1
|
Mishra L, Mishra M. Ribose-induced advanced glycation end products reduce the lifespan in Drosophila melanogaster by changing the redox state and down-regulating the Sirtuin genes. Biogerontology 2024; 26:28. [PMID: 39702854 DOI: 10.1007/s10522-024-10172-0] [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: 11/01/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Advanced Glycation End (AGE) products are one such factor that accumulates during aging and age-related diseases. However, how exogenous AGE compounds cause aging is an area that needs to be explored. Specifically, how an organ undergoes aging and aging-related phenomena that need further investigation. The intestine is the most exposed area to food substances. How AGEs affect the intestine in terms of aging need to be explored. Drosophila melanogaster, a well-known model organism, is used to decode aging and age-associated phenomena. In this study, we fed Ribose induced Advanced Glycation End products (Rib-AGE) to D. melanogaster to study the aging mechanism. The Rib-AGE-induced aging was checked in Drosophila. We found a series of changes in Rib-AGE-fed flies. Reactive oxygen species (ROS) and nitric oxide species (NOs) were higher in the Rib-AGE-fed flies, and the antioxidant level was lower. The intestinal permeability was altered. The microorganism load was higher inside the gut. The structural arrangement of the gut's microfilament was found to be damaged, and the nuclear shape was found to be irregular. Cell death within the gut was elevated in comparison to control. The food intake was found to be reduced. The relative mRNA expression of the Sirtuin 2 and Sirtuin 6 gene of D. melanogaster was downregulated in Rib-AGE-fed flies compared to the control. All these findings strongly suggest that Rib-AGE accelerates aging and age-related disorders in D. melanogaster.
Collapse
Affiliation(s)
- Lokanath Mishra
- Neural Developmental Biology Lab, Department of Life Science, NIT Rourkela, Rourkela, 769008, India
| | - Monalisa Mishra
- Neural Developmental Biology Lab, Department of Life Science, NIT Rourkela, Rourkela, 769008, India.
| |
Collapse
|
2
|
Beust C, Valdeolivas A, Baptista A, Brière G, Lévy N, Ozisik O, Baudot A. The Molecular Landscape of Premature Aging Diseases Defined by Multilayer Network Exploration. Adv Biol (Weinh) 2024; 8:e2400134. [PMID: 39123285 DOI: 10.1002/adbi.202400134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/26/2024] [Indexed: 08/12/2024]
Abstract
Premature Aging (PA) diseases are rare genetic disorders that mimic some aspects of physiological aging at an early age. Various causative genes of PA diseases have been identified in recent years, providing insights into some dysfunctional cellular processes. However, the identification of PA genes also revealed significant genetic heterogeneity and highlighted the gaps in this understanding of PA-associated molecular mechanisms. Furthermore, many patients remain undiagnosed. Overall, the current lack of knowledge about PA diseases hinders the development of effective diagnosis and therapies and poses significant challenges to improving patient care. Here, a network-based approach to systematically unravel the cellular functions disrupted in PA diseases is presented. Leveraging a network community identification algorithm, it is delved into a vast multilayer network of biological interactions to extract the communities of 67 PA diseases from their 132 associated genes. It is found that these communities can be grouped into six distinct clusters, each reflecting specific cellular functions: DNA repair, cell cycle, transcription regulation, inflammation, cell communication, and vesicle-mediated transport. That these clusters collectively represent the landscape of the molecular mechanisms that are perturbed in PA diseases, providing a framework for better understanding their pathogenesis is proposed. Intriguingly, most clusters also exhibited a significant enrichment in genes associated with physiological aging, suggesting a potential overlap between the molecular underpinnings of PA diseases and natural aging.
Collapse
Affiliation(s)
- Cécile Beust
- Aix Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Alberto Valdeolivas
- Aix Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Anthony Baptista
- Aix Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Galadriel Brière
- Aix Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
- Aix Marseille Univ, CNRS, I2M, Marseille, France
| | - Nicolas Lévy
- Aix Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Ozan Ozisik
- Aix Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Anaïs Baudot
- Aix Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
- CNRS, Marseille, France
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
| |
Collapse
|
3
|
Tiwari V, Alam MJ, Bhatia M, Navya M, Banerjee SK. The structure and function of lamin A/C: Special focus on cardiomyopathy and therapeutic interventions. Life Sci 2024; 341:122489. [PMID: 38340979 DOI: 10.1016/j.lfs.2024.122489] [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: 11/11/2023] [Revised: 01/21/2024] [Accepted: 02/03/2024] [Indexed: 02/12/2024]
Abstract
Lamins are inner nuclear membrane proteins that belong to the intermediate filament family. Lamin A/C lie adjacent to the heterochromatin structure in polymer form, providing skeletal to the nucleus. Based on the localization, lamin A/C provides nuclear stability and cytoskeleton to the nucleus and modulates chromatin organization and gene expression. Besides being the structural protein making the inner nuclear membrane in polymer form, lamin A/C functions as a signalling molecule involved in gene expression as an enhancer inside the nucleus. Lamin A/C regulates various cellular pathways like autophagy and energy balance in the cytoplasm. Its expression is highly variable in differentiated tissues, higher in hard tissues like bone and muscle cells, and lower in soft tissues like the liver and brain. In muscle cells, including the heart, lamin A/C must be expressed in a balanced state. Lamin A/C mutation is linked with various diseases, such as muscular dystrophy, lipodystrophy, and cardiomyopathies. It has been observed that a good number of mutations in the LMNA gene impact cardiac activity and its function. Although several works have been published, there are still several unexplored areas left regarding the lamin A/C function and structure in the cardiovascular system and its pathological state. In this review, we focus on the structural organization, expression pattern, and function of lamin A/C, its interacting partners, and the pathophysiology associated with mutations in the lamin A/C gene, with special emphasis on cardiovascular diseases. With the recent finding on lamin A/C, we have summarized the possible therapeutic interventions to treat cardiovascular symptoms and reverse the molecular changes.
Collapse
Affiliation(s)
- Vikas Tiwari
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
| | - Md Jahangir Alam
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India; Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Madhavi Bhatia
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
| | - Malladi Navya
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
| | - Sanjay K Banerjee
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India.
| |
Collapse
|
4
|
Mu X, Gerhard-Herman MD, Zhang YS. Building Blood Vessel Chips with Enhanced Physiological Relevance. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201778. [PMID: 37693798 PMCID: PMC10489284 DOI: 10.1002/admt.202201778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Indexed: 09/12/2023]
Abstract
Blood vessel chips are bioengineered microdevices, consisting of biomaterials, human cells, and microstructures, which recapitulate essential vascular structure and physiology and allow a well-controlled microenvironment and spatial-temporal readouts. Blood vessel chips afford promising opportunities to understand molecular and cellular mechanisms underlying a range of vascular diseases. The physiological relevance is key to these blood vessel chips that rely on bioinspired strategies and bioengineering approaches to translate vascular physiology into artificial units. Here, we discuss several critical aspects of vascular physiology, including morphology, material composition, mechanical properties, flow dynamics, and mass transport, which provide essential guidelines and a valuable source of bioinspiration for the rational design of blood vessel chips. We also review state-of-art blood vessel chips that exhibit important physiological features of the vessel and reveal crucial insights into the biological processes and disease pathogenesis, including rare diseases, with notable implications for drug screening and clinical trials. We envision that the advances in biomaterials, biofabrication, and stem cells improve the physiological relevance of blood vessel chips, which, along with the close collaborations between clinicians and bioengineers, enable their widespread utility.
Collapse
Affiliation(s)
- Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marie Denise Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
5
|
Yeter Doğan B, Günay N, Ada Y, Doğan ME. A novel MTX2 gene splice site variant resulting in exon skipping, causing the recently described mandibuloacral dysplasia progeroid syndrome. Am J Med Genet A 2023; 191:173-182. [PMID: 36269149 DOI: 10.1002/ajmg.a.63010] [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: 06/22/2022] [Revised: 09/09/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022]
Abstract
Until recently, mandibuloacral dysplasia (MAD) with type A and type B lipodystrophy was the first to come to mind in the association of mandibular hypoplasia, lipodystrophy, and acro-osteolysis. However, it has recently been added to the differential diagnosis of MAD, a newly defined syndrome, called MDPS. MDPS is a skeletal dysplasia characterized by postnatal growth retardation, hypotonia, generalized lipodystrophy, skin changes, progeroid traits, and dysmorphic facial features, including prominent eyes, long pinched nose, mandibular hypoplasia, and a small mouth. Biallelic null variants of the MTX2 gene are responsible for this syndrome. We performed whole-exome sequencing (WES) in a 6-year-old patient with skeletal dysplasia. WES revealed a novel homozygous c.543+1G>T splice site variant in the MTX2 gene. We also extracted total RNA from peripheral blood and used reverse transcription-polymerase chain reaction to generate cDNA. Sanger sequencing from cDNA showed that exon 8 of MTX2 was skipped. This study adds to the genetics and phenotype of MDPS and underlines the importance of comprehensive clinical and molecular research.
Collapse
Affiliation(s)
- Burcu Yeter Doğan
- Division of Pediatric Genetics, Department of Pediatrics, Kayseri City Education and Research Hospital, Kayseri, Turkey
| | - Neslihan Günay
- Division of Pediatric Nephrology, Department of Pediatrics, Kayseri City Education and Research Hospital, Kayseri, Turkey
| | - Yasin Ada
- Department of Medical Genetics, Kayseri City Education and Research Hospital, Kayseri, Turkey
| | - Muhammet Ensar Doğan
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
6
|
Structural basis for the interaction between unfarnesylated progerin and the Ig-like domain of lamin A/C in premature aging disorders. Biochem Biophys Res Commun 2022; 637:210-217. [DOI: 10.1016/j.bbrc.2022.10.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 10/20/2022] [Indexed: 11/24/2022]
|
7
|
Lamis A, Siddiqui SW, Ashok T, Patni N, Fatima M, Aneef AN. Hutchinson-Gilford Progeria Syndrome: A Literature Review. Cureus 2022; 14:e28629. [PMID: 36196312 PMCID: PMC9524302 DOI: 10.7759/cureus.28629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2022] [Indexed: 11/05/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging condition that involves genetic mutations, resulting in debilitating phenotypic features. The present state of knowledge on the molecular pathways that contribute to the pathophysiology of HGPS and the techniques being tested in vitro and in vivo to combat progerin toxicity have been discussed here. Nuclear morphological abnormalities, dysregulated gene expression, DNA repair deficiencies, telomere shortening, and genomic instability are all caused by progerin accumulation, all of which impair cellular proliferative capability. In addition, HGPS cells and preclinical animal models have revealed new information about the disease's molecular and cellular pathways and putative mechanisms involved in normal aging. This article has discussed the understanding of the molecular pathways by which progerin expression leads to HGPS and how the advanced therapy options for HGPS patients can help us understand and treat the condition.
Collapse
|
8
|
Chiang MC, Chern E. Current Development, Obstacle and Futural Direction of Induced Pluripotent Stem Cell and Mesenchymal Stem Cell Treatment in Degenerative Retinal Disease. Int J Mol Sci 2022; 23:ijms23052529. [PMID: 35269671 PMCID: PMC8910526 DOI: 10.3390/ijms23052529] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 11/26/2022] Open
Abstract
Degenerative retinal disease is one of the major causes of vision loss around the world. The past several decades have witnessed emerging development of stem cell treatment for retinal disease. Nevertheless, sourcing stem cells remains controversial due to ethical concerns and their rarity. Furthermore, induced pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs) are both isolated from patients’ mature tissues; thus, issues such as avoiding moral controversy and adverse events related to immunosuppression and obtaining a large number of cells have opened a new era in regenerative medicine. This review focuses on the current application and development, clinical trials, and latest research of stem cell therapy, as well as its limitations and future directions.
