Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (38)

Search Parameters:
Keywords = progerin

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 2988 KB  
Article
Microhand Platform Equipped with Plate-Shaped End-Effectors Enables Precise Probing of Intracellular Structure Contribution to Whole-Cell Mechanical Properties
by Masahiro Kawakami, Masaru Kojima, Toshihiko Ogura, Atsushi Kubo, Tatsuo Arai and Shinji Sakai
Micromachines 2025, 16(11), 1272; https://doi.org/10.3390/mi16111272 - 12 Nov 2025
Abstract
Cellular mechanical properties are critical indicators of cellular state and promising disease biomarkers. This study introduces a novel microhand system, featuring chopstick-like plate-shaped end-effectors, designed for stable and high-precision single-cell mechanical characterization. First, we automated the force sensor calibration to overcome the inefficiency [...] Read more.
Cellular mechanical properties are critical indicators of cellular state and promising disease biomarkers. This study introduces a novel microhand system, featuring chopstick-like plate-shaped end-effectors, designed for stable and high-precision single-cell mechanical characterization. First, we automated the force sensor calibration to overcome the inefficiency and unreliability of conventional manual methods. To validate the system’s sensitivity, we precisely quantified the mechanical contributions of subcellular components, such as the actin cytoskeleton and chromatin, by measuring stiffness reductions after treatment with Cytochalasin D and Trichostatin A, respectively. Notably, when applied to a cellular model of Hutchinson–Gilford progeria syndrome, we successfully captured disease-induced mechanical alterations. A distinct population of high-stiffness cells was detected in progerin-overexpressing cells, a feature not observed in the control groups. Furthermore, by controlling the indentation speed and depth, the mechanical properties of the cytoplasm and nucleus could be distinctly evaluated. These results demonstrate that our microhand system is a highly sensitive and robust platform, capable of detecting subtle, disease-related changes and elucidating the contributions of specific subcellular structures to cell mechanics. Full article
(This article belongs to the Special Issue Next-Generation Biomedical Devices)
Show Figures

Figure 1

12 pages, 3335 KB  
Article
Analysis of Beta-Dystroglycan in Different Cell Models of Senescence
by Guadalupe Elizabeth Jimenez-Gutierrez, Tania Ivette Zavaleta-Vásquez, Jessica Alexandra Lizcano-Meneses, Ian Alain Garcia-Aguirre, Marco Samuel Laredo-Cisneros, Jonathan J. Magaña, Steve J Winder, Joaquín Cordero-Martínez and Bulmaro Cisneros
Int. J. Mol. Sci. 2025, 26(16), 7726; https://doi.org/10.3390/ijms26167726 - 10 Aug 2025
Viewed by 683
Abstract
The functional diversity of β-dystroglycan is attributable to its dual distribution, the plasma membrane, and the nucleus. In the plasma membrane, β-DG is a component of the dystrophin-associated protein complex. In the nucleus, β-DG assembles with the nuclear lamina and emerin. Recent findings [...] Read more.
The functional diversity of β-dystroglycan is attributable to its dual distribution, the plasma membrane, and the nucleus. In the plasma membrane, β-DG is a component of the dystrophin-associated protein complex. In the nucleus, β-DG assembles with the nuclear lamina and emerin. Recent findings indicate a role for β-DG in senescence, as its knockout in C2C12 myoblasts induces genomic instability and promotes the senescent state. This study analyzed the behavior of β-DG in three distinct models of senescence: chronologically aged fibroblasts, sodium butyrate (NaBu)-induced senescent fibroblasts, and fibroblasts from a Hutchinson–Gilford progeria syndrome (HGPS) patient. β-DG was found mainly in the nucleus in all the senescent cell types, with a certain mislocalization to the cytoplasm in HGPS and NaBu-treated fibroblasts. Furthermore, the full-length β-DG (43 kDa) and the cleaved intracellular domain (ICD; ~26 kDa) were identified. The ICD level increased in aged fibroblasts, but its yield was poor or virtually nonexistent in NaBU-induced and HGPS fibroblasts, respectively. Remarkably, β-DG was sequestered by progerin in HGPS cells, hindering its interaction with lamin A. In summary, the observed alterations in β-DG may be associated with the senescent state, and such findings will serve for future studies aimed at elucidating its role in senescence. Full article
(This article belongs to the Special Issue Molecular Mechanisms of the Aging Process)
Show Figures

