to WT mice, with a compensatory elevation in mRNA transcripts associated with several H2S production enzymes, including cystathionine-γ-lyase (CSE). H2S levels and CSE protein were partially rescued in HFD fed G609G mice. As current treatments for patients with HGPS have failed to confer significant improvements to symptoms or longevity, the need for novel therapeutic targets is acute and the regulation of H2S through dietary or pharmacological means may be a promising new avenue for research.
Collapse
Affiliation(s)
- Stephen E. Wilkie
- Glasgow Ageing Research Network (GARNER), School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Division of Molecular Metabolism, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna 171 65, Sweden
| | - Diana E. Marcu
- Glasgow Ageing Research Network (GARNER), School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Roderick N. Carter
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Nicholas M. Morton
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Susana Gonzalo
- Department of Biochemistry and Molecular Biology, Edward A. Doisy Research Center, Saint Louis University School of Medicine, MO 63104, USA
| | - Colin Selman
- Glasgow Ageing Research Network (GARNER), School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
32
|
Hartinger R, Lederer EM, Schena E, Lattanzi G, Djabali K. Impact of Combined Baricitinib and FTI Treatment on Adipogenesis in Hutchinson-Gilford Progeria Syndrome and Other Lipodystrophic Laminopathies. Cells 2023; 12:1350. [PMID: 37408186 DOI: 10.3390/cells12101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 07/07/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disease that causes premature aging symptoms, such as vascular diseases, lipodystrophy, loss of bone mineral density, and alopecia. HGPS is mostly linked to a heterozygous and de novo mutation in the LMNA gene (c.1824 C > T; p.G608G), resulting in the production of a truncated prelamin A protein called "progerin". Progerin accumulation causes nuclear dysfunction, premature senescence, and apoptosis. Here, we examined the effects of baricitinib (Bar), an FDA-approved JAK/STAT inhibitor, and a combination of Bar and lonafarnib (FTI) treatment on adipogenesis using skin-derived precursors (SKPs). We analyzed the effect of these treatments on the differentiation potential of SKPs isolated from pre-established human primary fibroblast cultures. Compared to mock-treated HGPS SKPs, Bar and Bar + FTI treatments improved the differentiation of HGPS SKPs into adipocytes and lipid droplet formation. Similarly, Bar and Bar + FTI treatments improved the differentiation of SKPs derived from patients with two other lipodystrophic diseases: familial partial lipodystrophy type 2 (FPLD2) and mandibuloacral dysplasia type B (MADB). Overall, the results show that Bar treatment improves adipogenesis and lipid droplet formation in HGPS, FPLD2, and MADB, indicating that Bar + FTI treatment might further ameliorate HGPS pathologies compared to lonafarnib treatment alone.
Collapse
Affiliation(s)
- Ramona Hartinger
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Eva-Maria Lederer
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Elisa Schena
- Unit of Bologna, CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", 40136 Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Giovanna Lattanzi
- Unit of Bologna, CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", 40136 Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Karima Djabali
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| |
Collapse
|
33
|
Kang SM, Seo S, Song EJ, Kweon O, Jo AH, Park S, Woo TG, Kim BH, Oh GT, Park BJ. Progerinin, an Inhibitor of Progerin, Alleviates Cardiac Abnormalities in a Model Mouse of Hutchinson-Gilford Progeria Syndrome. Cells 2023; 12:cells12091232. [PMID: 37174632 PMCID: PMC10177486 DOI: 10.3390/cells12091232] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is an ultra-rare human premature aging disorder that precipitates death because of cardiac disease. Almost all cases of HGPS are caused by aberrant splicing of the LMNA gene that results in the production of a mutant Lamin A protein termed progerin. In our previous study, treatment with Progerinin has been shown to reduce progerin expression and improve aging phenotypes in vitro and in vivo HGPS models. In this record, cardiac parameters (stroke volume (SV), ejection fraction (EF), fractional shortening (FS), etc.) were acquired in LmnaWT/WT and LmnaG609G/WT mice fed with either a vehicle diet or a Progerinin diet by echocardiography (from 38 weeks to 50 weeks at various ages), and then the cardiac function was analyzed. We also acquired the tissue samples and blood serum of LmnaWT/WT and LmnaG609G/WT mice for pathological analysis at the end of echocardiography. From these data, we suggest that the administration of Progerinin in the HGPS model mouse can restore cardiac function and correct arterial abnormalities. These observations provide encouraging evidence for the efficacy of Progerinin for cardiac dysfunction in HGPS.