Collapse
|
9
|
MG132 Induces Progerin Clearance and Improves Disease Phenotypes in HGPS-like Patients’ Cells. Cells 2022; 11:cells11040610. [PMID: 35203262 PMCID: PMC8870437 DOI: 10.3390/cells11040610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Progeroid syndromes (PS), including Hutchinson-Gilford Progeria Syndrome (HGPS), are premature and accelerated aging diseases, characterized by clinical features mimicking physiological aging. Most classical HGPS patients carry a de novo point mutation within exon 11 of the LMNA gene encoding A-type lamins. This mutation activates a cryptic splice site, leading to the production of a truncated prelamin A, called prelamin A ∆50 or progerin, that accumulates in HGPS cell nuclei and is a hallmark of the disease. Some patients with PS carry other LMNA mutations and are named “HGPS-like” patients. They produce progerin and/or other truncated prelamin A isoforms (∆35 and ∆90). We previously found that MG132, a proteasome inhibitor, induced progerin clearance in classical HGPS through autophagy activation and splicing regulation. Here, we show that MG132 induces aberrant prelamin A clearance and improves cellular phenotypes in HGPS-like patients’ cells other than those previously described in classical HGPS. These results provide preclinical proof of principle for the use of a promising class of molecules toward a potential therapy for children with HGPS-like or classical HGPS.
Collapse
|
10
|
Frankel D, Delecourt V, Novoa-del-Toro EM, Robin JD, Airault C, Bartoli C, Carabalona A, Perrin S, Mazaleyrat K, De Sandre-Giovannoli A, Magdinier F, Baudot A, Lévy N, Kaspi E, Roll P. miR-376a-3p and miR-376b-3p overexpression in Hutchinson-Gilford progeria fibroblasts inhibits cell proliferation and induces premature senescence. iScience 2022; 25:103757. [PMID: 35118365 PMCID: PMC8800101 DOI: 10.1016/j.isci.2022.103757] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/10/2021] [Accepted: 01/07/2022] [Indexed: 11/29/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder, in which an abnormal and toxic protein called progerin, accumulates in cell nuclei, leading to major cellular defects. Among them, chromatin remodeling drives gene expression changes, including miRNA dysregulation. In our study, we evaluated miRNA expression profiles in HGPS and control fibroblasts. We identified an enrichment of overexpressed miRNAs belonging to the 14q32.2-14q32.3 miRNA cluster. Using 3D FISH, we demonstrated that overexpression of these miRNAs is associated with chromatin remodeling at this specific locus in HGPS fibroblasts. We then focused on miR-376b-3p and miR-376a-3p, both overexpressed in HGPS fibroblasts. We demonstrated that their induced overexpression in control fibroblasts decreases cell proliferation and increases senescence, whereas their inhibition in HGPS fibroblasts rescues proliferation defects and senescence and decreases progerin accumulation. By targeting these major processes linked to premature aging, these two miRNAs may play a pivotal role in the pathophysiology of HGPS. Several miRNAs are deregulated in HGPS fibroblasts compared with controls Progerin leads to overexpression of miRNAs belonging to the 14q32.2-14q32.3 cluster miR-376a and miR-376b overexpression decreases cell proliferation and increases senescence
Collapse
Affiliation(s)
- Diane Frankel
- Aix Marseille Univ, APHM, INSERM, MMG, Hôpital la Timone, Service de Biologie Cellulaire, 27 Bd Jean Moulin, Marseille, France
| | | | | | | | | | | | | | | | | | - Annachiara De Sandre-Giovannoli
- Aix Marseille Univ, APHM, INSERM, MMG, Hôpital la Timone, Département de Génétique Médicale, Biological Resource Center (CRB-TAC), Marseille, France
| | | | - Anaïs Baudot
- Aix Marseille Univ, INSERM, MMG, Marseille, France
| | - Nicolas Lévy
- Aix Marseille Univ, APHM, INSERM, MMG, Hôpital la Timone, Département de Génétique Médicale, Biological Resource Center (CRB-TAC), Marseille, France
| | - Elise Kaspi
- Aix Marseille Univ, APHM, INSERM, MMG, Hôpital la Timone, Service de Biologie Cellulaire, 27 Bd Jean Moulin, Marseille, France
| | - Patrice Roll
- Aix Marseille Univ, APHM, INSERM, MMG, Hôpital la Timone, Service de Biologie Cellulaire, 27 Bd Jean Moulin, Marseille, France
- Corresponding author
| |
Collapse
|
11
|
Pongbangli N, Pitipakorn K, Jai-aue S, Sirijanchune P, Pongpittayut S, Wongcharoen W. A 13-Year-Old Boy from Thailand with Hutchinson-Gilford Progeria Syndrome with Coronary Artery and Aortic Calcification and Non-ST-Segment Elevation Myocardial Infarction (NSTEMI). AMERICAN JOURNAL OF CASE REPORTS 2021; 22:e928969. [PMID: 33414362 PMCID: PMC7805248 DOI: 10.12659/ajcr.928969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/17/2020] [Accepted: 11/03/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Hutchinson-Gilford progeria syndrome (HGPS), also known as progeria, is due to a mutation in the LMNA gene, resulting in a life expectancy of no more than 13 years, and a high mortality rate due to cardiovascular disease. We report the case of a 13-year-old boy from Thailand with Hutchinson-Gilford progeria syndrome with coronary artery and aortic calcification and non-ST-segment elevation myocardial infarction (NSTEMI). CASE REPORT A 13-year-old Thai boy was diagnosed with progeria. His physical appearance included short stature and thin limbs with prominent joint stiffness. He had craniofacial disproportion, with the absence of earlobes and with micrognathia. His skin had a generalized scleroderma-like lesion and hair loss with prominent scalp veins. His mental and cognitive functions were normal. Unfortunately, the mutation status in the LMNA gene was not available for testing in Thailand. He was diagnosed as having NSTEMI based on clinical chest pain, 12-lead ECG, and elevated cardiac troponin level. The coronary calcium score reflected severe calcification of the aortic valve and coronary artery disease along the left main and left anterior descending arteries. The patient received treatment with medication and aggressive risk factor control. After 3 months of follow-up, the patient reported no recurrence of symptoms. CONCLUSIONS This case of Hutchinson-Gilford progeria syndrome is rare in that most patients do not live beyond 13 years of age. This patient presented with typical accelerated degenerative changes of the cardiovascular system, including NSTEMI.
Collapse
Affiliation(s)
- Natnicha Pongbangli
- Division of Cardiology, Department of Internal Medicine, Chiang-Rai Prachanukroh Hospital, Chiang-Rai, Thailand
| | - Kannika Pitipakorn
- Division of Cardiology, Department of Pediatric, Chiang-Rai Prachanukroh Hospital, Chiang-Rai, Thailand
| | - Sasivimon Jai-aue
- Division of Cardiology, Department of Internal Medicine, Chiang-Rai Prachanukroh Hospital, Chiang-Rai, Thailand
| | - Piyaporn Sirijanchune
- Division of Pulmonology, Department of Internal Medicine, Chiang-Rai Prachanukroh Hospital, Chiang-Rai, Thailand
| | - Sorawit Pongpittayut
- Division of Cardiology, Department of Pediatric, Chiang-Rai Prachanukroh Hospital, Chiang-Rai, Thailand
| | - Wanwarang Wongcharoen
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
12
|
Karabağ C, Jones ML, Peddie CJ, Weston AE, Collinson LM, Reyes-Aldasoro CC. Semantic segmentation of HeLa cells: An objective comparison between one traditional algorithm and four deep-learning architectures. PLoS One 2020; 15:e0230605. [PMID: 33006963 PMCID: PMC7531863 DOI: 10.1371/journal.pone.0230605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/06/2020] [Indexed: 12/21/2022] Open
Abstract
The quantitative study of cell morphology is of great importance as the structure and condition of cells and their structures can be related to conditions of health or disease. The first step towards that, is the accurate segmentation of cell structures. In this work, we compare five approaches, one traditional and four deep-learning, for the semantic segmentation of the nuclear envelope of cervical cancer cells commonly known as HeLa cells. Images of a HeLa cancer cell were semantically segmented with one traditional image-processing algorithm and four three deep learning architectures: VGG16, ResNet18, Inception-ResNet-v2, and U-Net. Three hundred slices, each 2000 × 2000 pixels, of a HeLa Cell were acquired with Serial Block Face Scanning Electron Microscopy. The first three deep learning architectures were pre-trained with ImageNet and then fine-tuned with transfer learning. The U-Net architecture was trained from scratch with 36, 000 training images and labels of size 128 × 128. The image-processing algorithm followed a pipeline of several traditional steps like edge detection, dilation and morphological operators. The algorithms were compared by measuring pixel-based segmentation accuracy and Jaccard index against a labelled ground truth. The results indicated a superior performance of the traditional algorithm (Accuracy = 99%, Jaccard = 93%) over the deep learning architectures: VGG16 (93%, 90%), ResNet18 (94%, 88%), Inception-ResNet-v2 (94%, 89%), and U-Net (92%, 56%).
Collapse
Affiliation(s)
- Cefa Karabağ
- Research Centre for Biomedical Engineering School of Mathematics, Computer Science and Engineering, Department of Electrical & Electronic Engineering, City, University of London, London, United Kingdom
| | - Martin L. Jones
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Christopher J. Peddie
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Anne E. Weston
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Lucy M. Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Constantino Carlos Reyes-Aldasoro
- giCentre, Department of Computer Science, School of Mathematics, Computer Science and Engineering, City, University of London, London, United Kingdom
| |
Collapse
|
13
|
Loss of MTX2 causes mandibuloacral dysplasia and links mitochondrial dysfunction to altered nuclear morphology. Nat Commun 2020; 11:4589. [PMID: 32917887 PMCID: PMC7486921 DOI: 10.1038/s41467-020-18146-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 07/27/2020] [Indexed: 11/08/2022] Open
Abstract
Mandibuloacral dysplasia syndromes are mainly due to recessive LMNA or ZMPSTE24 mutations, with cardinal nuclear morphological abnormalities and dysfunction. We report five homozygous null mutations in MTX2, encoding Metaxin-2 (MTX2), an outer mitochondrial membrane protein, in patients presenting with a severe laminopathy-like mandibuloacral dysplasia characterized by growth retardation, bone resorption, arterial calcification, renal glomerulosclerosis and severe hypertension. Loss of MTX2 in patients' primary fibroblasts leads to loss of Metaxin-1 (MTX1) and mitochondrial dysfunction, including network fragmentation and oxidative phosphorylation impairment. Furthermore, patients' fibroblasts are resistant to induced apoptosis, leading to increased cell senescence and mitophagy and reduced proliferation. Interestingly, secondary nuclear morphological defects are observed in both MTX2-mutant fibroblasts and mtx-2-depleted C. elegans. We thus report the identification of a severe premature aging syndrome revealing an unsuspected link between mitochondrial composition and function and nuclear morphology, establishing a pathophysiological link with premature aging laminopathies and likely explaining common clinical features.
Collapse
|
14
|
Lessel I, Chen MJ, Lüttgen S, Arndt F, Fuchs S, Meien S, Thiele H, Jones JR, Shaw BR, Crossman DK, Nürnberg P, Korf BR, Kubisch C, Lessel D. Two novel cases further expand the phenotype of TOR1AIP1-associated nuclear envelopathies. Hum Genet 2020; 139:483-498. [PMID: 32055997 PMCID: PMC7078146 DOI: 10.1007/s00439-019-02105-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 12/22/2019] [Indexed: 12/19/2022]
Abstract
Biallelic variants in TOR1AIP1, encoding the integral nuclear membrane protein LAP1 (lamina-associated polypeptide 1) with two functional isoforms LAP1B and LAP1C, have initially been linked to muscular dystrophies with variable cardiac and neurological impairment. Furthermore, a recurrent homozygous nonsense alteration, resulting in loss of both LAP1 isoforms, was identified in seven likely related individuals affected by multisystem anomalies with progeroid-like appearance and lethality within the 1st decade of life. Here, we have identified compound heterozygosity in TOR1AIP1 affecting both LAP1 isoforms in two unrelated individuals affected by congenital bilateral hearing loss, ventricular septal defect, bilateral cataracts, mild to moderate developmental delay, microcephaly, mandibular hypoplasia, short stature, progressive muscular atrophy, joint contractures and severe chronic heart failure, with much longer survival. Cellular characterization of primary fibroblasts of one affected individual revealed absence of both LAP1B and LAP1C, constitutively low lamin A/C levels, aberrant nuclear morphology including nuclear cytoplasmic channels, and premature senescence, comparable to findings in other progeroid forms of nuclear envelopathies. We additionally observed an abnormal activation of the extracellular signal-regulated kinase 1/2 (ERK 1/2). Ectopic expression of wild-type TOR1AIP1 mitigated these cellular phenotypes, providing further evidence for the causal role of identified genetic variants. Altogether, we thus further expand the TOR1AIP1-associated phenotype by identifying individuals with biallelic loss-of-function variants who survived beyond the 1st decade of life and reveal novel molecular consequences underlying the TOR1AIP1-associated disorders.