Figure 1

23 pages, 5695 KB  
Article
Impact of miR-181a on SIRT1 Expression and Senescence in Hutchinson–Gilford Progeria Syndrome
by Eva-Maria Lederer, Felix Quirin Fenzl, Peter Krüger, Moritz Schroll, Ramona Hartinger and Karima Djabali
Diseases 2025, 13(8), 245; https://doi.org/10.3390/diseases13080245 - 4 Aug 2025
Viewed by 702
Abstract
Background/Objectives: Hutchinson–Gilford progeria syndrome (HGPS) is a rare and fatal genetic disease caused by a silent mutation in the LMNA gene, leading to the production of progerin, a defective prelamin A variant. Progerin accumulation disrupts nuclear integrity, alters chromatin organization, and drives systemic [...] Read more.
Background/Objectives: Hutchinson–Gilford progeria syndrome (HGPS) is a rare and fatal genetic disease caused by a silent mutation in the LMNA gene, leading to the production of progerin, a defective prelamin A variant. Progerin accumulation disrupts nuclear integrity, alters chromatin organization, and drives systemic cellular dysfunction. While autophagy and inflammation are key dysregulated pathways in HGPS, the role of microRNAs (miRNAs) in these processes remains poorly understood. Methods: We performed an extensive literature review to identify miRNAs involved in autophagy and inflammation. Through stem-loop RT-qPCR in aging HGPS and control fibroblast strains, we identified significant miRNAs and focused on the most prominent one, miR-181a-5p, for in-depth analysis. We validated our in vitro findings with miRNA expression studies in skin biopsies from an HGPS mouse model and conducted functional assays in human fibroblasts, including immunofluorescence staining, β-Galactosidase assay, qPCR, and Western blot analysis. Transfection studies were performed using an miR-181a-5p mimic and its inhibitor. Results: We identified miR-181a-5p as a critical regulator of premature senescence in HGPS. miR-181a-5p was significantly upregulated in HGPS fibroblasts and an HGPS mouse model, correlating with Sirtuin 1 (SIRT1) suppression and induction of senescence. Additionally, we demonstrated that TGFβ1 induced miR-181a-5p expression, linking inflammation to miRNA-mediated senescence. Inhibiting miR-181a-5p restored SIRT1 levels, increased proliferation, and alleviated senescence in HGPS fibroblasts, supporting its functional relevance in disease progression. Conclusions: These findings highlight the important role of miR-181a-5p in premature aging and suggest its potential as a therapeutic target for modulating senescence in progeroid syndromes. Full article
(This article belongs to the Section Rare Syndrome)
Show Figures

Figure 1

23 pages, 2645 KB  
Article
Baricitinib and Lonafarnib Synergistically Target Progerin and Inflammation, Improving Lifespan and Health in Progeria Mice
by Peter Krüger, Moritz Schroll, Felix Quirin Fenzl, Ramona Hartinger, Eva-Maria Lederer, Agnes Görlach, Leslie B. Gordon, Paola Cavalcante, Nicola Iacomino, Birgit Rathkolb, Juan Antonio Aguilar Pimentel, Manuela Östereicher, Nadine Spielmann, Cordula Maria Wolf, Martin Hrabe de Angelis and Karima Djabali
Int. J. Mol. Sci. 2025, 26(10), 4849; https://doi.org/10.3390/ijms26104849 - 19 May 2025
Cited by 2 | Viewed by 2325
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a rare, fatal, and premature aging disorder caused by progerin, a truncated form of lamin A that disrupts nuclear architecture, induces systemic inflammation, and accelerates senescence. While the farnesyltransferase inhibitor lonafarnib extends the lifespan by limiting progerin farnesylation, [...] Read more.
Hutchinson–Gilford progeria syndrome (HGPS) is a rare, fatal, and premature aging disorder caused by progerin, a truncated form of lamin A that disrupts nuclear architecture, induces systemic inflammation, and accelerates senescence. While the farnesyltransferase inhibitor lonafarnib extends the lifespan by limiting progerin farnesylation, it does not address the chronic inflammation or the senescence-associated secretory phenotype (SASP), which worsens disease progression. In this study, we investigated the combined effects of baricitinib (BAR), a JAK1/2 inhibitor, and lonafarnib (FTI) in a LmnaG609G/G609G mouse model of HGPS. BAR + FTI therapy synergistically extended the lifespan by 25%, surpassing the effects of either monotherapy. Treated mice showed improved health, as evidenced by reduced kyphosis, better fur quality, decreased incidence of cataracts, and less severe dysgnathia. Histological analyses indicated reduced fibrosis in the dermal, hepatic, and muscular tissues, restored cellularity and thickness in the aortic media, and improved muscle fiber integrity. Mechanistically, BAR decreased the SASP and inflammatory markers (e.g., IL-6 and PAI-1), complementing the progerin-targeting effects of FTI. This preclinical study demonstrates the synergistic potential of BAR + FTI therapy in addressing HGPS systemic and tissue-specific pathologies, offering a promising strategy for enhancing both lifespan and health. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