Collapse
Affiliation(s)
- So-Mi Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea
| | - Seungwoon Seo
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul 03760, Republic of Korea
- Imvastech, 305, Science Building C, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Eun Ju Song
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Okhee Kweon
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Ah-Hyeon Jo
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea
| | - Soyoung Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea
| | - Tae-Gyun Woo
- PRG S & Tech Inc., Geumjeong-gu, Busan 46274, Republic of Korea
| | - Bae-Hoon Kim
- PRG S & Tech Inc., Geumjeong-gu, Busan 46274, Republic of Korea
| | - Goo Taeg Oh
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul 03760, Republic of Korea
- Imvastech, 305, Science Building C, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Bum-Joon Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea
- PRG S & Tech Inc., Geumjeong-gu, Busan 46274, Republic of Korea
| |
Collapse
|
34
|
ZP3 and AIPL1 participate in GVBD of mouse oocytes by affecting the nuclear membrane localization and maturation of farnesylated prelamin A. ZYGOTE 2023; 31:140-148. [PMID: 36533678 DOI: 10.1017/s0967199422000612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The low maturation rate of oocytes is an important reason for female infertility and failure of assisted pregnancy. The germinal vesicle breakdown (GVBD) is a landmark event of oocyte maturation. In our previous studies, we found that zona pellucida 3 (ZP3) was strongly concentrated in the nuclear region of germinal vesicle (GV) oocytes and interacted with aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) and lamin A to promote GVBD. In the current study, we found that lamin A is mainly concentrated in the nuclear membrane. When ZP3 is knocked down, lamin A will be partially transferred to the nucleus of oocytes. The prelamin A is increased in both the nuclear membrane and nucleus, while phosphorylated lamin A (p-lamin A) is significantly reduced. AIPL1 was also proved to accumulate in the GV region of oocytes, and ZP3 deletion can significantly inhibit the aggregation of AIPL1 in the nuclear region. Similar to ZP3 knockdown, the absence of AIPL1 resulted in a decrease in the occurrence of GVBD, an increase in the amount of prelamin A, and a significant decrease in p-lamin A in oocytes developed in vitro. Finally, we propose the hypothesis that ZP3 can stabilize farnesylated prelamin A on the nuclear membrane of AIPL1, and promote its further processing into mature lamin A, therefore promoting the occurrence of GVBD. This study may be an important supplement for the mechanism of oocyte meiotic resumption and provide new diagnostic targets and treatment clues for infertility patients with oocyte maturation disorder.
Collapse
|
35
|
Kim J, Hwang Y, Kim S, Chang Y, Kim Y, Kwon Y, Kim J. Transcriptional activation of endogenous Oct4 via the CRISPR/dCas9 activator ameliorates Hutchinson-Gilford progeria syndrome in mice. Aging Cell 2023:e13825. [PMID: 36964992 DOI: 10.1111/acel.13825] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 02/13/2023] [Accepted: 02/19/2023] [Indexed: 03/27/2023] Open
Abstract
Partial cellular reprogramming via transient expression of Oct4, Sox2, Klf4, and c-Myc induces rejuvenation and reduces aged-cell phenotypes. In this study, we found that transcriptional activation of the endogenous Oct4 gene by using the CRISPR/dCas9 activator system can efficiently ameliorate hallmarks of aging in a mouse model of Hutchinson-Gilford progeria syndrome (HGPS). We observed that the dCas9-Oct4 activator induced epigenetic remodeling, as evidenced by increased H3K9me3 and decreased H4K20me3 levels, without tumorization. Moreover, the progerin accumulation in HGPS aorta was significantly suppressed by the dCas9 activator-mediated Oct4 induction. Importantly, CRISPR/dCas9-activated Oct4 expression rescued the HGPS-associated vascular pathological features and lifespan shortening in the mouse model. These results suggest that partial rejuvenation via CRISPR/dCas9-mediated Oct4 activation can be used as a novel strategy in treating geriatric diseases.