Collapse
Affiliation(s)
- Ivana Lessel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Mei-Jan Chen
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 36394, USA
| | - Sabine Lüttgen
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Florian Arndt
- Department for Pediatric Cardiology, University Heart Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Sigrid Fuchs
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Stefanie Meien
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, 50931, Cologne, Germany
| | - Julie R Jones
- Molecular Diagnostic Laboratory, Greenwood Genetic Center, Greenwood, SC, 29646, USA
| | - Brandon R Shaw
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 36394, USA
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 36394, USA
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, 50931, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, 50931, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50931, Cologne, Germany
| | - Bruce R Korf
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 36394, USA
| | - Christian Kubisch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Davor Lessel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| |
Collapse
|
15
|
Grelet M, Blanck V, Sigaudy S, Philip N, Giuliano F, Khachnaoui K, Morel G, Grotto S, Sophie J, Poirsier C, Lespinasse J, Alric L, Calvas P, Chalhoub G, Layet V, Molin A, Colson C, Marsili L, Edery P, Lévy N, De Sandre-Giovannoli A. Outcomes of 4 years of molecular genetic diagnosis on a panel of genes involved in premature aging syndromes, including laminopathies and related disorders. Orphanet J Rare Dis 2019; 14:288. [PMID: 31829210 PMCID: PMC6907233 DOI: 10.1186/s13023-019-1189-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/30/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Segmental progeroid syndromes are a heterogeneous group of rare and often severe genetic disorders that have been studied since the twentieth century. These progeroid syndromes are defined as segmental because only some of the features observed during natural aging are accelerated. METHODS Since 2015, the Molecular Genetics Laboratory in Marseille La Timone Hospital proposes molecular diagnosis of premature aging syndromes including laminopathies and related disorders upon NGS sequencing of a panel of 82 genes involved in these syndromes. We analyzed the results obtained in 4 years on 66 patients issued from France and abroad. RESULTS Globally, pathogenic or likely pathogenic variants (ACMG class 5 or 4) were identified in about 1/4 of the cases; among these, 9 pathogenic variants were novel. On the other hand, the diagnostic yield of our panel was over 60% when the patients were addressed upon a nosologically specific clinical suspicion, excepted for connective tissue disorders, for which clinical diagnosis may be more challenging. Prenatal testing was proposed to 3 families. We additionally detected 16 variants of uncertain significance and reclassified 3 of them as benign upon segregation analysis in first degree relatives. CONCLUSIONS High throughput sequencing using the Laminopathies/ Premature Aging disorders panel allowed molecular diagnosis of rare disorders associated with premature aging features and genetic counseling for families, representing an interesting first-level analysis before whole genome sequencing may be proposed, as a future second step, by the National high throughput sequencing platforms ("Medicine France Genomics 2025" Plan), in families without molecular diagnosis.
Collapse
Affiliation(s)
- Maude Grelet
- Department of Medical Genetics, Assistance Publique Hopitaux de Marseille, Marseille, France
- Aix Marseille Univ, INSERM, MMG, Marseille, France
| | - Véronique Blanck
- Department of Medical Genetics, Assistance Publique Hopitaux de Marseille, Marseille, France
| | - Sabine Sigaudy
- Department of Medical Genetics, Assistance Publique Hopitaux de Marseille, Marseille, France
- Aix Marseille Univ, INSERM, MMG, Marseille, France
| | - Nicole Philip
- Department of Medical Genetics, Assistance Publique Hopitaux de Marseille, Marseille, France
- Aix Marseille Univ, INSERM, MMG, Marseille, France
| | | | | | - Godelieve Morel
- Hospices Civils de Lyon, Genetic Department and National HHT Reference Center, Femme-Mère-Enfants Hospital, F-69677 Bron, France
- Université Claude Bernard Lyon 1, F-69100 Villeurbanne, France
| | - Sarah Grotto
- Genetics Department, AP-HP, Robert-Debré University Hospital, Paris, France
| | - Julia Sophie
- Department of Medical Genetics, CHU Toulouse, Purpan Hospital, 31059 Toulouse, France
| | - Céline Poirsier
- Department of Genetics, Reims University Hospital, Reims, France
| | - James Lespinasse
- Department of Genetics, Centre Hospitalier de Chambéry- Hôtel-dieu, Chambery, France
| | - Laurent Alric
- Internal Medicine, CHU Toulouse, Rangueil Hospital, Toulouse 3 University Hospital Center, Toulouse, France
| | - Patrick Calvas
- Department of Medical Genetics, CHU Toulouse, Purpan Hospital, 31059 Toulouse, France
| | | | - Valérie Layet
- Department of Genetics, Le Havre Hospital, F76600 Le Havre, France
| | - Arnaud Molin
- Department of Genetics, CHU de Caen, Avenue de la Cote de Nacre, 14000 Caen, France
| | - Cindy Colson
- Department of Genetics, CHU de Caen, Avenue de la Cote de Nacre, 14000 Caen, France
| | - Luisa Marsili
- Department of Clinical Genetics, Lille University Hospital, CHU, Lille, France
| | - Patrick Edery
- Hospices Civils de Lyon, Genetic Department and National HHT Reference Center, Femme-Mère-Enfants Hospital, F-69677 Bron, France
- Université Claude Bernard Lyon 1, F-69100 Villeurbanne, France
| | - Nicolas Lévy
- Department of Medical Genetics, Assistance Publique Hopitaux de Marseille, Marseille, France
- Aix Marseille Univ, INSERM, MMG, Marseille, France
- CRB-TAC (Biological Ressource Center-Tissues, DNA, Cells), Assistance Publique Hopitaux de Marseille, Marseille, France
| | - Annachiara De Sandre-Giovannoli
- Department of Medical Genetics, Assistance Publique Hopitaux de Marseille, Marseille, France
- Aix Marseille Univ, INSERM, MMG, Marseille, France
- CRB-TAC (Biological Ressource Center-Tissues, DNA, Cells), Assistance Publique Hopitaux de Marseille, Marseille, France
| |
Collapse
|
16
|
Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, Campisi J, Collado M, Evangelou K, Ferbeyre G, Gil J, Hara E, Krizhanovsky V, Jurk D, Maier AB, Narita M, Niedernhofer L, Passos JF, Robbins PD, Schmitt CA, Sedivy J, Vougas K, von Zglinicki T, Zhou D, Serrano M, Demaria M. Cellular Senescence: Defining a Path Forward. Cell 2019; 179:813-827. [PMID: 31675495 DOI: 10.1016/j.cell.2019.10.005] [Citation(s) in RCA: 1827] [Impact Index Per Article: 304.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/20/2019] [Accepted: 10/03/2019] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a cell state implicated in various physiological processes and a wide spectrum of age-related diseases. Recently, interest in therapeutically targeting senescence to improve healthy aging and age-related disease, otherwise known as senotherapy, has been growing rapidly. Thus, the accurate detection of senescent cells, especially in vivo, is essential. Here, we present a consensus from the International Cell Senescence Association (ICSA), defining and discussing key cellular and molecular features of senescence and offering recommendations on how to use them as biomarkers. We also present a resource tool to facilitate the identification of genes linked with senescence, SeneQuest (available at http://Senequest.net). Lastly, we propose an algorithm to accurately assess and quantify senescence, both in cultured cells and in vivo.
Collapse
Affiliation(s)
- Vassilis Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Biomedical Research Foundation, Academy of Athens, Athens, Greece; Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK; Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Peter D Adams
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK; CRUK Beatson Institute, Glasgow G61 1BD, UK; Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, Lugano, Switzerland; Department of Medicine, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy
| | - Dorothy C Bennett
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Oliver Bischof
- Laboratory of Nuclear Organization and Oncogenesis, Department of Cell Biology and Infection, Inserm U993, Institute Pasteur, Paris, France
| | - Cleo Bishop
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark St, London E1 2AT, UK
| | | | - Manuel Collado
- Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), Santiago de Compostela, Spain
| | - Konstantinos Evangelou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerardo Ferbeyre
- Faculty of Medicine, Department of Biochemistry, Université de Montréal and CRCHUM, Montreal, QC, Canada
| | - Jesús Gil
- MRC London Institute of Medical Sciences (LMS), Du Cane Road, London, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, UK
| | - Eiji Hara
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Valery Krizhanovsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Diana Jurk
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Andrea B Maier
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit, Amsterdam, the Netherlands; Department of Medicine and Aged Care, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, MN, USA
| | - João F Passos
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, University of Minnesota, MN, USA
| | - Clemens A Schmitt
- Charité - University Medical Center, Department of Hematology, Oncology and Tumor Immunology, Virchow Campus, and Molekulares Krebsforschungszentrum, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Kepler University Hospital, Department of Hematology and Oncology, Johannes Kepler University, Linz, Austria
| | - John Sedivy
- Department of Molecular Biology, Cell Biology and Biochemistry, and Center for the Biology of Aging, Brown University, Providence, RI, USA
| | | | - Thomas von Zglinicki
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biology, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Daohong Zhou
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Manuel Serrano
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Marco Demaria
- University of Groningen (RUG), European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, the Netherlands.
| |
Collapse
|
17
|
Folgueras AR, Freitas-Rodríguez S, Velasco G, López-Otín C. Mouse Models to Disentangle the Hallmarks of Human Aging. Circ Res 2019; 123:905-924. [PMID: 30355076 DOI: 10.1161/circresaha.118.312204] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Model organisms have provided fundamental evidence that aging can be delayed and longevity extended. These findings gave rise to a new era in aging research aimed at elucidating the pathways and networks controlling this complex biological process. The identification of 9 hallmarks of aging has established a framework to evaluate the relative contribution of each hallmark and the interconnections among them. In this review, we revisit these hallmarks with the information obtained exclusively through the generation of genetically modified mouse models that have a significant impact on the aging process. We discuss within each hallmark those interventions that accelerate aging or that have been successful at increasing lifespan, with the final goal of identifying the most promising antiaging avenues based on the current knowledge provided by in vivo models.
Collapse
Affiliation(s)
- Alicia R Folgueras
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Sandra Freitas-Rodríguez
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Gloria Velasco
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Carlos López-Otín
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| |
Collapse
|
18
|
Harhouri K, Frankel D, Bartoli C, Roll P, De Sandre-Giovannoli A, Lévy N. An overview of treatment strategies for Hutchinson-Gilford Progeria syndrome. Nucleus 2019; 9:246-257. [PMID: 29619863 PMCID: PMC5973194 DOI: 10.1080/19491034.2018.1460045] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a sporadic, autosomal dominant disorder characterized by premature and accelerated aging symptoms leading to death at the mean age of 14.6 years usually due to cardiovascular complications. HGPS is caused by a de novo point mutation in the LMNA gene encoding the intermediate filament proteins lamins A and C which are structural components of the nuclear lamina. This mutation leads to the production of a truncated toxic form of lamin A, issued from aberrant splicing and called progerin. Progerin accumulates in HGPS cells' nuclei and is a hallmark of the disease. Small amounts of progerin are also produced during normal aging. HGPS cells and animal preclinical models have provided insights into the molecular and cellular pathways that underlie the disease and have also highlighted possible mechanisms involved in normal aging. This review reports recent medical advances and treatment approaches for patients affected with HGPS.