12 pages, 1284 KB  
Case Report
Mesenchymal Stem Cell Therapy for Hutchinson–Gilford Progeria: Improvements in Arterial Stiffness and Bone Mineral Density in a Single Case
by Eun-Young Joo, Ji-Sun Park, Hyun-Tae Shin, Myungji Yoo, Su-Jin Kim, Ji-Eun Lee and Gwang-Seong Choi
Children 2025, 12(4), 523; https://doi.org/10.3390/children12040523 - 18 Apr 2025
Cited by 1 | Viewed by 1636
Abstract
Background/Objectives: Hutchinson–Gilford progeria syndrome (HGPS) is a rare genetic disorder that cause premature aging due to LMNA mutations and progerin accumulation. Although lonafarnib, an FDA-approved farnesyltransferase inhibitor, offers modest extension of life, the disease remains progressive. As progeria is associated with stem cell [...] Read more.
Background/Objectives: Hutchinson–Gilford progeria syndrome (HGPS) is a rare genetic disorder that cause premature aging due to LMNA mutations and progerin accumulation. Although lonafarnib, an FDA-approved farnesyltransferase inhibitor, offers modest extension of life, the disease remains progressive. As progeria is associated with stem cell depletion and mesenchymal stem cell (MSC) therapy has shown efficacy in treating atherosclerosis, we aimed to evaluate its efficacy and safety in HGPS. Methods: A 7-year-old male with classic HGPS and preexisting severe cerebrovascular disease received four intravenous infusion of bone marrow-derived MSCs (2.5 × 10⁵ cells/kg) over 8 months. Growth, metabolic, cardiovascular, musculoskeletal, auditory, and inflammatory cytokines were monitored throughout the study. Prophylactic enoxaparin was administered to prevent vascular complications. Results: MSC therapy was associated with improved lean body mass (11.5%), bone mineral density (L-spine z-score: 0.55 → 2.03), reduced arterial stiffness (9.98% reductionin pulse wave velocity), joint range of motion, dentition, and decreased sICAM-1 levels. However, Cardiovascular deterioration continued, and the patient passed away 10 months after the fourth dose, likely due to progression of the underlying vascular disease. No severe adverse effects were attributed to MSC therapy. Conclusions: MSC therapy may offer short-term benefits in arterial stiffness, bone health and inflammation in HGPS without notable safety concerns. Further studies are warranted to validate these findings, explore earlier intervention, and determine long-term efficacy and optimal dosing strategies. Full article
(This article belongs to the Section Translational Pediatrics)
Show Figures