Collapse
Affiliation(s)
- Junyeop Kim
- Laboratory of Stem Cells & Cell reprogramming, Department of Chemistry, Dongguk University, 100-715, Seoul, Korea
| | - Yerim Hwang
- Laboratory of Stem Cells & Cell reprogramming, Department of Chemistry, Dongguk University, 100-715, Seoul, Korea
| | - Sumin Kim
- Laboratory of Stem Cells & Cell reprogramming, Department of Chemistry, Dongguk University, 100-715, Seoul, Korea
| | - Yujung Chang
- Laboratory of Stem Cells & Cell reprogramming, Department of Chemistry, Dongguk University, 100-715, Seoul, Korea
| | - Yunkyung Kim
- Laboratory of Stem Cells & Cell reprogramming, Department of Chemistry, Dongguk University, 100-715, Seoul, Korea
| | - Youngeun Kwon
- Laboratory of Protein Engineering, Department of Biomedical Engineering, Dongguk University, 04620, Seoul, Korea
| | - Jongpil Kim
- Laboratory of Stem Cells & Cell reprogramming, Department of Chemistry, Dongguk University, 100-715, Seoul, Korea
| |
Collapse
|
36
|
Clinical Spectrum of LMNA-Associated Type 2 Familial Partial Lipodystrophy: A Systematic Review. Cells 2023; 12:cells12050725. [PMID: 36899861 PMCID: PMC10000975 DOI: 10.3390/cells12050725] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/03/2023] Open
Abstract
Type 2 familial partial lipodystrophy (FPLD2) is a laminopathic lipodystrophy due to pathogenic variants in the LMNA gene. Its rarity implies that it is not well-known. The aim of this review was to explore the published data regarding the clinical characterisation of this syndrome in order to better describe FPLD2. For this purpose, a systematic review through a search on PubMed until December 2022 was conducted and the references of the retrieved articles were also screened. A total of 113 articles were included. FPLD2 is characterised by the loss of fat starting around puberty in women, affecting limbs and trunk, and its accumulation in the face, neck and abdominal viscera. This adipose tissue dysfunction conditions the development of metabolic complications associated with insulin resistance, such as diabetes, dyslipidaemia, fatty liver disease, cardiovascular disease, and reproductive disorders. However, a great degree of phenotypical variability has been described. Therapeutic approaches are directed towards the associated comorbidities, and recent treatment modalities have been explored. A comprehensive comparison between FPLD2 and other FPLD subtypes can also be found in the present review. This review aimed to contribute towards augmenting knowledge of the natural history of FPLD2 by bringing together the main clinical research in this field.
Collapse
|
37
|
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: 2.0] [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
|
38
|
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
|
39
|
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: 3.0] [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
|
40
|
Mitochondrial Dysfunction and Oxidative Stress in Hereditary Ectopic Calcification Diseases. Int J Mol Sci 2022; 23:ijms232315288. [PMID: 36499615 PMCID: PMC9738718 DOI: 10.3390/ijms232315288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 11/26/2022] [Accepted: 12/01/2022] [Indexed: 12/08/2022] Open
Abstract
Ectopic calcification (EC) is characterized by an abnormal deposition of calcium phosphate crystals in soft tissues such as blood vessels, skin, and brain parenchyma. EC contributes to significant morbidity and mortality and is considered a major health problem for which no effective treatments currently exist. In recent years, growing emphasis has been placed on the role of mitochondrial dysfunction and oxidative stress in the pathogenesis of EC. Impaired mitochondrial respiration and increased levels of reactive oxygen species can be directly linked to key molecular pathways involved in EC such as adenosine triphosphate homeostasis, DNA damage signaling, and apoptosis. While EC is mainly encountered in common diseases such as diabetes mellitus and chronic kidney disease, studies in rare hereditary EC disorders such as pseudoxanthoma elasticum or Hutchinson-Gilford progeria syndrome have been instrumental in identifying the precise etiopathogenetic mechanisms leading to EC. In this narrative review, we describe the current state of the art regarding the role of mitochondrial dysfunction and oxidative stress in hereditary EC diseases. In-depth knowledge of aberrant mitochondrial metabolism and its local and systemic consequences will benefit the research into novel therapies for both rare and common EC disorders.