Collapse
Affiliation(s)
- Karim Harhouri
- a Aix Marseille Univ, INSERM, MMG - U1251 , Marseille , France
| | - Diane Frankel
- a Aix Marseille Univ, INSERM, MMG - U1251 , Marseille , France.,b APHM, Hôpital la Timone, Service de Biologie Cellulaire , Marseille , France
| | | | - Patrice Roll
- a Aix Marseille Univ, INSERM, MMG - U1251 , Marseille , France.,b APHM, Hôpital la Timone, Service de Biologie Cellulaire , Marseille , France
| | - Annachiara De Sandre-Giovannoli
- a Aix Marseille Univ, INSERM, MMG - U1251 , Marseille , France.,c APHM, Hôpital la Timone , Département de Génétique Médicale , Marseille , France
| | - Nicolas Lévy
- a Aix Marseille Univ, INSERM, MMG - U1251 , Marseille , France.,c APHM, Hôpital la Timone , Département de Génétique Médicale , Marseille , France
| |
Collapse
|
19
|
Flex E, Martinelli S, Van Dijck A, Ciolfi A, Cecchetti S, Coluzzi E, Pannone L, Andreoli C, Radio FC, Pizzi S, Carpentieri G, Bruselles A, Catanzaro G, Pedace L, Miele E, Carcarino E, Ge X, Chijiwa C, Lewis MES, Meuwissen M, Kenis S, Van der Aa N, Larson A, Brown K, Wasserstein MP, Skotko BG, Begtrup A, Person R, Karayiorgou M, Roos JL, Van Gassen KL, Koopmans M, Bijlsma EK, Santen GWE, Barge-Schaapveld DQCM, Ruivenkamp CAL, Hoffer MJV, Lalani SR, Streff H, Craigen WJ, Graham BH, van den Elzen APM, Kamphuis DJ, Õunap K, Reinson K, Pajusalu S, Wojcik MH, Viberti C, Di Gaetano C, Bertini E, Petrucci S, De Luca A, Rota R, Ferretti E, Matullo G, Dallapiccola B, Sgura A, Walkiewicz M, Kooy RF, Tartaglia M. Aberrant Function of the C-Terminal Tail of HIST1H1E Accelerates Cellular Senescence and Causes Premature Aging. Am J Hum Genet 2019; 105:493-508. [PMID: 31447100 DOI: 10.1016/j.ajhg.2019.07.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 07/10/2019] [Indexed: 02/03/2023] Open
Abstract
Histones mediate dynamic packaging of nuclear DNA in chromatin, a process that is precisely controlled to guarantee efficient compaction of the genome and proper chromosomal segregation during cell division and to accomplish DNA replication, transcription, and repair. Due to the important structural and regulatory roles played by histones, it is not surprising that histone functional dysregulation or aberrant levels of histones can have severe consequences for multiple cellular processes and ultimately might affect development or contribute to cell transformation. Recently, germline frameshift mutations involving the C-terminal tail of HIST1H1E, which is a widely expressed member of the linker histone family and facilitates higher-order chromatin folding, have been causally linked to an as-yet poorly defined syndrome that includes intellectual disability. We report that these mutations result in stable proteins that reside in the nucleus, bind to chromatin, disrupt proper compaction of DNA, and are associated with a specific methylation pattern. Cells expressing these mutant proteins have a dramatically reduced proliferation rate and competence, hardly enter into the S phase, and undergo accelerated senescence. Remarkably, clinical assessment of a relatively large cohort of subjects sharing these mutations revealed a premature aging phenotype as a previously unrecognized feature of the disorder. Our findings identify a direct link between aberrant chromatin remodeling, cellular senescence, and accelerated aging.
Collapse
Affiliation(s)
- Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, 00161 Italy; Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, 00161 Italy
| | - Anke Van Dijck
- Department of Medical Genetics, University of Antwerp, Edegem, 2650 Belgium; Department of Neurology, Antwerp University Hospital, Edegem, 2650 Belgium
| | - Andrea Ciolfi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Serena Cecchetti
- Microscopy Area, Core Facilities, Istituto Superiore di Sanità, Rome, 00161 Italy
| | - Elisa Coluzzi
- Department of Science, University Roma Tre, Rome, 00146 Italy
| | - Luca Pannone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, 00161 Italy; Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Cristina Andreoli
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, 00161 Italy
| | - Francesca Clementina Radio
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Simone Pizzi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Giovanna Carpentieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, 00161 Italy; Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, 00161 Italy
| | | | - Lucia Pedace
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146 Italy
| | - Evelina Miele
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146 Italy
| | - Elena Carcarino
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146 Italy; Current affiliation: Cordeliers Research Centre, Inserm 1138, Sorbonne Université, Paris, 75006 France
| | - Xiaoyan Ge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Current affiliation: Department of Genetics and Genomic Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chieko Chijiwa
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada
| | - M E Suzanne Lewis
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada
| | - Marije Meuwissen
- Department of Medical Genetics, University of Antwerp, Edegem, 2650 Belgium
| | - Sandra Kenis
- Department of Neurology, Antwerp University Hospital, Edegem, 2650 Belgium
| | | | - Austin Larson
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kathleen Brown
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Melissa P Wasserstein
- Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Brian G Skotko
- Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02114, USA
| | | | | | - Maria Karayiorgou
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - J Louw Roos
- Department of Psychiatry, University of Pretoria, Weskoppies Hospital, Pretoria, 0001 South Africa
| | - Koen L Van Gassen
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 AB the Netherlands
| | - Marije Koopmans
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 AB the Netherlands
| | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2300 RC the Netherlands
| | - Gijs W E Santen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2300 RC the Netherlands
| | | | - Claudia A L Ruivenkamp
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2300 RC the Netherlands
| | - Mariette J V Hoffer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2300 RC the Netherlands
| | - Seema R Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haley Streff
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - William J Craigen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brett H Graham
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Daan J Kamphuis
- Departement of Neurology, Reinier de Graaf Ziekenhuis, Delft, 2600 GA the Netherlands
| | - Katrin Õunap
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, 50406 Estonia; Institute of Clinical Medicine, University of Tartu, Tartu, 50406 Estonia
| | - Karit Reinson
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, 50406 Estonia; Institute of Clinical Medicine, University of Tartu, Tartu, 50406 Estonia
| | - Sander Pajusalu
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, 50406 Estonia; Institute of Clinical Medicine, University of Tartu, Tartu, 50406 Estonia; Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Monica H Wojcik
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Clara Viberti
- Department of Medical Sciences, University of Turin, Turin, 10126 Italy; Italian Institute for Genomic Medicine, Turin, 10126 Italy
| | - Cornelia Di Gaetano
- Department of Medical Sciences, University of Turin, Turin, 10126 Italy; Italian Institute for Genomic Medicine, Turin, 10126 Italy
| | - Enrico Bertini
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Simona Petrucci
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, 00189 Italy; Division of Medical Genetics, Casa Sollievo della Sofferenza Hospital, IRCCS, San Giovanni Rotondo, 71013 Italy
| | - Alessandro De Luca
- Division of Medical Genetics, Casa Sollievo della Sofferenza Hospital, IRCCS, San Giovanni Rotondo, 71013 Italy
| | - Rossella Rota
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146 Italy
| | - Elisabetta Ferretti
- Department of Experimental Medicine, Sapienza University, Rome, 00161 Italy; Istituto Neuromed, IRCCS, Pozzilli, 86077 Italy
| | - Giuseppe Matullo
- Department of Medical Sciences, University of Turin, Turin, 10126 Italy; Italian Institute for Genomic Medicine, Turin, 10126 Italy
| | - Bruno Dallapiccola
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Antonella Sgura
- Department of Science, University Roma Tre, Rome, 00146 Italy
| | - Magdalena Walkiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Current affiliation: National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA
| | - R Frank Kooy
- Department of Medical Genetics, University of Antwerp, Edegem, 2650 Belgium.
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy.
| |
Collapse
|
20
|
Clements CS, Bikkul MU, Ofosu W, Eskiw C, Tree D, Makarov E, Kill IR, Bridger JM. Presence and distribution of progerin in HGPS cells is ameliorated by drugs that impact on the mevalonate and mTOR pathways. Biogerontology 2019; 20:337-358. [PMID: 31041622 PMCID: PMC6535420 DOI: 10.1007/s10522-019-09807-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/29/2019] [Indexed: 12/12/2022]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare, premature ageing syndrome in children. HGPS is normally caused by a mutation in the LMNA gene, encoding nuclear lamin A. The classical mutation in HGPS leads to the production of a toxic truncated version of lamin A, progerin, which retains a farnesyl group. Farnesyltransferase inhibitors (FTI), pravastatin and zoledronic acid have been used in clinical trials to target the mevalonate pathway in HGPS patients to inhibit farnesylation of progerin, in order to reduce its toxicity. Some other compounds that have been suggested as treatments include rapamycin, IGF1 and N-acetyl cysteine (NAC). We have analysed the distribution of prelamin A, lamin A, lamin A/C, progerin, lamin B1 and B2 in nuclei of HGPS cells before and after treatments with these drugs, an FTI and a geranylgeranyltransferase inhibitor (GGTI) and FTI with pravastatin and zoledronic acid in combination. Confirming other studies prelamin A, lamin A, progerin and lamin B2 staining was different between control and HGPS fibroblasts. The drugs that reduced progerin staining were FTI, pravastatin, zoledronic acid and rapamycin. However, drugs affecting the mevalonate pathway increased prelamin A, with only FTI reducing internal prelamin A foci. The distribution of lamin A in HGPS cells was improved with treatments of FTI, pravastatin and FTI + GGTI. All treatments reduced the number of cells displaying internal speckles of lamin A/C and lamin B2. Drugs targeting the mevalonate pathway worked best for progerin reduction, with zoledronic acid removing internal progerin speckles. Rapamycin and NAC, which impact on the MTOR pathway, both reduced both pools of progerin without increasing prelamin A in HGPS cell nuclei.
Collapse
Affiliation(s)
- Craig S Clements
- Progeria Research Team, Ageing Studies Theme, Institute for Environment, Health and Societies, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK
| | - Mehmet U Bikkul
- Progeria Research Team, Ageing Studies Theme, Institute for Environment, Health and Societies, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK
| | - Wendy Ofosu
- Progeria Research Team, Ageing Studies Theme, Institute for Environment, Health and Societies, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK.,Department of Biomedical Sciences, University of Westminster, 115 New Cavendish Street, London, W1W 6UW, UK
| | - Christopher Eskiw
- Food and Bioproduct Sciences, College of Agriculture and Bioresources, University of Saskatchewan, 51 Campus Drive, Saskatoon, SK, S7B 5A8, Canada
| | - David Tree
- Progeria Research Team, Ageing Studies Theme, Institute for Environment, Health and Societies, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK
| | - Evgeny Makarov
- Progeria Research Team, Ageing Studies Theme, Institute for Environment, Health and Societies, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK
| | - Ian R Kill
- Progeria Research Team, Ageing Studies Theme, Institute for Environment, Health and Societies, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK
| | - Joanna M Bridger
- Progeria Research Team, Ageing Studies Theme, Institute for Environment, Health and Societies, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK. .,Genome Engineering and Maintenance Network, Ageing Studies Theme, Institute of Environment, Health and Societies, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK.
| |
Collapse
|
21
|
Haydont V, Neiveyans V, Fortunel NO, Asselineau D. Transcriptome profiling of human papillary and reticular fibroblasts from adult interfollicular dermis pinpoints the ‘tissue skeleton’ gene network as a component of skin chrono-ageing. Mech Ageing Dev 2019; 179:60-77. [DOI: 10.1016/j.mad.2019.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/23/2018] [Accepted: 01/07/2019] [Indexed: 10/27/2022]
|
22
|
Osanai T, Tanaka M, Izumiyama K, Mikami K, Kitajima M, Tomisawa T, Magota K, Tomita H, Okumura K. Intracellular protons accelerate aging and switch on aging hallmarks in mice. J Cell Biochem 2018; 119:9825-9837. [PMID: 30129099 DOI: 10.1002/jcb.27302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 06/27/2018] [Indexed: 12/31/2022]
Abstract
Diet-induced metabolic acidosis is associated with the impairment of bone metabolism and an increased risk of a number of chronic noncommunicable diseases, such as type 2 diabetes mellitus and hypertension. The serum bicarbonate level is an independent predictor of chronic kidney disease progression. We investigated whether proton accelerates aging by analyzing both coupling factor 6-overexpressing transgenic (TG) and high salt-fed mice which display sustained intracellular acidosis, due to enhanced proton import through ecto-F1 Fo complex and/or reduced proton export through Na+ -K+ ATPase inhibition. Both types of mice displayed shortened lifespan and early senescence-associated phenotypes such as signs of hair greying and alopecia, weight loss, and/or reduced organ mass. In chronic intracellular acidosis mice, autophagy was impaired by regression of Atg7, an increase in nuclear acetylated LC3 II, and acetylation of Atg7. The increase in histone 3 trimethylation at lysine 4 (H3K4me3) and H4K20me3 and the decrease in H3K9me3 and H3K27me3 were observed in the heart and kidney obtained from both TG and high salt-fed mice. The decrease in lamin A/C, emerin, and heterochromatin protein 1α without changes in barrier-to-autointegration factor and high-mobility group box 1 was confirmed in TG and high salt-fed mice. Suppression of nuclear histone deacetylase 3-emerin system is attributable to epigenetic regression of Atg7 and H4K5 acetylation. These findings will shed light on novel aging and impaired autophagy mechanism, and provide implications in a target for antiaging therapy.