Figure 1

15 pages, 1860 KB  
Article
Progerin mRNA Is Associated with Smoking and Signs of Increased Microvascular Damage in Patients with Diabetic Macular Edema
by Marc-Michael Zaruba, Reinhard Angermann, Simon Staggl, Vivek Jeyakumar, Sofia Mair, Victoria Stöckl, Julia Neyer, Thomas Maurer, Maria Ungericht, Jasmina Gavranovic-Novakovic, Axel Bauer, Claus Zehetner and Moritz Messner
Int. J. Mol. Sci. 2025, 26(5), 2099; https://doi.org/10.3390/ijms26052099 - 27 Feb 2025
Viewed by 1402
Abstract
The premature aging disease Hutchinson–Gilford Syndrome (HGPS) is caused by defined mutations in the LMNA gene, resulting in the activation of a cryptic splice donor site, which leads to a defective truncated prelamin A protein called progerin. Notably, progerin expression has also been [...] Read more.
The premature aging disease Hutchinson–Gilford Syndrome (HGPS) is caused by defined mutations in the LMNA gene, resulting in the activation of a cryptic splice donor site, which leads to a defective truncated prelamin A protein called progerin. Notably, progerin expression has also been detected in non-mutated healthy individuals, and therefore, its involvement in the physiological aging process has been widely discussed. Since diabetes mellitus is associated with premature aging and increased cardiovascular mortality, we aimed to investigate the role of progerin expression in patients with diabetic retinopathy (DR). mRNA expression of progerin was analyzed in blood samples from 140 patients with DR who received anti-vascular endothelial growth factor (VEGF) therapy. Progerin mRNA levels were significantly lower in female compared to male patients (n = 42 vs. n = 98; 0.67 ± 0.19 vs. 0.89 ± 0.51, p = 0.006) and higher in patients with non-proliferative (NP)DR (n = 87 vs. n = 53; 0.9 ± 0.51 vs. 0.71 ± 0.29, p = 0.013) compared to those with proliferative (P)DR. Additionally, a positive correlation was found between progerin mRNA expression and the number of intravitreal anti-VEGF applications (n = 139, r = 0.21, p = 0.015), central macula thickness (CMT), (n = 137, r = 0.18, p = 0.036) and nicotine consumption (n = 105, r = 0.235, p = 0.002). The nuclear localization and significant upregulation of progerin mRNA and protein levels in dermal fibroblasts from HGPS donors emphasize its role in cellular aging mechanisms. Progerin mRNA levels were higher in patients with NPDR. CMT, number of intravitreal anti-VEGF therapy treatments, and cigarette consumption were positively related to progerin mRNA, suggesting an association with disease progression and premature aging. Full article
(This article belongs to the Special Issue Gene Regulation in Endocrine Disease)
Show Figures

Figure 1

19 pages, 1408 KB  
Review
Angiopoietin-2: A Therapeutic Target for Vascular Protection in Hutchinson–Gilford Progeria Syndrome
by Sahar Vakili and Kan Cao
Int. J. Mol. Sci. 2024, 25(24), 13537; https://doi.org/10.3390/ijms252413537 - 18 Dec 2024
Cited by 1 | Viewed by 2797
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a pediatric condition characterized by clinical features that resemble accelerated aging. The abnormal accumulation of a toxic form of the lamin A protein known as progerin disrupts cellular functions, leading to various complications, including growth retardation, loss of [...] Read more.
Hutchinson–Gilford progeria syndrome (HGPS) is a pediatric condition characterized by clinical features that resemble accelerated aging. The abnormal accumulation of a toxic form of the lamin A protein known as progerin disrupts cellular functions, leading to various complications, including growth retardation, loss of subcutaneous fat, abnormal skin, alopecia, osteoporosis, and progressive joint contractures. Death primarily occurs as the result of complications from progressive atherosclerosis, especially from cardiac disease, such as myocardial infarction or heart failure, or cerebrovascular disease like stroke. Despite the availability of lonafarnib, the only US Food and Drug Administration-approved treatment for HGPS, cardiovascular complications remain the leading cause of morbidity and mortality in affected patients. Defective angiogenesis—the process of forming new blood vessels from existing ones—plays a crucial role in the development of cardiovascular disease. A recent study suggests that Angiopoietin-2 (Ang2), a pro-angiogenic growth factor that regulates angiogenesis and vascular stability, may offer therapeutic potential for the treatment of HGPS. In this review, we describe the clinical features and key cellular processes impacted by progerin and discuss the therapeutic potential of Ang2 in addressing these challenges. Full article
Show Figures