Collapse
|
41
|
Monnerat G, Kasai-Brunswick TH, Asensi KD, Silva dos Santos D, Barbosa RAQ, Cristina Paccola Mesquita F, Calvancanti Albuquerque JP, Raphaela PF, Wendt C, Miranda K, Domont GB, Nogueira FCS, Bastos Carvalho A, Campos de Carvalho AC. Modelling premature cardiac aging with induced pluripotent stem cells from a hutchinson-gilford Progeria Syndrome patient. Front Physiol 2022; 13:1007418. [PMID: 36505085 PMCID: PMC9726722 DOI: 10.3389/fphys.2022.1007418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare genetic disorder that causes accelerated aging and a high risk of cardiovascular complications. However, the underlying mechanisms of cardiac complications of this syndrome are not fully understood. This study modeled HGPS using cardiomyocytes (CM) derived from induced pluripotent stem cells (iPSC) derived from a patient with HGPS and characterized the biophysical, morphological, and molecular changes found in these CM compared to CM derived from a healthy donor. Electrophysiological recordings suggest that the HGPS-CM was functional and had normal electrophysiological properties. Electron tomography showed nuclear morphology alteration, and the 3D reconstruction of electron tomography images suggests structural abnormalities in HGPS-CM mitochondria, however, there was no difference in mitochondrial content as measured by Mitotracker. Immunofluorescence indicates nuclear morphological alteration and confirms the presence of Troponin T. Telomere length was measured using qRT-PCR, and no difference was found in the CM from HGPS when compared to the control. Proteomic analysis was carried out in a high-resolution system using Liquid Chromatography Tandem Mass Spectrometry (LC-MS/MS). The proteomics data show distinct group separations and protein expression differences between HGPS and control-CM, highlighting changes in ribosomal, TCA cycle, and amino acid biosynthesis, among other modifications. Our findings show that iPSC-derived cardiomyocytes from a Progeria Syndrome patient have significant changes in mitochondrial morphology and protein expression, implying novel mechanisms underlying premature cardiac aging.
Collapse
Affiliation(s)
- Gustavo Monnerat
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil,Laboratory of Proteomics, LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tais Hanae Kasai-Brunswick
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil,National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karina Dutra Asensi
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danubia Silva dos Santos
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | - Pires Ferreira Raphaela
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Camila Wendt
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kildare Miranda
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilberto Barbosa Domont
- Proteomic Unit, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio César Sousa Nogueira
- Laboratory of Proteomics, LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil,Proteomic Unit, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriana Bastos Carvalho
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Antonio Carlos Campos de Carvalho
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil,National Science and Technology Institute in Regenerative Medicine, Rio de Janeiro, Brazil,*Correspondence: Antonio Carlos Campos de Carvalho,
| |
Collapse
|
42
|
Ghosh DK, Pande S, Kumar J, Yesodharan D, Nampoothiri S, Radhakrishnan P, Reddy CG, Ranjan A, Girisha KM. The E262K mutation in Lamin A links nuclear proteostasis imbalance to laminopathy-associated premature aging. Aging Cell 2022; 21:e13688. [PMID: 36225129 PMCID: PMC9649601 DOI: 10.1111/acel.13688] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/09/2022] [Accepted: 07/25/2022] [Indexed: 01/25/2023] Open
Abstract
Deleterious, mostly de novo, mutations in the lamin A (LMNA) gene cause spatio-functional nuclear abnormalities that result in several laminopathy-associated progeroid conditions. In this study, exome sequencing in a sixteen-year-old male with manifestations of premature aging led to the identification of a mutation, c.784G>A, in LMNA, resulting in a missense protein variant, p.Glu262Lys (E262K), that aggregates in nucleoplasm. While bioinformatic analyses reveal the instability and pathogenicity of LMNAE262K , local unfolding of the mutation-harboring helical region drives the structural collapse of LMNAE262K into aggregates. The E262K mutation also disrupts SUMOylation of lysine residues by preventing UBE2I binding to LMNAE262K , thereby reducing LMNAE262K degradation, aggregated LMNAE262K sequesters nuclear chaperones, proteasomal proteins, and DNA repair proteins. Consequently, aggregates of LMNAE262K disrupt nuclear proteostasis and DNA repair response. Thus, we report a structure-function association of mutant LMNAE262K with toxicity, which is consistent with the concept that loss of nuclear proteostasis causes early aging in laminopathies.