Collapse
Affiliation(s)
- Tomohiro Osanai
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Makoto Tanaka
- Department of Hypertension and Stroke Internal Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kei Izumiyama
- Department of Cardiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kasumi Mikami
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Maiko Kitajima
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Toshiko Tomisawa
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Koji Magota
- Daiichi Sankyo Co, Ltd, Biologics Technology Research Laboratories Group 1, Pharmaceutical Technology Division, Gunma, Japan
| | - Hirofumi Tomita
- Department of Cardiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Ken Okumura
- Saiseikai Kumamoto Hospital, Division of Cardiology, Kumamoto, Japan
| |
Collapse
|
23
|
Frankel D, Delecourt V, Harhouri K, De Sandre-Giovannoli A, Lévy N, Kaspi E, Roll P. MicroRNAs in hereditary and sporadic premature aging syndromes and other laminopathies. Aging Cell 2018; 17:e12766. [PMID: 29696758 PMCID: PMC6052405 DOI: 10.1111/acel.12766] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2018] [Indexed: 12/11/2022] Open
Abstract
Hereditary and sporadic laminopathies are caused by mutations in genes encoding lamins, their partners, or the metalloprotease ZMPSTE24/FACE1. Depending on the clinical phenotype, they are classified as tissue‐specific or systemic diseases. The latter mostly manifest with several accelerated aging features, as in Hutchinson–Gilford progeria syndrome (HGPS) and other progeroid syndromes. MicroRNAs are small noncoding RNAs described as powerful regulators of gene expression, mainly by degrading target mRNAs or by inhibiting their translation. In recent years, the role of these small RNAs has become an object of study in laminopathies using in vitro or in vivo murine models as well as cells/tissues of patients. To date, few miRNAs have been reported to exert protective effects in laminopathies, including miR‐9, which prevents progerin accumulation in HGPS neurons. The recent literature has described the potential implication of several other miRNAs in the pathophysiology of laminopathies, mostly by exerting deleterious effects. This review provides an overview of the current knowledge of the functional relevance and molecular insights of miRNAs in laminopathies. Furthermore, we discuss how these discoveries could help to better understand these diseases at the molecular level and could pave the way toward identifying new potential therapeutic targets and strategies based on miRNA modulation.
Collapse
Affiliation(s)
- Diane Frankel
- Aix Marseille Univ; INSERM; MMG; Marseille France
- APHM, Hôpital la Timone; Service de Biologie Cellulaire; Marseille France
| | | | | | - Annachiara De Sandre-Giovannoli
- Aix Marseille Univ; INSERM; MMG; Marseille France
- APHM, Hôpital la Timone; Département de Génétique Médicale; Marseille France
| | - Nicolas Lévy
- Aix Marseille Univ; INSERM; MMG; Marseille France
- APHM, Hôpital la Timone; Département de Génétique Médicale; Marseille France
| | - Elise Kaspi
- Aix Marseille Univ; INSERM; MMG; Marseille France
- APHM, Hôpital la Timone; Service de Biologie Cellulaire; Marseille France
| | - Patrice Roll
- Aix Marseille Univ; INSERM; MMG; Marseille France
- APHM, Hôpital la Timone; Service de Biologie Cellulaire; Marseille France
| |
Collapse
|
24
|
Guinde J, Frankel D, Perrin S, Delecourt V, Lévy N, Barlesi F, Astoul P, Roll P, Kaspi E. Lamins in Lung Cancer: Biomarkers and Key Factors for Disease Progression through miR-9 Regulation? Cells 2018; 7:E78. [PMID: 30012957 PMCID: PMC6071028 DOI: 10.3390/cells7070078] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 02/08/2023] Open
Abstract
Lung cancer represents the primary cause of cancer death in the world. Malignant cells identification and characterization are crucial for the diagnosis and management of patients with primary or metastatic cancers. In this context, the identification of new biomarkers is essential to improve the differential diagnosis between cancer subtypes, to select the most appropriate therapy, and to establish prognostic correlations. Nuclear abnormalities are hallmarks of carcinoma cells and are used as cytological diagnostic criteria of malignancy. Lamins (divided into A- and B-types) are localized in the nuclear matrix comprising nuclear lamina, where they act as scaffolding protein, involved in many nuclear functions, with regulatory effects on the cell cycle and differentiation, senescence and apoptosis. Previous studies have suggested that lamins are involved in tumor development and progression with opposite results concerning their prognostic role. This review provides an overview of lamins expression in lung cancer and the relevance of these findings for disease diagnosis and prognosis. Furthermore, we discuss the link between A-type lamins expression in lung carcinoma cells and nuclear deformability, epithelial to mesenchymal transition, and metastatic potential, and which mechanisms could regulate A-type lamins expression in lung cancer, such as the microRNA miR-9.
Collapse
Affiliation(s)
- Julien Guinde
- Aix Marseille Université, INSERM, MMG, 13385 Marseille, France.
- APHM, Hôpital Nord, Department of Thoracic Oncology-Pleural Diseases-Interventional Pulmonology, CEDEX 5, 13385 Marseille, France.
| | - Diane Frankel
- Aix Marseille Université, APHM, INSERM, MMG, Hôpital la Timone, Service de Biologie Cellulaire, 13385 Marseille, France.
| | - Sophie Perrin
- Aix Marseille Université, INSERM, MMG, 13385 Marseille, France.
- ProGeLife, 13385 Marseille, France.
| | | | - Nicolas Lévy
- Aix Marseille Université, APHM, INSERM, MMG, Hôpital la Timone, Département de Génétique Médicale, 13385 Marseille, France.
| | - Fabrice Barlesi
- Aix Marseille Université, APHM, CNRS, INSERM, CRCM, Multidisciplinary Oncology & Therapeutic Innovations Department, 13385 Marseille, France.
| | - Philippe Astoul
- APHM, Hôpital Nord, Department of Thoracic Oncology-Pleural Diseases-Interventional Pulmonology, CEDEX 5, 13385 Marseille, France.
| | - Patrice Roll
- Aix Marseille Université, APHM, INSERM, MMG, Hôpital la Timone, Service de Biologie Cellulaire, 13385 Marseille, France.
| | - Elise Kaspi
- Aix Marseille Université, APHM, INSERM, MMG, Hôpital la Timone, Service de Biologie Cellulaire, 13385 Marseille, France.
| |
Collapse
|
25
|
Osanai T, Tanaka M, Mikami K, Kitajima M, Magota K, Tomita H, Okumura K. Mitochondrial inhibitory factor protein 1 attenuates coupling factor 6-induced aging signal. J Cell Biochem 2018; 119:6194-6203. [PMID: 29575130 DOI: 10.1002/jcb.26828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/28/2018] [Indexed: 12/16/2022]
Abstract
Coupling factor 6 (CF6) forces a counter-clockwise rotation of plasma membrane F1 Fo complex, resulting in proton import and accelerated aging. Inhibitory factor peptide 1 (IF1) suppresses a unidirectional counter-clockwise rotation of F1 Fo complex without affecting ATP synthesis. We tested the hypothesis that IF1 may attenuate CF6-induced aging signaling in CF6-overexpressing transgenic (TG) cells. In IF1-GFP overexpressing wild type (WT) cells, the diffuse peripheral staining of tubular mitochondria was observed with a dense widely distributed network around the nucleus. In TG cells, however, the only peri-nuclear network of fragmented mitochondria was observed at 24 h, but it was developed to a widely distributed mitochondrial network of tubular mitochondria at 72 h. TG cells displayed aging hallmarks of telomere attrition, epigenetic alterations, defective proteostasis, and genomic instability with a decrease in emerin and lamin and loss of heterochromatin. IF1 induction rescued TG cells from telomere attrition, expression of genomic instability with the increase in emerin and lamin, and that of epigenetic alterations with recovery of heterochromatin. In defective proteostasis, IF1 induction restored a potent peri-nuclear staining of autolysosomes compared with the baseline weak staining. The decrease in Atg7 was restored, whereas the increase in P62 was abolished. We conclude that genetic disruption of proton signals by IF1 induction suppressed CF6-induced expression of aging hallmarks such as telomere attrition, epigenetic alterations, defective proteostasis, and genomic instability. Given the widespread biological actions of CF6, the physiological and pathological actions of IF1 may be complex.
Collapse
Affiliation(s)
- Tomohiro Osanai
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Makoto Tanaka
- Department of Hypertension and Stroke Internal Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kasumi Mikami
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Maiko Kitajima
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Koji Magota
- Daiichi Sankyo Co., Ltd., Biologics Technology Research Laboratories Group1, Pharmaceutical Technology Division, 2716-1, Kurakake, Akaiwa, Chiyoda-machi, Oura-gun, Gunma, Japan
| | - Hirofumi Tomita
- Department of Cardiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Ken Okumura
- Division of Cardiology, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| |
Collapse
|
26
|
Burla R, La Torre M, Merigliano C, Vernì F, Saggio I. Genomic instability and DNA replication defects in progeroid syndromes. Nucleus 2018; 9:368-379. [PMID: 29936894 PMCID: PMC7000143 DOI: 10.1080/19491034.2018.1476793] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Progeroid syndromes induced by mutations in lamin A or in its interactors – named progeroid laminopathies – are model systems for the dissection of the molecular pathways causing physiological and premature aging. A large amount of data, based mainly on the Hutchinson Gilford Progeria syndrome (HGPS), one of the best characterized progeroid laminopathy, has highlighted the role of lamins in multiple DNA activities, including replication, repair, chromatin organization and telomere function. On the other hand, the phenotypes generated by mutations affecting genes directly acting on DNA function, as mutations in the helicases WRN and BLM or in the polymerase polδ, share many of the traits of progeroid laminopathies. These evidences support the hypothesis of a concerted implication of DNA function and lamins in aging. We focus here on these aspects to contribute to the comprehension of the driving forces acting in progeroid syndromes and premature aging.
Collapse
Affiliation(s)
- Romina Burla
- a Dipartimento di Biologia e Biotecnologie "C. Darwin" , Sapienza Università di Roma , Roma , Italy.,b Istituto di Biologia e Patologia Molecolari del CNR , Rome , Italy
| | - Mattia La Torre
- a Dipartimento di Biologia e Biotecnologie "C. Darwin" , Sapienza Università di Roma , Roma , Italy.,b Istituto di Biologia e Patologia Molecolari del CNR , Rome , Italy
| | - Chiara Merigliano
- a Dipartimento di Biologia e Biotecnologie "C. Darwin" , Sapienza Università di Roma , Roma , Italy
| | - Fiammetta Vernì
- a Dipartimento di Biologia e Biotecnologie "C. Darwin" , Sapienza Università di Roma , Roma , Italy
| | - Isabella Saggio
- a Dipartimento di Biologia e Biotecnologie "C. Darwin" , Sapienza Università di Roma , Roma , Italy.,b Istituto di Biologia e Patologia Molecolari del CNR , Rome , Italy.,c Istituto Pasteur Fondazione Cenci Bolognetti , Rome , Italy
| |
Collapse
|
27
|
Paci M, Elkhatib R, Longepied G, Bourgeois P, Ray PF, Levy N, Mitchell MJ, Metzler-Guillemain C. The involvement of the nuclear lamina in human and rodent spermiogenesis: a systematic review. Basic Clin Androl 2018; 28:7. [PMID: 29946470 PMCID: PMC6008938 DOI: 10.1186/s12610-018-0072-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/19/2018] [Indexed: 12/11/2022] Open
Abstract
The nuclear lamina (NL) is a filamentous protein meshwork, composed essentially of lamins, situated between the inner nuclear membrane and the chromatin. The NL is a component of the nuclear envelope, interacts with a wide range of proteins and is required for normal nuclear structure and physiological development. During spermiogenesis the spermatid nucleus is elongated, and dramatically reduced in size with protamines replacing histones to produce a highly compacted chromatin. There is mounting evidence from studies in human and rodent, that the NL plays an important role in mammalian spermatid differentiation during spermiogenesis. In this review, we summarize and discuss the data available in the literature regarding the involvement of lamins and their direct or indirect partners in normal and abnormal human spermiogenesis.