Figure 1

23 pages, 20158 KB  
Article
Inflammation and Fibrosis in Progeria: Organ-Specific Responses in an HGPS Mouse Model
by Peter Krüger, Moritz Schroll, Felix Fenzl, Eva-Maria Lederer, Ramona Hartinger, Rouven Arnold, Deniz Cagla Togan, Runjia Guo, Shiyu Liu, Andreas Petry, Agnes Görlach and Karima Djabali
Int. J. Mol. Sci. 2024, 25(17), 9323; https://doi.org/10.3390/ijms25179323 - 28 Aug 2024
Cited by 9 | Viewed by 3781
Abstract
Hutchinson–Gilford Progeria Syndrome (HGPS) is an extremely rare genetic disorder that causes accelerated aging, due to a pathogenic variant in the LMNA gene. This pathogenic results in the production of progerin, a defective protein that disrupts the nuclear lamina’s structure. In our study, [...] Read more.
Hutchinson–Gilford Progeria Syndrome (HGPS) is an extremely rare genetic disorder that causes accelerated aging, due to a pathogenic variant in the LMNA gene. This pathogenic results in the production of progerin, a defective protein that disrupts the nuclear lamina’s structure. In our study, we conducted a histopathological analysis of various organs in the LmnaG609G/G609G mouse model, which is commonly used to study HGPS. The objective of this study was to show that progerin accumulation drives systemic but organ-specific tissue damage and accelerated aging phenotypes. Our findings show significant fibrosis, inflammation, and dysfunction in multiple organ systems, including the skin, cardiovascular system, muscles, lungs, liver, kidneys, spleen, thymus, and heart. Specifically, we observed severe vascular fibrosis, reduced muscle regeneration, lung tissue remodeling, depletion of fat in the liver, and disruptions in immune structures. These results underscore the systemic nature of the disease and suggest that chronic inflammation and fibrosis play crucial roles in the accelerated aging seen in HGPS. Additionally, our study highlights that each organ responds differently to the toxic effects of progerin, indicating that there are distinct mechanisms of tissue-specific damage. Full article
(This article belongs to the Special Issue Inflammation and Cellular Senescence in Age-Related Diseases)
Show Figures

Figure 1

15 pages, 7476 KB  
Article
Use of Farnesyl Transferase Inhibitors in an Ageing Model in Drosophila
by Annely Brandt, Roman Petrovsky, Maria Kriebel and Jörg Großhans
J. Dev. Biol. 2023, 11(4), 40; https://doi.org/10.3390/jdb11040040 - 29 Oct 2023
Cited by 3 | Viewed by 2692
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 [...] Read more.
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. Full article
Show Figures

Figure 1

17 pages, 737 KB  
Review
Progerin, an Aberrant Spliced Form of Lamin A, Is a Potential Therapeutic Target for HGPS
by Bae-Hoon Kim, Yeon-Ho Chung, Tae-Gyun Woo, So-Mi Kang, Soyoung Park and Bum-Joon Park
Cells 2023, 12(18), 2299; https://doi.org/10.3390/cells12182299 - 18 Sep 2023
Cited by 5 | Viewed by 5669
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is an extremely rare genetic disorder caused by the mutant protein progerin, which is expressed by the abnormal splicing of the LMNA gene. HGPS affects systemic levels, with the exception of cognition or brain development, in children, showing that [...] Read more.
Hutchinson–Gilford progeria syndrome (HGPS) is an extremely rare genetic disorder caused by the mutant protein progerin, which is expressed by the abnormal splicing of the LMNA gene. HGPS affects systemic levels, with the exception of cognition or brain development, in children, showing that cellular aging can occur in the short term. Studying progeria could be useful in unraveling the causes of human aging (as well as fatal age-related disorders). Elucidating the clear cause of HGPS or the development of a therapeutic medicine could improve the quality of life and extend the survival of patients. This review aimed to (i) briefly describe how progerin was discovered as the causative agent of HGPS, (ii) elucidate the puzzling observation of the absence of primary neurological disease in HGPS, (iii) present several studies showing the deleterious effects of progerin and the beneficial effects of its inhibition, and (iv) summarize research to develop a therapy for HGPS and introduce clinical trials for its treatment. Full article
(This article belongs to the Special Issue Advances in Treatments for Hutchinson-Gilford Progeria Syndrome)
Show Figures