Collapse
Affiliation(s)
- Debasish Kumar Ghosh
- Department of Medical Genetics, Manipal Academy of Higher Education, Kasturba Medical College, Manipal, Manipal, India
| | - Shruti Pande
- Department of Medical Genetics, Manipal Academy of Higher Education, Kasturba Medical College, Manipal, Manipal, India
| | - Jeevan Kumar
- Department of Medical Genetics, Manipal Academy of Higher Education, Kasturba Medical College, Manipal, Manipal, India
| | - Dhanya Yesodharan
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences & Research Centre, Cochin, India
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences & Research Centre, Cochin, India
| | - Periyasamy Radhakrishnan
- Suma Genomics Private Limited, Manipal Center for Biotherapeutics Research and Department of Reproductive Science, Manipal Academy of Higher Education, Manipal, India
| | - Chilakala Gangi Reddy
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Akash Ranjan
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, India
| | - Katta M Girisha
- Department of Medical Genetics, Manipal Academy of Higher Education, Kasturba Medical College, Manipal, Manipal, India
| |
Collapse
|
43
|
Inesta-Vaquera F, Weiland F, Henderson CJ, Wolf CR. In vivo stress reporters as early biomarkers of the cellular changes associated with progeria. J Cell Mol Med 2022; 26:5463-5472. [PMID: 36201626 PMCID: PMC9639039 DOI: 10.1111/jcmm.17574] [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: 06/28/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022] Open
Abstract
Age‐related diseases account for a high proportion of the total global burden of disease. Despite recent advances in understanding their molecular basis, there is a lack of suitable early biomarkers to test selected compounds and accelerate their translation to clinical trials. We have investigated the utility of in vivo stress reporter systems as surrogate early biomarkers of the degenerative disease progression. We hypothesized that cellular stress observed in models of human degenerative disease preceded overt cellular damage and at the same time will identify potential cytoprotective pathways. To test this hypothesis, we generated novel accelerated ageing (progeria) reporter mice by crossing the LmnaG609G mice into our oxidative stress/inflammation (Hmox1) and DNA damage (p21) stress reporter models. Histological analysis of reporter expression demonstrated a time‐dependent and tissue‐specific activation of the reporters in tissues directly associated with Progeria, including smooth muscle cells, the vasculature and gastrointestinal tract. Importantly, reporter expression was detected prior to any perceptible deleterious phenotype. Reporter expression can therefore be used as an early marker of progeria pathogenesis and to test therapeutic interventions. This work also demonstrates the potential to use stress reporter approaches to study and find new treatments for other degenerative diseases.
Collapse
Affiliation(s)
- Francisco Inesta-Vaquera
- Division of Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, UK
| | - Florian Weiland
- Department of Microbial and Molecular Systems (M2S), Centre for Food and Microbial Technology (CLMT), Laboratory of Enzyme, Fermentation and Brewing Technology (EFBT), Technology Campus Ghent, Ghent, Belgium
| | - Colin J Henderson
- Division of Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, UK
| | - Charles Roland Wolf
- Division of Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, UK
| |
Collapse
|
44
|
Chen X, Yao H, Andrés V, Bergo MO, Kashif M. Status of treatment strategies for Hutchinson-Gilford progeria syndrome with a focus on prelamin: A posttranslational modification. Basic Clin Pharmacol Toxicol 2022; 131:217-223. [PMID: 35790078 PMCID: PMC9795874 DOI: 10.1111/bcpt.13770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 12/30/2022]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder characterized by premature ageing and early death at a mean age of 14.7 years. At the molecular level, HGPS is caused by a de novo heterozygous mutation in LMNA, the gene encoding A-type lamins (mainly lamin A and C) and nuclear proteins, which have important cellular functions related to structure of the nuclear envelope. The LMNA mutation leads to the synthesis of a truncated prelamin A protein (called progerin), which cannot undergo normal processing to mature lamin A. In normal cells, prelamin A processing involves four posttranslational processing steps catalysed by four different enzymes. In HGPS cells, progerin accumulates as a farnesylated and methylated intermediate in the nuclear envelope where it is toxic and causes nuclear shape abnormalities and senescence. Numerous efforts have been made to target and reduce the toxicity of progerin, eliminate its synthesis and enhance its degradation, but as of today, only the use of farnesyltransferase inhibitors is approved for clinical use in HGPS patients. Here, we review the main current strategies that are being evaluated for treating HGPS, and we focus on efforts to target the posttranslational processing of progerin.