Collapse
Affiliation(s)
- Marine Paci
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France.,APHM Hôpital La Conception, Pôle femmes-Parents-enfants, Centre Clinico-Biologique d'Assistance Médicale à la Procréation-CECOS, 13385 Marseille Cedex 5, France
| | - Razan Elkhatib
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Guy Longepied
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Patrice Bourgeois
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Pierre F Ray
- 3Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, Université Grenoble Alpes, CHU Grenoble Alpes, F-38000 Grenoble, France
| | - Nicolas Levy
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Michael J Mitchell
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Catherine Metzler-Guillemain
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France.,APHM Hôpital La Conception, Pôle femmes-Parents-enfants, Centre Clinico-Biologique d'Assistance Médicale à la Procréation-CECOS, 13385 Marseille Cedex 5, France
| |
Collapse
|
28
|
Pathological modelling of pigmentation disorders associated with Hutchinson-Gilford Progeria Syndrome (HGPS) revealed an impaired melanogenesis pathway in iPS-derived melanocytes. Sci Rep 2018; 8:9112. [PMID: 29904107 PMCID: PMC6002548 DOI: 10.1038/s41598-018-27165-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/29/2018] [Indexed: 12/21/2022] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare genetic disorder that leads to premature aging. In this study, we used induced pluripotent stem cells to investigate the hypopigmentation phenotypes observed in patients with progeria. Accordingly, two iPS cell lines were derived from cells from HGPS patients and differentiated into melanocytes. Measurements of melanin content revealed a lower synthesis of melanin in HGPS melanocytes as compared to non-pathologic cells. Analysis of the melanosome maturation process by electron microscopy revealed a lower percentage of mature, fully pigmented melanosomes. Finally, a functional rescue experiment revealed the direct role of progerin in the regulation of melanogenesis. Overall, these results report a new dysregulated pathway in HGPS and open up novel perspectives in the study of pigmentation phenotypes that are associated with normal and pathological aging.
Collapse
|
29
|
Spectral comparisons of mammalian cells and intact organelles by solid-state NMR. J Struct Biol 2018; 206:49-54. [PMID: 29859329 DOI: 10.1016/j.jsb.2018.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/15/2018] [Accepted: 05/25/2018] [Indexed: 01/18/2023]
Abstract
Whole-cell protein profiling, spatial localization, and quantification of activities such as gene transcription and protein translation are possible with modern biochemical and biophysical techniques. Yet, addressing questions of overall compositional changes within a cell - capturing the relative amounts of protein and ribosomal RNA levels and lipid content simultaneously - would require extractions and purifications with caveats due to isolation yields and detection methods. A holistic view of cellular composition would aid in the study of cellular composition and function. Here, solid state NMR is used to identify 13C NMR signatures for cellular organelles in HeLa cells without the use of any isotopic labeling. Comparisons are made with carbon spectra of subcellular assemblies including DNA, lipids, ribosomes, nuclei and mitochondria. Whole-cell comparisons are made with different mammalian cells lines, with red blood cells that lack nuclei and organelles, and with Gram-negative and Gram-positive bacteria. Furthermore, treatment of mammalian cells with cycloheximide, a commonly used protein synthesis inhibitor, revealed unanticipated changes consistent with a significant increase in protein glycosylation, obvious at the whole cell level. Thus, we demonstrate that solid-state NMR serves as a unique analytical tool to catalog and compare the ratios of distinct carbon types in cells and serves as a discovery tool to reveal the workings of inhibitors such as cycloheximide on whole-cell biochemistry.
Collapse
|
30
|
Zhang S, Duan E. Fighting against Skin Aging: The Way from Bench to Bedside. Cell Transplant 2018; 27:729-738. [PMID: 29692196 PMCID: PMC6047276 DOI: 10.1177/0963689717725755] [Citation(s) in RCA: 366] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 07/05/2017] [Accepted: 07/14/2017] [Indexed: 12/31/2022] Open
Abstract
As the most voluminous organ of the body that is exposed to the outer environment, the skin suffers from both intrinsic and extrinsic aging factors. Skin aging is characterized by features such as wrinkling, loss of elasticity, laxity, and rough-textured appearance. This aging process is accompanied with phenotypic changes in cutaneous cells as well as structural and functional changes in extracellular matrix components such as collagens and elastin. In this review, we summarize these changes in skin aging, research advances of the molecular mechanisms leading to these changes, and the treatment strategies aimed at preventing or reversing skin aging.
Collapse
Affiliation(s)
- Shoubing Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Central laboratory of Molecular and Cellular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Enkui Duan
- State Key Lab of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
31
|
Carrero D, Soria-Valles C, López-Otín C. Hallmarks of progeroid syndromes: lessons from mice and reprogrammed cells. Dis Model Mech 2017; 9:719-35. [PMID: 27482812 PMCID: PMC4958309 DOI: 10.1242/dmm.024711] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ageing is a process that inevitably affects most living organisms and involves the accumulation of macromolecular damage, genomic instability and loss of heterochromatin. Together, these alterations lead to a decline in stem cell function and to a reduced capability to regenerate tissue. In recent years, several genetic pathways and biochemical mechanisms that contribute to physiological ageing have been described, but further research is needed to better characterize this complex biological process. Because premature ageing (progeroid) syndromes, including progeria, mimic many of the characteristics of human ageing, research into these conditions has proven to be very useful not only to identify the underlying causal mechanisms and identify treatments for these pathologies, but also for the study of physiological ageing. In this Review, we summarize the main cellular and animal models used in progeria research, with an emphasis on patient-derived induced pluripotent stem cell models, and define a series of molecular and cellular hallmarks that characterize progeroid syndromes and parallel physiological ageing. Finally, we describe the therapeutic strategies being investigated for the treatment of progeroid syndromes, and their main limitations. Summary: This Review defines the molecular and cellular hallmarks of progeroid syndromes according to the main cellular and animal models, and discusses the therapeutic strategies developed to date.
Collapse
Affiliation(s)
- Dido Carrero
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo 33006, Spain
| | - Clara Soria-Valles
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo 33006, Spain
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo 33006, Spain
| |
Collapse
|
32
|
Kaspi E, Frankel D, Guinde J, Perrin S, Laroumagne S, Robaglia-Schlupp A, Ostacolo K, Harhouri K, Tazi-Mezalek R, Micallef J, Dutau H, Tomasini P, De Sandre-Giovannoli A, Lévy N, Cau P, Astoul P, Roll P. Low lamin A expression in lung adenocarcinoma cells from pleural effusions is a pejorative factor associated with high number of metastatic sites and poor Performance status. PLoS One 2017; 12:e0183136. [PMID: 28806747 PMCID: PMC5555706 DOI: 10.1371/journal.pone.0183136] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/31/2017] [Indexed: 11/29/2022] Open
Abstract
The type V intermediate filament lamins are the principal components of the nuclear matrix, including the nuclear lamina. Lamins are divided into A-type and B-type, which are encoded by three genes, LMNA, LMNB1, and LMNB2. The alternative splicing of LMNA produces two major A-type lamins, lamin A and lamin C. Previous studies have suggested that lamins are involved in cancer development and progression. A-type lamins have been proposed as biomarkers for cancer diagnosis, prognosis, and/or follow-up. The aim of the present study was to investigate lamins in cancer cells from metastatic pleural effusions using immunofluorescence, western blotting, and flow cytometry. In a sub-group of lung adenocarcinomas, we found reduced expression of lamin A but not of lamin C. The reduction in lamin A expression was correlated with the loss of epithelial membrane antigen (EMA)/MUC-1, an epithelial marker that is involved in the epithelial to mesenchymal transition (EMT). Finally, the lamin A expression was inversely correlated with the number of metastatic sites and the WHO Performance status, and association of pleural, bone and lung metastatic localizations was more frequent when lamin A expression was reduced. In conclusion, low lamin A but not lamin C expression in pleural metastatic cells could represent a major actor in the development of metastasis, associated with EMT and could account for a pejorative factor correlated with a poor Performance status.
Collapse
Affiliation(s)
- Elise Kaspi
- Aix Marseille Univ, INSERM, GMGF, Marseille, France
- APHM, Hôpital la Timone, Service de Biologie Cellulaire, Marseille, France
| | - Diane Frankel
- Aix Marseille Univ, INSERM, GMGF, Marseille, France
- APHM, Hôpital la Timone, Service de Biologie Cellulaire, Marseille, France
| | - Julien Guinde
- Aix Marseille Univ, INSERM, GMGF, Marseille, France
- APHM, Hôpital Nord, Department of Thoracic Oncology–Pleural diseases–Interventional pulmonology, Marseille, France
| | | | - Sophie Laroumagne
- APHM, Hôpital Nord, Department of Thoracic Oncology–Pleural diseases–Interventional pulmonology, Marseille, France
| | - Andrée Robaglia-Schlupp
- Aix Marseille Univ, INSERM, GMGF, Marseille, France
- APHM, Hôpital la Timone, Service de Biologie Cellulaire, Marseille, France
- APHM, Hôpital la Timone, Département de Génétique Médicale et Centre de Ressources Biologiques, Marseille, France
| | | | | | - Rachid Tazi-Mezalek
- APHM, Hôpital Nord, Department of Thoracic Oncology–Pleural diseases–Interventional pulmonology, Marseille, France
| | - Joelle Micallef
- APHM, Hôpital la Timone, Service de Pharmacologie Clinique & Centre d’Investigation Clinique—CPCET, Marseille, France
- Aix Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France
| | - Hervé Dutau
- APHM, Hôpital Nord, Department of Thoracic Oncology–Pleural diseases–Interventional pulmonology, Marseille, France
| | - Pascale Tomasini
- Aix Marseille Univ, APHM, Marseille Early Phases Cancer Trials Center CLIP, Marseille, France
| | - Annachiara De Sandre-Giovannoli
- Aix Marseille Univ, INSERM, GMGF, Marseille, France
- APHM, Hôpital la Timone, Département de Génétique Médicale et Centre de Ressources Biologiques, Marseille, France
| | - Nicolas Lévy
- Aix Marseille Univ, INSERM, GMGF, Marseille, France
- APHM, Hôpital la Timone, Département de Génétique Médicale et Centre de Ressources Biologiques, Marseille, France
| | - Pierre Cau
- Aix Marseille Univ, INSERM, GMGF, Marseille, France
| | - Philippe Astoul
- APHM, Hôpital Nord, Department of Thoracic Oncology–Pleural diseases–Interventional pulmonology, Marseille, France
- Aix Marseille Univ, Marseille, France
| | - Patrice Roll
- Aix Marseille Univ, INSERM, GMGF, Marseille, France
- APHM, Hôpital la Timone, Service de Biologie Cellulaire, Marseille, France
- * E-mail:
| |
Collapse
|
33
|
Mitotic Dysfunction Associated with Aging Hallmarks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1002:153-188. [DOI: 10.1007/978-3-319-57127-0_7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
34
|
Ribas J, Zhang YS, Pitrez PR, Leijten J, Miscuglio M, Rouwkema J, Dokmeci MR, Nissan X, Ferreira L, Khademhosseini A. Biomechanical Strain Exacerbates Inflammation on a Progeria-on-a-Chip Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:10.1002/smll.201603737. [PMID: 28211642 PMCID: PMC5545787 DOI: 10.1002/smll.201603737] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/02/2017] [Indexed: 05/22/2023]
Abstract
Organ-on-a-chip platforms seek to recapitulate the complex microenvironment of human organs using miniaturized microfluidic devices. Besides modeling healthy organs, these devices have been used to model diseases, yielding new insights into pathophysiology. Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disease showing accelerated vascular aging, leading to the death of patients due to cardiovascular diseases. HGPS targets primarily vascular cells, which reside in mechanically active tissues. Here, a progeria-on-a-chip model is developed and the effects of biomechanical strain are examined in the context of vascular aging and disease. Physiological strain induces a contractile phenotype in primary smooth muscle cells (SMCs), while a pathological strain induces a hypertensive phenotype similar to that of angiotensin II treatment. Interestingly, SMCs derived from human induced pluripotent stem cells of HGPS donors (HGPS iPS-SMCs), but not from healthy donors, show an exacerbated inflammatory response to strain. In particular, increased levels of inflammation markers as well as DNA damage are observed. Pharmacological intervention reverses the strain-induced damage by shifting gene expression profile away from inflammation. The progeria-on-a-chip is a relevant platform to study biomechanics in vascular biology, particularly in the setting of vascular disease and aging, while simultaneously facilitating the discovery of new drugs and/or therapeutic targets.