Figure 1

22 pages, 19445 KB  
Article
Impact of Combined Baricitinib and FTI Treatment on Adipogenesis in Hutchinson–Gilford Progeria Syndrome and Other Lipodystrophic Laminopathies
by Ramona Hartinger, Eva-Maria Lederer, Elisa Schena, Giovanna Lattanzi and Karima Djabali
Cells 2023, 12(10), 1350; https://doi.org/10.3390/cells12101350 - 9 May 2023
Cited by 13 | Viewed by 3423
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 [...] Read more.
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. Full article
(This article belongs to the Collection Lamins and Laminopathies)
Show Figures

Figure 1

13 pages, 2001 KB  
Communication
Progerinin, an Inhibitor of Progerin, Alleviates Cardiac Abnormalities in a Model Mouse of Hutchinson–Gilford Progeria Syndrome
by So-mi Kang, Seungwoon Seo, Eun Ju Song, Okhee Kweon, Ah-hyeon Jo, Soyoung Park, Tae-Gyun Woo, Bae-Hoon Kim, Goo Taeg Oh and Bum-Joon Park
Cells 2023, 12(9), 1232; https://doi.org/10.3390/cells12091232 - 24 Apr 2023
Cited by 9 | Viewed by 3890
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 [...] Read more.
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. Full article
(This article belongs to the Special Issue Advances in Treatments for Hutchinson-Gilford Progeria Syndrome)
Show Figures

Figure 1

9 pages, 2420 KB  
Case Report
Acute Coronary Syndrome Treated with Percutaneous Coronary Intervention in Hutchinson–Gilford Progeria
by Luciano De Simone, Serena Chiellino, Gaia Spaziani, Giulio Porcedda, Giovan Battista Calabri, Sergio Berti, Silvia Favilli, Laura Stefani and Giuseppe Santoro
Children 2023, 10(3), 526; https://doi.org/10.3390/children10030526 - 8 Mar 2023
Cited by 1 | Viewed by 2619
Abstract
Hutchinson–Gilford progeria syndrome is an extremely rare genetic disease caused by a de novo mutation in the LMNA gene, leading to an accumulation of a form of Lamin A, called Progerin, which results in a typical phenotype and a marked decrease in life [...] Read more.
Hutchinson–Gilford progeria syndrome is an extremely rare genetic disease caused by a de novo mutation in the LMNA gene, leading to an accumulation of a form of Lamin A, called Progerin, which results in a typical phenotype and a marked decrease in life expectancy, due to early atherosclerosis and cardiovascular disease. We report the case of a fourteen-year-old Chinese boy with Hutchinson–Gilford progeria syndrome admitted to the emergency room because of precordial pain. Physical examination showed tachycardia 130 beats/min and arterial hypertension: 170/120 mmHg, normal respiratory rate, no neurological impairment; ECG evidenced sinus tachycardia, left ventricular hypertrophy, horizontal ST-segment depression in I, aVL, II, III, aVF leads, and V4–V6 and ST-segment elevation in aVR and V1 leads. Echocardiography highlighted preserved global left ventricular function with concentric hypertrophy, altered diastolic flow pattern, mitral valve insufficiency, and minimal aortic regurgitation. Blood tests evidenced an increase in high-sensitivity troponin T level (335 pg/mL). NSTEMI diagnosis was performed, and the patient was admitted to the intensive care unit. A coronary CT angiography showed a severe obstruction of the common trunk of the left coronary artery, for which an urgent percutaneous coronary intervention (PCI) was proposed. A selective coronary angiography imaged complete chronic occlusion of the left main coronary artery as well as severe stenosis at the origin of a very enlarged right coronary artery that vascularized the left coronary artery through collaterals. Afterwards, the right coronary artery was probed using an Amplatz right (AR1) guiding catheter, through which a large 3.5 mm drug-eluting coronary stent (Xience Sierra, Abbott, Abbott Park, IL, USA) was implanted. At the end of the procedure, no residual stenosis was imaged and improved vascularization of the left coronary artery distribution segments was observed. Dual antiplatelet therapy (DAPT) consisting of aspirin (75 mg daily) and clopidogrel (37.5 mg daily) and anti-hypertensive therapy were started. At the one-year follow-up, the patient had not reported any occurrence of anginal chest pain. Full article
(This article belongs to the Special Issue Cardiovascular Disease in Children)
Show Figures