Collapse
Affiliation(s)
- Xue Chen
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina,Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Haidong Yao
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)MadridSpain,CIBER de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Martin O. Bergo
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Muhammad Kashif
- Center for Hematology and Regenerative Medicine, Department of Medicine, HuddingeKarolinska InstitutetHuddingeSweden
| |
Collapse
|
45
|
Caliskan A, Crouch SAW, Giddins S, Dandekar T, Dangwal S. Progeria and Aging-Omics Based Comparative Analysis. Biomedicines 2022; 10:2440. [PMID: 36289702 PMCID: PMC9599154 DOI: 10.3390/biomedicines10102440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 10/21/2023] Open
Abstract
Since ancient times aging has also been regarded as a disease, and humankind has always strived to extend the natural lifespan. Analyzing the genes involved in aging and disease allows for finding important indicators and biological markers for pathologies and possible therapeutic targets. An example of the use of omics technologies is the research regarding aging and the rare and fatal premature aging syndrome progeria (Hutchinson-Gilford progeria syndrome, HGPS). In our study, we focused on the in silico analysis of differentially expressed genes (DEGs) in progeria and aging, using a publicly available RNA-Seq dataset (GEO dataset GSE113957) and a variety of bioinformatics tools. Despite the GSE113957 RNA-Seq dataset being well-known and frequently analyzed, the RNA-Seq data shared by Fleischer et al. is far from exhausted and reusing and repurposing the data still reveals new insights. By analyzing the literature citing the use of the dataset and subsequently conducting a comparative analysis comparing the RNA-Seq data analyses of different subsets of the dataset (healthy children, nonagenarians and progeria patients), we identified several genes involved in both natural aging and progeria (KRT8, KRT18, ACKR4, CCL2, UCP2, ADAMTS15, ACTN4P1, WNT16, IGFBP2). Further analyzing these genes and the pathways involved indicated their possible roles in aging, suggesting the need for further in vitro and in vivo research. In this paper, we (1) compare "normal aging" (nonagenarians vs. healthy children) and progeria (HGPS patients vs. healthy children), (2) enlist genes possibly involved in both the natural aging process and progeria, including the first mention of IGFBP2 in progeria, (3) predict miRNAs and interactomes for WNT16 (hsa-mir-181a-5p), UCP2 (hsa-mir-26a-5p and hsa-mir-124-3p), and IGFBP2 (hsa-mir-124-3p, hsa-mir-126-3p, and hsa-mir-27b-3p), (4) demonstrate the compatibility of well-established R packages for RNA-Seq analysis for researchers interested but not yet familiar with this kind of analysis, and (5) present comparative proteomics analyses to show an association between our RNA-Seq data analyses and corresponding changes in protein expression.