Collapse
Affiliation(s)
- João Ribas
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Doctoral Program in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Yu Shrike Zhang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Patrícia R. Pitrez
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Jeroen Leijten
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Mario Miscuglio
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Mehmet Remzi Dokmeci
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Xavier Nissan
- INSERM U861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex 91030, France
| | | | | |
Collapse
|
35
|
A High Throughput Phenotypic Screening reveals compounds that counteract premature osteogenic differentiation of HGPS iPS-derived mesenchymal stem cells. Sci Rep 2016; 6:34798. [PMID: 27739443 PMCID: PMC5064407 DOI: 10.1038/srep34798] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/12/2016] [Indexed: 12/19/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare fatal genetic disorder that causes systemic accelerated aging in children. Thanks to the pluripotency and self-renewal properties of induced pluripotent stem cells (iPSC), HGPS iPSC-based modeling opens up the possibility of access to different relevant cell types for pharmacological approaches. In this study, 2800 small molecules were explored using high-throughput screening, looking for compounds that could potentially reduce the alkaline phosphatase activity of HGPS mesenchymal stem cells (MSCs) committed into osteogenic differentiation. Results revealed seven compounds that normalized the osteogenic differentiation process and, among these, all-trans retinoic acid and 13-cis-retinoic acid, that also decreased progerin expression. This study highlights the potential of high-throughput drug screening using HGPS iPS-derived cells, in order to find therapeutic compounds for HGPS and, potentially, for other aging-related disorders.
Collapse
|
36
|
Robin JD, Magdinier F. Physiological and Pathological Aging Affects Chromatin Dynamics, Structure and Function at the Nuclear Edge. Front Genet 2016; 7:153. [PMID: 27602048 PMCID: PMC4993774 DOI: 10.3389/fgene.2016.00153] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 08/08/2016] [Indexed: 01/29/2023] Open
Abstract
Lamins are intermediate filaments that form a complex meshwork at the inner nuclear membrane. Mammalian cells express two types of Lamins, Lamins A/C and Lamins B, encoded by three different genes, LMNA, LMNB1, and LMNB2. Mutations in the LMNA gene are associated with a group of phenotypically diverse diseases referred to as laminopathies. Lamins interact with a large number of binding partners including proteins of the nuclear envelope but also chromatin-associated factors. Lamins not only constitute a scaffold for nuclear shape, rigidity and resistance to stress but also contribute to the organization of chromatin and chromosomal domains. We will discuss here the impact of A-type Lamins loss on alterations of chromatin organization and formation of chromatin domains and how disorganization of the lamina contributes to the patho-physiology of premature aging syndromes.
Collapse
Affiliation(s)
- Jérôme D Robin
- IRCAN, CNRS UMR 7284/INSERM U1081, Faculté de Médecine Nice, France
| | | |
Collapse
|
37
|
All-trans retinoic acid and rapamycin normalize Hutchinson Gilford progeria fibroblast phenotype. Oncotarget 2016; 6:29914-28. [PMID: 26359359 PMCID: PMC4745772 DOI: 10.18632/oncotarget.4939] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/31/2015] [Indexed: 11/25/2022] Open
Abstract
Hutchinson Gilford progeria syndrome is a fatal disorder characterized by accelerated aging, bone resorption and atherosclerosis, caused by a LMNA mutation which produces progerin, a mutant lamin A precursor. Progeria cells display progerin and prelamin A nuclear accumulation, altered histone methylation pattern, heterochromatin loss, increased DNA damage and cell cycle alterations. Since the LMNA promoter contains a retinoic acid responsive element, we investigated if all-trans retinoic acid administration could lower progerin levels in cultured fibroblasts. We also evaluated the effect of associating rapamycin, which induces autophagic degradation of progerin and prelamin A. We demonstrate that all-trans retinoic acid acts synergistically with low-dosage rapamycin reducing progerin and prelamin A, via transcriptional downregulation associated with protein degradation, and increasing the lamin A to progerin ratio. These effects rescue cell dynamics and cellular proliferation through recovery of DNA damage response factor PARP1 and chromatin-associated nuclear envelope proteins LAP2α and BAF. The combined all-trans retinoic acid-rapamycin treatment is dramatically efficient, highly reproducible, represents a promising new approach in Hutchinson-Gilford Progeria therapy and deserves investigation in ageing-associated disorders.
Collapse
|
38
|
Antisense-Based Progerin Downregulation in HGPS-Like Patients' Cells. Cells 2016; 5:cells5030031. [PMID: 27409638 PMCID: PMC5040973 DOI: 10.3390/cells5030031] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/15/2016] [Accepted: 07/04/2016] [Indexed: 12/28/2022] Open
Abstract
Progeroid laminopathies, including Hutchinson-Gilford Progeria Syndrome (HGPS, OMIM #176670), are premature and accelerated aging diseases caused by defects in nuclear A-type Lamins. Most HGPS patients carry a de novo point mutation within exon 11 of the LMNA gene encoding A-type Lamins. This mutation activates a cryptic splice site leading to the deletion of 50 amino acids at its carboxy-terminal domain, resulting in a truncated and permanently farnesylated Prelamin A called Prelamin A Δ50 or Progerin. Some patients carry other LMNA mutations affecting exon 11 splicing and are named “HGPS-like” patients. They also produce Progerin and/or other truncated Prelamin A isoforms (Δ35 and Δ90) at the transcriptional and/or protein level. The results we present show that morpholino antisense oligonucleotides (AON) prevent pathogenic LMNA splicing, markedly reducing the accumulation of Progerin and/or other truncated Prelamin A isoforms (Prelamin A Δ35, Prelamin A Δ90) in HGPS-like patients’ cells. Finally, a patient affected with Mandibuloacral Dysplasia type B (MAD-B, carrying a homozygous mutation in ZMPSTE24, encoding an enzyme involved in Prelamin A maturation, leading to accumulation of wild type farnesylated Prelamin A), was also included in this study. These results provide preclinical proof of principle for the use of a personalized antisense approach in HGPS-like and MAD-B patients, who may therefore be eligible for inclusion in a therapeutic trial based on this approach, together with classical HGPS patients.
Collapse
|
39
|
Soria-Valles C, Carrero D, Gabau E, Velasco G, Quesada V, Bárcena C, Moens M, Fieggen K, Möhrcken S, Owens M, Puente DA, Asensio Ó, Loeys B, Pérez A, Benoit V, Wuyts W, Lévy N, Hennekam RC, De Sandre-Giovannoli A, López-Otín C. NovelLMNAmutations cause an aggressive atypical neonatal progeria without progerin accumulation. J Med Genet 2016; 53:776-785. [DOI: 10.1136/jmedgenet-2015-103695] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 05/12/2016] [Accepted: 05/26/2016] [Indexed: 11/04/2022]
|
40
|
Soria-Valles C, López-Otín C. iPSCs: On the Road to Reprogramming Aging. Trends Mol Med 2016; 22:713-724. [PMID: 27286740 DOI: 10.1016/j.molmed.2016.05.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/11/2016] [Accepted: 05/17/2016] [Indexed: 01/01/2023]
Abstract
Aging is characterized by irreversible loss of physiological integrity, often accompanied by an organism's loss of function and increased vulnerability to death. Defects in the mechanisms preserving cellular homeostasis over time may give rise to accelerated aging. Somatic cell reprogramming of aged cells can be associated with rejuvenation, erasing certain age-associated features, and illustrating the reversibility potential of aging. Here, we focus on recent advances in the generation of human induced pluripotent stem cells from progeroid syndromes and late-onset diseases such as Alzheimer's or Parkinson's. These cellular models have contributed to a better understanding of such pathologies, as well as to the development of novel therapeutic approaches. We also discuss different strategies to identify and target age-associated reprogramming barriers to facilitate the treatment of age-related disorders.
Collapse
Affiliation(s)
- Clara Soria-Valles
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain.
| |
Collapse
|
41
|
Osorio FG, Soria-Valles C, Santiago-Fernández O, Freije JMP, López-Otín C. NF-κB signaling as a driver of ageing. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:133-74. [PMID: 27572128 DOI: 10.1016/bs.ircmb.2016.04.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
NF-κB signaling exerts essential roles in immunity and cellular stress responses, regulating many functions related with organism innate defense. Besides, NF-κB altered signaling has been causally linked to ageing and diverse pathological conditions. We discuss herein the functional involvement of this signaling pathway in ageing, visiting recent experimental evidence about NF-κB activation in this complex process, its functional consequences and the novel biological functions raised from these works. Moreover, we discuss ageing intervention strategies based on NF-κB inhibition, which have demonstrated to be effective at delaying and even reverting different ageing manifestations in human and mouse models of both normal and accelerated ageing. Altogether, the current evidence supports that NF-κB activation constitutes a driving force of the ageing process and a preferential target for rejuvenation-aimed approaches.
Collapse
Affiliation(s)
- F G Osorio
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Oviedo, Oviedo, Spain
| | - C Soria-Valles
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Oviedo, Oviedo, Spain
| | - O Santiago-Fernández
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Oviedo, Oviedo, Spain
| | - J M P Freije
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Oviedo, Oviedo, Spain
| | - C López-Otín
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Oviedo, Oviedo, Spain.
| |
Collapse
|
42
|
Pitrez P, Rosa S, Praça C, Ferreira L. Vascular disease modeling using induced pluripotent stem cells: Focus in Hutchinson-Gilford Progeria Syndrome. Biochem Biophys Res Commun 2016; 473:710-8. [DOI: 10.1016/j.bbrc.2015.10.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/02/2015] [Indexed: 02/03/2023]
|
43
|
A Heterozygous ZMPSTE24 Mutation Associated with Severe Metabolic Syndrome, Ectopic Fat Accumulation, and Dilated Cardiomyopathy. Cells 2016; 5:cells5020021. [PMID: 27120622 PMCID: PMC4931670 DOI: 10.3390/cells5020021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/01/2016] [Accepted: 04/18/2016] [Indexed: 12/31/2022] Open
Abstract
ZMPSTE24 encodes the only metalloprotease, which transforms prelamin into mature lamin A. Up to now, mutations in ZMPSTE24 have been linked to Restrictive Dermopathy (RD), Progeria or Mandibulo-Acral Dysplasia (MAD). We report here the phenotype of a patient referred for severe metabolic syndrome and cardiomyopathy, carrying a mutation in ZMPSTE24. The patient presented with a partial lipodystrophic syndrome associating hypertriglyceridemia, early onset type 2 diabetes, and android obesity with truncal and abdominal fat accumulation but without subcutaneous lipoatrophy. Other clinical features included acanthosis nigricans, liver steatosis, dilated cardiomyopathy, and high myocardial and hepatic triglycerides content. Mutated fibroblasts from the patient showed increased nuclear shape abnormalities and premature senescence as demonstrated by a decreased Population Doubling Level, an increased beta-galactosidase activity and a decreased BrdU incorporation rate. Reduced prelamin A expression by siRNA targeted toward LMNA transcripts resulted in decreased nuclear anomalies. We show here that a central obesity without subcutaneous lipoatrophy is associated with a laminopathy due to a heterozygous missense mutation in ZMPSTE24. Given the high prevalence of metabolic syndrome and android obesity in the general population, and in the absence of familial study, the causative link between mutation and phenotype cannot be formally established. Nevertheless, altered lamina architecture observed in mutated fibroblasts are responsible for premature cellular senescence and could contribute to the phenotype observed in this patient.