Figure 1

24 pages, 1183 KB  
Review
The Molecular and Cellular Basis of Hutchinson–Gilford Progeria Syndrome and Potential Treatments
by Noelle J. Batista, Sanket G. Desai, Alexis M. Perez, Alexa Finkelstein, Rachel Radigan, Manrose Singh, Aaron Landman, Brian Drittel, Daniella Abramov, Mina Ahsan, Samantha Cornwell and Dong Zhang
Genes 2023, 14(3), 602; https://doi.org/10.3390/genes14030602 - 27 Feb 2023
Cited by 16 | Viewed by 11946
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a rare, autosomal-dominant, and fatal premature aging syndrome. HGPS is most often derived from a de novo point mutation in the LMNA gene, which results in an alternative splicing defect and the generation of the mutant protein, progerin. [...] Read more.
Hutchinson–Gilford progeria syndrome (HGPS) is a rare, autosomal-dominant, and fatal premature aging syndrome. HGPS is most often derived from a de novo point mutation in the LMNA gene, which results in an alternative splicing defect and the generation of the mutant protein, progerin. Progerin behaves in a dominant-negative fashion, leading to a variety of cellular and molecular changes, including nuclear abnormalities, defective DNA damage response (DDR) and DNA repair, and accelerated telomere attrition. Intriguingly, many of the manifestations of the HGPS cells are shared with normal aging cells. However, at a clinical level, HGPS does not fully match normal aging because of the accelerated nature of the phenotypes and its primary effects on connective tissues. Furthermore, the epigenetic changes in HGPS patients are of great interest and may play a crucial role in the pathogenesis of HGPS. Finally, various treatments for the HGPS patients have been developed in recent years with important effects at a cellular level, which translate to symptomatic improvement and increased lifespan. Full article
(This article belongs to the Special Issue DNA Replication/Repair, and the DNA Damage Response in Human Disease)
Show Figures

Figure 1

20 pages, 7115 KB  
Article
Rescue of Mitochondrial Function in Hutchinson-Gilford Progeria Syndrome by the Pharmacological Modulation of Exportin CRM1
by Feliciano Monterrubio-Ledezma, Fernando Navarro-García, Lourdes Massieu, Ricardo Mondragón-Flores, Luz Adriana Soto-Ponce, Jonathan J. Magaña and Bulmaro Cisneros
Cells 2023, 12(2), 275; https://doi.org/10.3390/cells12020275 - 10 Jan 2023
Cited by 12 | Viewed by 5111
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disorder caused by the expression of progerin, a mutant variant of Lamin A. Recently, HGPS studies have gained relevance because unraveling its underlying mechanism would help to understand physiological aging. We previously reported that [...] Read more.
Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disorder caused by the expression of progerin, a mutant variant of Lamin A. Recently, HGPS studies have gained relevance because unraveling its underlying mechanism would help to understand physiological aging. We previously reported that the CRM1-mediated nuclear protein export pathway is exacerbated in HGPS cells, provoking the mislocalization of numerous protein targets of CRM1. We showed that normalization of this mechanism by pharmacologically inhibiting CRM1 with LMB (specific CRM1 inhibitor), mitigates the senescent phenotype of HGPS cells. Since mitochondrial dysfunction is a hallmark of HGPS, in this study we analyze the effect of LMB on mitochondrial function. Remarkably, LMB treatment induced the recovery of mitochondrial function in HGPS cells, as shown by the improvement in mitochondrial morphology, mitochondrial membrane potential, and ATP levels, which consequently impeded the accumulation of ROS but not mitochondrial superoxide. We provide evidence that the beneficial effect of LMB is mechanistically based on a combinatory effect on mitochondrial biogenesis via upregulation of PGC-1α expression (master transcription cofactor of mitochondrial genes), and mitophagy through the recovery of lysosomal content. The use of exportin CRM1 inhibitors constitutes a promising strategy to treat HGPS and other diseases characterized by mitochondrial impairment. Full article
Show Figures

Graphical abstract

Back to TopTop