Collapse
Affiliation(s)
- Aylin Caliskan
- Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Samantha A. W. Crouch
- Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Sara Giddins
- Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Seema Dangwal
- Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
46
|
Pang JKS, Chia S, Zhang J, Szyniarowski P, Stewart C, Yang H, Chan WK, Ng SY, Soh BS. Characterizing arrhythmia using machine learning analysis of Ca 2+ cycling in human cardiomyocytes. Stem Cell Reports 2022; 17:1810-1823. [PMID: 35839773 PMCID: PMC9391413 DOI: 10.1016/j.stemcr.2022.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/18/2022] Open
Abstract
Accurate modeling of the heart electrophysiology to predict arrhythmia susceptibility remains a challenge. Current electrophysiological analyses are hypothesis-driven models drawing conclusions from changes in a small subset of electrophysiological parameters because of the difficulty of handling and understanding large datasets. Thus, we develop a framework to train machine learning classifiers to distinguish between healthy and arrhythmic cardiomyocytes using their calcium cycling properties. By training machine learning classifiers on a generated dataset containing a total of 3,003 healthy derived cardiomyocytes and their various arrhythmic states, the multi-class models achieved >90% accuracy in predicting arrhythmia presence and type. We also demonstrate that a binary classifier trained to distinguish cardiotoxic arrhythmia from healthy electrophysiology could determine the key biological changes associated with that specific arrhythmia. Therefore, machine learning algorithms can be used to characterize underlying arrhythmic patterns in samples to improve in vitro preclinical models and complement current in vivo systems. Calcium reporter enabled analysis of hPSC-derived CM electrophysiology Calcium electrophysiology can be decomposed to train machine learning classifiers Multi-class classifier distinguishes different forms of arrhythmias Binary-class classifiers identify key electrophysiological changes a priori
Collapse
Affiliation(s)
- Jeremy K S Pang
- Disease Modeling and Therapeutics Laboratory, A(∗)STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore 138673, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Sabrina Chia
- Disease Modeling and Therapeutics Laboratory, A(∗)STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore 138673, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Jinqiu Zhang
- A(∗)STAR Skin Research Labs, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Piotr Szyniarowski
- A(∗)STAR Skin Research Labs, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Colin Stewart
- A(∗)STAR Skin Research Labs, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Woon-Khiong Chan
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Shi Yan Ng
- Neurotherapeutics Laboratory, A(∗)STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore 138673, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore; National Neuroscience Institute, Singapore 308433, Singapore
| | - Boon-Seng Soh
- Disease Modeling and Therapeutics Laboratory, A(∗)STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore 138673, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore.
| |
Collapse
|
47
|
Salvador J, Iruela-Arispe ML. Nuclear Mechanosensation and Mechanotransduction in Vascular Cells. Front Cell Dev Biol 2022; 10:905927. [PMID: 35784481 PMCID: PMC9247619 DOI: 10.3389/fcell.2022.905927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Vascular cells are constantly subjected to physical forces associated with the rhythmic activities of the heart, which combined with the individual geometry of vessels further imposes oscillatory, turbulent, or laminar shear stresses on vascular cells. These hemodynamic forces play an important role in regulating the transcriptional program and phenotype of endothelial and smooth muscle cells in different regions of the vascular tree. Within the aorta, the lesser curvature of the arch is characterized by disturbed, oscillatory flow. There, endothelial cells become activated, adopting pro-inflammatory and athero-prone phenotypes. This contrasts the descending aorta where flow is laminar and endothelial cells maintain a quiescent and atheroprotective phenotype. While still unclear, the specific mechanisms involved in mechanosensing flow patterns and their molecular mechanotransduction directly impact the nucleus with consequences to transcriptional and epigenetic states. The linker of nucleoskeleton and cytoskeleton (LINC) protein complex transmits both internal and external forces, including shear stress, through the cytoskeleton to the nucleus. These forces can ultimately lead to changes in nuclear integrity, chromatin organization, and gene expression that significantly impact emergence of pathology such as the high incidence of atherosclerosis in progeria. Therefore, there is strong motivation to understand how endothelial nuclei can sense and respond to physical signals and how abnormal responses to mechanical cues can lead to disease. Here, we review the evidence for a critical role of the nucleus as a mechanosensor and the importance of maintaining nuclear integrity in response to continuous biophysical forces, specifically shear stress, for proper vascular function and stability.