Collapse
|
44
|
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare premature aging disease presenting many features resembling the normal aging process. HGPS patients die before the age of 20 years due to cardiovascular problems and heart failure. HGPS is linked to mutations in the LMNA gene encoding the intermediate filament protein lamin A. Lamin A is a major component of the nuclear lamina, a scaffold structure at the nuclear envelope that defines mechanochemical properties of the nucleus and is involved in chromatin organization and epigenetic regulation. Lamin A is also present in the nuclear interior where it fulfills lamina-independent functions in cell signaling and gene regulation. The most common LMNA mutation linked to HGPS leads to mis-splicing of the LMNA mRNA and produces a mutant lamin A protein called progerin that tightly associates with the inner nuclear membrane and affects the dynamic properties of lamins. Progerin expression impairs many important cellular processes providing insight into potential disease mechanisms. These include changes in mechanosignaling, altered chromatin organization and impaired genome stability, and changes in signaling pathways, leading to impaired regulation of adult stem cells, defective extracellular matrix production and premature cell senescence. In this review, we discuss these pathways and their potential contribution to the disease pathologies as well as therapeutic approaches used in preclinical and clinical tests.
Collapse
Affiliation(s)
- Sandra Vidak
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Vienna Biocenter (VBC), Medical University Vienna, Dr. Bohr-Gasse 9/3, 1030, Vienna, Austria
| | - Roland Foisner
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Vienna Biocenter (VBC), Medical University Vienna, Dr. Bohr-Gasse 9/3, 1030, Vienna, Austria.
| |
Collapse
|
45
|
Jaross W. Are Molecular Vibration Patterns of Cell Structural Elements Used for Intracellular Signalling? Open Biochem J 2016; 10:12-6. [PMID: 27073582 PMCID: PMC4807408 DOI: 10.2174/1874091x01610010012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 09/04/2015] [Accepted: 09/22/2015] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND To date the manner in which information reaches the nucleus on that part within the three-dimensional structure where specific restorative processes of structural components of the cell are required is unknown. The soluble signalling molecules generated in the course of destructive and restorative processes communicate only as needed. HYPOTHESIS All molecules show temperature-dependent molecular vibration creating a radiation in the infrared region. Each molecule species has in its turn a specific frequency pattern under given specific conditions. Changes in their structural composition result in modified frequency patterns of the molecules in question. The main structural elements of the cell membrane, of the endoplasmic reticulum, of the Golgi apparatus, and of the different microsomes representing the great variety of polar lipids show characteristic frequency patterns with peaks in the region characterised by low water absorption. These structural elements are very dynamic, mainly caused by the creation of signal molecules and transport containers. By means of the characteristic radiation, the area where repair or substitution services are needed could be identified; this spatial information complements the signalling of the soluble signal molecules. Based on their resonance properties receptors located on the outer leaflet of the nuclear envelope should be able to read typical frequencies and pass them into the nucleus. Clearly this physical signalling must be blocked by the cell membrane to obviate the flow of information into adjacent cells. CONCLUSION If the hypothesis can be proved experimentally, it should be possible to identify and verify characteristic infrared frequency patterns. The application of these signal frequencies onto cells would open entirely new possibilities in medicine and all biological disciplines specifically to influence cell growth and metabolism. Similar to this intracellular system, an extracellular signalling system with many new therapeutic options has to be discussed.
Collapse
Affiliation(s)
- Werner Jaross
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| |
Collapse
|
46
|
Blondel S, Egesipe AL, Picardi P, Jaskowiak AL, Notarnicola M, Ragot J, Tournois J, Le Corf A, Brinon B, Poydenot P, Georges P, Navarro C, Pitrez PR, Ferreira L, Bollot G, Bauvais C, Laustriat D, Mejat A, De Sandre-Giovannoli A, Levy N, Bifulco M, Peschanski M, Nissan X. Drug screening on Hutchinson Gilford progeria pluripotent stem cells reveals aminopyrimidines as new modulators of farnesylation. Cell Death Dis 2016; 7:e2105. [PMID: 26890144 PMCID: PMC5399184 DOI: 10.1038/cddis.2015.374] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/19/2015] [Accepted: 11/20/2015] [Indexed: 11/09/2022]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder characterized by a dramatic appearance of premature aging. HGPS is due to a single-base substitution in exon 11 of the LMNA gene (c.1824C>T) leading to the production of a toxic form of the prelamin A protein called progerin. Because farnesylation process had been shown to control progerin toxicity, in this study we have developed a screening method permitting to identify new pharmacological inhibitors of farnesylation. For this, we have used the unique potential of pluripotent stem cells to have access to an unlimited and relevant biological resource and test 21,608 small molecules. This study identified several compounds, called monoaminopyrimidines, which target two key enzymes of the farnesylation process, farnesyl pyrophosphate synthase and farnesyl transferase, and rescue in vitro phenotypes associated with HGPS. Our results opens up new therapeutic possibilities for the treatment of HGPS by identifying a new family of protein farnesylation inhibitors, and which may also be applicable to cancers and diseases associated with mutations that involve farnesylated proteins.
Collapse
Affiliation(s)
- S Blondel
- INSERM U861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France.,UEVE, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - A-L Egesipe
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - P Picardi
- Department of Medicine and Surgery, University of Salerno, Via Allende, Baronissi Salerno 84081, Italy
| | - A-L Jaskowiak
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - M Notarnicola
- Laboratory of Nutritional Biochemistry, National Institute for Digestive Diseases "S. de Bellis", Castellana Grotte, Bari 70013, Italy
| | - J Ragot
- INSERM U861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France.,UEVE, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - J Tournois
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - A Le Corf
- INSERM U861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France.,UEVE, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - B Brinon
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - P Poydenot
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - P Georges
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - C Navarro
- Aix Marseille Université, UMR S 910: Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine Timone, Marseille, France.,INSERM, UMR S 910: Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, Marseille, France
| | - P R Pitrez
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Largo Marques de Pombal, Coimbra 3004-517, Portugal
| | - L Ferreira
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Largo Marques de Pombal, Coimbra 3004-517, Portugal
| | - G Bollot
- SYNSIGHT, a/s IncubAlliance 86 rue de Paris Orsay 91400, France
| | - C Bauvais
- SYNSIGHT, a/s IncubAlliance 86 rue de Paris Orsay 91400, France
| | - D Laustriat
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - A Mejat
- Ecole Normale Supérieure de Lyon, Laboratoire de Biologie Moléculaire de la Cellule, UMR 5239 CNRS/ENS Lyon/UCBL, 46 Allée d'Italie, Lyon, France
| | - A De Sandre-Giovannoli
- Aix Marseille Université, UMR S 910: Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine Timone, Marseille, France.,INSERM, UMR S 910: Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, Marseille, France
| | - N Levy
- Aix Marseille Université, UMR S 910: Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine Timone, Marseille, France.,INSERM, UMR S 910: Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, Marseille, France
| | - M Bifulco
- Department of Medicine and Surgery, University of Salerno, Via Allende, Baronissi Salerno 84081, Italy
| | - M Peschanski
- INSERM U861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France.,UEVE, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France.,CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| | - X Nissan
- INSERM U861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France.,UEVE, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France.,CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, Evry Cedex 91030, France
| |
Collapse
|
47
|
Ghosh S, Liu B, Wang Y, Hao Q, Zhou Z. Lamin A Is an Endogenous SIRT6 Activator and Promotes SIRT6-Mediated DNA Repair. Cell Rep 2015; 13:1396-1406. [PMID: 26549451 DOI: 10.1016/j.celrep.2015.10.006] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 07/13/2015] [Accepted: 10/02/2015] [Indexed: 01/08/2023] Open
Abstract
The nuclear lamins are essential for various molecular events in the nucleus, such as chromatin organization, DNA replication, and provision of mechanical support. A specific point mutation in the LMNA gene creates a truncated prelamin A termed progerin, causing Hutchinson-Gilford progeria syndrome (HGPS). SIRT6 deficiency leads to defective genomic maintenance and accelerated aging similar to HGPS, suggesting a potential link between lamin A and SIRT6. Here, we report that lamin A is an endogenous activator of SIRT6 and facilitates chromatin localization of SIRT6 upon DNA damage. Lamin A promotes SIRT6-dependent DNA-PKcs (DNA-PK catalytic subunit) recruitment to chromatin, CtIP deacetylation, and PARP1 mono-ADP ribosylation in response to DNA damage. The presence of progerin jeopardizes SIRT6 activation and compromises SIRT6-mediated molecular events in response to DNA damage. These data reveal a critical role for lamin A in regulating SIRT6 activities, suggesting that defects in SIRT6 functions contribute to impaired DNA repair and accelerated aging in HGPS.
Collapse
Affiliation(s)
- Shrestha Ghosh
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pok Fu Lam, Hong Kong; Shenzhen Institute of Innovation and Research, The University of Hong Kong, Nanshan, Shenzhen 518000, China
| | - Baohua Liu
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pok Fu Lam, Hong Kong; School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Yi Wang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pok Fu Lam, Hong Kong
| | - Quan Hao
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pok Fu Lam, Hong Kong
| | - Zhongjun Zhou
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pok Fu Lam, Hong Kong; Shenzhen Institute of Innovation and Research, The University of Hong Kong, Nanshan, Shenzhen 518000, China.
| |
Collapse
|
48
|
Affiliation(s)
- Dorota Skowronska-Krawczyk
- a Howard Hughes Medical Institute; Department of Medicine; School of Medicine ; University of California San Diego ; La Jolla , CA USA
| | | |
Collapse
|
49
|
Abstract
Ageing constitutes a critical impediment to somatic cell reprogramming. We have explored the regulatory mechanisms that constitute age-associated barriers, through derivation of induced pluripotent stem cells (iPSCs) from individuals with premature or physiological ageing. We demonstrate that NF-κB activation blocks the generation of iPSCs in ageing. We also show that NF-κB repression occurs during cell reprogramming towards a pluripotent state. Conversely, ageing-associated NF-κB hyperactivation impairs the generation of iPSCs by eliciting the reprogramming repressor DOT1L, which reinforces senescence signals and downregulates pluripotency genes. Genetic and pharmacological NF-κB inhibitory strategies significantly increase the reprogramming efficiency of fibroblasts from Néstor-Guillermo progeria syndrome and Hutchinson-Gilford progeria syndrome patients, as well as from normal aged donors. Finally, we demonstrate that DOT1L inhibition in vivo extends lifespan and ameliorates the accelerated ageing phenotype of progeroid mice, supporting the interest of studying age-associated molecular impairments to identify targets of rejuvenation strategies.
Collapse
|
50
|
Gonzalo S, Kreienkamp R. DNA repair defects and genome instability in Hutchinson-Gilford Progeria Syndrome. Curr Opin Cell Biol 2015; 34:75-83. [PMID: 26079711 DOI: 10.1016/j.ceb.2015.05.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 05/27/2015] [Accepted: 05/29/2015] [Indexed: 12/11/2022]
Abstract
The integrity of the nuclear lamina has emerged as an important factor in the maintenance of genome stability. In particular, mutations in the LMNA gene, encoding A-type lamins (lamin A/C), alter nuclear morphology and function, and cause genomic instability. LMNA gene mutations are associated with a variety of degenerative diseases and devastating premature aging syndromes such as Hutchinson-Gilford Progeria Syndrome (HGPS) and Restrictive Dermopathy (RD). HGPS is a severe laminopathy, with patients dying in their teens from myocardial infarction or stroke. HGPS patient-derived cells exhibit nuclear shape abnormalities, changes in epigenetic regulation and gene expression, telomere shortening, genome instability, and premature senescence. This review highlights recent advances in identifying molecular mechanisms that contribute to the pathophysiology of HGPS, with a special emphasis on DNA repair defects and genome instability.
Collapse
Affiliation(s)
- Susana Gonzalo
- Edward A. Doisy Department of Biochemistry and Molecular Biology, St Louis University School of Medicine, St. Louis, MO 63104, USA.
| | - Ray Kreienkamp
- Edward A. Doisy Department of Biochemistry and Molecular Biology, St Louis University School of Medicine, St. Louis, MO 63104, USA
| |
Collapse
|