Collapse
Affiliation(s)
| | - M. Luisa Iruela-Arispe
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
48
|
Meqbel BRM, Gomes M, Omer A, Gallouzi IE, Horn HF. LINCing Senescence and Nuclear Envelope Changes. Cells 2022; 11:1787. [PMID: 35681483 PMCID: PMC9179861 DOI: 10.3390/cells11111787] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 01/27/2023] Open
Abstract
The nuclear envelope (NE) has emerged as a nexus for cellular organization, signaling, and survival. Beyond its role as a barrier to separate the nucleoplasm from the cytoplasm, the NE's role in supporting and maintaining a myriad of other functions has made it a target of study in many cellular processes, including senescence. The nucleus undergoes dramatic changes in senescence, many of which are driven by changes in the NE. Indeed, Lamin B1, a key NE protein that is consistently downregulated in senescence, has become a marker for senescence. Other NE proteins have also been shown to play a role in senescence, including LINC (linker of nucleoskeleton and cytoskeleton) complex proteins. LINC complexes span the NE, forming physical connections between the cytoplasm to the nucleoplasm. In this way, they integrate nuclear and cytoplasmic mechanical signals and are essential not only for a variety of cellular functions but are needed for cell survival. However, LINC complex proteins have been shown to have a myriad of functions in addition to forming a LINC complex, often existing as nucleoplasmic or cytoplasmic soluble proteins in a variety of isoforms. Some of these proteins have now been shown to play important roles in DNA repair, cell signaling, and nuclear shape regulation, all of which are important in senescence. This review will focus on some of these roles and highlight the importance of LINC complex proteins in senescence.
Collapse
Affiliation(s)
- Bakhita R. M. Meqbel
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar;
| | - Matilde Gomes
- KAUST Smart-Health Initiative and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Jeddah 21589, Saudi Arabia; (M.G.); (I.E.G.)
| | - Amr Omer
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada;
| | - Imed E. Gallouzi
- KAUST Smart-Health Initiative and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Jeddah 21589, Saudi Arabia; (M.G.); (I.E.G.)
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada;
| | - Henning F. Horn
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar;
| |
Collapse
|
49
|
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
|
50
|
Ribeiro R, Macedo JC, Costa M, Ustiyan V, Shindyapina AV, Tyshkovskiy A, Gomes RN, Castro JP, Kalin TV, Vasques-Nóvoa F, Nascimento DS, Dmitriev SE, Gladyshev VN, Kalinichenko VV, Logarinho E. In vivo cyclic induction of the FOXM1 transcription factor delays natural and progeroid aging phenotypes and extends healthspan. NATURE AGING 2022; 2:397-411. [PMID: 37118067 DOI: 10.1038/s43587-022-00209-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/15/2022] [Indexed: 04/30/2023]
Abstract
The FOXM1 transcription factor exhibits pleiotropic C-terminal transcriptional and N-terminal non-transcriptional functions in various biological processes critical for cellular homeostasis. We previously found that FOXM1 repression during cellular aging underlies the senescence phenotypes, which were vastly restored by overexpressing transcriptionally active FOXM1. Yet, it remains unknown whether increased expression of FOXM1 can delay organismal aging. Here, we show that in vivo cyclic induction of an N-terminal truncated FOXM1 transgene on progeroid and naturally aged mice offsets aging-associated repression of full-length endogenous Foxm1, reinstating both transcriptional and non-transcriptional functions. This translated into mitigation of several cellular aging hallmarks, as well as molecular and histopathological progeroid features of the short-lived Hutchison-Gilford progeria mouse model, significantly extending its lifespan. FOXM1 transgene induction also reinstated endogenous Foxm1 levels in naturally aged mice, delaying aging phenotypes while extending their lifespan. Thus, we disclose that FOXM1 genetic rewiring can delay senescence-associated progeroid and natural aging pathologies.
Collapse
Affiliation(s)
- Rui Ribeiro
- Aging and Aneuploidy Laboratory, i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Graduate Program in Areas of Basic and Applied Biology (GABBA), ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Joana C Macedo
- Aging and Aneuploidy Laboratory, i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Madalena Costa
- Anatomy Department, Unit for Multidisciplinary Biomedical Research, ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Vladimir Ustiyan
- Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anastasia V Shindyapina
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Rita N Gomes
- INEB - Instituto Nacional de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - José Pedro Castro
- Aging and Aneuploidy Laboratory, i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Tanya V Kalin
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Francisco Vasques-Nóvoa
- INEB - Instituto Nacional de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Cardiovascular Research and Development Center, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Diana S Nascimento
- INEB - Instituto Nacional de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sergey E Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Elsa Logarinho
- Aging and Aneuploidy Laboratory, i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
| |
Collapse
|